key: cord-0877260-0g0c3uag authors: Menter, David G.; Afshar-Kharghan, Vahid; Shen, John Paul; Martch, Stephanie L.; Maitra, Anirban; Kopetz, Scott; Honn, Kenneth V.; Sood, Anil K. title: Of vascular defense, hemostasis, cancer, and platelet biology: an evolutionary perspective date: 2022-01-12 journal: Cancer Metastasis Rev DOI: 10.1007/s10555-022-10019-5 sha: 1c337d9fc1c501395f8551e1e49e5a531ce1836c doc_id: 877260 cord_uid: 0g0c3uag We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2. We have acquired an informed perspective and extensive expertise in the field investigating the role of platelets in cancer biology [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] . One of the key discoveries that led to a better understanding of the links between coagulation and cancer was that of Trousseau's syndrome, which is defined as a migratory thrombophlebitis found typically in patients with an underlying malignancy. Among other discoveries throughout the years, one of the key discoveries linking platelets to tumor cell interactions was made by Gasic et al. [11] . Our efforts in this field greatly expanded the knowledge base regarding platelet-tumor cell interactions and the role of prostaglandins in particular during cancer metastasis [2, 12] . Fast forward, this study is for expanding our current knowledge base in identifying unique aspects of platelet function and platelet biology in the growth and progression of ovarian cancer [4, 10] . It is from this unique cancer-centric perspective that we recognized that the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. This perspective led us to examine the evolutionary connections placing platelets at the epicenter of hemostasis, immunity, and adaptive phylogeny. All of these novel perspectives may provide new leads to better understand not only the role of platelets in vascular protection and immune function but also a keener insight into the role of platelets in cancer and metastasis. Positive sense, single-stranded RNA viruses are thought to be among the earliest forms of life; lineages have been traced through the conservation of RNA-dependent RNA polymerases within the picornavirus super-group [13] [14] [15] [16] . Over the past century, multiple single-stranded RNA viruses, including human immunodeficiency virus-1, influenza A, and severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2), have emerged to infect humans on a large scale-a modern consequence of ancient virus evolution [17, 18] . Endogenous retroviruses help to record past RNA retroviral infections and are ubiquitous in vertebrate genomes [19] . Coronaviruses are among the class of positive-strand RNA viruses that infect mammals and birds and can achieve zoonosis [20] [21] [22] . Evidence of a SARS-like coronavirus arising in bats shows zoonotic implications of a direct ancestor of SARS coronavirus [23, 24] . Bats can carry viruses that are deadly to other mammals without having serious symptoms, possibly involving unique immune-and rapid cellular-spreading properties [25] . During the current SARS-CoV-2 pandemic, key viral mutations may have also occurred via person-to-person transfer complicated by zoonotic passage, highlighting the rapid evolution of this virus [26] [27] [28] [29] [30] [31] [32] . Overall, the phylogeny of vertebrate RNA viruses reflects that of their vertebrate hosts coevolving with a transition from ocean-to-land and exhibits origins that date back to the time of early terrestrial animals [18, 33, 34]. Many aspects of platelet biology and evolution remain unrecognized, especially regarding their rapid responsiveness to viruses [35] [36] [37] . Platelets may have coevolved with viruses to provide speedy responses but this possibility remains unexplored. However, platelets have been shown to endocytose viral particles and are activated via toll-like receptor signaling [38] . The severity of low-platelet count (thrombocytopenia), a common complication of influenzavirus infection, predicts the clinical outcome of critically ill patients [39] . These platelet responses are biochemically and biologically rapid and may also involve integrin receptors [40] . In the case of viral diseases like COVID-19, the responses may remain unnoticed during asymptomatic and early phases [41, 42] . As there is an expression gradient of entry factors for SARS-CoV-2 along the airways, these are the likely sites of initial viral infection, spread, and clearance [43] . General symptoms can consist of cough, myalgia, loss of appetite, and gastrointestinal discomfort. Olfactory and gastrointestinal disorders are prevalent with sudden anosmia (loss of smell), ageusia (loss of taste), or dysgeusia (metallic taste), but not always fever, as informative symptoms of the COVID-19 infection [44] [45] [46] [47] [48] . Given the one-to-two, cell-layer separation between many airway passages and the microcirculation coupled with the evolutionary characteristics of platelets, this suggests that they are the first thrombocyte/immunocyte that virions such as SARS-CoV-2 may encounter and thereby rapidly respond to in the bloodstream [38, 49] . For example, with the human immunodeficiency virus, platelets have been shown to reduce viral load [50] and harbor viral particles [51]. Coincident activation of innate immunity and the coagulation system after an injury is a phylogenetically ancient, adaptive response that can be traced back to the early stages of eukaryotic and chordate evolution [52] [53] [54] [55] [56] [57] . Most invertebrate species like the horseshoe crab (Limulus polyphemus) lack differentiated phagocytic cells and platelets, but they do have large nucleated amoeboid granular hemocytes [58-60] (Fig. 1) . Once their open circulatory system is breached, they respond to microbial threats via a common amoebocyte-and humoral pathway of inflammation and clotting [58] [59] [60] . Lysates prepared from the amebocytes of Limulus have long been known to undergo coagulation upon exposure to endotoxin and have been used to detect that endotoxin [61] . Archetypal functions of Limulus amebocytes are found in human platelets, including aggregation, adhesion, spreading, and granule-based release of coagulation factors [62] [63] [64] . These properties are found throughout phylogenetic evolution as common antimicrobial and proinflammatory responses to stimuli that activate both the clotting cascade and the phagocytic effector cells. Non-mammalian vertebrates-that include early chordates, fish, birds, amphibians, and reptiles-have large, nucleated, Throughout evolution, the cell size and dimensions of thrombocyte nuclei have gradually decreased; this process accelerated as species became more terrestrial and began to thermoregulate (Fig. 1 , a more important function of platelets in mammals may be the lack of nuclei and limited replication machinery to support long-term viral replication, providing a more highly evolved defense and, potentially, a sink against these infections. The vertebrate transition from aquatic to terrestrial environments applied significant selective pressure to the coevolution of viruses and animals. This was accompanied by multiple geophysical, external, selective pressures, which include changes in luminosity, oxygen, carbon dioxide, and asteroid-based impacts (Fig. 2) . Co-evolution is a complex interaction of co-divergence between virus and host over an extended time and includes frequent cross-species transmissions coupled with external survival pressures severe enough to elicit the change [18, 33] . In this manner, SARS-CoV-2 evolution has accelerated in conjunction with zoonotic exchanges with humans over the past 100 years (on the time scale in Fig. 2 , this would be approximately the size of a period). The evolution of platelets includes multiple adaptations to deal with viral infections. As one evolutionary mechanism for dealing with viral pathogens, large, multinucleated megakaryocytes evolved in the bone marrow to protect and enable the production of high numbers of platelets [89-91]. These mechanisms have avian roots and enabled eutherian placentalization [92, 93] . There is another significant advantage over the 2-N thrombocytes of lower vertebrates found in mammalian megakaryocytes. Significantly enlarged megakaryocytes provide a major amplification response to bleeding or viral immune threats incurred by increasing DNA content up to 128 N [93] No studies have yet determined whether these megakaryocytes serve and monoclonal source of targeted platelets responding to a specific threat. The adaptation of increasing DNA content significantly increases the capacity to produce even more platelets with increased receptor density; more organelles per unit cellular volume; and more prothrombotic proteins to reduce bleeding time or adapt to viral threats. This process culminates in mammalian platelet evolution and hemochorial placentation because mammals are the only vertebrate group that has evolved with a highly effective and unique system of hemostasis [93] . This hemostasis effectiveness is essential at parturition where even minimally invasive placentae can hemorrhage [93] . Platelet evolution has enabled the prevention of rapid blood loss and damage from massive injury or microinjury, which is particularly important when the brain is more advanced. Because microbes and viruses have a short ten-day life span, platelets must have co-evolved to create survival advantages with adaptive mechanisms to meet and sequester rapidly cycled microbial-and viral threats and pathogen response-based turnover. The sheer number and biophysical properties of megakaryocyte-derived mammalian platelets make them more likely to be the first blood component to encounter virus particles and limit spread. Human platelet genesis is well studied and primarily occurs by membrane-bound, organelle-containing cytoplasmic extrusion into the blood stream of numerous small size-and volume blebs ranging between 9.7 and 12.8 femtoliters or in plate-like structures of 2.6 to 2.9 μm in diameter [90] . This blebbing, subcellular, genesis process occurs primarily in the bone marrow from surfaces of megakaryocytes-the largest cell in the body (ranging between 40 and 100 μm in diameter). Normal human-platelet concentrations range between 150,000 and 400,000 per microliter. The roughly 10 11 platelets produced each day are responsible for coagulation-specifically for creating a fibrin plug at the site of blood vessel injury. However, emerging data suggest a greater impact on viral interdiction, pathogen surveillance, cancer biology, and immune function. As part of the . Platelet surface changes that occur after taking in viral particles can also mark the platelets for clearance by other organs including the liver and spleen. Other key aspects of adaptive evolution in the mammalian platelet are size decreases and improvements in biophysical shape properties to enhance encounters with virus-infected vascular surfaces. In lower vertebrates, nucleated thrombocytes are less plate-like and10 to 20 times larger than platelets causing them to primarily distribute in the center of laminar shear fields [36, 70, 72] (Fig. 1 ). In humans, resting platelets are plate-like discs designed to maximize planarsurface interactions [112] [113] [114] that, due to their small size and shape, biophysically concentrate toward the outer fluidshear fields of flowing blood [115] [116] [117] [118] [119] [120] [121] [122] . Taken together, these properties enhance encounters and recognition of any vascular-wall lesions, wounds, or tumors. Should platelets encounter basement membranes or their underlying matrix, the platelets undergo receptor-mediated activation [2, [123] [124] [125] [126] [127] . This activation is connected to rapid cytoskeletal-and membrane changes to form filopodia leading to adhesion [128] [129] [130] [131] [132] [133] [134] [135] . This is a process that occurs within seconds along with a shape change from exocytic degranulation. Degranulation, in turn, encompasses the release of proteins, growth factors, cytokines, and lysozymes along with a variety of bioactive lipids, small molecules, and other factors. Successful sequential recruitment of additional platelets and immune cells coupled with thrombogenesis ultimately seals any tissue gaps and sterilizes the lesion. Since platelets segregate within shear fields near the endothelial cell surfaces, they have a high probability of encountering viral particles that enter the bloodstream. This behavior also affords platelet endothelial-cell interactions that can occur with an aggressive, influenza-like cytokine storm that triggers cell damage in lung capillaries [136] . Compared to any other form of circulating microvesicle, exocytic platelets generate the majority of microparticles and exosomes in circulation [137] [138] [139] [140] [141] [142] -possibly another form of platelet evolution to deal with viral threats. In lower vertebrates, microparticle generation by thrombocytes plays a key role in thrombocyte function [70] . During laser-injury experiments in zebrafish, thrombocyte microparticles were the first cell-based entities to arrive at the injury site, even before the young thrombocytes [143, 144] . In the case of viral infections like SARS-CoV-2, platelet microparticle and exosome shedding would significantly increase the platelet-membrane surface area and possibly mitigate the threat potential through target decoration and further immunesystem stimulation [145] . A phylogenetic adaptive loss of platelet nuclei may also enable rapid endothelial transmigration into perivascular spaces. Platelets readily undergo tissue migration because of their small size, minimal displacement volume, and highly active cytoskeletal responses linked to activation, adhesion, and aggregation [146] [147] [148] . Even greater is the vascular-transmigration advantage of being unencumbered by the presence of nuclei, which limit the migration of other immune cells to interdict viral threats [146] [147] [148] [149] [150] . This nuclei absence may also limit the distance that platelets can move into tissue, which limits nucleotide and protein-synthesis capabilities. Due to limited nucleotide and protein-synthesis capabilities, platelets cannot indefinitely maintain protein replacement [80, 151] . These immune-surveillance and rapid viral-response properties facilitate speedy sterilization and quickly initiate the repair process during a virus-induced, tissue-injury response. With viral infections, platelets have been shown to influence viral invasion of the central nervous system in canine distemper-virus infection [152, 153] ; however, few studies exist that examine platelet invasion or extravasation as part of a viral immune response. Alternatively, because viral infection may lead to endothelial damage and platelet extravasation, activation and granule release may promote the attraction of other immune cells that produce inflammation in the affected tissues. Various factors in response to various stimuli can contribute to platelet transmigration across leaky or inflamed vessel walls to aid in wound sterilization and tissue regeneration [154] [155] [156] [157] [158] [159] [160] . Platelet inflammatory responses can also cause them to undergo vascular transmigration and granule release that engages various additional immune cells [161] [162] [163] . Within the extravascular microenvironment, the release of the stromal cell-derived factor-1 named C-X-C chemokine-ligand 12 (CXCL12), along with other cytokines, can also provide a potent stimulus for platelet migration into extravascular spaces [10, 150, 164-167] [168] [169] [170] [171] . Among the many response factors expressed by platelets are C-X-C chemokine-receptors 4 and 7 (CXCR-4, CXCR7), which are the cognate receptors for CXCL12. These platelet receptors may be involved in inflammatory-or allergic responses or in platelet activation in human immunodeficiency virus and other viral infections [172] . Along with those involving lymphatic biology, these thrombo-immune responses and wound healing characteristics were conserved from lower vertebrates [107, [173] [174] [175] [176] [177] [178]. Cyclooxygenase and prostaglandin production, associated with gene duplications and losses, have been retained throughout metazoan and vertebrate evolution [179, 180] that began in teleosts and resulted in a divergence between cyclooxygenase-1 and cyclooxygenase cyclooxygenase-2 (COX-1 and COX-2) [180] . A release of the COX-2-associated prostacyclin PGI 2 from vascular endothelial cells helps to maintain the resting state of circulating platelets through G-protein-coupled receptors (GPCR) [181] [182] [183] [184] [185] (Fig. 3 ). Should platelets become activated during the virus uptake, responses are rapidly amplified by activating molecules such as thrombin or adenosine diphosphate (ADP). These molecules are released by platelets and trigger their cognate GPCRs at the cell surface or as a result of the release reaction. Similarly, COX-1-associated and endogenously synthesized thromboxane A 2 (TXA 2 ) amplifies the local platelet response to recruit additional platelets through TXA 2 -GPCRs that accelerate the thrombogenesis/clotting process (Fig. 4) . Platelet TXA 2 levels increase following virus infections and can be attenuated by aspirin treatment [186] [187] [188] [189] [190] . The production of TXA 2 and other lipids initiates vasoconstriction [191, 192] . As a primary stimulant of platelet recruitment and aggregation, TXA 2 is among the shorterlived prostaglandins due to molecular epoxide bond strain in the active part of the molecule that is prone to rapid (< 30 s) hydrolysis [193, 194] . Because these rapid changes accelerate over a matter of minutes, a fibrin network is generated to trap and activate more platelets within the forming clot in a cyclic fashion to limit blood loss. This amplification process occurs over 20 min or less, while clot maturation and solidification continue for about an hour to initiate-over days to weeks-prompting immune-cell recruitment and inflammation. Recruitment of fibroblasts and immune cells through platelet-initiated, angiogenic repair mechanisms stimulates further wound healing and resolution [195] . The release of dense, δ-granule components influences the vascular microenvironment of the platelet (Fig. 3) . The release of calcium and magnesium ions promotes platelet activation and aggregation. The release of nucleotides such as ADP activates platelets through P2Y 1 and P2Y 12 receptors [196, 197] and stimulates vasoconstriction [198] (Fig. 4) . ADP can accumulate in the ischemic vascular microenvironment as a consequence of circulatory stasis or thrombus formation. The release of neurotransmitters such as serotonin (5-hydroxytryptamine), epinephrine, and histamine can potentiate ADP-induced platelet activation and aggregation [196, 197, 199, 200] . Platelets may serve as neuronal and innate immune cells to help mediate T cells during tissue inflammation [201] . Epinephrine is also likely to worsen COVID-19-induced stress-related effects on platelets and platelet-mediated cytokine responses [202] . By contrast, lower-vertebrate thrombocytes do not aggregate in response to ADP or epinephrine; neither do they accumulate or produce serotonin [65, 203] . Conversely, platelet release of alpha (α)-granules in serum results in substantial increases in multiple proteins responsible for most of the coagulation, growth stimulation, angiogenesis, immune function, and wound-repair factors. Adhesion molecules released from α-granules help to stimulate the rapid arrest of platelets in circulation. This is a process that is mediated by adhesion factors such as the von Willebrand factor (vWF), fibrinogen, thrombospondin, and fibronectin as well as integrins and other adhesion receptors αIIbβ3, αvβ3, and P selectin. These adhesion proteins regulate platelet interactions with other platelets, and with endothelial cells, exposed basement-membrane extracellular matrix, leukocytes, neutrophils, monocytes, and tumor cells. These same storage α-granules release the prothrombin, fibrinogen, factor V, and factor VIII that stimulate and promote coagulation and fibrin formation [3, 9, [204] [205] [206] [207] [208] [209] [210] [211] [212] . At the top, virus replication within vascular endothelial cells is initiated by SARS-CoV-2 spike-protein binding to angiotensinconverting enzyme (ACE) and enzymatic processing by transmembrane protease, serine 2 (TMPRSS2). Viral uptake results in viralparticle uncoating, viral reverse transcriptase activity of the positive single-stranded (+ ssRNA) genome, and replication. The endoplasmic reticulum of the host cell supports protein synthesis, assembly in the Golgi apparatus, and delivery to the exterior by exosomal transport or host-cell disruption, death, and retraction. The platelet depicted at the center illustrates the numerous receptors that bind to various virus types. Those that bind coronavirus species are indicated by largerfont text and include (alphabetically) coxsackie-adenovirus receptor (CAR), CCL chemokine Cys-Cys (C-C motif) ligand, cluster of differentiation (CD), C-type lectin domain family 2 and 5 (CLEC-2 and CLEC-5), complement receptor (CR 3a and CR 5a), Cys-X-Cys (C-X-C) motif chemokine receptor type (CXCR1, CXCR1-2, and CXCR1-4), dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), Fc gamma receptor II (FcγRII), heparan sulfate proteoglycan (HSP), integrin (α v β 3 ), and toll-like receptor (TLR). Also depicted are the phosphate-transferring nucleotide-binding domain-like receptor protein 3 (NRLP3), the inflammasome-response switch serine/threonine-protein kinase (NEK7), and the pro-inflammatory cytokine caspase 1 that mediates the enzymatic processing of interleukin-1β (IL-1β) prior to microvesicle release. Also shown are the large numbers of G-protein-coupled receptors involved in platelet signal transduction, activation, and granule release Endothelial cells express angiotensin-converting enzyme (ACE) and transmembrane protease, serine 2 (TMPRSS2) that are involved in the SARS-CoV-2 invasion via its spikesurface protein binding and processing [213] [214] [215] [216] [217] [218] (Fig. 4) . After uptake, SARS-CoV-2 undergoes uncoating and viral-RNA reverse transcription and replication in the rough endoplasmic reticulum followed by new SARS-CoV-2 assembly and release (Fig. 4) . Should the virus infect endothelial cells, endotheliitis, complement activation, and endothelial-cell death can occur [219] [220] [221] [222] [223] [224] . Endothelial-cell retraction, whether virus-or inflammation-induced, can lead to matrix exposure that triggers platelet activation and granule release. Blood-vessel injury can amplify platelet responses and may help trigger heightened immune responses during the second phases of COVID-19 infection [219, 222, 225] . Analysis of megakaryocytes in bone marrow data from the Human Cell Atlas did not identify expression of these receptors [226, 227] ; however, the detection of ACE2-and TMPRSS2 receptors in platelets requires direct protein-characterization technology, not RNA sequencing, because the platelets lack nuclei and the sequencing technique does not provide adequate sensitivity. As part of their immune-surveillance properties, platelets can recognize foreign bodies or invading pathogens [9, [228] [229] [230] . Along with other platelet receptors, the toll-like receptors expressed by megakaryocytes [231] may be involved in innate immune responses to viruses, may take part in endocytosis of a variety of pathogens by platelets [38, [232] [233] [234] , and may release factors that can influence antiviral responses [157, [235] [236] [237] [238] [239] [240] [241] [242] [243] [244] [245] [246] (Fig. 4 ). Normally, platelets serve to rapidly respond during the wounding process and hemostasis. When virally damaged, blood-vessel endothelial cells die, are lost, or retract; this exposes platelets to subendothelial elements that initiate receptor-based recognition of the extracellular matrix. These interactions are driven by a variety of platelet glycoprotein receptors that bind to endothelial cells and extracellular matrix factors including proteoglycans, laminin, fibronectin, vitronectin, and various isoforms of collagen. vWF binds to exposed collagen I, collagen III, and collagen VI fibers. vWF is also recruited into matrix networks by forming tethering fiber strands [247] [248] [249] [250] [251] . Many vWF characteristics of hemostatic proteins are conserved during ancestral vertebrate evolution, but the hagfish (Myxine glutinosa) jawless vertebrate (a cyclostome) lacks functional domains required for primary hemostasis under high flow [66] . Many of the typical platelet-surface receptors also bind to viruses [34] but the role of platelet binding that may sequester viral particles in the vasculature has not been studied in detail. Platelets contextually encounter numerous bloodstreamcirculating ligands, cells, pathogens, and extracellularmatrix ligands. Immediate platelet responses are governed by a variety of integrin receptors through transmembrane glycoprotein-α and glycoprotein-β heterodimers once ligands are engaged (Fig. 4) . Resting platelets express low-affinity conformation integrins that are bent to protect binding sites and to form high-affinity-or open-binding states that, once activated, efficiently interact with ligands. Rolling platelet behavior is initiated by surface-level, multimeric GPIb-complex interactions with vWF that activate a key stabilization integrin α IIb β 3 [252] [253] [254] [255] to bind a variety of RGD (arginine-glycine-aspartic acid)-containing ligands [252] [253] [254] [255] including fibrin, fibrinogen, fibronectin, vitronectin, thrombospondin, or vWF complexes. Platelets also interact with hanta-and adenoviruses through α IIb β 3 receptors [94, 98, [256] [257] [258] [259] [260] . Certain integrins bind to echo-and rotaviruses [261] while vitronectin is engaged by platelet α v β 3 integrin heterodimers that also bind to echovirus9; coxsackieviruses A9 and -A16; and hanta-, parecho-, and coronaviruses [98, 256, 257] . Platelet C-type lectin-like receptors CLEC-2 and CLEC-5A initiate platelet activation and trigger signal transduction through molecular multimerization [262] [263] [264] [265] [266] . CLEC-2 binds to the transmembrane sialomucin-glycoprotein podoplanin among other molecules [267] [268] [269] . Podoplanin is expressed on cells of the lymphatic endothelia, type I lung epithelia, choroid plexus epithelia, kidney podocytes, and lymph-node stroma to potentiate migration and invasion [267] [268] [269] [270] . CLEC-2 expression is maintained on activated platelets and on platelet microparticles to substantially increase the probability of platelet-mediated interactions with virus particles [271] . Platelet CLEC-2 and CLEC-5A receptors bind to the human immunodeficiency-and coronaviruses [271] [272] [273] . Platelet P-selectin, also known as membrane glycoprotein GMP-140 [274, 275] , binds to P-selectin glycoprotein ligand 1 (PSGL-1) [276] , neutrophil leukocyte-endothelial cell-adhesion molecule 1 (LECAM-1) [277] , endothelial cell-leukocyte adhesion-molecule 1 (ELAM-1) [278] , and Sialyl Lewis (x) oligosaccharide [279] . Along with a variety of immune cells and endothelial cells, P-selectin interaction is important in mediating inflammation, autoimmunity, and wound healing, and plays a role in coxsackievirus-induced myocarditis [280] [281] [282] [283] [284] . P-, L-, and E-selectins help with the tethering and rolling of cells that flow past the vascular luminal surfaces during the initial phases of intravascular, adhesive interactions. These processes are stabilized by other receptors [285] [286] [287] [288] [289] [290] [291] and may contribute to interactions with virus-laden platelets. Platelets often serve as foci for coordinating thrombus initiation and formation. Tissue factor initiates platelet activation and aggregation [292] and may be linked to some virusinduced prothrombotic states associated with septic shock, myocarditis, and respiratory viral infections [293] [294] [295] [296] . Thromboemboli and thrombocytopenia can also lead to COVID-19-related complications as reported in a number of publications [297] [298] [299] [300] [301] [302] [303] [304] [305] [306] . In vivo, the viral threat mitigation process is likely to be rapid, as platelets aggregate in response to circulating threats. In the case of platelet interactions with virions such as SARS-CoV-2, intravital observations are unlikely due to the small, average-viral-particle size. Histologically, these changes are difficult to track microscopically due to the lack of nuclei as platelet reference points; the post-activation, morphological platelet changes; and dramatic, aggregation-related shape changes that create an amorphous mass. Rapid thrombocyte/platelet responses have been conserved phylogenetically in vertebrates [307] . Depending on the agonist, the initiation of zebrafish thrombocyte/platelet aggregation occurs within 20 s, plateaus over 1-3 min, and involves conserved α IIb receptor subunits [307] . Thrombotic plug formation is stabilized and counterbalanced by the disaggregation of platelets and fibrinolysis. Platelets migrate and help to initiate immune sterilization, tissue repair, and scar formation [195, [308] [309] [310] [311] . In critically ill patients with a significant viral load, these plateletmediated immune responses may be more rapid and severe. Major and life-threatening events associated with SARS-CoV-2 infections are common. A number of coagulation/ thrombotic disorders are linked to COVID-19 infections including stroke, disseminated intravascular coagulation, pulmonary embolisms, acute respiratory distress syndrome (ARDS), sepsis-induced coagulopathy, local microthrombi, venous thromboembolism, arterial thrombotic complications, and thrombo-inflammation connected to disease progress, severity, or mortality [312] [313] [314] [315] . One tragic example in the young is the association between COVID-19-related strokes and amputations [316] [317] [318] [319] [320] [321] [322] [323] [324] [325] [326] . Platelets can play an integral role at any stage of coagulation/thrombotic disorders. Like other viral infections, asymptomatic disease is present in a significant but hard-to-identify fraction of affected individuals. In most symptomatic patients, a 1-week, self-limiting, viral respiratory disease often occurs [221, [327] [328] [329] [330] [331] [332] [333] . This phase of disease may end when neutralizing-, anti-viral-, T-cell-, and antibody immunity is engaged. If cross-reactivity occurs with other serum-based coronavirus antigens, immunoglobulin (Ig)M-, IgA-, and IgG-type virus-specific antibody levels can be important indicators of population immunity against this disease. In the case of extreme viral load during the first infection course or repeated exposure to virus that can occur in healthcare workers, the rate and extent of this response can be an important factor for severity and localization of disease [221, [327] [328] [329] [330] [331] [332] . These stage-and extent-of-disease responses will influence the development of any high-specificity and a high-accuracy serological assay that is easy to use, reliable, reproducible, and critical to the successful measurement of COVID-19 immunity in our global population [334] [335] [336] [337] [338] [339] [340] [341] [342] [343] [344] [345] [346] [347] [348] [349] [350] [351] [352] [353] . As COVID-19 progression becomes more severe, antibodies that platelet FcγRIIA (CD32a) receptors can recognize may be generated [259, 354] (Fig. 4) . FcγRIIA-known to recognize aggregated IgG complexes [355] -can contribute to α IIb β 3 activation and aggregation [356] . Aggregated IgG complexes can trigger the release of microvesicles, as can happen with H1N1-virus (swine flu) exposure [354, 356] . FcγRIIA receptors contribute to the production of COVID-19-related autoantigens also amplified by platelet microparticle release. This may underlie the formation of potent inflammatory components: the microparticle-associated immune complexes [357] . Similarly, the complexes appear to play a role in heparin-induced thrombocytopenia, a prothrombotic disorder mediated by complexes between platelet factor 4 (PF4) and heparin or other polyanions [358] . This risk of thrombosis may extend beyond exposure to heparin implicating other PF4 partners as well [358] . Platelet FcγRIIA receptors are capable of recognizing multiple antibody subclasses that include IgM, IgA, and IgG [359] [360] [361] [362] [363] [364] -all produced by patients with COVID-19 [221, 327, 339, 342, 365] . Thrombocytopenia initially has been associated with severe COVID-19 adverse outcomes and mortality [297, 300, [304] [305] [306] 366 ]-possibly due, in part, to the consumption of platelets during the ongoing platelet-virus interaction process. However, others have not found thrombocytopenia to be an infallible predictor of disease progression or adverse outcome [367] . Decreases in platelet numbers have been associated with poor outcomes in multiple studies [306, [368] [369] [370] [371] [372] [373] [374] . Platelet changes can be associated with neutrophil extracellular traps-extracellular webs of chromatin, microbicidal proteins, and oxidant enzymes that are released by neutrophils to contain infections [242, 375, 376] . Some of these conserved higher-orders in response to microorganisms can lead to clustering around pathogens that encapsulates them, properties which are also observed in thrombocytes of lower vertebrates [377] [378] [379] . Using ultrasound-guided, minimally invasive autopsy to assess COVID-19 pulmonary involvement revealed the presence of fibrinous thrombi in alveolar arterioles along with a high density of alveolar megakaryocytes that likely contribute to thrombocytosis [222] [380] . With accompanying elevations in interleukin (IL)-1 and IL-6 in patients with COVID-19 [297, 370, [381] [382] [383] [384] [385] [386] [387] [388] [389] [390] [391] [392] [393] , this may lead to thrombocytosis due to inflammation-induced platelet activation, inflammasome formation, and IL-1-laden microvesicle release (Fig. 4) , all of which are processes that associate with consumption-based feedback. IL-6 elevates platelet production in the metastatic ovarian-cancer setting when generated by tumors that stimulate liver-based thrombopoietin production [4, 10] . Thrombopoietin, in turn, stimulates megakaryocyte/platelet thrombocytosis in the bone marrow [4, 10] . By contrast with COVID-19, pathogenic T cells and inflammatory monocytes associated with large amounts of IL-6 secretion can incite an inflammatory (cytokine) storm, which may be curbed through treatment with tocilizumab, a monoclonal antibody treatment that targets IL-6 pathways [380] . Controversy exists over the use of aspirin in patients with COVID-19 particularly in those who are pregnant and display skin reactions [394] [395] [396] . In other studies, aspirin has shown benefit when combined with rivaroxaban (dual pathway inhibition) for the prevention of ischemic events in patients with cardiovascular-and peripheral-artery disease [397] [398] . Whether platelet consumption is the body's way of isolating these diseases or immediately mitigating the spread of COVID-19 remains to be determined. In other studies, anti-coagulation effects were associated with improved COVID-19 survival after adjusting for mechanical ventilation. ARDS is among the most worrisome of all responses to SARS-CoV-2 infections [220, 221] (Fig. 5 ). Platelets can promote or contribute to all stages of ARDS development and progression, including thrombosis, inflammation, angiogenesis, and fibrosis [219-221, 223, 313, 366, 399-402] . Platelets can also help initiate many of the inflammatory responses in the lung. Endothelial-cell death and retraction can trigger platelet activation and granule release [219] [220] [221] 403] , the latter of which includes many of the proinflammatory molecules that contribute to immune-cell infiltration and cytokine storming [27, [404] [405] [406] [407] [408] [409] [410] [411] [412] . Autopsies of COVID-19 patients have been difficult to achieve but they reveal many details of ARDS [220-222, 224, 413-418] . Certain reports suggest that in severe COVID-19 cases, platelets are centered within thrombotic responses with an absence of protective immune states [403, 419, 420] . As ARDS progresses, exudative, diffuse alveolar damage (DAD) occurs and can intensify the release of alveolar exudates to stimulate hyaline membranes, septal edema, and mild/moderate lymphocytic infiltration. This can lead to pleomorphic, alveolar, epithelial-cell alterations resulting from SARS-CoV-2 cytopathic effects, which then can cause diffuse epithelial desquamation. Epithelial cells can also develop distorted cytoplasm, large nuclei, eosinophilic nucleoli, giant cells, and squamous alveolar metaplasia. The release of platelet α-granule proteins can stimulate epithelial-cell-and endothelial proliferation and fibroblast invasion. Proliferative DAD has been characterized by disorganized fibrous tissue within alveolar septa while alveolar lumens can become severe with fibrinous thrombi [222] . These pulmonary changes are the result of severe epithelial injury and microthrombotic vascular phenomena [222] . In patients with COVID-19, platelets can associate with SARS-CoV-2 RNA to become hyperactivated and can contribute to an eicosanoid storm [421] [422] . Hyperactivated platelets are characterized by proinflammatory granule release which includes C-X-C motif chemokines such as CXCL1 (GRO-α), CXCL4 (PF4), CXCL5 (ENA-78), CXCL7 (PBP, β-TG, CTAP-III, and NAP-2), CXCL8 (IL-8), and CXCL12 (also called stromal cell-derived factor-α) [171] (Fig. 4) . Platelet α-granules that are C-C motif chemokines include CCL2 (MCP-1), CCL3 (MIP-1α), CCL5 (RANTES), CCL7 (MCP-3), and CCL17 (TARC); along with IL1-β, the platelet-activating factor acetylhydrolase, and lysophosphatidic acid [423] . In some cases, SARS-CoV-2-infected lung tissue may not be heavily infiltrated by CD20 + B cells, CD57 + NK cells, or lymphoid aggregates. By contrast, the number of CD4 + and CD8 + T cells may be few-to-moderate and may form small aggregates in patients with fibroproliferative DAD [222] . CD68 + macrophages populate alveolar spaces and sites of tissue remodeling in fibroproliferative areas. Some multinucleated, atypical giantcells (CD68 + alveolar macrophages) [222] can amplify the production and release of factors that contribute to a cytokine storm [424] [425] [426] [427] . In SARS-CoV-2-infected lung tissue ex vivo, platelets combined with resident and infiltrating immune cells led to the increased production of interferons IFNβ, IFNγ, IFNλ1, IFNλ2, and IFNλ3 as well as interleukins IL-1β, IL-6, IL-8, MCP1, MIP1α, RANTES, CXCL1, CXCL2, and CXCL5 [427] . Platelets can also contribute to many of the extrapulmonary comorbidities related to ARDS [220-222, 402, 413-417] . In many cases, the extrapulmonary disease attributed to thrombotic axis comorbidities (such as hypertension and diabetes mellitus) often shows renal arteriolosclerosis, cardiomyocytes hypertrophy, myocardial fibrosis, focal glomerular sclerosis, liver steatosis, and cerebral microvascular disease [222] . COVID-19-induced shock and thrombotic lesion formation may also be associated with liver-based centrilobular congestion and acute kidney-tubular lesions. Syndromes secondary to SARS-COV-2 infection include systemic inflammation or shock linked to platelet activity associated with myositis, orchitis, lymphomononuclear myocarditis, and superficial perivascular mononuclear infiltrates in the skin. Secondary platelet endothelial-cell-mediated changes in small vessels can include cell tumefaction, vessel wall edema, and fibrinoid alteration. Other secondary changes include fibrin microthrombi in the glomeruli, skin, testis, liver sinusoids, and heart; mesangial glomerulopathy; liver-based hyperreactive Kupffer cells; spleen-based lymphoid hypoplasia; and brain-based reactive gliosis. In contrast to type 1 pneumocytes, type 2 pneumocytes contain high levels of angiotensin-converting enzyme (ACE) and highly expressed transmembrane protease, serine 2 (TMPRSS2) -key targets for SARS-CoV-2 infection. The ensuing viral replication and vascular spread can trigger both vascular endothelial-and pneumocyte-cell death and matrix exposure that activates platelets. Pulmonary injury in SARS-CoV-2-induced COVID-19 can be amplified by many platelet-derived factors and bioactive processes [440] . Complete occlusion of blood vessels leads to hypoxia and additional damage. Acute respiratory distress and diffuse alveolar damage (DAD) can be induced by platelet factors and amplified by residential macrophages, neutrophils, and lymphocyte apoptosis. Platelets release cytokines and chemokines that also stimulate the production of residential, alveolar immune cells; recruitment of additional macrophages and neutro-phils; lymphocyte apoptosis; formation of reactive oxygen species (ROS); and increased inflammation. Vascular endothelial-cell damage resulting from viral damage and complement activation amplifies the platelet activation and increases permeability and inflammatory thrombus formation. Fibrin formation and fibrinolysis can also be activated, releasing fibrin degradation fragments. Blood-vessel changes can dominate while damage in the alveolar space remains relatively mild. Platelet-neutrophil extracellular traps (NETs) may also play a role in severe disease [441] . Damage that accelerates to the alveolar space becomes more severe with additional immune-celldeath-related NET formation. Platelets, along with increased permeability and edema, can contribute to any cytokine storm that ensues. Platelets can also contribute to recruitment and activation of fibroblasts and myofibroblasts, formation of fibrous tissue, and scarring 6 Summary and future directions This paper describes the importance of platelet evolution and responses secondary to SARS-CoV-2 infection along with their likely role in the thrombotic and immune function of patients with COVID-19. As primordial life forms, viruses (particularly single-stranded-RNA viruses such as SARS-CoV-2) coevolved and diverged as did vertebrates as they adapted to major environmental selective pressures while emerging from an aquatic to a terrestrial environment. Invertebrate and vertebrate thrombocytes and immunocytes retained aggregation capabilities, coagulation enzymes, and phagocytic properties involved in virus immobilization and uptake; these evolved into an active open-canalicular system. Retention of a highly active and responsive cytoskeletal system of granule release and rapid migratory capacity sustained cellular responsiveness to microbial threats. Thrombocyte/platelet evolution developed and maintained receptor-based, pathogen-directed, recognition capabilities to identify viruses. This evolution was coupled with active, membranous vesicle production to increase surface contacts and the probability of interacting with virus particles. As receptor-based response times decreased significantly and diverged over time, a finely regulated balance between endothelial COX2/PGI 2 -and platelet COX1/TxA 2 pathway enzymes emerged. Targeting platelet adenosine receptors may also have an impact on the COVID disease state [428] . Selective serotonin reuptake inhibitors (SSRIs) are also candidates for potential intervention [429] . As vertebrates coevolved with viruses, their platelets typically remained elongated and retained morphological support by a circular ring of microtubules that ultimately formed the mammalian platelet. As they gradually became smaller and evolved into plate-like discs, this maximized planar-surface interactions and provided a biophysical advantage by concentrating the numerous platelets produced each day toward the outer fluid-shear fields of flowing blood. Management of severe COVID-19 is likely to require case-specific considerations depending on viral-load exposure, clinical presentation, and assessment of circulating platelets, megakaryocytes, cytokines, immune cells, and antibodies [430] [431] [432] [433] [434] [435] [436] [437] [438] [439] . Much remains to be discovered regarding the evolution and biological function of platelets including a consideration of the many emerging selective pressures involved with SARS-CoV-2and other viral infections. We continue to evolve in a globally connected world of rapid transit and exchange, which allows for the rapid dissemination of diseases that can outpace our ability to quickly respond with therapeutic interventions. Having learned so much from our experience studying the role of platelets in cancer and metastasis, we have tried to bring new insights into their role in immune defense mechanisms. From this perspective, we recognize that a better understanding of the platelet biology involved in the viral immune response is critical to being prepared for the rapid development of lethal viral threats such as SARS-CoV-2-and those yet to evolve. Tumor cell-platelet interactions in vitro and their relationship to in vivo arrest of hematogenously circulating tumor cells Inhibition of tumor cell induced platelet aggregation by prostacyclin and carbacyclin: An ultrastructural study Possible strategies for antimetastastic therapy The platelet lifeline to cancer: Challenges and opportunities Platelet "first responders" in wound response, cancer, and metastasis Role of platelet-derived tgfbeta1 in the progression of ovarian cancer Role of ADP receptors on platelets in the growth of ovarian cancer FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal Platelets and cancer: A casual or A shear gradient-dependent platelet aggregation mechanism drives thrombus formation Morphological study of the interaction of intravascular tumor cells with endothelial cells and subendothelial matrix The functional role of platelets in the regulation of angiogenesis Real-time imaging of heterotypic platelet-neutrophil interactions on the activated endothelium during vascular inflammation and thrombus Formation in live mice Platelets selectively enhance lymphocyte adhesion on subendothelial matrix under arterial flow conditions Platelet Adhesion Platelets in thrombotic and non-thrombotic disorders A phenomenological particle-based platelet model for simulating filopodia formation during early activation The microtubule modulator RanBP10 plays a critical role in regulation of platelet discoid shape and degranulation Dynamics of platelet thrombus formation Mechanisms and implications of platelet discoid shape The elegant platelet: Signals controlling actin assembly Microtubule coils versus the surface membrane cytoskeleton in maintenance and restoration of platelet discoid shape Platelet adhesion to collagen via the alpha 2 beta 1 integrin under arterial flow conditions causes rapid tyrosine phosphorylation of pp125FAK Anatomy of the Platelet Cytoskeleton Platelets in thrombotic and non-thrombotic disorders Endothelial-platelet interactions in influenzainduced pneumonia: A potential therapeutic target Novel aspects of extracellular vesicles as mediators of cancer-associated thrombosis Correlative analysis of nanoparticle tracking, flow cytometric and functional measurements for circulating microvesicles in normal subjects Circulating microvesicle number, function and small RNA content vary with age, gender, smoking status, lipid and hormone profiles Immobilization and detection of platelet-derived extracellular vesicles on functionalized silicon substrate: Cytometric and spectrometric approach Microparticles in cancer Platelets, circulating tumor cells, and the circulome Role of zebrafish thrombocyte and non-thrombocyte microparticles in hemostasis Laser-induced thrombosis in zebrafish Platelet serotonin aggravates myocardial ischemia/reperfusion injury via neutrophil degranulation A microfluidic imaging chamber for the direct observation of chemotactic transmigration Migration of mononuclear cells in the modified Boyden chamber as evaluated by DNA quantification and flow cytometry Nuclear mechanics during cell migration Platelet-specific chemokines contribute to the pathogenesis of acute lung injury SGK1 sensitivity of platelet migration Age and biosynthesis and breakdown of thrombocyte proteins Establishment of central nervous system infection by canine distemper virus: Breach of the blood-brain barrier and facilitation by antiviral antibody Effects of induced thrombocytopenia on viral invasion of the central nervous system in canine distemper virus infection The migration of platelets and their interaction with other migrating cells Platelets in Thrombotic and Non-Thrombotic Disorders Platelet lysate embedded scaffolds for skin regeneration Platelets in liver disease, cancer and regeneration Platelets: Much more than bricks in a breached wall Personalized plasma-based medicine to treat age-related diseases The impact of autologous platelet concentrates on endodontic healing: A systematic review Platelet-Rich Plasma (PRP) in Orthopedic Sports Medicine The role of platelets in mediating a response to human influenza infection Micro RNAs from DNA viruses are found widely in plasma in a large observational human population Circulating platelets as mediators of immunity, inflammation, and thrombosis Platelets effects on tumor growth The potential role of HIF on tumour progression and dissemination PI3 kinase-dependent stimulation of platelet migration by stromal cell-derived factor 1 (SDF-1) Platelet-derived CXC chemokines: Old players in new games High shear flow induces migration of adherent human platelets Distinct platelet packaging, release, and surface expression of proangiogenic and antiangiogenic factors on different platelet stimuli Differential response of platelets to chemokines: RANTES non-competitively inhibits stimulatory effect of SDF-1 alpha Platelet chemokines in vascular disease Functional expression of CCR1, CCR3, CCR4, and CXCR4 chemokine receptors on human platelets Characterisation of thrombocytes in osteichthyes Differences in responses of grass carp to different types of grass carp reovirus (GCRV) and the mechanism of hemorrhage revealed by transcriptome sequencing The contribution of Aeromonas salmonicida extracellular products to the induction of inflammation in Atlantic salmon (Salmo salar L.) following vaccination with oil-based vaccines A comparative immunopathological study of injection site reactions in salmonids following intraperitoneal injection with oil-adjuvanted vaccines Inflammatory cells of teleostean fish: A review focusing on mast cells/eosinophilic granule cells and rodlet cells Matrix solid-phase dispersion combined with HPLC-DAD for simultaneous determination of nine lignans in Saururus chinensis Ancestral genetic complexity of arachidonic acid metabolism in Metazoa Gene duplications and losses within the cyclooxygenase family of teleosts and other chordates Nitric oxide-sensitive guanylyl cyclase is the only nitric oxide receptor mediating platelet inhibition Analysis of CD39/ATP diphosphohydrolase (ATPDase) expression in endothelial cells, platelets and leukocytes An enzyme isolated from arteries transforms prostaglandin endoperoxides to an unstable substance that inhibits platelet aggregation Synergism between nitric oxide and hydrogen peroxide in the inhibition of platelet function: The roles of soluble guanylyl cyclase and vasodilator-stimulated phosphoprotein Nucleotides and cd39: Principal modulatory players in hemostasis and thrombosis A randomized placebo controlled trial of aspirin effects on immune activation in chronically human immunodeficiency virus-infected adults on virologically suppressive antiretroviral therapy Aspirin attenuates platelet activation and immune activation in HIV-1-infected subjects on antiretroviral therapy: A pilot study The HIV protease inhibitor, ritonavir, dysregulates human platelet function in vitro Biosynthesis and biological actions of prostaglandins and thromboxanes Arachidonate cascade in the platelets of influenza virus infected mice Vasoconstrictor prostanoids EETs promote hypoxic pulmonary vasoconstriction via constrictor prostanoids Synthesis and structure of the platelet aggregation factor thromboxane A2 Thromboxanes: A new group of biologically active compounds derived from prostaglandin endoperoxides Platelets in tissue repair: Control of apoptosis and interactions with regenerative cells Human platelet storage organelles. A review Advances in platelet granule biology ADP receptor P2Y12 is expressed in vascular smooth muscle cells and stimulates contraction in human blood vessels. Arteriosclerosis Potentiation of ADP-induced aggregation in human platelet-rich plasma by 5-hydroxytryptamine and adrenaline Blockade of platelet alpha2B-adrenergic receptors: A novel antiaggregant mechanism Fresh evidence for platelets as neuronal and innate immune cells: Their role in the activation, differentiation, and deactivation of Th1, Th17, and Tregs during tissue inflammation In vitro studies of aggregation of non-mammalian thrombocytes Comprehensive analysis of lysine crotonylation in proteome of maintenance hemodialysis patients Twinfilin 2a regulates platelet reactivity and turnover in mice Imaging flow cytometry in the assessment of leukocyte-platelet aggregates Podoplanin and CLEC-2 drive cerebrovascular patterning and integrity during development Platelet mechanosensing of substrate stiffness during clot formation mediates adhesion, spreading, and activation Platelet activation by C1q results in the induction of alpha IIb/beta 3 integrins (GPIIb-IIIa) and the expression of P-selectin and procoagulant activity Endothelial leukocyte adhesion molecule 1: An inducible receptor for neutrophils related to complement regulatory proteins and lectins Role of platelet membrane in enhancement of tumor cell adhesion to endothelial cell extracellular matrix Platelet enhancement of tumor cell adhesion to subendothelial matrix: Role of platelet cytoskeleton and platelet membrane SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor Structural basis of receptor recognition by SARS-CoV-2 Abnormalities of peripheral blood system in patients with COVID-19 in Wenzhou, China Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation Structural basis for the recognition of SARS-CoV-2 by fulllength human ACE2 Endothelial cell infection and endotheliitis in COVID-19 Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in Covid-19 Immune response to SARS-CoV-2 and mechanisms of immunopathological changes in COVID-19 Pulmonary and systemic involvement of COVID-19 assessed by ultrasound-guided minimally invasive autopsy Endothelial cells orchestrate COVID-19 coagulopathy COVID-19 Autopsies Blood vessel injury may spur disease's fatal second phase The Human Cell Atlas bone marrow singlecell interactive web portal The bone marrow microenvironment at single-cell resolution Platelets in leucocyte recruitment and function Pathogen sensing, subsequent signalling, and signalosome in human platelets Platelets as immune cells: Bridging inflammation and cardiovascular disease Expression and functionality of Toll-like receptor 3 in the megakaryocytic lineage Platelets modulate innate immune response against human respiratory syncytial virus in vitro Platelet toll-like receptors in thromboinflammation T granules in human platelets function in TLR9 organization and signaling Platelet surface expression of stromal cellderived factor-1 receptors CXCR4 and CXCR7 is associated with clinical outcomes in patients with coronary artery disease Platelet-derived SDF-1 primes the pulmonary capillary vascular niche to drive lung alveolar regeneration SDF-1alpha induces differential trafficking of CXCR4-CXCR7 involving cyclophilin A, CXCR7 ubiquitination and promotes platelet survival Expression of stromal cell-derived factor-1 receptors CXCR4 and CXCR7 on circulating platelets of patients with acute coronary syndrome and association with left ventricular functional recovery Platelet-mediated modulation of adaptive immunity Endothelial cell-immune cell interaction in IBD Inflammatory contribution of platelets revisited: New players in the arena of inflammation Platelets: New bricks in the building of neutrophil extracellular traps Platelets and their interactions with other immune cells Nouvelle cuisine: Platelets served with inflammation. The Journal of Immunology The inflammatory role of platelets via their TLRs and Siglec receptors Role of chemokine receptors CXCR4 and CXCR7 for platelet function GPIbalpha-vWF rolling under shear stress shows differences between type 2B and 2M von Willebrand disease Direct observation of von Willebrand factor elongation and fiber formation on collagen during acute whole blood exposure to pathological flow. Arteriosclerosis Shear-dependent rolling on von Willebrand factor of mammalian cells expressing the platelet glycoprotein Ib-IX-V complex Platelets and the injured vessel wall-"rolling into action": Focus on glycoprotein Ib/V/IX and the platelet cytoskeleton Platelet glycoprotein Ibalpha forms catch bonds with human WT vWF but not with type 2B von Willebrand disease vWF The GPIIb/IIIa (integrin alphaIIbbeta3) odyssey: A technology-driven saga of a receptor with twists, turns, and even a bend Differences in alpha-beta transmembrane domain interactions among integrins enable diverging integrin signaling Interactions of platelet integrin alphaIIb and beta3 transmembrane domains in mammalian cell membranes and their role in integrin activation The beta3 integrin cytoplasmic tail: Protein scaffold and control freak Human cytomegalovirusplatelet interaction triggers toll-like receptor 2-dependent proinflammatory and proangiogenic responses Integrin alpha(v)beta3 (vitronectin receptor) is a candidate receptor for the virulent echovirus 9 strain Barty Adenoviral transduction efficiency of ovarian cancer cells can be limited by loss of integrin beta3 subunit expression and increased by reconstitution of integrin alphavbeta3 Human platelets and their capacity of binding viruses: Meaning and challenges Platelet Activating Factor (PAF) Receptor Deletion or Antagonism Attenuates Severe HSV-1 Meningoencephalitis Determinants of the specificity of rotavirus interactions with the alpha2beta1 integrin Platelet ITAM signaling Platelet receptors activated via mulitmerization: Glycoprotein VI, GPIb-IX-V, and CLEC-2 The future of glycoprotein VI as an antithrombotic target A critical role of platelet TGF-beta release in podoplanin-mediated tumour invasion and metastasis Tetraspanin family member CD9 inhibits Aggrus/podoplanin-induced platelet aggregation and suppresses pulmonary metastasis The physiological and pathophysiological roles of platelet CLEC-2 A novel Syk-dependent mechanism of platelet activation by the C-type lectin receptor CLEC-2 Involvement of the snake toxin receptor CLEC-2, in podoplanin-mediated platelet activation, by cancer cells Significance of podoplanin expression in cancerassociated fibroblasts: A comprehensive review CLEC-2 expression is maintained on activated platelets and on platelet microparticles Infection with human coronavirus NL63 enhances streptococcal adherence to epithelial cells CLEC2 and CLEC5A: Pathogenic host factors in acute viral infections Cloning of GMP-140, a granule membrane protein of platelets and endothelium: Sequence similarity to proteins involved in cell adhesion and inflammation A platelet alpha-granule membrane protein (GMP-140) is expressed on the plasma membrane after activation PSGL-1-dependent myeloid leukocyte activation The neutrophil selectin LECAM-1 presents carbohydrate ligands to the vascular selectins ELAM-1 and GMP-140 CD62 and endothelial cellleukocyte adhesion molecule 1 (ELAM-1) recognize the same carbohydrate ligand, sialyl-Lewis x The three members of the selectin receptor family recognize a common carbohydrate epitope, the sialyl Lewis(x) oligosaccharide Platelets and autoimmunity Platelet function in pre-eclampsia Platelets, inflammation and tissue regeneration Platelets interact with Coxsackieviruses B and have a critical role in the pathogenesis of virus-induced myocarditis GMP-140 binds to a glycoprotein receptor on human neutrophils: Evidence for a lectin-like interaction The role of platelet activation in tumor metastasis Cancer-related coagulopathy (Trousseau's syndrome): Review of the literature and experience of a single center of internal medicine Deadly allies: The fatal interplay between platelets and metastasizing cancer cells Contribution of platelets to tumour metastasis Platelets alter tumor cell attributes to propel metastasis: Programming in transit Assessing the risk and prognosis of thrombotic complications in cancer patients Selectin-carbohydrate interactions during inflammation and metastasis Haemostasis Double-stranded DNA induces a prothrombotic phenotype in the vascular endothelium Viral myocarditis and coagulopathy: Increased tissue factor expression and plasma thrombogenicity Inhibition of lipopolysaccharide-induced interferon regulatory factor 3 activation and protection from septic shock by hydroxystilbenes Procoagulant activity of endothelial cells after infection with respiratory viruses Exploring possible mechanisms for COVID-19 induced thrombocytopenia: Unanswered questions A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: A study of a family cluster Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past Thrombocytopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: A meta-analysis Rheumatologists' perspective on coronavirus disease 19 (COVID-19) and potential therapeutic targets Coronavirus disease 2019 (COVID-19) and pregnancy: What obstetricians need to know A review of salivary diagnostics and its potential implication in detection of Covid-19 Hematological findings and complications of COVID-19 Mechanism of thrombocytopenia in COVID-19 patients Thrombocytopenia and its association with mortality in patients with COVID-19 Zebrafish thrombocyte aggregation by whole blood aggregometry and flow cytometry Platelets exit venules by a transcellular pathway at sites of F-met peptide-induced acute inflammation in guinea pigs Factors which influence blood platelet migration The migration of human platelets in vitro Ion channels in the regulation of platelet migration Coagulopathy of Coronavirus Disease Coagulation abnormalities and thrombosis in patients with COVID-19 COVID-19 coagulopathy: an evolving story Clinical features of patients infected with 2019 novel coronavirus in Wuhan All Feet On Deck-The Role of Podiatry During the COVID-19 Pandemic: Preventing hospitalizations in an overburdened healthcare system, reducing amputation and death in people with diabetes Acute stroke management pathway during Coronavirus-19 pandemic Characteristics of ischaemic stroke associated with COVID-19 Rising evidence for neurological involvement in COVID-19 pandemic Collateral effect of Covid-19 on stroke evaluation in the United States COVID-19 and the cerebrocardiovascular systems: What do we know so far? Large-vessel stroke as a presenting feature of Covid-19 in the young Severe acute respiratory syndrome coronavirus 2 infection and ischemic stroke Acute ischemic stroke complicating common carotid artery thrombosis during a severe COVID-19 infection Intelligent classification of platelet aggregates by agonist type Eleven faces of coronavirus disease Prolonged SARS-CoV-2 RNA shedding: Not a rare phenomenon Antibody responses to SARS-CoV-2 in patients with COVID-19 Profile of IgG and IgM antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Antibody detection and dynamic characteristics in patients with COVID-19 Longitudinal change of SARS-Cov2 antibodies in patients with COVID-19 SARS-CoV-2 infections and serologic responses from a sample of U.S. Navy Service Members -USS Theodore Roosevelt A peptide-based magnetic chemiluminescence enzyme immunoassay for serological diagnosis of coronavirus disease 2019 (COVID-19) Performance of VivaDiag IgM/IgG rapid test is inadequate for diagnosis of COVID-19 in acute patients referring to emergency room department COVID-19: A meta-analysis of diagnostic test accuracy of commercial assays registered in Brazil SARS-Cov-2 infection: Response of human immune system and possible implications for the rapid test and treatment Rapid point-of-care testing for SARS-CoV-2 in a community screening setting shows low sensitivity Detection of IgM and IgG antibodies in patients with coronavirus disease 2019 Diagnostic accuracy of an automated chemiluminescent immunoassay for anti-SARS-CoV-2 IgM and IgG antibodies: an Italian experience Diagnostic value and dynamic variance of serum antibody in coronavirus disease 2019 Development and clinical application of a rapid IgM-IgG combined antibody test for SARS-CoV-2 infection diagnosis Assessment of immune response to SARS-CoV-2 with fully automated MAGLUMI 2019-nCoV IgG and IgM chemiluminescence immunoassays Evaluation of nucleocapsid and spike protein-based ELISAs for detecting antibodies against SARS-CoV-2 Analytical performances of a chemiluminescence immunoassay for SARS-CoV-2 IgM/IgG and antibody kinetics Clinical evaluation of a rapid colloidal gold immunochromatography assay for SARS-Cov-2 IgM/IgG Reliability and usefulness of a rapid IgM-IgG antibody test for the diagnosis of SARS-CoV-2 infection: A preliminary report Vitro Diagnostic Assays for COVID-19: Recent Advances and Emerging Trends. Diagnostics (Basel) A method to prevent SARS-CoV-2 IgM false positives in gold immunochromatography and enzyme-linked immunosorbent assays Characteristics of patients with coronavirus disease (COVID-19) confirmed using an IgM-IgG antibody test Evaluation of the auxiliary diagnostic value of antibody assays for the detection of novel coronavirus (SARS-CoV-2) Diagnostic performance of COVID-19 serology assays Detection of serum IgM and IgG for COVID-19 diagnosis Human platelet IgG Fc receptor FcgammaRIIA in immunity and thrombosis Platelet FcgammaRIIA binds and internalizes IgG-containing complexes Influenza virus H1N1 activates platelets through FcgammaRIIA signaling and thrombin generation The exposure of autoantigens by microparticles underlies the formation of potent inflammatory components: The microparticle-associated immune complexes Recognition of PF4-VWF complexes by heparin-induced thrombocytopenia antibodies contributes to thrombus propagation Coexistence of (partial) immune defects and risk of recurrent respiratory infections Clinical, virological and epidemiological characterization of dengue outbreak in Myanmar Risk factors in HIV-1-infected patients developing repetitive bacterial infections: Toxicological, clinical, specific antibody class responses, opsonophagocytosis and Fc(gamma) RIIa polymorphism characteristics High prevalence of dengue antibodies and the arginine variant of the FcgammaRIIa polymorphism in asymptomatic individuals in a population of Minas Gerais State Southeast Brazil An atypical IgM class platelet cold agglutinin induces GPVI-dependent aggregation of human platelets Specific detection of antibodies to different flaviviruses using a new immune complex ELISA Antibody responses to SARS-CoV-2 in patients of novel coronavirus disease 2019 Platelets in Coronavirus Disease Clinical characteristics of coronavirus disease 2019 in China The hemocyte counts as a potential biomarker for predicting disease progression in COVID-19: A retrospective study Longitudinal hematologic and immunologic variations associated with the progression of COVID-19 patients in China Hematologic, biochemical and immune biomarker abnormalities associated with severe illness and mortality in coronavirus disease 2019 (COVID-19): A metaanalysis COVID-19 and its implications for thrombosis and anticoagulation Clinical features and chest CT manifestations of coronavirus disease 2019 (COVID-19) in a single Clinical and laboratory findings of middle east respiratory syndrome coronavirus infection Middle East respiratory syndrome coronavirus in pediatrics: A report of seven cases from Saudi Arabia. Frontiers in Medicine Neutrophil extracellular traps in COVID-19 Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood Circulating plateletneutrophil complexes represent a subpopulation of activated neutrophils primed for adhesion, phagocytosis and intracellular killing Recent advances in platelet-polymorphonuclear leukocyte interaction Inflammation in fish as seen from a morphological point of view with special reference to the vascular compartment 2020 #11}Effective treatment of severe COVID-19 patients with tocilizumab Elevated Interleukin-6 and Severe COVID-19: A Meta-Analysis Detectable serum SARS-CoV-2 viral load (RNAaemia) is closely correlated with drastically elevated interleukin 6 (IL-6) level in critically ill COVID-19 patients COVID-19 and the cardiovascular system: Implications for risk assessment, diagnosis, and treatment options 2019 novel coronavirus disease (COVID-19) in hemodialysis patients: A report of two cases Can we use interleukin-6 (IL-6) blockade for coronavirus disease 2019 (COVID-19)-induced cytokine release syndrome (CRS)? A cluster of health care workers with COVID-19 pneumonia caused by SARS-CoV-2 Immune environment modulation in pneumonia patients caused by coronavirus: SARS-CoV, MERS-CoV and SARS-CoV-2 First case of COVID-19 complicated with fulminant myocarditis: A case report and insights Predictive factors for disease progression in hospitalized patients with coronavirus disease 2019 in Wuhan Clinical and autoimmune characteristics of severe and critical cases with COVID-19 How to reduce the likelihood of coronavirus-19 (CoV-19 or SARS-CoV-2) infection and lung inflammation mediated by IL-1 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 Hemostatic laboratory derangements in COVID-19 with a focus on platelet count Meta-analysis of the effect of aspirin on mortality in COVID-19 Intermediate-dose anticoagulation, aspirin, and in-hospital mortality in COVID-19: A propensity score-matched analysis Why we should not stop giving aspirin to pregnant women during the COVID-19 pandemic Cost-effectiveness of rivaroxaban plus aspirin (dual pathway inhibition) for prevention of ischaemic events in patients with cardiovascular disease: on top optimisation of secondary prevention medication in the context of COVID-19 pandemia Rivaroxaban and aspirin in peripheral vascular disease: A review of implementation strategies and management of common clinical scenarios COVID-19 pneumonia with hemoptysis: Acute segmental pulmonary emboli associated with novel coronavirus infection Difference of coagulation features between severe pneumonia induced by SARS-CoV2 and non-SARS-CoV2 Coronavirus blood-clot mystery intensifies SARS-CoV-2 and viral sepsis: Observations and hypotheses Platelet and endothelial activation as potential mechanisms behind the thrombotic complications of COVID-19 patients SARS-CoV-2 infection: The role of cytokines in COVID-19 disease Genetic gateways to COVID-19 infection: Implications for risk, severity, and outcomes COVID-19: Consider cytokine storm syndromes and immunosuppression Novel paediatric presentation of COVID-19 with ARDS and cytokine storm syndrome without respiratory symptoms SARS-CoV-2: A storm is raging Cytokine storm induced by SARS-CoV-2 Neutrophils and neutrophil extracellular traps drive necroinflammation in COVID-19 In the eye of the COVID-19 cytokine storm Prevention of the cytokine storm in COVID-19 Targeting potential drivers of COVID-19: Neutrophil extracellular traps Postmortem lung findings in an asthmatic patient with coronavirus disease 2019 Gross and histopathological pulmonary findings in a COVID-19 associated death during self-isolation Tissue plasminogen activator (tPA) treatment for COVID-19 associated acute respiratory distress syndrome (ARDS): A case series Autopsy report with clinical pathological correlation COVID-19 autopsy in people who died in community settings: The first series Thromboembolic involvement and its possible pathogenesis in COVID-19 mortality: Lesson from post-mortem reports Global absence and targeting of protective immune states in severe COVID-19 Platelets Can Associate With SARS-CoV-2 RNA and Are Hyperactivated in COVID-19 Inflammation resolution: A dual-pronged approach to averting cytokine storms in COVID-19 Platelet alpha-granules: Basic biology and clinical correlates SARS unique domain (SUD) of severe acute respiratory syndrome coronavirus induces NLRP3 inflammasome-dependent CXCL10-mediated pulmonary inflammation Tropism, replication competence, and innate immune responses of the coronavirus SARS-CoV-2 in human respiratory tract and conjunctiva: an analysis in ex-vivo and in-vitro cultures Focus on receptors for coronaviruses with special reference to angiotensinconverting enzyme 2 as a potential drug target -A perspective Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19 Harnessing adenosine A2A receptors as a strategy for suppressing the lung inflammation and thrombotic complications of COVID-19: Potential of pentoxifylline and dipyridamole SSRIs: Applications in inflammatory lung disease and implications for COVID-19 Preventing the development of severe COVID-19 by modifying immunothrombosis Understanding the andromeda strain -The role of cytokine release, coagulopathy and antithrombin III in SARS-CoV2 critical illness Coronavirus 2019, Microthromboses, and Platelet Activating Factor Platelet functions and activities as potential hematologic parameters related to Coronavirus Disease Practical guidance for the management of adults with immune thrombocytopenia during the COVID-19 pandemic Megakaryocytes: Masters of innate immunity? Arteriosclerosis, Thrombosis, and Vascular COVID-19-associated coagulopathy: An exploration of mechanisms Potential role for tissue factor in the pathogenesis of hypercoagulability associated with in COVID-19 Natural killer cells associated with SARS-CoV-2 viral RNA shedding, antibody response and mortality in COVID-19 patients Antibody-induced procoagulant platelets in severe COVID-19 infection A descriptive and quantitative immunohistochemical study demonstrating a spectrum of platelet recruitment patterns across pulmonary infections including COVID-19 Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome Publisher's note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations