Open Access

Thrombocytopenia in COVID‑19 and vaccine‑induced thrombotic thrombocytopenia

  • Authors:
    • Styliani A. Geronikolou
    • Işil Takan
    • Athanasia Pavlopoulou
    • Marina Mantzourani
    • George P. Chrousos
  • View Affiliations

  • Published online on: January 21, 2022     https://doi.org/10.3892/ijmm.2022.5090
  • Article Number: 35
  • Copyright: © Geronikolou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The highly heterogeneous symptomatology and unpredictable progress of COVID‑19 triggered unprecedented intensive biomedical research and a number of clinical research projects. Although the pathophysiology of the disease is being progressively clarified, its complexity remains vast. Moreover, some extremely infrequent cases of thrombotic thrombocytopenia following vaccination against SARS‑CoV‑2 infection have been observed. The present study aimed to map the signaling pathways of thrombocytopenia implicated in COVID‑19, as well as in vaccine‑induced thrombotic thrombocytopenia (VITT). The biomedical literature database, MEDLINE/PubMed, was thoroughly searched using artificial intelligence techniques for the semantic relations among the top 50 similar words (>0.9) implicated in COVID‑19‑mediated human infection or VITT. Additionally, STRING, a database of primary and predicted associations among genes and proteins (collected from diverse resources, such as documented pathway knowledge, high‑throughput experimental studies, cross‑species extrapolated information, automated text mining results, computationally predicted interactions, etc.), was employed, with the confidence threshold set at 0.7. In addition, two interactomes were constructed: i) A network including 119 and 56 nodes relevant to COVID‑19 and thrombocytopenia, respectively; and ii) a second network containing 60 nodes relevant to VITT. Although thrombocytopenia is a dominant morbidity in both entities, three nodes were observed that corresponded to genes (AURKA, CD46 and CD19) expressed only in VITT, whilst ADAM10, CDC20, SHC1 and STXBP2 are silenced in VITT, but are commonly expressed in both COVID‑19 and thrombocytopenia. The calculated average node degree was immense (11.9 in COVID‑19 and 6.43 in VITT), illustrating the complexity of COVID‑19 and VITT pathologies and confirming the importance of cytokines, as well as of pathways activated following hypoxic events. In addition, PYCARD, NLP3 and P2RX7 are key potential therapeutic targets for all three morbid entities, meriting further research. This interactome was based on wild‑type genes, revealing the predisposition of the body to hypoxia‑induced thrombosis, leading to the acute COVID‑19 phenotype, the ‘long‑COVID syndrome’, and/or VITT. Thus, common nodes appear to be key players in illness prevention, progression and treatment.

Introduction

The current SARS-CoV-2-induced pandemic has raised a number of public health policy and scientific queries, related to the virus origin, transmission, activity, contamination, pathophysiologic effects and treatment. As of May 3, 2021, almost 188 million cases had been confirmed, while 4.05 million deaths had been registered under the cause of death: 'COVID-19'. Although this may underline an apogee of the third phase of the pandemic in some countries, or may have been the result of certain interventions. Public health policy approaches, communication campaigns, pharmacological approaches, surveillance, and prevention practices have been suggested.

The highly varying symptomatology and the unpredictable global progress of COVID-19 have triggered an unprecedentedly intensive activity in biomedical research and public policy decisions. Furthermore, although the pathophysiology of the disease is being progressively clarified, its complexity remains vast, and preventive care approaches or treatments, although both have significantly improved, remain unsatisfactory.

Notably, the extremely rare yet highly unpredictable and occasionally lethal vaccination-induced thrombotic thrombocytopenia (VITT) syndrome has emphasized the gaps in the current knowledge of certain unsuspected pathophysiological pathways. The VITT morbid entity is of particular importance given the generally mild and to a certain extent expected vaccination side-effects, namely chills, fever, diarrhea, fatigue, muscle pain, headache and mildly increased blood coagulability (1,2). As of April 2021, 16 vaccination options were available: Two RNA vaccines [BNT162b2 (Comirnaty) by Pfizer-BioNTech, mRNA.1273 (Spikevax) by Moderna], seven conventional inactivated ones (CoronaVac, Covaxin, BBIBP-CorV, WIBP-CorV, Minhai-Kangtai, QazVac, CovIran Bakerat), five viral vector-employing ones (Covishield and Vaxzevria by Oxford Astra-Zeneca, the Janssen COVID-19 vaccine by Johnson & Johnson, the Sputnik V and Sputnik Light by the Gamaleya Research Institute of Epidemiology and Microbiology in Russia, and the AD5-nCOV-Convidencia by CanSino Biologics Inc.), and two protein subunit vaccines (EpiVacCorona and RDB-dimer). Vaccination programs have been implemented so as to reach 'herd immunity', in every country. According to national health authority reports, as of August 30, 2021, 5.27 billion doses had been administered globally. This is equal to 39.7% of the population on the planet (where, however, only 1.6% of individuals in the low-income countries had received at least one dose), having been fully vaccinated (3). As of August 30, 2021, 55.15% of the Greek population had been fully vaccinated (3).

The aim of the present study was to illustrate the signaling pathways implicated in SARS-CoV-2 infection, including those of the extremely rare, yet severe VITT syndrome.

Data and methods

The scientific literature database, MEDLINE/PubMed (https://pubmed.ncbi.nlm.nih.gov/), was searched thoroughly for genes or gene products implicated in COVID-19 infection and VITT syndrome. Searches were conducted in the PubTator article collection (4) (https://www.ncbi.nlm.nih.gov/research/pubtator/) from the LitCovid database (5), using i) ('COVID19' OR 'SARS-CoV-2') AND ('VITT' OR 'vaccine-induced thrombotic thrombocytopenia'); ii) ('COVID19' OR 'SARS-CoV-2') AND ('thrombocytopenia' OR 'thrombopenia') key words to obtain relevant articles. Of the 495 candidate articles, 190 met the inclusion criteria which were as follows: i) written in English; ii) include an abstract; and iii) contain adequate information in their text for processing (Fig. 1).

The natural language toolkit (NLTK: https://www.nltk.org/), a freely accessible Python platform, was used for text processing, including tokenization, parsing and stemming. Word2vec embeddings module in the open-source Python library Gensim (https://pypi.org/project/gensim/) was implemented to train word vectors of processed text. A list of all word-to-word distances was extracted. To calculate the similarity distances between each word pair, the Word2Vec.most_similar function in Gensim Word2vec model was used. The top 50 detected entries were included in the present study. The work flow is presented in Fig. 1. The search results are illustrated in Fig. 2.

Furthermore, the interactions among the retrieved genes/proteins were investigated by employing the Search Tool for Retrieval of Interacting Genes/Proteins (STRING) database v11.0 (6,7), a database containing both primary and predicted, physical and functional association data among genes or proteins. These data are collected from diverse resources, such as documented pathway knowledge, high-throughput experimental studies, cross-species extrapolated information, automated text mining results, computationally predicted interactions, etc. The confidence threshold value for displaying interactions was set to 'high' (i.e., 0.7). The interactions in the generated network were manipulated and visualized through Cytoscape (http://www.cytoscape.org/) (8), a software platform for network processing and statistical analyses; the Edge Betweenness mode was used to detect the number of the shortest paths that pass-through a given edge in the COVID-19 network.

Results

Main findings

The constructed networks presented in Fig. 2 provide noteworthy information on how diverse terms are closely interlinked within the context of thrombocytopenia induced by SARS-CoV-2 infection or through vaccination. The term thrombocytopenia appears with a rather high frequency in the COVID-19/VITT network (Fig. 2A). Similarly, the term VITT is included in the COVID-19/thrombocytopenia network (Fig. 2B). COVID-19 and VITT share several comorbidities implicating vascular and epithelial dysfunction and thrombocytopenia. The nodes represent the top 50 words with a cosine similarity score of each word vector >0.9.

Interactome construction

Subsequently, two interactomes were constructed: The first one involving 119 nodes is described in Table I and illustrated in Fig. 3. Collectively, 119 nodes are involved in COVID-19, while 57 are implicated in thrombocytopenia [the latter profits from an unpublished work of ours (unpublished data). Of these, 14 nodes were common in both entities (Figs. 3 and 4), namely AIM2, IFI16, NOD2, CD8A, IL-1B, 1L-6, JAK2, NCAM1, HLA-DRB1, SERPINE1, TGFB1, TLR2, TNF and VWF. The major hubs detected are displayed in the center of the constructed circular network, while the less connected nodes are shown at the periphery of the circle (Fig. 3). The thrombocytopenia-related nodes are represented in square bullets, and the COVID-19-related ones are presented in circles, whilst the common nodes are depicted in rhomboids. The calculated average node degree of the entire interactome was extremely high (11.9).

Table I

Genes included in the molecular networks depicted in Figs. 3 and 4.

Table I

Genes included in the molecular networks depicted in Figs. 3 and 4.

Gene symbolGene nameMain function with brief description (Refs.)Figure(s)Entitya
ACE2Angiotensin I converting enzyme 2Transmembrane protein-entry point of SARS-CoV-2 (22-24,28)3C
ADAM10ADAM metallopeptidase domain 10Sheddase with strong specificity for peptide hydrolysis reactions (68-70)3T
ADAM17ADAM metallopeptidase domain 17Sheddase triggering release of cytokines receptors, ligands, etc. (68,69,71)3,4V, T
ADAMTS13ADAM metallopeptidase with throm bospondin type 1 motif 13Enzyme that cleaves von Willebrand factor (68,69)3,4V, T
ADRA2CAdrenoceptor alpha 2CMediators in catecholamine-induced inhibition of adenylate cyclase through the action of G proteins (72)3C
ADRB1Adrenoceptor beta 1Renin release/lipolysis/Increases heart rate with chrono/inotropic effect (73)3C
ADRB2Adrenoceptor beta 2Facilitating respiration (74)3C
AGERAdvanced glycosylation end-product specific receptorMediates interactions of advanced glycosylation end products (75)3C
AIM2 Interferon-inducible protein AIM2AIM2 inflammasome plays a crucial role in the defense against viral infection (76)3C, T
ANGPT1Angiopoietin 1Receptor of advanced glycosylation end products of proteins, mediating amyloid beta peptide effect on neurons and microglia (77)3C
ANGPT2Angiopoietin 2Binds to TEK/TIE2, competing for the ANGPT1 binding site, and modulating ANGPT1 signaling (78)3C
AURKA Serine/threonine-protein kinase 6Orchestrate an exit from mitosis by contributing to the completion of cytokinesis the process through which the cytoplasm of the parent cell is split into two daughter cells (79)4V
C4BComplement C4B (Chido blood group)Mediator of local inflammatory process, inducing the contraction of smooth muscle, increasing vascular permeability and causing histamine release from mast cells and basophilic leukocytes (80)3,4V, T
C5Complement C5Involved in the complement system (81)3,4V, T
C6Complement C6Causes cell lysis (82)3,4V, T
C7Complement C7Creates a hole on pathogen surfaces leading to cell lysis (82)3,4V, T
C9Complement C9Cell lysis and death contributor (82)3,4V, T
CASP1Caspase 1Inflammatory response initiator (83)3,4C, V
CASP10Caspase 10Cell apoptosis (84)3C
CASP9Caspase 9Innate immunity, mitochondrial apoptosis (85)3C
CCL2C-C motif chemokine ligand 2Induces a strong chemotactic response and mobilization of intracellular calcium ions (86,87)3C
CCL3Chemokine (C-C motif) ligand 3Pyrogenic, attracting macrophages, monocytes and neutrophils (88)3C
CCN2Cellular communication network factor 2Cell adhesion, apoptosis, migration, proliferation, differentiation, apoptosis, survival and senescence (89)3C
CD3DCD3d moleculeCell differentiation and adaptive immune response (90)3C
CD3ECD3e moleculeCell differentiation and adaptive immune response (90)3C
CD3GCD3g moleculeCell differentiation and adaptive immune response (90)3C
CD4CD4 moleculeCell differentiation and adaptive immune response (91)3C
CD40LGCD40 ligandActs as a ligand for integrins which have cell-type dependent effects, such as B-cell activation, NF-κB signaling and anti-apoptotic signaling (92,93)3T
CD8ACD8a moleculeMultiple functions in responses against both ex/internal offenses (91)3C, T
CD19B-lymphocyte antigen CD19Decreases B-cell receptor pathways (94,95)4V
CD40LGCluster of differentiation 40Mediates many immune and inflammatory responses including T-cell-dependent immunoglobulin class switching, memory B cell development, and germinal center formation (96)3,4T, V
CD46CD46 complement regulatory proteinActivates T-lymphocytes following vaccination (97,98)4V
CDC20Cell division cycle 20Regulates the formation of synaptic vesicle clustering at active zone to the presynaptic membrane in post-mitotic neurons; Cdc20-apc/ c-induced degradation of neurod2 induces presynaptic differentiation (91)3,4V, T
CDCA3Cell division cycle associated 3Involves in protein ubiquitination (99)3,4V, T
CRPC-reactive proteinMitotic initiator (100)3C
CSF1RColony stimulating factor 1 receptorControls the production, differentiation, and function macrophages (93,101)3,4V, T
CSF2Colony stimulating factor 2Cytokine affecting mostly eosinophils and macrophages (102)3,4V, T
CXCL10C-X-C motif chemokine ligand 10Chemoattraction for T- and NK cells, monocytes (87,93,103,104)3C
CXCL8C-X-C motif chemokine ligand 8Neutrophil chemotactic factor increasing respiratory burst (87,105)3,4C, V
CYP11B2Cytochrome P450 family 11 subfamily B member 2Aldosterone synthesis (87,106)3C
CYP2C19Cytochrome P450 family 2 subfamily C member 19Part of cytochrome P450, involved in drug and lipid metabolism (107)3 C
CYP2C9Cytochrome P450 family 2 subfamily C member 9Part of cytochrome P450, involved in drug and lipid metabolism (107)3 C
DDX58Retinoic acid-inducible gene IActivates interferon and cytokines production after viral infection (108)3C
EDN1Endothelin 1Potent vasoconstrictor (106,109)3C
EPOErythropoietinStimulation of erythropoiesis, vasoconstriction, angiogenesis (106)3,4V, T
F2Coagulation factor II, thrombinActivates the coagulation cascade inhibition (110)3,4V, T
FCGR1AFc fragment of IgG receptor IaComplex activation or inhibitory effects on cell functions (111)3,4V, T
FCGR1BFc fragment of IgG receptor IbHumoral immune response (112)3,4V, T
FCGR2AFc fragment of IgG receptor IIaHumoral immune response to pathogens, phagocytosis of opsonized antigens (113)3,4V, T
FCGR2BFc fragment of IgG receptor IIbPhagocytosis of immune complexes and regulation of antibody production (114)3,4V, T
FCGR3AFc fragment of IgG receptor IIIaMediates antibody-dependent cellular cytotoxicity and phagocytosis (115)3,4V, T
FCGR3BFc fragment of IgG receptor IIIbCaptures immune complexes in the peripheral circulation (116)3,4V, T
FGF7Fibroblast growth factor 7Cell growth, morphogenesis and tissue repair (117)3,4C, V
FKBP1AFKBP prolyl isomerase 1AImmunoregulation and basic cellular processes involving protein folding and trafficking (118)3,4V, T
FN1Fibronectin 1Cell growth, morphogenesis and tissue repair (70)3C
FOSFos proto-oncogene, AP-1 transcription factor subunitSignal transduction, cell proliferation and differentiation (119)3C
GNB3G protein subunit beta 3Integrates signals between receptor and effector proteins (120)3C
GZMAGranzyme ACommon component necessary for lysis of target cells by cytotoxic T-lymphocytes and natural killer cells (24)3C
GZMBGranzyme BRecognize specific infected target cells (121)3C
GZMHGranzyme HSuppresses viral replication (122)3C
HLA-AMajor histocompatibility complex, class I, ASole link between the immune system and what happens inside cells (123)3,4C, V
HLA-BMajor histocompatibility complex, class I, BHelps the immune system distinguish the endo-from exogenous proteins (123)3,4C, V
HLA-DRB1HLA class II histocompatibility antigen, DRB1 beta chainTriggers response to viral infections (41)3,4C, V, T
ICAM1Intercellular adhesion molecule 1Signal transduction (92,93)3,4V, T
IFI16Interferon gamma inducible protein 16Recognizes RNA viral infection, enhancing DDX58 production (124)3C, T
IFNA1Interferon alpha 1Antiviral and immunomodulator (125)3C
IFNG (IFN-γ)Interferon gammaAntiviral antibacterial and immunomodulatory effects (104)3,4V, T
IFNL1Interferon lambda 1Antiviral antibacterial and immunomodulatory effects (126)3C
IFNL2Interferon lambda 2Antiviral antibacterial and immunomodulatory effects (126)3C
IFNL3Interferon lambda 3Antiviral antibacterial and immunomodulatory effects (126)3C
IFNLR1Interferon lambda receptor 1Antiviral antibacterial and immunomodulatory effects (126)3C
IKBKGInhibitor of nuclear factor kappa B kinase regulatory subunit gammaAntiviral activity through JAK/STAT signaling activation (127)3C
IL10Interleukin 10Multiple, pleiotropic effects in immunoregulation, limits excessive infected tissue disruption (92)3C
IL10RBInterleukin 10 receptor subunit betaJAK1 and STAT2-mediated phosphorylation of STAT3 (128)3C
IL12AInterleukin 12AInduces IFNG (92)3C
IL12BInterleukin 12BInduces IFNG by resting PBMC (92)3C
IL17AInterleukin 17AMediates protective innate immunity to pathogens or contributes to pathogenesis of inflammatory diseases (87)3C
IL18Interleukin 18Potent inducer of inflammatory cytokines, especially IFNG (129)3C
IL1AInterleukin 1 alphaPromotion of intimal inflammation, fever, sepsis and atherogenesis (41)3C
IL1BInterleukin 1 betaPromotion of fever, development of diabetes mellitus, apoptosis of pancreatic β-cells (87,105)3,4C, V, T
IL1RAPInterleukin 1 receptor accessory proteinInduces synthesis of acute phase and proinflammatory proteins during infection, tissue damage, or stress (130)3C
IL3Interleukin 3Growth and differentiation of hematopoietic progenitor cells regulator and functional activator of mature neutrophils or macrophages (131)3,4V, T
IL33Interleukin 33Gene transcription regulator, released after cell necrosis triggering immune response and stress (132)3C
IL36GInterleukin 36 gammaInflammasome dependent, involved in systemic inflammation (133)3C
IL4Interleukin 4Hematopoiesis, antibody production, inflammation response (117)3,4V, T
IL5Interleukin 5Eosinophil migration, activation survival (134)3,4V, T
IL6Interleukin 6Innate and adaptive immune response to infections (135)3,4C, V, T
INSInsulinBlood sugar regulator (136)3C
ITGA2BIntegrin subunit alpha 2bCoagulation (137,138)3,4V, T
JAK1Janus kinase 1Cell growth survival, development differentiation of various cell types (139)3C
JAK2Janus kinase 2Cell growth and proliferation (139)3C, T
JUNJun proto-oncogene, AP-1 transcription factor subunitGene expression regulator (92)3C
KCNE1Potassium voltage-gated channel subfamily E regulatory subunit 1Potassium channels regulator (140,141)3C
KCNH2Potassium voltage-gated channel subfamily H member 2Electrical signals transmission (141)3C
KCNJ2Potassium inwardly rectifying channel subfamily J member 2Muscle movement (heart or skeletal) (142)3C
KCNQ1Potassium voltage-gated channel subfamily Q member 1Electrical signals generation and transmission (143)3C
LCN2Lipocalin 2Sequesters iron and preventing its use by bacteria, thus limiting their growth (144)3C
MMP1Matrix metallopeptidase 1Degrades collagen type I and II (145,146)3C
MMP2Matrix metallopeptidase 2Extracellular matrix (146)3C
MPLMPL proto-oncogene, thrombopoietin receptorProliferator of cells involved in blood clotting (147)3,4V, T
MS4A1Membrane spanning 4-domains A1Regulator of cellular calcium influx necessary for the B-lymphocytes activation (148)3,4C, V
MS4A3Membrane spanning 4-domains A3Marker of immature circulating neutrophils, a cellular population associated to COVID-19 severe disease (148)3C
MUC1Mucin 1, cell surface associatedHigh viscosity of airway mucus and sputum retention in the small airway of COVID-19 patients (149)3C
MYD88MYD88 innate immune signal transduction adaptorInitiates early immune responses (150)3C
NCAM1Neural cell adhesion molecule 1Molecular mimicry between NCAM-1 and the SARS-CoV-2 envelope protein (151)3C, T
NFAT5Nuclear factor of activated T-cells 5Protects cells against harmful effects of stress (137)3C
NFATC1Nuclear factor of activated T-cells 1Transcription factor (137)3,4C, V
NFATC2Nuclear factor of activated T-cells 2Neuroinflammatory factor (137)3C
NFATC3Nuclear factor of activated T-cells 3Involved in proliferation of human pulmonary fibroblasts after hypoxic stimulus (137)3C
NFATC4Nuclear factor of activated T-cells 4Transcriptional regulator in naive T-cells and differentiated effector T-cells, dependent on calcium/PLCγ/calmodulin/calcineurin signaling (137)3C
NFKB1Nuclear factor kappa B subunit 1Activated by various intra/extra-cellular stimuli as viruses (92)3,4C, V
NLRP3NLR family pyrin domain containing 3Intracellular sensor that detects a broad range of pathogen motifs (59)3,4C, V
NOD2Inflammatory bowel disease protein 1Activates NFKB1, negatively regulates TLR2 (152,153)3C, T
NOS1Nitric oxide synthase 1Chemical messenger (154,155)3C
NOS1APNitric oxide synthase 1 adaptor proteinInhibitor of Nnos (156)3C
NOS3Nitric oxide synthase 3Regulating vascular tone, cellular proliferation leucocyte adhesion and platelet aggregation (157,158)3C
NTRK1Neurotrophic receptor tyrosine kinase 1Development and survival of neurons (159)3,4V, T
NTRK2Neurotrophic receptor tyrosine kinase 2Development and maturation of the central and the peripheral nervous systems (159)3,4V, T
NTRK3Neurotrophic receptor tyrosine kinase 3Development of heart and nervous (159)3,4V, T
OLFM4Olfactomedin 4Facilitates cell adhesion, most probably through interaction with cell surface lectins and cadherin (160)3C
P2RX1Purinergic receptor P2X 1Ligand-gated ion channel with relatively high calcium permeability (161)3C
P2RX7Purinergic receptor P2X 7Receptor for ATP that acts as a ligand-gated ion channel (162)3,4C, V
PDGFAPlatelet derived growth factor subunit AWound healing (163)3C
PECAM1Platelet and endothelial cell adhesion molecule 1Cell adhesion (164)3C
PLAURPlasminogen activator, urokinase receptorLocalizing and promoting plasmin formation (165)3C
PPP3CBProtein phosphatase 3 catalytic subunit betaTransduction of intracellular Ca(2+)-mediated signals (166)3C
PRF1Perforin 1Defense against virus-infected cells (122)3C
PTGS2 Prostaglandin-endoperoxide synthase 2Role in the inflammatory response (167)3C
PTPN11Protein tyrosine phosphatase non-receptor type 11Positively regulates MAPK signal transduction pathway (168,169)3C
PYCARDPYD and CARD domain containingKey mediator in apoptosis and inflammation (170,171)3,4C, V
RENReninAngiotensin I from angiotensinogen generator in the plasma, initiating a cascade of reactions that produce an elevation of blood pressure and increased sodium retention by the kidney (172,173)3C
SCL11A2Natural resistance-associated macrophage protein 2Important in metal transport and their insertion into mitochondria (174)3,4V, T
SCN5ASodium voltage-gated channel alpha subunit 5Responsible for the initial upstroke of the action potential in an electrocardiogram (175)3C
SELESelectin EImmunoadhesion (176)3,4V, T
SELPSelectin PMediates rapid rolling of leukocyte rolling over vascular surfaces during the initial steps in inflammation through interaction with SELPLG (177)3,4V, T
SERPINE1Serpin family E member 1Alveolar type 2 cells senescence in the lung (178)3C, T
SERPINE2Serpin family E member 2Serine protease inhibitor with activity toward thrombin, trypsin, and urokinase (40)3C
SFTPCSurfactant protein CLowering the surface tension at the air-liquid interface in the peripheral air spaces (179)3C
SFTPDSurfactant protein DMay participate in the extracellular reorganization or turnover of pulmonary surfactant, regulates immune response (180)3C
SHC1SHC adaptor protein 1Signaling adapter that couples activated growth factor receptors to signaling pathways (181)3T
SIGIRRSingle Ig and TIR domain containingInflammation immune, response modulator (182)3C
SLC11A2Solute carrier family 11-member 2Metal transporter (183)3T
SOCS1Suppressor of cytokine signaling 1Exerts the viral virulence effect via inhibition of type I and type II interferon (IFN) function (184)3,4V, T
STXBP2Syntaxin binding protein 2Involved in cytolytic pathway (185)3T
TBK1TANK binding kinase 1Regulator of inflammatory responses to foreign agents (186)3C
TFTransferrinTransports of iron from sites of absorption and heme degradation to those of storage and utilization (187)3,4V, T
TFPITissue factor pathway inhibitorAnticoagulant protein blocking the initiation of blood coagulation by inhibiting TF-f VIIa and early forms of prothrombinase (188)3,4V, T
TFRCTransferrin receptorErythropoiesis and neurologic development (189)3,4V, T
TGFB1Transforming growth factor beta 1Gene expression proliferation (70)3C, T
THPOThrombopoietinRegulates platelets and macrophages differentiation (190)3,4V, T
TICAM1Toll-like receptor adaptor molecule 1Native immunity against invading pathogens (191)3C
TLR2Toll-like receptor 2Pathogen recognition-potential therapeutic target (192-194)3C, T
TLR4Toll-like receptor 4Upregulated after SARS-CoV-2 infection (195)3C
TNFTumor necrosis factorBiomarker of COVID-19 severity (104)3,4C, V, T
TNFRSF1ATNF receptor superfamily member 1AContributes to the induction of non-cytocidal TNF effects including anti-viral state and activation of the acid sphingomyelinase (93,104)3C
TNFRSF1BTNF receptor superfamily member 1BRegulates TNF-α function by antagonizing its biological activity (93,104)3C
TRAF3TNF receptor associated factor 3Regulates pathways leading to a NFKB and MAP kinases activation, and B-cell survival (196)3C
TYK2Tyrosine kinase 2Antiviral activity (197)3C
VCAM1Vascular cell adhesion molecule 1Mediates the adhesion of lymphocytes, monocytes, eosinophils and basophils to vascular endothelium (198)3,4V, T
VEGFAVascular endothelial growth factor ADominant inducer to blood vessels growth (increases their permeability) (199)3C
VKORC1Vitamin K epoxide reductase complex subunit 1Reduces inactive vitamin K 2,3-epoxide to active vitamin K (200)3C
VWFvon Willebrand factorInvolved in hemostasis and thrombosis (201)3,4C, V, T

a Entities: C, COVID-19; V, vaccine-induced thrombotic thrombocytopenia; T, thrombocytopenia.

The second one including 61 molecules, is described in Table I and illustrated in Fig. 5. Of these, 47 are common with thrombocytopenia (indicated by a polygon), and 16 with COVID-19 (represented by circles). The VITT-related molecules are denoted with triangles.

Venn diagrams were further created to illustrate the nodes that are common between thrombocytopenia and COVID-19 or VITT (Fig. 4A and B, respectively), between COVID-19 and VITT (Fig. 4C), and amid the three morbid entities (Fig. 4D). The common nodes are listed in each diagram in detail.

All included molecules herein are listed in Table I. The figure (network) in which each molecule is implicated is also noted in a separate column in Table I.

Discussion

Epidemics were already identified as entities in antiquity by Hippocrates and named by him in his Treatises 'On Epidemics' (9,10). Viral epidemics were described therein and in other works of the Hippocratic Corpus (11,12). On the other hand, Aristotle, the ancient Greek physician and philosopher (4th century B.C.) wrote that 'the creativeness of nature focuses on qualities rather than quantities and description rather than measurements' (13,14). This concept was rejected by Newton's determinism and reductionism and was since forgotten, until it was re-established by Wulff in 1999 (15). Indeed, subtle change in qualities may trigger phase shift alterations with unpredictable consequences, as the Chaos theory of dynamic systems recently confirmed (16). According to this concept, the systems theory was coined as representing a rapid, cost and time-effective method of research (17). It may integrate basic, preclinical and clinical research, and both human and animal results to unravel new insights in complex and often unpredictable issues. In the case of the COVID-19 pandemic, the urgency, and certain ethical issues, make such an in silico approach a sine qua non research method.

The human-to-human transmission of SARS-CoV-2 is either mediated by respiratory droplets via sneezing/coughing or even just breathing, while the disease demonstrates an incubation period of 5-7 days (18). The clinical outcomes range from asymptomatic to influenza-like, or to even pneumonia and severe acute respiratory distress syndrome (ARDS) (19), and thromboembolic events (20,21), pointing to the lung tropism of this virus. Dissimilarities in patients' profiles are attributed to genetic and/or epigenetic variations and underlying pathologies. Dissimilarities in severity may be attributed to the aforementioned factors, but also to the size of the viral inoculum and/or viral mutations.

COVID-19 and the thrombocytopenia interactions network

Ariadne's thread appears to be the angiotensin I converting enzyme 2 (ACE2), which clearly plays a crucial role. SARS-CoV-2, via its spike S protein, a surface glycoprotein that surrounds the spherical virus, is attached to ACE2 and this is followed by entry into cells of the host (22-27). ACE2 is expressed in cells of a number of human organs (including the skin, nasal and oral mucosa, lung, nasopharynx, brain, lymph nodes, thymus, stomach, small intestine, colon, bone marrow, spleen, liver and kidneys). Additionally, its expression in lung alveoli (type 2 pneumonocytes) and small intestine endothelium, as well as in the arterial and other tissue smooth muscle epithelium (28), may trigger the release of anaphylatoxin (29). There is clinical evidence to confirm the aforementioned knowledge of COVID-19 (29).

In the generated network illustrated in Fig. 2, ACE2 interacts with CYP11B2 and with IL-6. The latter is the greatest hub in this vastly interconnected network, with 63 interactions, confirming that the progress of SARS-CoV-2-induced infection would profit from therapeutic blockade of IL-6. As noted by Mazzoni et al (24), blocking this mechanism would 'suppress noxious systemic inflammation but also restore the protective antiviral potential'. It has been established that innate immunity via natural killer (NK) cells exerts the frontline defense, with CD8+ T-lymphocytes being important for the long-term surveillance against viruses, while adaptive immune responses play a key role in the control of viral infections (28). Both responses are mediated either via cytotoxicity or by IF-γ, IL-12 and IL-18. Virus-induced cytotoxicity is primarily moderated by perforin and granzymes. Increased severity in viral infections may lead to dysregulated immunity and tissue/organ damage (30). Clinical evidence in SARS-CoV-2 infection has demonstrated that high IL-6 levels in patients in intensive care units, are inversely associated with the concentration of NK cells (24,31).

The network included dense interactions illustrating clearly that SARS-CoV-2-specific T-cells are critical for the extended damage caused by the 'cytokine storm' (or 'cytokine release syndrome') (30,32) (Fig. 3). This excessive inflammatory response may be lethal for some patients (29,33). Although the phenomenon may manifest in other inflammatory conditions, including bacterial sepsis, pneumonia, sterile inflammation, etc., the extent in the secretion of several specific cytokines is different in COVID-19-related storm (29). Of note, COVID-19 infection has been associated with changes in the blood coagulation mechanisms, with differing manifestations in different patients, in distinct phases of the disease, and independently of disease severity.

Autoimmune destruction of platelets, cytokine release and high consumption of coagulation factors and platelets have been observed in patients with SARS-CoV-2 infection (Geronikolou et al, unpublished data) and initial hypercoagulability (34). Thromboembolic events increase by 31% in patients with COVID-19 admitted in intensive care units (35,36); the phenomenon may be interpreted by the 'two way activation theory' (20,37), i.e., thrombogenesis via inflammation-relevant pathways, with parallel occurrence of release of VWF large polymers. The coagulation and platelet profiles of patients with COVID-19 are then rather normal, unlike in patients with sepsis where platelets are activated and consumed, with the occurrence of thrombocytopenia (38). Only a few patients may then survive, particularly of those with extensive disseminated intravascular coagulation (38). Thrombosis has been observed in situ in the lungs, as well as in a systemic manner, in a similar fashion with classic sepsis and acute respiratory distress syndrome. Reported thromboembolic complications include mostly venous pulmonary embolism (38), aortic graft thrombosis, and mesenteric ischemia; coronary and cerebral thrombosis cases have been reported, although these are rare. The so-called 'COVID toe' is a sign of thrombosis accompanied by arterial and venous clots, urgent oxygen demand and multiple organ dysfunction (20,36,39).

COVID-19 and thrombocytopenia interactomes share only 14 nodes (AIM2, IFI16, TLR2, NOD2, NKAM1, IL-6, TNF, JAK2, IL-1B, SERPINE1, HLA-DRB1, TGFB1, CD8A, and VWF) (Fig. 3), most of which serve as major hubs (IL-6, TNF, JAK2, IL-1B, SERPINE1, TGFB1, CD8A and VWF) in the herein presented interactome (Figs. 1 and 2).

Cytokines, such as IL-1B, 1L-6 and TNF contribute to the so-called cytokine storm, as aforementioned. Moreover, JAK2 is a kinase suspected to be implicated in thrombocytopenia via reduced levels of thrombopoietin or via decreased expression levels of their cognate receptors (cMpl receptors). JAK2 mutations (V617F) that are present in the majority of patients with myeloproliferative disease, may increase hematopoietic cell sensitivity to erythropoietin and thrombopoietin. NKAM1 or CD56 is a homophilic binding glycoprotein expressed on the surface of neurons, glia cells and skeletal muscles. NKAM1 is a prototypic marker of NK cells, also present in CD8+ T-cells. These cell types exhibit diminished antiviral ability and cytotoxic impairment during COVID-19 infection (24). CD8A1 is a cytotoxic marker for T-cell populations. SERPINE1 or plasminogen activator inhibitor-1 is a protein encoded by the SERPINE1 gene, which participates in both thrombosis and atherogenesis (40).

TGFB1 is a multifunctional peptide, with diverse activities, including the control of cell growth, proliferation, differentiation, and apoptosis. It can also down-regulate the activity of immune cells via decreasing the expression levels of cytokine receptors, such as that of IL-2. Several types of T-cells secrete TGFB1, so as to inhibit cytotoxicity and the secretion of certain cytokines, such as interferon-γ, TNF-α and various interleukins, such as IL-6. This makes this molecule a potential target of therapeutic value. On the other hand, the hemostatic VWF is detected in blood plasma, endothelium and megakaryocytes, as well as in subendothelial connective tissue. This factor appears to be also increased and implicated in autoimmune diseases, such as thrombotic thrombocytopenic purpura, as well as in stroke and atrial fibrillation, due to the platelet clots that are potentially formed when its levels are elevated.

Recent literature has further revealed that an HLA class I and II molecule, that is, HLA-DRB1, which is common in COVID-19 and in thrombocytopenia networks (Fig. 2), may play a role in the observed COVID-19 individual and ethnic diversity in clinical severity and/or response to therapy or vaccination (41-44). Of note, HLA-DRB1 is interconnected with the lymphocyte function markers CD3D, CD3E, CD3G, CD4, lymphocyte regulation positive FCGR1A, FCGR1B, HLA class I and II molecules, such as HLA-A, HLA-B, similar to the NCAM1, PTPN1, SHC1 and VCAM1 molecules that have been implicated in thrombosis and atherosclerosis. NCAM1 is involved in cell-cell adhesion in neural-muscle cells in the embryonic phase and later, and more notably, in the responsiveness to viral infections (rabies virus and papilloma virus) (45). PTPN1 is a potential therapeutic target of obesity and type 2 diabetes mellitus as well (46); SHC1 is implicated in reactive oxygen species regulation, thus, in the oxidative stress response (47), while VCAM1 is directly involved in thrombosis and atherogenesis and acute respiratory syndrome (48-51).

VITT and thrombocytopenia interactome

Various coagulation mechanisms have been implicated in VITT: High levels of D-dimers and low levels of fibrinogen have been observed in patients (2,52,53). On the other hand, early reports of VITT described a higher incidence of the syndrome in young women, exhibiting both age-dependence and sexual dimorphism. VITT, though very rare, is of utmost importance. Yet, in March, 2021, the European Medicines Agency (EMA) issued a statement noting that the thromboembolic events of VITT in vaccinated populations were not higher than in general population (54). Subsequently, the 'risk vs. benefit' equilibrium was weighed by the World Health Organization (WHO), promoting the benefit of the vaccination vs. the extremely low risk of thromboembolic risk of VITT in the general population (55).

VITT is currently termed 'thrombosis with thrombocytopenia syndrome (TTS)' by the Centers for Disease Control and Prevention (CDC) and the US Food and Drug Administration (FDA) (56), and is characterized by arterial and venous thrombosis at unexpected sites (i.e., cerebral venous sinuses, splanchnic vessels of variant severity and/or positive platelet factor (PF) 4-heparin ELISA ('HIT' ELISA) syndrome (52), exhibiting both age dependence and sexual dimorphism (more frequent in individuals <50 years old and of the female sex) (2). The laboratory and clinical features of this syndrome are similar to those of the heparin-induced thrombocytopenia (HIT) syndrome and/or the HIT-like autoimmune thrombosis with thrombocytopenia syndrome (2,52,53), both of which have already been reported following surgery, the uptake of certain pharmaceuticals, or during some infections in patients that are not being treated with heparin. The therapeutic suggestions of this recently coined syndrome include early initiation of non-heparin anticoagulation, high-dose IVIG, and/or prednisolone (57).

The genetic basis of the VITT syndrome appears to be closely intertwined with that of the COVID-19 disease and, as such, they share 16 nodes: CASP1, CXCL8, FGF7, HLA-A, HLA-B, IL1B, IL6, MS4A1, NFATC1, NFKB1, NLP3, P2RX7, PYCARD, TNF, TFP1, VWF (Figs. 3Figure 4-5). The purpose of the vaccine is to inhibit pathways that mediate this condition (52,58). More importantly, the relevant research is ongoing with the extremely rare cases of this syndrome, as VITT incidence is ~0.74-1 cases per 100,000 vaccinated subjects (52). Of note, the anti-COVID-19 vaccines do not cause illness and the two morbid entities (COVID-19 and VITT) are by no means identical, with the etiopathology of the latter being actually autoimmune, with auto-antibodies against platelet factor 4. More explicitly, COVID-19 network shares 14 nodes with thrombocytopenia (AIM2, CD8A, HLA-DRB1, IFI16, IL1B, IL6, JAK2, NCAM1, NOD2, SERPINE1, TGFB1, TLR2, TNF and VWF), while VITT (which is a type of thrombocytopenia) shares 46 nodes with thrombocytopenia (Figs. 3Figure 4-5). Notably, SHC1, STXBP2, CDC20 and ADAM10 are silenced in VITT, while AURKA, CD46, CD19 are uniquely expressed following vaccination (apparently not expressed in common thrombocytopenia or in COVID-19) (Figs. 3Figure 4-5). These molecules were not previously identified as VITT-related and are, thus, a novel finding, at least to the best of our knowledge.

It is known that the NLP3 inflammasome is implicated in both COVID-19 and VITT, apart from its participation in other inflammatory reactions (59). It has also been previously demonstrated that acute thrombotic events may manifest during hypoxia, as shown in COVID-19, due to an early proinflammatory state in the venous milieu, mediated by a HIF-induced NLP3 inflammasome complex (60,61). In the network constructed in the present study, NLP3 connects with CASP1, IL-IB, IL17A, CXCL8, IL-6, MYD88, NFKB1, P2RX7, PYCARD and TNF.

P2RX7 exhibits sexually dimorphic and contrasting roles in the pathogenesis of thrombosis, depending on the pathogen type, the severity of infection, the cell type infected and the level of tissue activation (62). In the thrombocytopenia/ COVID-19/VITT cases, the viral load, the cell-type infected and the infecting virus strain or certain vaccine types have been associated with NLP3 hyperactivation, which in the presence of comorbidities, such as liver, renal, gut or respiratory tract illnesses, diabetes mellitus, previous infections, exposure to pollutants, and/or lifestyle factors, such as smoking and obesity, may upend the roles of P2RX7 and PYCARD to those of tissue-damaging, or even lethal factors (62,63). More importantly, the persistent neurological effects ('long-COVID-19') observed in a large percentage of patients with COVID-19 may be explained via the activation of these pathways. Thus, P2RX7 antagonists may be promising therapeutics in the management of both VITT and 'long-COVID-19' (62,64), as P2RX7 receptor stimulation has been implicated in lung damage, psychiatric disorders and pathological inflammation (65,66). In the COVID-19 interactome, P2RX7 directly interacts with NLP3, CASP1 and P2RX1. On the contrary, in the VITT network, P2RX7 directly interacts only with NLP3, IL1B and CASP1. Accordingly, PYCARD interacts with NLP3, CASP1, IL1B, IL18 and IKBKG in COVID-19, and with NLP3, CASP1 and IL1B in the VITT syndrome (Table II). The common node in all possible combinations, as shown in Table II, is CASP1, a downstream event of the NLP3 inflammasome; CASP1 activation promotes IL1B production, which may be prevented by a pan-caspase inhibitor or by glyburide treatment (67).

Table II

Common direct connections between 'PYCARD' or 'P2RX7' and 'COVID-19' or 'VITT'.

Table II

Common direct connections between 'PYCARD' or 'P2RX7' and 'COVID-19' or 'VITT'.

GeneCOVID-19VITTCOMMON direct connections
PYCARDNLP3, CASP1, IL1B, IL18, IKBKGNLP3, CASP1, IL1BNLP3, CASP1, IL1B
P2RX7NLP3, CASP1, P2RX1CASP1, IL1BCASP1
COMMON direct connectionsNLP3, CASP1CASP1, IL1BCASP1

To this end, the present study investigated the aforementioned issues through the construction of molecular networks and the detection of at least one known COVID/VITT/thrombocytopenia molecule that confirmed that endothelial dysfunction and blood thrombosis are the key players of both COVID-19 and VITT morbid entities. One limitation of the present study is that it included only wild-type genes and their products. To the best of our knowledge, however, this is the first effort made at providing a comprehensive network map of the molecules involved in the underlying mechanisms of COVID-19, long COVID-19 and/or VITT pathophysiology.

In conclusion, the interactomes presented herein revealed therapeutic and vaccination modification targets (i.e., SHC1, NCAM1, HLAs, CD8A, PTPN1, VWF and TBP1). It was also demonstrated that: i) NCAM1 is involved in SARS-CoV-2 infection responsiveness, apart from papilloma and rabies virus infections, and may be responsible for relevant vaccination side effects; ii) NLP3, P2RX7 and PYCARD contribution may help explain (partly or mostly) VITT and/or 'long COVID-19 side-effects'; iii) furthermore, the antagonism of these latter nodes should focus on potential pharmacological targets in the context of SARS-CoV-2 infection and/or vaccine immunization responsiveness. In conclusion, network construction is a powerful tool, which may be used to elucidate the physiology and pathophysiology of different states in clinical investigation. The highly interconnected network presented herein highlights the complexity of COVID-19/VITT pathophysiology, mapping the key role of cytokines, enzymes and immune response markers (lymphocyte regulators and human leucocyte antigens) that may be potential drug or vaccine targets. It was constructed using wild-type genes and gene products, revealing the body's predisposition to COVID-19 infection or VITT. Of note, the COVID-19 and thrombocytopenia common nodes appear to be key players in the natural history of the illness.

Availability of data and materials

The datasets used and/or analyzed during the current study are available throughout the manuscript.

Authors' contributions

SAG and MM were involved in the conceptualization of the study. SAG was involved in the study methodology. SAG, AP and MM were involved in data validation. SAG and AP was involved in formal analysis and in the investigative aspects of the study. SAG was involved in the provision of resources (study material). SAG, IT and AP was involved in data curation. IT provided the software used in this study. SAG, IT, GPC and AP were involved in the interpretation of the data, and in the writing and preparation of the original draft. SAG, AP, MM and GPC were involved in the writing, reviewing and editing of the manuscript. MM and GPC supervised the study. SAG and GPC were involved in project administration. All authors confirm the authenticity of the raw data and have read and agreed to the published version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

No funding was received.

References

1 

World Health Organization (WHO): Coronovirus disease (COVID-19): Vaccines safety. WHO; Geneva: 2021

2 

Greinacher A, Thiele T, Warkentin TE, Weisser K, Kyrle PA and Eichinger S: Thrombotic thrombocytopenia after ChAdOx1 nCov-19 vaccination. N Engl J Med. 384:2092–2101. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Mathieu E, Ritchie H, Ortiz-Ospina E, Roser M, Hasell J, Appel C, Giattino C and Rodés-Guirao L: A global database of COVID-19 vaccinations. Nat Hum Behav. 5:947–953. 2021. View Article : Google Scholar

4 

Wei CH, Allot A, Leaman R and Lu Z: PubTator central: Automated concept annotation for biomedical full text articles. Nucleic Acids Res. 47(W1): W587–W593. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Chen Q, Allot A and Lu Z: LitCovid: An open database of COVID-19 literature. Nucleic Acids Res. 49(D1): D1534–D1540. 2021. View Article : Google Scholar :

6 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47(D1): D607–D613. 2019. View Article : Google Scholar

7 

Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, Doncheva NT, Legeay M, Fang T, Bork P, et al: The STRING database in 2021: Customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 49(D1): D605–D612. 2021. View Article : Google Scholar

8 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Hippocrates: Epidemics 2, 4-7. Smith Wesley D: Loeb Classical Library 477. Harvard University Press; Cambridge, MA: 1994

10 

Jouanna J: Hippocrates. John Hopkins University Press; Baltimore, MD: 1999

11 

Mammas IN and Spandidos DA: Paediatric virology in the Hippocratic corpus. Exp Ther Med. 12:541–549. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Pappas G, Kiriaze IJ and Falagas ME: Insights into infectious disease in the era of Hippocrates. Int J Infect Dis. 12:347–350. 2008. View Article : Google Scholar

13 

Misselbrook D: Aristotle, hume and the goals of medicine. J Eval Clin Pract. 22:544–549. 2016. View Article : Google Scholar

14 

Wulff HR: The concept of disease: From Newton back to Aristotle. Lancet. 354(Suppl): SIV501999. View Article : Google Scholar

15 

Wulff HR: The concept of disease: From Newton back to Aristotle. Lancet. 54:3541999.

16 

Lorenz EN: Deterministic nonperiodic flow. J Atmos Sci. 20:130–141. 1963. View Article : Google Scholar

17 

Barabási AL, Gulbahce N and Loscalzo J: Network medicine: A network-based approach to human disease. Nat Rev Genet. 12:56–68. 2011. View Article : Google Scholar :

18 

Li Q, Guan X, Wu P, Wang X, Zhou L, Tong Y, Ren R, Leung KSM, Lau EHY, Wong JY, et al: Early transmission dynamics in Wuhan, China, of novel coronavirus-infected pneumonia. N Engl J Med. 382:1199–1207. 2020. View Article : Google Scholar :

19 

Raoult D, Zumla A, Locatelli F, Ippolito G and Kroemer G: Coronavirus infections: Epidemiological, clinical and immunological features and hypotheses. Cell Stress. 4:66–75. 2020. View Article : Google Scholar : PubMed/NCBI

20 

Mondal S, Quintili AL, Karamchandani K and Bose S: Thromboembolic disease in COVID-19 patients: A brief narrative review. J Intensive Care. 8:702020. View Article : Google Scholar : PubMed/NCBI

21 

Xu P, Zhou Q and Xu J: Mechanism of thrombocytopenia in COVID-19 patients. Ann Hematol. 99:1205–1208. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, Somasundaran M, Sullivan JL, Luzuriaga K, Greenough TC, et al: Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 426:450–454. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Ge XY, Li JL, Yang XL, Chmura AA, Zhu G, Epstein JH, Mazet JK, Hu B, Zhang W, Peng C, et al: Isolation and characterization of a bat SARS-like coronavirus that uses the ACE2 receptor. Nature. 503:535–538. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Mazzoni A, Salvati L, Maggi L, Capone M, Vanni A, Spinicci M, Mencarini J, Caporale R, Peruzzi B, Antonelli A, et al: Impaired immune cell cytotoxicity in severe COVID-19 is IL-6 dependent. J Clin Invest. 130:4694–4703. 2020. View Article : Google Scholar :

25 

Sama IE, Ravera A, Santema BT, van Goor H, Ter Maaten JM, Cleland JGF, Rienstra M, Friedrich AW, Samani NJ, Ng LL, et al: Circulating plasma concentrations of angiotensin-converting enzyme 2 in men and women with heart failure and effects of renin-angiotensin-aldosterone inhibitors. Eur Heart J. 41:1810–1817. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Diaz JH: Hypothesis: Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers may increase the risk of severe COVID-19. J Travel Med. 27:taaa0412020. View Article : Google Scholar : PubMed/NCBI

27 

Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, et al: SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 18:271–280.e8. 2020. View Article : Google Scholar

28 

Hamming I, Timens W, Bulthuis ML, Lely AT, Navis G and van Goor H: Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J Pathol. 203:631–637. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Gao T, Hu M, Zhang X, Li H, Zhu L, Liu H, Dong Q, Zhang Z, Wang Z, Hu Y, et al: Highly pathogenic coronavirus N protein aggravates lung injury by MASP-2-mediated complement over-activation. medRxiv. ppmedrxiv-20041962. 2020.

30 

Cao X: COVID-19: Immunopathology and its implications for therapy. Nat Rev Immunol. 20:269–270. 2020. View Article : Google Scholar

31 

Channappanavar R and Perlman S: Pathogenic human coronavirus infections: Causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. 39:529–539. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Zhao J, Zhao J and Perlman S: T cell responses are required for protection from clinical disease and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. J Virol. 84:9318–9325. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Meduri GU, Kohler G, Headley S, Tolley E, Stentz F and Postlethwaite A: Inflammatory cytokines in the BAL of patients with ARDS. Persistent elevation over time predicts poor outcome. Chest. 108:1303–1314. 1995. View Article : Google Scholar : PubMed/NCBI

34 

Tang N, Li D, Wang X and Sun Z: Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost. 18:844–847. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, Delabranche X, Merdji H, Clere-Jehl R, Schenck M, Fagot Gandet F, et al: High risk of thrombosis in patients with severe SARS-CoV-2 infection: A multicenter prospective cohort study. Intensive Care Med. 46:1089–1098. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Klok FA, Kruip MJHA, van der Meer NJM, Arbous MS, Gommers DAMPJ, Kant KM, Kaptein FHJ, van Paassen J, Stals MAM, Huisman MV and Endeman H: Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res. 191:145–147. 2020. View Article : Google Scholar :

37 

Chang JC: Hemostasis based on a novel 'two-path unifying theory' and classification of hemostatic disorders. Blood Coagul Fibrinolysis. 29:573–584. 2018. View Article : Google Scholar

38 

Chang JC: Sepsis and septic shock: Endothelial molecular pathogenesis associated with vascular microthrombotic disease. Thromb J. 17:102019. View Article : Google Scholar : PubMed/NCBI

39 

Seirafianpour F, Sodagar S, Pour Mohammad A, Panahi P, Mozafarpoor S, Almasi S and Goodarzi A: Cutaneous manifestations and considerations in COVID-19 pandemic: A systematic review. Dermatol Ther. 33:e139862020. View Article : Google Scholar : PubMed/NCBI

40 

Vaughan DE: PAI-1 and atherothrombosis. J Thromb Haemost. 3:1879–1883. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Badary OA: Pharmacogenomics and COVID-19: Clinical implications of human genome interactions with repurposed drugs. Pharmacogenomics J. 21:275–284. 2021. View Article : Google Scholar : PubMed/NCBI

42 

Chen MR, Kuo HC, Lee YJ, Chi H, Li SC, Lee HC and Yang KD: Phenotype, susceptibility, autoimmunity, and immunotherapy between Kawasaki disease and coronavirus disease-19 associated multisystem inflammatory syndrome in children. Front Immunol. 12:6328902021. View Article : Google Scholar : PubMed/NCBI

43 

Romero-López JP, Carnalla-Cortés M, Pacheco-Olvera DL, Ocampo-Godínez JM, Oliva-Ramírez J, Moreno-Manjón J, Bernal-Alferes B, López-Olmedo N, García-Latorre E, Domínguez-López ML, et al: A bioinformatic prediction of antigen presentation from SARS-CoV-2 spike protein revealed a theoretical correlation of HLA-DRB1*01 with COVID-19 fatality in Mexican population: An ecological approach. J Med Virol. 93:2029–2038. 2021. View Article : Google Scholar

44 

Anzurez A, Naka I, Miki S, Nakayama-Hosoya K, Isshiki M, Watanabe Y, Nakamura-Hoshi M, Seki S, Matsumura T, Takano T, et al: Association of HLA-DRB1*09:01 with severe COVID-19. HLA. 98:37–42. 2021. View Article : Google Scholar : PubMed/NCBI

45 

Rotondo JC, Bosi S, Bassi C, Ferracin M, Lanza G, Gafà R, Magri E, Selvatici R, Torresani S, Marci R, et al: Gene expression changes in progression of cervical neoplasia revealed by microarray analysis of cervical neoplastic keratinocytes. J Cell Physiol. 230:806–812. 2015. View Article : Google Scholar

46 

Combs AP: Recent advances in the discovery of competitive protein tyrosine phosphatase 1B inhibitors for the treatment of diabetes, obesity, and cancer. J Med Chem. 53:2333–2344. 2010. View Article : Google Scholar

47 

Finkel T and Holbrook NJ: Oxidants, oxidative stress and the biology of ageing. Nature. 408:239–247. 2000. View Article : Google Scholar : PubMed/NCBI

48 

Choi YM, Kwon HS, Choi KM, Lee WY and Hong EG: Short-term effects of beraprost sodium on the markers for cardiovascular risk prediction in type 2 diabetic patients with microalbuminuria. Endocrinol Metab (Seoul). 34:398–405. 2019. View Article : Google Scholar

49 

Nomura S, Taniura T, Shouzu A, Omoto S, Suzuki M, Okuda Y and Ito T: Effects of sarpogrelate, eicosapentaenoic acid and pitavastatin on arterioslcerosis obliterans-related biomarkers in patients with type 2 diabetes (SAREPITASO study). Vasc Health Risk Manag. 14:225–232. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Zheng Y, Liu SQ, Sun Q, Xie JF, Xu JY, Li Q, Pan C, Liu L and Huang YZ: Plasma microRNAs levels are different between pulmonary and extrapulmonary ARDS patients: A clinical observational study. Ann Intensive Care. 8:232018. View Article : Google Scholar : PubMed/NCBI

51 

Attia EF, Jolley SE, Crothers K, Schnapp LM and Liles WC: Soluble vascular cell adhesion molecule-1 (sVCAM-1) is elevated in bronchoalveolar lavage fluid of patients with acute respiratory distress syndrome. PLoS One. 11:e01496872016. View Article : Google Scholar :

52 

Cines DB and Bussel JB: SARS-CoV-2 vaccine-induced immune thrombotic thrombocytopenia. N Engl J Med. 384:2254–2256. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Schultz NH, Sørvoll IH, Michelsen AE, Munthe LA, Lund-Johansen F, Ahlen MT, Wiedmann M, Aamodt AH, Skattør TH, Tjønnfjord GE and Holme PA: Thrombosis and thrombocytopenia after ChAdOx1 nCoV-19 vaccination. N Engl J Med. 384:2124–2130. 2021. View Article : Google Scholar : PubMed/NCBI

54 

European Medicines Agency (EMA): COVID-19 Vaccine AstraZeneca: PRAC investigating cases of thromboembolic events-vaccine's benefits currently still outweigh risks-update. 2021.

55 

World Health Organization (WHO): Statement of the WHO global advisory committee on vaccine safety (GACVS) COVID-19 subcommittee on safety signals related to the AstraZeneca COVID-19 vaccine. WHO; Geneva: 2021

56 

Bussel JB, Connors JM, Cines DB, Dunbar CE, Michaelis LC, Kreuziger LB, Lee AYY and Pabinger-Fasching I: Thrombosis with thrombocytopenia syndrome (also termed vaccine-induced thrombotic thrombocytopenia). American Society of Haematology; Washington, DC: 2021

57 

Thaler J, Ay C, Gleixner KV, Hauswirth AW, Cacioppo F, Grafeneder J, Quehenberger P, Pabinger I and Knöbl P: Successful treatment of vaccine-induced prothrombotic immune thrombocytopenia (VIPIT). J Thromb Haemost. 19:1819–1822. 2021. View Article : Google Scholar : PubMed/NCBI

58 

Smadja DM, Mentzer SJ, Fontenay M, Laffan MA, Ackermann M, Helms J, Jonigk D, Chocron R, Pier GB, Gendron N, et al: COVID-19 is a systemic vascular hemopathy: Insight for mechanistic and clinical aspects. Angiogenesis. 24:755–788. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Kashir J, Ambia AR, Shafqat A, Sajid MR, AlKattan K and Yaqinuddin A: Scientific premise for the involvement of neutrophil extracellular traps (NETs) in vaccine-induced thrombotic thrombocytopenia (VITT). J Leukoc Biol. Sep 1–2021.Epub ahead of prin. View Article : Google Scholar : PubMed/NCBI

60 

Gupta N, Sahu A, Prabhakar A, Chatterjee T, Tyagi T, Kumari B, Khan N, Nair V, Bajaj N, Sharma M and Ashraf MZ: Activation of NLRP3 inflammasome complex potentiates venous thrombosis in response to hypoxia. Proc Natl Acad Sci USA. 114:4763–4768. 2017. View Article : Google Scholar : PubMed/NCBI

61 

Salaro E, Rambaldi A, Falzoni S, Amoroso FS, Franceschini A, Sarti AC, Bonora M, Cavazzini F, Rigolin GM, Ciccone M, et al: Involvement of the P2X7-NLRP3 axis in leukemic cell proliferation and death. Sci Rep. 6:262802016. View Article : Google Scholar :

62 

Ribeiro DE, Oliveira-Giacomelli Á, Glaser T, Arnaud-Sampaio VF, Andrejew R, Dieckmann L, Baranova J, Lameu C, Ratajczak MZ and Ulrich H: Hyperactivation of P2X7 receptors as a culprit of COVID-19 neuropathology. Mol Psychiatry. 26:1044–1059. 2021. View Article : Google Scholar

63 

Savio LEB, de Andrade Mello P, da Silva CG and Coutinho-Silva R: The P2X7 receptor in inflammatory diseases: Angel or demon. Front Pharmacol. 9:522018. View Article : Google Scholar

64 

Pacheco PAF and Faria RX: The potential involvement of P2X7 receptor in COVID-19 pathogenesis: A new therapeutic target? Scand J Immunol. 93:e129602021. View Article : Google Scholar

65 

Ortiz GG, Pacheco-Moisés FP, Macías-Islas M, Flores-Alvarado LJ, Mireles-Ramírez MA, González-Renovato ED and Her nández-Nava r ro VE: Role of the blood-brain barrier in multiple sclerosis. Arch Med Res. 45:687–697. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Di Virgilio F, Tang Y, Sarti AC and Rossato M: A rationale for targeting the P2X7 receptor in coronavirus disease 19. Br J Pharmacol. 177:4990–4994. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Ferreira AC, Soares VC, de Azevedo-Quintanilha IG, Dias SDSG, Fintelman-Rodrigues N, Sacramento CQ, Mattos M, de Freitas CS, Temerozo JR, Teixeira L, et al: SARS-CoV-2 engages inflammasome and pyroptosis in human primary monocytes. Cell Death Discov. 7:432021. View Article : Google Scholar : PubMed/NCBI

68 

Moss ML and Bartsch JW: Therapeutic benefits from targeting of ADAM family members. Biochemistry. 43:7227–7235. 2004. View Article : Google Scholar : PubMed/NCBI

69 

Souza JSM, Lisboa ABP, Santos TM, Andrade MVS, Neves VBS, Teles-Souza J, Jesus HNR, Bezerra TG, Falcão VGO, Oliveira RC and Del-Bem LE: The evolution of ADAM gene family in eukaryotes. Genomics. 112:3108–3116. 2020. View Article : Google Scholar

70 

Xu J, Xu X, Jiang L, Dua K, Hansbro PM and Liu G: SARS-CoV-2 induces transcriptional signatures in human lung epithelial cells that promote lung fibrosis. Respir Res. 21:1822020. View Article : Google Scholar : PubMed/NCBI

71 

Katneni UK, Alexaki A, Hunt RC, Schiller T, DiCuccio M, Buehler PW, Ibla JC and Kimchi-Sarfaty C: Coagulopathy and thrombosis as a result of severe COVID-19 infection: A microvascular focus. Thromb Haemost. 120:1668–1679. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Tian J, Sun D, Xie Y, Liu K and Ma Y: Network pharmacology-based study of the molecular mechanisms of Qixuekang in treating COVID-19 during the recovery period. Int J Clin Exp Pathol. 13:2677–2690. 2020.PubMed/NCBI

73 

Boron WF and Boulpaep EL: Medical physiology: A cellular and molecular approach. Saunders Elsevier; Philadelphia, PA: 2012

74 

Fitzpatrick D, Purves D and Augustine G: Neuroscience. 3rd edition. Sinauer Associates, Inc; Sunderland, MA: 2004

75 

Wang Q, Zhu W, Xiao G, Ding M, Chang J and Liao H: Effect of AGER on the biological behavior of non-small cell lung cancer H1299 cells. Mol Med Rep. 22:810–818. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Man SM, Karki R and Kanneganti TD: AIM2 inflammasome in infection, cancer, and autoimmunity: Role in DNA sensing, inflammation, and innate immunity. Eur J Immunol. 46:269–280. 2016. View Article : Google Scholar

77 

Bafunno V, Firinu D, D'Apolito M, Cordisco G, Loffredo S, Leccese A, Bova M, Barca MP, Santacroce R, Cicardi M, et al: Mutation of the angiopoietin-1 gene (ANGPT1) associates with a new type of hereditary angioedema. J Allergy Clin Immunol. 141:1009–1017. 2018. View Article : Google Scholar

78 

PubMed Gene database: ANGPT2 angiopoietin 2 [Homo sapiens (human)]. https://www.ncbi.nlm.nih.gov/gene?Db=gene&Cmd=ShowDetailView&TermToSearch=285 Accessed December 12, 2020.

79 

Marumoto T, Honda S, Hara T, Nitta M, Hirota T, Kohmura E and Saya H: Aurora-A kinase maintains the fidelity of early and late mitotic events in HeLa cells. J Biol Chem. 278:51786–51795. 2003. View Article : Google Scholar : PubMed/NCBI

80 

Li N, Zhang J, Liao D, Yang L, Wang Y and Hou S: Association between C4, C4A, and C4B copy number variations and susceptibility to autoimmune diseases: A meta-analysis. Sci Rep. 7:426282017. View Article : Google Scholar : PubMed/NCBI

81 

Horiuchi T and Tsukamoto H: Complement-targeted therapy: Development of C5- and C5a-targeted inhibition. Inflamm Regen. 36:112016. View Article : Google Scholar : PubMed/NCBI

82 

Hobart MJ, Fernie BA and DiScipio RG: Structure of the human C7 gene and comparison with the C6, C8A, C8B, and C9 genes. J Immunol. 154:5188–5194. 1995.PubMed/NCBI

83 

Xia S, Zhang Z, Magupalli VG, Pablo JL, Dong Y, Vora SM, Wang L, Fu TM, Jacobson MP, Greka A, et al: Gasdermin D pore structure reveals preferential release of mature interleukin-1. Nature. 593:607–611. 2021. View Article : Google Scholar : PubMed/NCBI

84 

Cohen GM: Caspases: The executioners of apoptosis. Biochem J. 326:1–16. 1997. View Article : Google Scholar : PubMed/NCBI

85 

Avrutsky MI and Troy CM: Caspase-9: A multimodal therapeutic target with diverse cellular expression in human disease. Front Pharmacol. 12:7013012021. View Article : Google Scholar : PubMed/NCBI

86 

Singh S, Anshita D and Ravichandiran V: MCP-1: Function, regulation, and involvement in disease. Int Immunopharmacol. 101:1075982021. View Article : Google Scholar : PubMed/NCBI

87 

Coperchini F, Chiovato L, Ricci G, Croce L, Magri F and Rotondi M: The cytokine storm in COVID-19: Further advances in our understanding the role of specific chemokines involved. Cytokine Growth Factor Rev. 58:82–91. 2021. View Article : Google Scholar :

88 

Guan E, Wang J and Norcross MA: Identification of human macrophage inflammatory proteins 1alpha and 1beta as a native secreted heterodimer. J Biol Chem. 276:12404–12409. 2001. View Article : Google Scholar

89 

Charrier A and Brigstock DR: Regulation of pancreatic function by connective tissue growth factor (CTGF, CCN2). Cytokine Growth Factor Rev. 24:59–68. 2013. View Article : Google Scholar

90 

Garcillán B, Fuentes P, Marin AV, Megino RF, Chacon-Arguedas D, Mazariegos MS, Jiménez-Reinoso A, Muñoz-Ruiz M, Laborda RG, Cárdenas PP, et al: CD3G or CD3D knockdown in mature, but not immature, T lymphocytes similarly cripples the human TCRαβ complex. Front Cell Dev Biol. 9:6084902021. View Article : Google Scholar

91 

Heritable gene regulation in the CD4:CD8 T cell lineage choice. Front Immunol. 8:2912017.PubMed/NCBI

92 

Sharma P, Pandey AK and Bhattacharyya DK: Determining crucial genes associated with COVID-19 based on COPD findings✶,✶✶. Comput Biol Med. 128:1041262021. View Article : Google Scholar

93 

Zou M, Su X, Wang L, Yi X, Qiu Y, Yin X, Zhou Z, Niu X, Wang L and Su M: The molecular mechanism of multiple organ dysfunction and targeted intervention of COVID-19 based on time-order transcriptomic analysis. Front Immunol. 12:7297762021. View Article : Google Scholar : PubMed/NCBI

94 

Jing Y, Luo L, Chen Y, Westerberg LS, Zhou P, Xu Z, Herrada AA, Park CS, Kubo M, Mei H, et al: SARS-CoV-2 infection causes immunodeficiency in recovered patients by downregulating CD19 expression in B cells via enhancing B-cell metabolism. Signal Transduct Target Ther. 6:3452021. View Article : Google Scholar :

95 

Badbaran A, Mailer RK, Dahlke C, Woens J, Fathi A, Mellinghoff SC, Renné T, Addo MM, Riecken K and Fehse B: Digital PCR to quantify ChAdOx1 nCoV-19 copies in blood and tissues. Mol Ther Methods Clin Dev. 23:418–423. 2021. View Article : Google Scholar : PubMed/NCBI

96 

Grewal IS and Flavell RA: CD40 and CD154 in cell-mediated immunity. Annu Rev Immunol. 16:111–135. 1998. View Article : Google Scholar : PubMed/NCBI

97 

Riley-Vargas RC, Gill DB, Kemper C, Liszewski MK and Atkinson JP: CD46: Expanding beyond complement regulation. Trends Immunol. 25:496–503. 2004. View Article : Google Scholar : PubMed/NCBI

98 

Lundstrom K, Barh D, Uhal BD, Takayama K, Aljabali AAA, Abd El-Aziz TM, Lal A, Redwan EM, Adadi P, Chauhan G, et al: COVID-19 vaccines and thrombosis-roadblock or dead-end street? Biomolecules. 11:10202021. View Article : Google Scholar : PubMed/NCBI

99 

Chen J, Goyal N, Dai L, Lin Z, Del Valle L, Zabaleta J, Liu J, Post SR, Foroozesh M and Qin Z: Developing new ceramide analogs and identifying novel sphingolipid-controlled genes against a virus-associated lymphoma. Blood. 136:2175–2187. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Dementyeva E, Kryukov F, Kubiczkova L, Nemec P, Sevcikova S, Ihnatova I, Jarkovsky J, Minarik J, Stefanikova Z, Kuglik P and Hajek R: Clinical implication of centrosome amplification and expression of centrosomal functional genes in multiple myeloma. J Transl Med. 11:772013. View Article : Google Scholar :

101 

Martinez FO, Combes TW, Orsenigo F and Gordon S: Monocyte activation in systemic Covid-19 infection: Assay and rationale. EBioMedicine. 59:1029642020. View Article : Google Scholar : PubMed/NCBI

102 

Root RK and Dale DC: Granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor: Comparisons and potential for use in the treatment of infections in nonneutropenic patients. J Infect Dis. 179(Suppl 2): S342–S352. 1999. View Article : Google Scholar

103 

Zhang N, Zhao YD and Wang XM: CXCL10 an important chemokine associated with cytokine storm in COVID-19 infected patients. Eur Rev Med Pharmacol Sci. 24:7497–7505. 2020.PubMed/NCBI

104 

Bergamaschi C, Terpos E, Rosati M, Angel M, Bear J, Stellas D, Karaliota S, Apostolakou F, Bagratuni T, Patseas D, et al: Systemic IL-15, IFN-γ, and IP-10/CXCL10 signature associated with effective immune response to SARS-CoV-2 in BNT162b2 mRNA vaccine recipients. Cell Rep. 36:1095042021. View Article : Google Scholar

105 

Du HX, Zhu JQ, Chen J, Zhou HF, Yang JH and Wan HT: Revealing the therapeutic targets and molecular mechanisms of emodin-treated coronavirus disease 2019 via a systematic study of network pharmacology. Aging (Albany NY). 13:14571–14589. 2021. View Article : Google Scholar

106 

Lombardero M, Kovacs K and Scheithauer BW: Erythropoietin: A hormone with multiple functions. Pathobiology. 78:41–53. 2011. View Article : Google Scholar : PubMed/NCBI

107 

Petrović J, Pešić V and Lauschke VM: Frequencies of clinically important CYP2C19 and CYP2D6 alleles are graded across Europe. Eur J Hum Genet. 28:88–94. 2020. View Article : Google Scholar

108 

Kell AM and Gale M Jr: RIG-I in RNA virus recognition. Virology. 479-480:110–121. 2015. View Article : Google Scholar : PubMed/NCBI

109 

Boron WF and Boulpaep EL: Medical physiology: A cellular and molecular approach. 2nd edition. Saunders Elsevier; Philadelphia, PA: 2009

110 

Devreese KMJ: COVID-19-related laboratory coagulation findings. Int J Lab Hematol. 43(Suppl 1): S36–S42. 2021. View Article : Google Scholar

111 

Patel KR, Roberts JT and Barb AW: Multiple variables at the leukocyte cell surface impact Fc γ receptor-dependent mechanisms. Front Immunol. 10:2232019. View Article : Google Scholar

112 

Kelton JG, Smith JW, Santos AV, Murphy WG and Horsewood P: Platelet IgG Fc receptor. Am J Hematol. 25:299–310. 1987. View Article : Google Scholar : PubMed/NCBI

113 

Qiao J, Al-Tamimi M, Baker RI, Andrews RK and Gardiner EE: The platelet Fc receptor, FcγRIIa. Immunol Rev. 268:241–252. 2015. View Article : Google Scholar : PubMed/NCBI

114 

Fearon DT and Carroll MC: Regulation of B lymphocyte responses to foreign and self-antigens by the CD19/CD21 complex. Annu Rev Immunol. 18:393–422. 2000. View Article : Google Scholar : PubMed/NCBI

115 

Hartwig JH, Barkalow K, Azim A and Italiano J: The elegant platelet: Signals controlling actin assembly. Thromb Haemost. 82:392–398. 1999. View Article : Google Scholar

116 

Viertlboeck BC, Schweinsberg S, Hanczaruk MA, Schmitt R, Du Pasquier L, Herberg FW and Göbel TW: The chicken leukocyte receptor complex encodes a primordial, activating, high-affinity IgY Fc receptor. Proc Natl Acad Sci USA. 104:11718–11723. 2007. View Article : Google Scholar : PubMed/NCBI

117 

Tan Y and Tang F: SARS-CoV-2-mediated immune system activation and potential application in immunotherapy. Med Res Rev. 41:1167–1194. 2021. View Article : Google Scholar

118 

Hotchkiss KM, Clark NM and Olivares-Navarrete R: Macrophage response to hydrophilic biomaterials regulates MSC recruitment and T-helper cell populations. Biomaterials. 182:202–215. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Springer S, Menzel LM and Zieger M: Google trends provides a tool to monitor population concerns and information needs during COVID-19 pandemic. Brain Behav Immun. 87:109–110. 2020. View Article : Google Scholar : PubMed/NCBI

120 

Brockmeyer NH, Potthoff A, Kasper A, Nabring C, Jöckel KH and Siffert W: GNB3 C825T polymorphism and response to anti-retroviral combination therapy in HIV-1-infected patients-a pilot study. Eur J Med Res. 10:489–494. 2005.PubMed/NCBI

121 

Uddin MN, Akter R, Li M and Abdelrahman Z: Expression of SARS-COV-2 cell receptor gene ACE2 is associated with immunosuppression and metabolic reprogramming in lung adenocarcinoma based on bioinformatics analyses of gene expression profiles. Chem Biol Interact. 335:1093702021. View Article : Google Scholar

122 

Bieberich F, Vazquez-Lombardi R, Yermanos A, Ehling RA, Mason DM, Wagner B, Kapetanovic E, Di Roberto RB, Weber CR, Savic M, et al: A single-cell atlas of lymphocyte adaptive immune repertoires and transcriptomes reveals age-related differences in convalescent COVID-19 patients. Front Immunol. 12:7010852021. View Article : Google Scholar : PubMed/NCBI

123 

Fricke-Galindo I and Falfán-Valencia R: Genetics insight for COVID-19 susceptibility and severity: A review. Front Immunol. 12:6221762021. View Article : Google Scholar : PubMed/NCBI

124 

Jiang Z, Wei F, Zhang Y, Wang T, Gao W, Yu S, Sun H, Pu J, Sun Y, Wang M, et al: IFI16 directly senses viral RNA and enhances RIG-I transcription and activation to restrict influenza virus infection. Nat Microbiol. 6:932–945. 2021. View Article : Google Scholar : PubMed/NCBI

125 

Kennedy RB, Poland GA, Ovsyannikova IG, Oberg AL, Asmann YW, Grill DE, Vierkant RA and Jacobson RM: Impaired innate, humoral, and cellular immunity despite a take in smallpox vaccine recipients. Vaccine. 34:3283–3290. 2016. View Article : Google Scholar : PubMed/NCBI

126 

Kotenko SV: IFN-λs. Curr Opin Immunol. 23:583–590. 2011. View Article : Google Scholar :

127 

Wu UI and Holland SM: Host susceptibility to non-tuberculous mycobacterial infections. Lancet Infect Dis. 15:968–980. 2015. View Article : Google Scholar : PubMed/NCBI

128 

Voloudakis G, Hoffman G, Venkatesh S, Lee KM, Dobrindt K, Vicari JM, Zhang W, Beckmann ND, Jiang S, Hoagland D, et al: IL10RB as a key regulator of COVID-19 host susceptibility and severity. medRxiv. View Article : Google Scholar

129 

Vecchié A, Bonaventura A, Toldo S, Dagna L, Dinarello CA and Abbate A: IL-18 and infections: Is there a role for targeted therapies. J Cell Physiol. 236:1638–1657. 2021. View Article : Google Scholar

130 

Peters VA, Joesting JJ and Freund GG: IL-1 receptor 2 (IL-1R2) and its role in immune regulation. Brain Behav Immun. 32:1–8. 2013. View Article : Google Scholar :

131 

Bénard A, Jacobsen A, Brunner M, Krautz C, Klösch B, Swierzy I, Naschberger E, Podolska MJ, Kouhestani D, David P, et al: Interleukin-3 is a predictive marker for severity and outcome during SARS-CoV-2 infections. Nat Commun. 12:11122021. View Article : Google Scholar :

132 

Zizzo G and Cohen PL: Imperfect storm: Is interleukin-33 the Achilles heel of COVID-19? Lancet Rheumatol. 12:e779–e790. 2020. View Article : Google Scholar

133 

Walsh PT and Fallon PG: The emergence of the IL-36 cytokine family as novel targets for inflammatory diseases. Ann NY Acad Sci. 1417:23–34. 2018. View Article : Google Scholar

134 

Nussbaum JC, Van Dyken SJ, von Moltke J, Cheng LE, Mohapatra A, Molofsky AB, Thornton EE, Krummel MF, Chawla A, Liang HE and Locksley RM: Type 2 innate lymphoid cells control eosinophil homeostasis. Nature. 502:245–248. 2013. View Article : Google Scholar : PubMed/NCBI

135 

Coomes EA and Haghbayan H: Interleukin-6 in Covid-19: A systematic review and meta-analysis. Rev Med Virol. 30:1–9. 2020. View Article : Google Scholar : PubMed/NCBI

136 

Das UN: Bioactive lipids in COVID-19-further evidence. Arch Med Res. 52:107–120. 2021. View Article : Google Scholar

137 

Islam ABMMK, Khan MA, Ahmed R, Hossain MS, Kabir SMT, Islam MS and Siddiki AMAMZ: Transcriptome of nasopharyngeal samples from COVID-19 patients and a comparative analysis with other SARS-CoV-2 infection models reveal disparate host responses against SARS-CoV-2. J Transl Med. 19:322021. View Article : Google Scholar : PubMed/NCBI

138 

O'Brien JR: Shear-induced platelet aggregation. Lancet. 335:711–713. 1990. View Article : Google Scholar : PubMed/NCBI

139 

Langmuir P, Yeleswaram S, Smith P, Knorr B and Squier P: Design of clinical trials evaluating ruxolitinib, a JAK1/JAK2 inhibitor, for treatment of COVID-19-associated cytokine storm. Dela J Public Health. 6:50–54. 2020. View Article : Google Scholar : PubMed/NCBI

140 

Melman YF, Krummerman A and McDonald TV: KCNE regulation of KvLQT1 channels: Structure-function correlates. Trends Cardiovasc Med. 12:182–187. 2002. View Article : Google Scholar : PubMed/NCBI

141 

Gouas L, Nicaud V, Berthet M, Forhan A, Tiret L, Balkau B and Guicheney P; D.E.S.I.R. Study Group: Association of KCNQ1, KCNE1, KCNH2 and SCN5A polymorphisms with QTc interval length in a healthy population. Eur J Hum Genet. 13:1213–1222. 2005. View Article : Google Scholar : PubMed/NCBI

142 

Lazzerini PE, Acampa M, Laghi-Pasini F, Bertolozzi I, Finizola F, Vanni F, Natale M, Bisogno S, Cevenini G, Cartocci A, et al: Cardiac arrest risk during acute infections: Systemic inflammation directly prolongs QTc interval via cytokine-mediated effects on potassium channel expression. Circ Arrhythm Electrophysiol. 13:e0086272020. View Article : Google Scholar : PubMed/NCBI

143 

Szendrey M, Guo J, Li W, Yang T and Zhang S: COVID-19 drugs chloroquine and hydroxychloroquine, but not azithromycin and remdesivir, block hERG potassium channels. J Pharmacol Exp Ther. 377:265–272. 2021. View Article : Google Scholar : PubMed/NCBI

144 

Dahl SL, Woodworth JS, Lerche CJ, Cramer EP, Nielsen PR, Moser C, Thomsen AR, Borregaard N and Cowland JB: Lipocalin-2 functions as inhibitor of innate resistance to mycobacterium tuberculosis. Front Immunol. 9:27172018. View Article : Google Scholar :

145 

Vincenti MP and Brinckerhoff CE: Transcriptional regulation of collagenase (MMP-1, MMP-13) genes in arthritis: Integration of complex signaling pathways for the recruitment of gene-specific transcription factors. Arthritis Res. 4:157–164. 2002. View Article : Google Scholar : PubMed/NCBI

146 

Jabłońska-Trypuć A, Matejczyk M and Rosochacki S: Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. J Enzyme Inhib Med Chem. 31(Suppl 1): S177–S183. 2016. View Article : Google Scholar

147 

Pisano TJ, Hakkinen I and Rybinnik I: Large vessel occlusion secondary to COVID-19 hypercoagulability in a young patient: A case report and literature review. J Stroke Cerebrovasc Dis. 29:1053072020. View Article : Google Scholar : PubMed/NCBI

148 

Apostolidis SA, Kakara M, Painter MM, Goel RR, Mathew D, Lenzi K, Rezk A, Patterson KR, Espinoza DA, Kadri JC, et al: Cellular and humoral immune responses following SARS-CoV-2 mRNA vaccination in patients with multiple sclerosis on anti-CD20 therapy. Nat Med. 27:1990–2001. 2021. View Article : Google Scholar : PubMed/NCBI

149 

Lu W, Liu X, Wang T, Liu F, Zhu A, Lin Y, Luo J, Ye F, He J, Zhao J, et al: Elevated MUC1 and MUC5AC mucin protein levels in airway mucus of critical ill COVID-19 patients. J Med Virol. 93:582–584. 2021. View Article : Google Scholar

150 

Conti P, Ronconi G, Caraffa A, Gallenga CE, Ross R, Frydas I and Kritas SK: Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by Coronavirus-19 (COVI-19 or SARS-CoV-2): Anti-inflammatory strategies. J Biol Regul Homeost Agents. 34:327–331. 2020.PubMed/NCBI

151 

Morsy S: NCAM protein and SARS-COV-2 surface proteins: In-silico hypothetical evidence for the immunopathogenesis of Guillain-Barré syndrome. Med Hypotheses. 145:1103422020. View Article : Google Scholar

152 

Root-Bernstein R: Innate receptor activation patterns involving TLR and NLR synergisms in COVID-19, ALI/ARDS and sepsis cytokine storms: A review and model making novel predictions and therapeutic suggestions. Int J Mol Sci. 22:21082021. View Article : Google Scholar :

153 

Watanabe T, Kitani A, Murray PJ and Strober W: NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol. 5:800–808. 2004. View Article : Google Scholar : PubMed/NCBI

154 

Esposito E and Cuzzocrea S: The role of nitric oxide synthases in lung inflammation. Curr Opin Investig Drugs. 8:899–909. 2007.PubMed/NCBI

155 

Gamkrelidze M, Intskirveli N, Vardosanidze K, Goliadze L, Chikhladze KH and Ratiani L: Myocardial dysfunction during septic shock (review). Georgian Med News. 237:40–46. 2014.

156 

Zang X, Li S, Zhao Y, Chen K, Wang X, Song W, Ma J, Tu X, Xia Y, Zhang S and Gao C: Systematic meta-analysis of the association between a common NOS1AP genetic polymorphism, the QTc interval, and sudden death. Int Heart J. 60:1083–1090. 2019. View Article : Google Scholar : PubMed/NCBI

157 

Guan SP, Seet RCS and Kennedy BK: Does eNOS derived nitric oxide protect the young from severe COVID-19 complications. Ageing Res Rev. 64:1012012020. View Article : Google Scholar

158 

Thom SR, Fisher D, Xu YA, Garner S and Ischiropoulos H: Role of nitric oxide-derived oxidants in vascular injury from carbon monoxide in the rat. Am J Physiol. 276:H984–H992. 1999.

159 

Valent A, Danglot G and Bernheim A: Mapping of the tyrosine kinase receptors trkA (NTRK1), trkB (NTRK2) and trkC(NTRK3) to human chromosomes 1q22, 9q22 and 15q25 by fluorescence in situ hybridization. Eur J Hum Genet. 5:102–104. 1997. View Article : Google Scholar : PubMed/NCBI

160 

Liu W, Chen L, Zhu J and Rodgers GP: The glycoprotein hGC-1 binds to cadherin and lectins. Exp Cell Res. 312:1785–1797. 2006. View Article : Google Scholar : PubMed/NCBI

161 

Hennigs JK, Lüneburg N, Stage A, Schmitz M, Körbelin J, Harbaum L, Matuszcak C, Mienert J, Bokemeyer C, Böger RH, et al: The P2-receptor-mediated Ca2+ signalosome of the human pulmonary endothelium-implications for pulmonary arterial hypertension. Purinergic Signal. 15:299–311. 2019. View Article : Google Scholar : PubMed/NCBI

162 

Russo MV and McGavern DB: Immune surveillance of the CNS following infection and injury. Trends Immunol. 36:637–650. 2015. View Article : Google Scholar : PubMed/NCBI

163 

Tuuminen R, Nykänen A, Keränen MA, Krebs R, Alitalo K, Koskinen PK and Lemström KB: The effect of platelet-derived growth factor ligands in rat cardiac allograft vasculopathy and fibrosis. Transplant Proc. 38:3271–3273. 2006. View Article : Google Scholar : PubMed/NCBI

164 

Blum E, Margalit R, Levy L, Getter T, Lahav R, Zilber S, Bradfield P, Imhof BA, Alpert E and Gruzman A: A Potent leukocyte transmigration blocker: GT-73 showed a protective effect against LPS-induced ARDS in mice. Molecules. 26:45832021. View Article : Google Scholar : PubMed/NCBI

165 

Rovina N, Akinosoglou K, Eugen-Olsen J, Hayek S, Reiser J and Giamarellos-Bourboulis EJ: Soluble urokinase plasminogen activator receptor (suPAR) as an early predictor of severe respiratory failure in patients with COVID-19 pneumonia. Crit Care. 24:1872020. View Article : Google Scholar

166 

Kumar S, Jain A, Choi SW, da Silva GPD, Allers L, Mudd MH, Peters RS, Anonsen JH, Rusten TE, Lazarou M and Deretic V: Mammalian Atg8 proteins and the autophagy factor IRGM control mTOR and TFEB at a regulatory node critical for responses to pathogens. Nat Cell Biol. 22:973–985. 2020. View Article : Google Scholar : PubMed/NCBI

167 

Hoxha M: What about COVID-19 and arachidonic acid pathway. Eur J Clin Pharmacol. 76:1501–1504. 2020. View Article : Google Scholar : PubMed/NCBI

168 

Keikha M, Ghazvini K, Eslami M, Yousefi B, Casseb J, Yousefi M and Karbalaei M: Molecular targeting of PD-1 signaling pathway as a novel therapeutic approach in HTLV-1 infection. Microb Pathog. 144:1041982020. View Article : Google Scholar : PubMed/NCBI

169 

Coggeshall KM: Negative signaling in health and disease. Immunol Res. 19:47–64. 1999. View Article : Google Scholar : PubMed/NCBI

170 

de Souza JG, Starobinas N and Ibañez O: Unknown/enigmatic functions of extracellular ASC. Immunology. 163:377–388. 2021. View Article : Google Scholar

171 

Larabi A, Barnich N and Nguyen HTT: New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy. 16:38–51. 2020. View Article : Google Scholar :

172 

Brown MJ: Renin: Friend or foe? Heart. 93:1026–1033. 2007. View Article : Google Scholar

173 

Amraei R and Rahimi N: COVID-19, renin-angiotensin system and endothelial dysfunction. Cells. 9:16522020. View Article : Google Scholar :

174 

Yanatori I, Yasui Y, Noguchi Y and Kishi F: Inhibition of iron uptake by ferristatin II is exerted through internalization of DMT1 at the plasma membrane. Cell Biol Int. 39:427–434. 2015. View Article : Google Scholar

175 

Denham NC, Pearman CM, Ding WY, Waktare J, Gupta D, Snowdon R, Hall M, Cooper R, Modi S, Todd D and Mahida S: Systematic re-evaluation of SCN5A variants associated with Brugada syndrome. J Cardiovasc Electrophysiol. 30:118–127. 2019. View Article : Google Scholar

176 

Smadja DM, Guerin CL, Chocron R, Yatim N, Boussier J, Gendron N, Khider L, Hadjadj J, Goudot G, Debuc B, et al: Angiopoietin-2 as a marker of endothelial activation is a good predictor factor for intensive care unit admission of COVID-19 patients. Angiogenesis. 23:611–620. 2020. View Article : Google Scholar : PubMed/NCBI

177 

Bongiovanni D, Klug M, Lazareva O, Weidlich S, Biasi M, Ursu S, Warth S, Buske C, Lukas M, Spinner CD, et al: SARS-CoV-2 infection is associated with a pro-thrombotic platelet phenotype. Cell Death Dis. 12:502021. View Article : Google Scholar : PubMed/NCBI

178 

Wu D and Yang XO: Dysregulation of pulmonary responses in severe COVID-19. Viruses. 13:9572021. View Article : Google Scholar :

179 

Katzen J and Beers MF: Contributions of alveolar epithelial cell quality control to pulmonary fibrosis. J Clin Invest. 130:5088–5099. 2020. View Article : Google Scholar : PubMed/NCBI

180 

Nandy D, Sharma N and Senapati S: Systematic review and meta-analysis confirms significant contribution of surfactant protein D in chronic obstructive pulmonary disease. Front Genet. 10:3392019. View Article : Google Scholar : PubMed/NCBI

181 

Di Lisa F, Kaludercic N, Carpi A, Menabò R and Giorgio M: Mitochondria and vascular pathology. Pharmacol Rep. 61:123–130. 2009. View Article : Google Scholar

182 

Dinarello CA, Nold-Petry C, Nold M, Fujita M, Li S, Kim S and Bufler P: Suppression of innate inflammation and immunity by interleukin-37. Eur J Immunol. 46:1067–1081. 2016. View Article : Google Scholar : PubMed/NCBI

183 

Montalbetti N, Simonin A, Kovacs G and Hediger MA: Mammalian iron transporters: families SLC11 and SLC40. Mol Aspects Med. 34:270–287. 2013. View Article : Google Scholar

184 

Schulert GS, Blum SA and Cron RQ: Host genetics of pediatric SARS-CoV-2 COVID-19 and multisystem inflammatory syndrome in children. Curr Opin Pediatr. 33:549–555. 2021. View Article : Google Scholar : PubMed/NCBI

185 

Pachlopnik Schmid J and de Saint Basile G: Angeborene hämophagozytische lymphohistiozytose (HLH). Klin Padiatr. 222:345–350. 2010. View Article : Google Scholar : PubMed/NCBI

186 

Shi JH, Xie X and Sun SC: TBK1 as a regulator of autoimmunity and antitumor immunity. Cell Mol Immunol. 15:743–745. 2018. View Article : Google Scholar : PubMed/NCBI

187 

Zimecki M, Actor JK and Kruzel ML: The potential for Lactoferrin to reduce SARS-CoV-2 induced cytokine storm. Int Immunopharmacol. 95:1075712021. View Article : Google Scholar :

188 

Chabot PR, Raiola L, Lussier-Price M, Morse T, Arseneault G, Archambault J and Omichinski JG: Structural and functional characterization of a complex between the acidic transactivation domain of EBNA2 and the Tfb1/p62 subunit of TFIIH. PLoS Pathog. 10:e10040422014. View Article : Google Scholar : PubMed/NCBI

189 

Speeckaert MM, Speeckaert R and Delanghe JR: Biological and clinical aspects of soluble transferrin receptor. Crit Rev Clin Lab Sci. 47:213–228. 2010. View Article : Google Scholar

190 

Bg S, Gosavi S, Ananda Rao A, Shastry S, Raj SC, Sharma A, Suresh A and Noubade R: Neutrophil-to-lymphocyte, lymphocyte-to-monocyte, and platelet-to-lymphocyte ratios: Prognostic significance in COVID-19. Cureus. 13:e126222021.PubMed/NCBI

191 

Campbell GR, To RK, Hanna J and Spector SA: SARS-CoV-2, SARS-CoV-1, and HIV-1 derived ssRNA sequences activate the NLRP3 inflammasome in human macrophages through a non-classical pathway. iScience. 24:1022952021. View Article : Google Scholar :

192 

Borrello S, Nicolò C, Delogu G, Pandolfi F and Ria F: TLR2: a crossroads between infections and autoimmunity. Int J Immunopathol Pharmacol. 24:549–556. 2011. View Article : Google Scholar

193 

Zheng M, Karki R, Williams EP, Yang D, Fitzpatrick E, Vogel P, Jonsson CB and Kanneganti TD: TLR2 senses the SARS-CoV-2 envelope protein to produce inflammatory cytokines. Nat Immunol. 22:829–838. 2021. View Article : Google Scholar : PubMed/NCBI

194 

Khan S, Shafiei M, Longoria C, Schoggins JW, Savani R and Zaki H: SARS-CoV-2 spike protein induces inflammation via TLR2-dependent activation of the NF-κB pathway. Elife. 10:e685632021. View Article : Google Scholar

195 

Sohn KM, Lee SG, Kim HJ, Cheon S, Jeong H, Lee J, Kim IS, Silwal P, Kim YJ, Paik S, et al: COVID-19 patients upregulate toll-like receptor 4-mediated inflammatory signaling that mimics bacterial sepsis. J Korean Med Sci. 35:e3432020. View Article : Google Scholar : PubMed/NCBI

196 

Guven-Maiorov E, Keskin O, Gursoy A, VanWaes C, Chen Z, Tsai CJ and Nussinov R: TRAF3 signaling: Competitive binding and evolvability of adaptive viral molecular mimicry. Biochim Biophys Acta. 1860:2646–2655. 2016. View Article : Google Scholar

197 

Callaway E: The quest to find genes that drive severe COVID. Nature. 595:346–348. 2021. View Article : Google Scholar : PubMed/NCBI

198 

Kaur S, Tripathi DM and Yadav A: The enigma of endothelium in COVID-19. Front Physiol. 11:9892020. View Article : Google Scholar :

199 

Rovas A, Osiaevi I, Buscher K, Sackarnd J, Tepasse PR, Fobker M, Kühn J, Braune S, Göbel U, Thölking G, et al: Microvascular dysfunction in COVID-19: The MYSTIC study. Angiogenesis. 24:145–157. 2021. View Article : Google Scholar

200 

Holcomb D, Alexaki A, Hernandez N, Hunt R, Laurie K, Kames J, Hamasaki-Katagiri N, Komar AA, DiCuccio M and Kimchi-Sarfaty C: Gene variants of coagulation related proteins that interact with SARS-CoV-2. PLoS Comput Biol. 17:e10088052021. View Article : Google Scholar : PubMed/NCBI

201 

Shahidi M: Thrombosis and von Willebrand factor. Adv Exp Med Biol. 906:285–306. 2017. View Article : Google Scholar

Related Articles

Journal Cover

March-2022
Volume 49 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Geronikolou SA, Takan I, Pavlopoulou A, Mantzourani M and Chrousos GP: Thrombocytopenia in COVID‑19 and vaccine‑induced thrombotic thrombocytopenia. Int J Mol Med 49: 35, 2022
APA
Geronikolou, S.A., Takan, I., Pavlopoulou, A., Mantzourani, M., & Chrousos, G.P. (2022). Thrombocytopenia in COVID‑19 and vaccine‑induced thrombotic thrombocytopenia. International Journal of Molecular Medicine, 49, 35. https://doi.org/10.3892/ijmm.2022.5090
MLA
Geronikolou, S. A., Takan, I., Pavlopoulou, A., Mantzourani, M., Chrousos, G. P."Thrombocytopenia in COVID‑19 and vaccine‑induced thrombotic thrombocytopenia". International Journal of Molecular Medicine 49.3 (2022): 35.
Chicago
Geronikolou, S. A., Takan, I., Pavlopoulou, A., Mantzourani, M., Chrousos, G. P."Thrombocytopenia in COVID‑19 and vaccine‑induced thrombotic thrombocytopenia". International Journal of Molecular Medicine 49, no. 3 (2022): 35. https://doi.org/10.3892/ijmm.2022.5090