Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI

Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI

Accepted Manuscript Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI Alan T. Nurden PII: ...

2MB Sizes 0 Downloads 33 Views

Accepted Manuscript Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI

Alan T. Nurden PII: DOI: Article Number: Reference:

S0268-960X(19)30069-4 https://doi.org/10.1016/j.blre.2019.100592 100592 YBLRE 100592

To appear in:

Blood Reviews

Please cite this article as: A.T. Nurden, Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI, Blood Reviews, https://doi.org/ 10.1016/j.blre.2019.100592

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Clinical Significance of Altered Collagen-Receptor Functioning in Platelets with Emphasis on Glycoprotein VI Alan T Nurden

NU

SC RI P

T

Institut de Rhythmologie et de Modélisation Cardiaque, Hôpital Xavier Arnozan, Pessac, France

MA

Correspondence: Alan T Nurden

ED

IHU-LIRYC, PTIB, Hôpital Xavier Arnozan, 33600 Pessac, France Tel: +33.5.56.55.19.52

PT

Fax: +33.5.57.10.28.64

AC

CE

E-mail: [email protected]

ACCEPTED MANUSCRIPT Abstract Much interest surrounds the receptors α2β1 and glycoprotein VI (GPVI) whose synchronized action mediates the attachment and activation of platelets on collagen, essential for preventing blood loss but also the most thrombogenic component of the vessel wall. Subject to density variations on platelets through natural polymorphisms, the absence of α2β1 or GPVI uniquely

T

leads to a substantial block of hemostasis without causing major bleeding. Specific to the

SC RI P

megakaryocyte lineage, GPVI and its signaling pathways are most promising targets for antithrombotic therapy. This review looks at the clinical consequences of the loss of collagen receptor function with emphasis on both the inherited and acquired loss of GPVI with brief

NU

mention of mouse models when necessary. A detailed survey of rare case reports of patients with inherited disease-causing variants of the GP6 gene is followed by an assessment of the

MA

causes and clinical consequences of acquired GPVI deficiency, a more frequent finding most often due to antibody-induced platelet GPVI shedding. Release of soluble GPVI is brought

ED

about by platelet metalloproteinases; a process induced by ligand or antibody binding to GPVI or even high shear forces. Also included is an assessment of the clinical importance of GPVI-

PT

mediated platelet interactions with fibrin and of the promise shown by the pharmacological

CE

inhibition of GPVI in a cardiovascular context. The role for GPVI in platelet function in inflammation and in the evolution and treatment of major illnesses such as rheumatoid arthritis,

AC

cancer and sepsis is also discussed.

Keywords: Glycoprotein VI; inherited defects; acquired defects, polymorphisms; soluble GPVI shedding; mouse models; inflammation; anti-thrombotic therapy.

ACCEPTED MANUSCRIPT 1. Introduction Platelets maintain blood circulation by preventing bleeding through hemostatic plug formation at sites of vessel injury and by preserving blood vessel integrity [1,2]. A source of active metabolites, cytokines and growth factors, platelets also promote angiogenesis and tissue repair [3]. Collagen is the main substrate for platelets in subendothelium and its exposure in

T

atherosclerotic plaques initiates occlusion in myocardial infarction and stroke [4-6]. Under

SC RI P

flow, platelets first interact through glycoprotein (GP) Ibα which binds to von Willebrand factor (VWF) multimers pre-adsorbed to collagen; the platelets then translocate along the collagen fibers favoring the interactions that permit firm adhesion [7,8]. Platelet interaction

NU

with collagen is a multistep process that principally involves two receptors, integrin α2β1 and the immunoglobulin superfamily member, GPVI that act in synergy in a density-dependent

MA

manner with GPVI principally governing the signaling responses [9-12]. GPVI (58-62 kDa) has a single chain and is co-expressed with the promiscuous Fc

ED

receptor chain (FcR). Formed by non-covalent transmembrane contact, this complex is exclusive to platelets and megakaryocytes (MKs). Three groups independently cloned the GP6

PT

gene some 20 years ago [13-15]. Located on the long arm of chromosome 19 (19q.13.42), GP6

CE

has 8 exons that encode 2 terminal disulfide- linked immunoglobulin (Ig)-homology loops (IgC1 and Ig-C2), a short glycosylated stalk, a transmembrane domain and a 51 amino acid (aa)

AC

cytoplasmic tail in the principally located splice form (Fig. 1). The protein has two extracellular N- and many O-glycosylation sites that are important for function [16,17]. The Ig domains contain distinct but overlapping binding sites for collagen, collagen-related peptide (CRP) and the snake venom protein convulxin (Cvx), two ligands used to mimic collagen activation of platelets in diagnostic and research [18]. The GPVI cytoplasmic tail includes binding sites for calmodulin and the tyrosine kinases Fyn/Lyn [19,20]. Ligand binding to GPVI activates Fyn or Lyn allowing them to mediate phosphorylation of the two FcR cytoplasmic tail immuno-

ACCEPTED MANUSCRIPT tyrosine based activation motifs (ITAMs). This favors the phosphorylation of Syk and the formation of a signaling complex containing adaptor proteins such as Linker of Activation of T cells (LAT) and SLP76 with the involvement of Src, phosphatidylinositol 3-kinases (PI3kinases), phospholipase C2 (PLC2) and Ca2+-mobilization; steps that initiate platelet spreading and aggregation as well as other functional responses (reviewed in [21]). ITAM-

T

mediated signaling is essential for collagen- induced activation of αIIbβ3 itself exhibiting

SC RI P

functional interplay with α2β1 [22-24]. At the same time negative regulation of GPVI signaling is mediated by G6b-B, PECAM and the protein tyrosine phosphatase, CD148 [25-27]. The relative role of monomeric or dimeric forms of GPVI in mediating the platelet

NU

response was the object of early debate [28-31]. Clustering and association with lipid rafts increases GPVI avidity for collagen and promotes downstream signaling. GPVI is promiscuous

MA

with laminin, extracellular matrix metalloproteinase inducer (EMMPRIN), fibronectin (Fn) and vitronectin (Vn) other potential ligands in the vessel wall [32-36] (Fig. 1). The hormone

ED

adiponectin and histones are plasma ligands [37,38]. Then the role of GPVI was transformed by the late discovery that GPVI also mediates platelet interactions with fibrin [39,40]. Binding of

PT

GPVI to fibrin also leads to platelet activation with phosphatidylserine (PS) exposure

CE

promoting more thrombin generation in a feedback loop. But controversy persists with regard to the relative roles of monomeric versus dimeric GPVI in mediating GP function. Thus, while

AC

Onselaer et al [41] showed that monomeric GPVI binds to the fibrin D-dimer domain and that soluble D-dimer blocked platelet activation by both fibrin and collagen, Ebrahim et al [42] using recombinant technology found that dimeric GPVI binds to collagen but not fibrin under static conditions and arterial flow. Then, Induruwa et al [43] confirmed that collagen-binding dimeric GPVI recognized soluble D-dimer, monomeric and polymerized fibrin and extended the results to Fg. Mangin et al [44] extended these findings by showing that binding of GPVI to immobilized fibrinogen (Fg) under flow is followed by a time-dependent formation of

ACCEPTED MANUSCRIPT lamellipodial sheets and stress fibers. In a recent review, Slater et al [45] show how technical issues related to the use of recombinant GPVI protein may account for some of the above discrepancies. In summary, GPVI is now recognized as a pluripotent receptor whose influence extends beyond the initial platelet-collagen interaction to thrombus build up and coagulation. Such observations suggest that it is a player not only in thrombosis but also in inflammatory

T

states including sepsis. GPVI deficiency therefore has implications far beyond that of a

SC RI P

potential bleeding diathesis. The object of this review is to assess the clinical effects of inherited and acquired deficiencies of GPVI in human disease with brief mention of mouse

NU

models and when appropriate of the α2β1 integrin, its companion receptor for collagen.

2. Natural polymorphis ms and variations in collagen receptor expression

MA

Early studies established that the platelet response to collagen was linked to the surface density of platelet collagen receptors. Historically, findings were first reported for α2β1,

ED

polymorphisms in the coding regions of the ITGA2 gene (5q23-31) being linked to a 5-fold variation in platelet α2β1 density in a control human population [46]. Three dimorphic allelic

PT

coding variants were highlighted: A1 (807T/1648G) associated with high, A2 (807C/1648G)

CE

and A3 (807C/1648A) with lower α2β1 densities. Importantly, the rate of attachment of platelets to collagen at high shear (1,500/sec) in a flow chamber correlated with the α2β1 copy

AC

number. The genetics of α2β1 expression were extended when further polymorphisms within the ITGA2 proximal 5’-regulatory regions and linked to decreased binding of the Sp1 transcription factor were also shown to influence α2β1density [47]. At this time, Furihata et al [48] in the Kunicki laboratory described a 5-fold range in GPVI expression in platelets of normal donors and linked Cvx or CRP-induced prothrombinase activity (and therefore thrombin generation) to GPVI density. A series of 10 SNPs (5 synonymous and 5 nonsynonymous affecting coding regions) were found in the GP6 gene [49,50]. Highlighted were 2

ACCEPTED MANUSCRIPT common haplotypes affecting 5 aa, 3 in the extracellular domain and two in the cytoplasmic tail of GPVI (“a” allele, SKTQH; “b” allele PEALN) with allelic frequencies of 0.85 and 0.13 respectively (Fig. 2). The “b” allele associated with lower GPVI density on platelets and reduced platelet functional responses to collagen or CRP [49,50]. However, recent studies on larger cohorts underscore a somewhat lower variability. For example, Best et al [51] showed

T

GPVI levels to vary 1.5 fold and α2β1 levels 3-fold among 102 healthy donors. Platelets of

SC RI P

donors with the “b” allele of GPVI representing 22% of the population had 10% lower surface expression of GPVI. Later, Watkins et al [52] generated a high resolution SNP map by sequencing the promoter, exons and splice sites of GP6 for 96 unrelated Caucasian individuals.

NU

The SKTQH and PEALN isoforms were confirmed as major alleles. Among 18 SNPs was a rare non-synonymous SNP (103Leu>Val) (MAF=0.005) giving reduced ligand binding. Their

MA

study population was then extended to 2000 individuals with the identification of 12 additional isoforms and significant changes in GP6 haplotype expression between people of different

ED

geographical and ethnic backgrounds.

The degree by which the above haplotypes controlled GPVI density and platelet

PT

function continued to be less striking in larger cohorts. Jones et al [53] studied 506 subjects

CE

identifying individuals who were hyper- or hypo-responders to CRP; sequence variations at the GP6 locus, primarily the SKTQH and PEALN haplotypes now accounted for only 35% of the

AC

variation. The Kunicki group turned to expression studies and showed that the presence of the cytoplasmic domain LN substitutions in the “b” allele significantly diminished signal transduction, an effect that over-rode ligand binding or GPVI copy number [54]. As discussed earlier (1. Introduction), the GPVI cytoplasmic domain binds calmodulin and the tyrosine kinases Fyn/Lyn. Trifiro et al [54] showed that L317 conferred an increased affinity for calmodulin while N322 attenuated binding to Fyn/Lyn and consequently Syk phosphorylation.

ACCEPTED MANUSCRIPT Do variations in GPVI expression and haplotype-related functional differences of platelets have clinical consequences? Kunicki et al [55] compared secondary platelet candidate gene polymorphisms with bleeding severity in pedigrees of 14 index cases with type I von Willebrand disease (VWD) where patients have low VWF levels. Increased bleeding scores were linked to ITGA2 (807C), ITGA2B (Ile843) and GP6 (PEALN) haplotypes but no

T

associations were found for 6 other candidate genes. When extended to an analysis of pedigrees

SC RI P

with VWD type 2A, 2B and 2M, of 5 selected platelet genes only an ITGA2 promoter haplotype (-52T) was associated with an increased bleeding severity and no correlations were seen for GP6 [56]. In fact, the 807C and -52T variants are in strong linkage disequilibrium and

NU

both associate with low α2β1density. Finally, Cheli et al [57] performed a genome-wide SNP scan using Gp6-/- mice, and revealed significant linkage of animals with a prolonged tail

MA

bleeding time to a locus on chromosome 4 designated as modifier of hemostasis 4 (Mh4). The authors concluded that one or more genes at this locus control the extent to which thrombus

ED

formation is disrupted in the absence of GPVI. No follow-up studies on Mh4 have been

PT

reported.

CE

3. Inherited defects of GPVI

The first report of a possible inherited deficiency of GPVI (OMIM:614201) came when Moroi

AC

et al [58] described a Japanese woman with lifelong mild mucocutaneous bleeding and platelets that failed to adhere, secrete or aggregate with type I and type III collagen despite a full response to ADP, epinephrine, arachidonic acid (AA), thrombin and ristocetin (Table 1). Significantly, clot retraction was normal and her bleeding time only slightly prolonged. Radiolabeling of surface GPs showed a deficit of GPVI but a normal presence of other membrane receptors. Her platelets failed to bind a human antibody to GPVI (see also Sugiyama et al. [59] and Section 6). Platelets of her non-consanguineous parents had 50% GPVI

ACCEPTED MANUSCRIPT expression and responded well to collagen. Arai et al [60] then reported a Japanese woman with a congenital mild bleeding syndrome whose severity decreased with age. Her bleeding time was only moderately prolonged. Nevertheless, extensive bleeding accompanied hysterectomy. Her platelet count was moderately reduced (Table 1). While her platelets were refractory with collagen they responded well to other agonists. Platelet adhesion to collagen under static

T

conditions was mildly reduced. Clot retraction was normal. Western blotting (WB) using

SC RI P

human antibody to GPVI (see Sugiyama et al. [59]) revealed about 10% residual GPVI in her platelets. Other membrane receptors including integrin α2β1 were normally present. Thus 10% residual GPVI was insufficient to support platelet activation by collagen. Enigmatic, was the

NU

report from Goto et al [61] describing how platelets from the above patients also showed less firm adhesion on vWF.

MA

The first reported GP6 gene mutations came from Dumont et al [62]. The patient was a young girl with mild bleeding since infancy, a PFA-100 closure time prolonged to 210 seconds

ED

for collagen/epinephrine and poorly responsive to collagen and CRP (Table 1). Noteworthy is that secretion in response to collagen and collagen- induced thrombin generation was also

PT

impaired. Flow cytometry showed GPVI at 540 copies per platelet versus 3076 for the controls;

CE

WB confirmed the incomplete deficiency with a smeared migration of the residual protein. FcR was normally present. DNA sequencing revealed compound heterozygozity for two

AC

predicted disease-causing GP6 mutations (Fig. 2): (i) a c.172C>T transition giving an Arg38Cys (R38C) mutation within exon 3 and affecting the first Ig loop of GPVI and (ii) a duplication of 5 nucleotides (c.356_360) in exon 4 resulting in a frameshift (fs), a premature stop 30 codons downstream, an absence of the corresponding mRNA probably indicated mRNA decay. Introduction of Arg38Cys into recombinant GPVI reproduced the abnormal migration as seen by WB and a marked loss of collagen binding. The smeared migration was hypothesized to result from mismatched disulfides caused by the extra Cys residue. The ability

ACCEPTED MANUSCRIPT of the patient’s platelets to form thrombi on collagen under flow was reduced by 43%. In a parallel report, Hermans et al [63] described a young woman with moderate lifelong bleeding marked by several episodes of post-traumatic and post-surgery bleeding. Reduced platelet function responses were selective to collagen and its mimetics. Although thrombus formation on collagen under flow was poor, more single platelets attached. Enigmatically, the PFA-100

T

closure time with the collagen/ADP membrane was shortened while that for the

SC RI P

collagen/epinephrine cartridge was unchanged. Her platelet count and platelet ultrastructure were normal. GPVI expression was much reduced with residual protein normally migrating on WB. DNA sequencing revealed two heterozygous GP6 mutations: (i) an out-of frame 16bp del

NU

predicting a stop codon after 56 aa and (ii) a missense Ser176Asn substitution within the second Ig domain of GPVI (Fig. 2). Ser176 is highly conserved between species and its

MA

substitution was predicted to disrupt H-bonding; the second short truncated variant was not expressed. Both parents were heterozygous carriers and asymptomatic; in fact the mother with

ED

GPVIAsn176 unexpectedly had platelets normally expressing GPVI. Studies in HEK293 cells confirmed a normal posttranslational processing of GPVIAsn176 suggesting a functional

PT

defect.

CE

My group in Bordeaux set up sequencing of the GP6 gene in response to a request from Dr. Diego Mezzano who was investigating unrelated patients (2 female and 2 male) belonging

AC

to 4 non-consanguineous Chilean families [64]. All propositi presented with lifelong mild mucocutaneous bleeding (Table 1). Platelets of each were specifically refractory to collagen or, when tested, CRP or Cvx. When measured, the bleeding time was moderately prolonged. Flow cytometry failed to locate GPVI on the platelets of each patient. Sequencing of GP6 revealed an adenine insertion in exon 6 (c.711_712insA) causing a frameshift and generating a stop codon at aa242 of the mature protein; this homozygous variant was common to all patients who all expressed the high density “a” allele for GP6 (Fig. 2). There was no family history of

ACCEPTED MANUSCRIPT bleeding and the families are from different rural regions of Chile and while a founder effect can be hypothesized there is currently no clear explanation. The mutation predicts the synthesis of a soluble form of GPVI truncated extracellular to the trans-membrane domain. Indeed, WB detected a limited presence of truncated protein (around 49 kDa) in the platelets of each patient but no full-length GPVI. Obligate heterozygotes were asymptomatic and aggregated well with

T

collagen. Intriguingly a sister of one of the patients was also homozygous for the mutation but

SC RI P

has never shown signs of bleeding despite platelets unresponsive to collagen and lacking surface GPVI. Later, Onselaer et al [41] reported two more patients and showed that platelet adhesion to fibrin was also reduced by 40%, with αIIbβ3 the obvious candidate as the second

NU

fibrin receptor. Those platelets that did attach showed markedly reduced spreading, only occasional platelets showed signs of activation. Then, Mangin et al [44] studying platelets from

MA

two of the patients confirmed reduced adhesion on immobilized Fg, the platelets failing to form lamellipodal sheets and stress fibres. Blocking αIIbβ3 of GPVI negative platelets resulted in a

ED

total absence of adhesion on Fg. They also confirmed that clot retraction was normal, presumably mediated by αIIbβ3.

PT

The results so far accumulated from inherited GPVI deficiency support the conclusion

CE

that the hemorrhagic tendency is mostly moderate, contrasting for example, with the more severe bleeding that accompanies GPIb-IX deficiency in Bernard-Soulier syndrome and αIIbβ3

AC

deficiency in Glanzmann thrombasthenia (GT) [65,66]. Notwithstanding, the number of patients is small and care is needed not to over-interpret the available data for more information is required – especially for situations such as childbirth and trauma-related bleeding including surgery where two of the above patients did bleed excessively (Table 1).

4. Mouse models for α2β1 and GPVI deficiency

ACCEPTED MANUSCRIPT Mouse models have proved essential in understating the dynamics of the platelet collagen interaction and the roles of α2β1 and GPVI. First, mice genetically depleted of β1 integrins were shown to retain a normal bleeding time despite delayed but substantial thrombus formation on collagen fibrils and a lack of platelet aggregation with soluble collagen [67]. As these mice also lack α5β1 and α6β1 receptors for Fn and laminin, efforts turned to generating

T

α2-deficient mice [68,69]. Höltkotter et al [68] showed that the mice developed normally with

SC RI P

no obvious fertility or other physiological defects and that their platelets showed the specific defects in the collagen response seen for β1 knockout. Again there was no bleeding phenotype and the tail bleeding time was unchanged. Chen et al [69] confirmed that α2β1 knockout mice

NU

were viable with normal wound healing but detected a multifaceted phenotype with defects extending to epithelial cells and branching morphogenesis. Significantly, a blocking

MA

monoclonal antibody (MoAb) to GPVI totally abrogated the adhesion of the α2β1-deficient platelets to collagen [68]. Mice whose platelets were immunologically depleted of GPVI

ED

(detailed in Section 5) also only had a mild disruption of primary hemostasis despite the platelets showing reduced thrombus formation on collagen under flow [70,71]. It was

PT

significant then when platelets from mice genetically engineered to lack GPVI or FcR (also

CE

leading to GPVI deficiency) showed a severe loss of platelet adhesion, spreading and thrombus formation on collagen but again showed little bleeding [72]. Interestingly, mouse platelets

AC

genetically modified to express 5-fold lower levels of GPVI showed comparable thrombus formation on a high-density collagen coated surface to controls in so doing raising questions about the clinical significance of human GP6 polymorphisms [51]. In summary, α2β1 and GPVI clearly act in synergy and have complementary roles.

5. Shedding of GPVI from mice and human platelets

ACCEPTED MANUSCRIPT Shedding of the bulk of the soluble extracellular domain of GPVI (sGPVI) from circulating platelets causes loss of the collagen- interacting domains thereby regulating platelet function. Recognizing and understanding the mechanisms behind GPVI shedding is key to understanding acquired GPVI pathology. a) Antibody and ligand-induced release of sGPVI. Pioneering studies from Bernhard Nieswandt

T

and his group showed that injecting rat MoAbs to GPVI (the JAQ series) into mice led not only

SC RI P

to a transient thrombocytopenia but also to a long-term (at least 10 days) immune-depletion of GPVI from platelets with thromboembolic protection [67,70,71,73,74]. Studies performed with human platelet-rich plasma showed that adding mouse MoAbs against GPVI not only resulted

NU

in sGPVI shedding, the majority also induced platelet aggregation [75]. Release of sGPVI also occurred with monovalent Fab fragments (showing that Fc-receptor interactions were not

MA

necessary) and also occurred with MoAbs against both ligand and non-ligand-binding domains of GPVI. Enigmatically, no loss of GPVI occurred when JAQ antibodies were incubated with

ED

mouse platelets in vitro. But in vivo in mice both antibody-induced GPVI shedding and the thrombocytopenia are rapid (maximum at 30 min with return of the platelet count to normal

PT

after 48h); it is interesting to ask whether within this short time scale the bleeding risk was

CE

more severe [71]. Intracellular signaling is required for GPVI loss; shedding was prevented when elements of FcR-mediated ITAM signaling were interfered with. For example, GPVI

AC

shedding did not occur in LAT-/- mice (LAT is part of the GPVI signalosome); instead JAQ1GPVI complexes were internalized by the platelets followed by a slow intracellular degradation. The LAT-/- platelets failed to respond to collagen, but increases in bleeding time were modest [76]. Boylan et al [77] introduced human platelets into the circulation of non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice; Fab fragments of a MoAb to human GPVI were then injected. Shedding of sGPVI occurred from the human but not the

ACCEPTED MANUSCRIPT mouse platelets. But dampened platelet responses to ADP and thrombin were also seen. In a more classic mouse model, others also noted that JAQ1-induced GPVI shedding was accompanied by a short transient decrease in the platelet response to thrombin [78]. Such a loss would also accentuate bleeding risk. Important but enigmatic is that not all MoAbs to GPVI cause in vivo GPVI shedding or depletion although they may block function [79,80]. Clearly

T

antibodies to GPVI in patients have to be tested individually. As GPVI also contributes to

SC RI P

platelet aggregate formation under flow by interacting with platelet-bound Fg or fibrin, thrombus formation is also affected by GPVI loss [43,44]. One study reports that newly formed fibrin promotes shedding of GPVI from human platelets independently of ITAM signaling and

NU

that this occurs in pathology, for example in patients with severe burns [81]. b) Mechanisms for GPVI loss. Shedding of sGPVI is by proteolysis through the action

MA

of metalloproteinases with the release of a large 55 kDa fragment leaving a short transmembrane and intracellular fragment attached to the platelet (Fig. 1). Most studies have

ED

identified “A Disintegrin and Metalloproteinase Domain-containing protein 10” (ADAM10) as the protease that cleaves GPVI although ADAM17 and other metalloproteinases may also

PT

intervene [74,82,83]. The process appears under intracellular control, agents blocking the

CE

interaction of calmodulin with the GPVI cytoplasmic tail stimulate GPVI cleavage. GPVI can also be removed from the platelet surface by intracellular clearing and both shedding and

AC

internalization are abrogated in mice with a point mutation in the FcR ITAM domain again showing a role for intracellular signaling [76,84]. Release of sGPVI also occurs during coagulation by a process highly dependent on FXa that was speculated to activate ADAM10 directly and, as explained above, through direct interaction with fibrin [81,85]. A key question is how ADAM10 or other proteases in platelets find and cleave ligandor antibody-bound GPVI so rapidly. Localization using fluorochrome- labeled antibodies showed that the principle ADAM10 pools of platelets are intracellular but are organized

ACCEPTED MANUSCRIPT independently of P-selectin and α-granules (see Fig. 4 of ref [86]). So presumably a specific surface translocation mechanism is necessary. While beyond the scope of this review, recent studies suggest that several mechanisms can lead to sGPVI release and metalloproteinase activation with a role for membrane tetraspanin family as mediators of GPVI lateral diffusion and/or carriers or activators of metalloproteinases [87,88].

T

c) Importance of shear. As well as ligand binding, transient exposure to mechanical

SC RI P

force can lead to metalloproteinase activation and sGPVI release [89]. This is very relevant as high shear occurs in atherosclerosed coronary vessels or after ischemic events, and indeed high levels of circulating sGPVI have been detected in pilot studies on patients with stable angina

NU

pectoris and stroke [89,90]. Similar situations occur in acquired von Willebrand disease in a cardiovascular context and with the use of ventricular assist devices and extracorporeal

MA

membrane oxygenation where loss of platelet receptors (GPIbα and GPVI) and loss of platelet

ED

function can be a cause of bleeding [91].

6. Pathology of α2β1 deficiency and less well-known collagen receptors on platelets

PT

Early reports described inherited defects of α2β1 (originally known as Very Late Antigen

CE

(VLA)-2 and then GPIa-IIa) in a mild to severe bleeding disorder linked to a much reduced platelet response to collagen [92,93]. The patients had excessive post-traumatic bleeding and

AC

for women, menorrhagia. Other reports described GPIa deficiency associated with a lack of thrombospondin or in myeloproliferative disorders [51,94,95]. The relatively severe bleeding experienced by the patients reported by Nieuwenhuis et al [92,93] contrasts to the absence of bleeding in mice with platelets genetically modified to lack α2β1 (see Section 4). The genetic defect in these families needs elucidating. Interestingly, Noris et al [96] described inherited α2β1deficiency but in the context of autosomal dominant thrombocytopenia and mild bleeding later shown to be associated with variants in the ANKRD26 gene [97]. Another purported

ACCEPTED MANUSCRIPT collagen receptor on platelets is CD36 (GPIV) [98]. However, patients genetically lacking CD36 respond normally to collagen and at best it has a minor role [99]. Another purported collagen receptor is leukocyte-associated immunoglobulin- like receptor (LAIR-1) but it has not been linked to a specific pathology [100]. Autoantibodies to α2β1 are a cause of ITP [101,102] but little is known of their potential blocking of the platelet-collagen response. One exception is

T

an inhibitory IgM antibody cloned and sequenced by Deckmyn et al [103]. A MoAb binding to

integrin in the signaling response to collagen [104].

SC RI P

αIIb also specifically blocks the platelet/collagen interaction underlining a potential role for this

NU

7. Acquired defects associated with antibodies to GPVI in human pathology Historically, Sugiyama et al [59] reported the first acquired defect of GPVI in a Japanese

MA

woman with ITP associated with Grave’s disease (Table 2). Thrombocytopenia was severe and associated with spontaneous mucocutaneous bleeding and the patient received platelet

ED

transfusions. Corticotherapy restored her platelet count to near normal but mild bleeding was still occasionally seen. Her platelets aggregated well to ADP, epinephrine, AA, thrombin and

PT

ristocetin but were refractory to collagen and this included dense granule secretion and

CE

thromboxane B2 synthesis. Although platelet adhesion to collagen fibrils was reduced, some platelets did attach. Significantly, both her plasma and isolated IgG induced the aggregation of

AC

control platelets, a response shown to depend on dense granule secretion. Her antibody primarily immunoprecipitated a membrane GP in the 50-60 kDa range from detergent solubilized control platelets later shown to be GPVI. Takahashi & Moroi [105] described a middle-aged Japanese woman with systemic lupus erythematosus (SLE), chronic thyroiditis and mild bleeding whose platelets showed similar characteristics. Her thrombocytopenia was initially severe but her platelet count normalized after corticosteroid therapy but again bleeding continued. Platelet-associated IgG was elevated and an antibody to GPVI reactive in WB

ACCEPTED MANUSCRIPT present in her serum; platelet GPVI was much reduced. Her antibody aggregated control platelets; cross-linking GPVI in the platelet membrane was a proposed mechanism. Significantly, F(ab’)2 fragments of the antibody of Sugiyama et al [59], now identified as an anti-GPVI, induced signaling pathways identical to those used by collagen and its mimetics Cvx and CRP, with tyrosine phosphorylation of Syk and Src [106]. Platelets from the Japanese

T

patients deficient in GPVI responded to collagen with a normal α2β1-dependent tyrosine

SC RI P

phosphorylation of Src but GPVI-dependent phosphorylation of Syk and activation of PLC2, Vav and FAK (focal adhesion kinase) was severely reduced [107].

Boylan et al [108] then reported a young woman in the USA with ITP and mild bleeding

NU

and moderate thrombocytopenia that reproduced the findings of the Japanese patients. Adherence and thrombus formation on collagen under flow were shown to be defective.

MA

Despite a much-reduced platelet expression of GPVI and FcRher platelets contained mRNA encoding GPVI and no mutations were found in the GP6 gene. The patient’s plasma possessed

ED

an IgG antibody to GPVI speculated to cause the rapid clearance of sGPVI from the plasma.

PT

An unusual case was an elderly woman from Bordeaux with gray platelet syndrome (GPS) and an antibody to GPVI [108]. GPS is an inherited platelet disorder caused by mutations in

CE

NBEAL2, a gene essential for the packaging of proteins during -granule synthesis in MKs [110]. GPS is associated with mild to severe mucocutaneous bleeding, macrothrombocytopenia

AC

an enlarged spleen and myelofibrosis that was strongly suspected although a confirmatory bone marrow biopsy was not performed [111]. Unlike typical GPS, platelet aggregation with collagen or Cvx was virtually absent and there was a severe reduction of GPVI in her platelets. Autoantibodies to GPVI were not detected either on her platelets or in her serum; sequencing GP6 revealed no potential disease-causing mutations (the patient had the high density “a” allele). It was speculated but not proven that endogenous metalloprote inase activity in the marrow was responsible for GPVI loss.

ACCEPTED MANUSCRIPT Studies continued with two more unrelated female Japanese patients with a mild bleeding diathesis, moderate thrombocytopenia and platelet function typical of acquired GPVI deficiency (cases 5 and 6, Table 2). For both, the platelet count gradually recovered but occasional mild bleeding continued; for the first platelets attached poorly to collagen under static conditions with spreading profoundly affected [112,113]. In both cases, plasma antibody

T

to GPVI was not detected but platelet-associated IgG was elevated and eluates contained

SC RI P

antibodies to GPVI as shown in a dot-blot assay using recombinant protein. Testing against transfected αIIbβ3-expressing heterologous cells also detected antibodies to αIIbβ3 in the second patient [113]. Significantly, as the patient’s platelet count increased, platelet expression

NU

of GPVI improved and the bleeding tendency diminished as antibody titers in platelet eluates decreased. Although GPVI shedding is a tempting hypothesis for these patients, sGPVI was not

MA

elevated and neither was the 10 kDa membrane-bound remnant of GPVI observed in WB. Not least, these cases show the importance of careful antibody testing.

ED

Gardiner et al [114] continued the reports of severe thrombocytopenia with ITP linked to anti-GPVI antibody. Corticotherapy again restored her platelet count but spontaneous

PT

pathological bruising continued. The collagen/epinephrine PFA-100 closure time was markedly

CE

prolonged. Here, platelet-associated IgG was elevated and plasma autoantibody to GPVI present. The antibody aggregated control platelets, an activity blocked by sGPVI and in part by

AC

a MoAb (IV.3) binding

. Decreased GPVI on her platelets and analyses for sGPVI

and of the residual transmembrane domain and cytoplasmic tail of GPVI strongly suggested shedding of GPVI, and this was confirmed when her plasma caused GPVI loss from control platelets. Interestingly, the authors later showed that 6-month treatment with the TPO receptor mimetic, romiplostim, restored both the platelet response to collagen and GPVI expression on her platelets but enigmatically, sGPVI in her plasma remained elevated and plasma autoantibody to GPVI continued to be detected [115]. How these results are explained is

ACCEPTED MANUSCRIPT unclear although previous studies have shown that TPO up-regulates GPVI expression in MKs by c-Mpl-mediated demethylation of a cytosine phosphate-guanosine-rich island within the GP6 promoter [116]. A young French woman with an ITP-like condition came to our attention in 2005 with severe thrombocytopenia (case 8, Table 2); corticotherapy restored her platelet count but within

T

a year a lupus nephropathy led to the diagnosis of SLE with antibody to DNA [117]. She

SC RI P

improved after treatment but later her condition again worsened and she received transfusions as a precaution prior to biopsies. In 2006, platelet function testing revealed a selective absence of platelet responses to collagen and Cvx and a specific platelet deficiency of GVI and FcR;

NU

her PFA-100 closure time (collagen + epinephrine) was markedly prolonged. Testing throughout subsequent treatment revealed a progressive restoration of the platelet responses to

MA

collagen and increases in platelet GPVI expression and her PFA-100 closure time normalized. Plasma samples analyzed for anti-GPVI antibodies employing a specially developed ELISA

ED

with recombinant sGPVI showed that antibody to GPVI was initially strong but that its titer fell as platelet GPVI levels improved. Plasma sGPVI remained low. DNA sequencing showed no

PT

potential disease-causing mutations and that the patient’s GP6 possessed the “a” allele

CE

associated with higher GPVI levels. The plasma antibody was of the IgG3 subtype and when titers were strong it activated control platelets with long incubations resulting in sGPVI release

AC

and a loss of the collagen response. Significantly, restoration of platelet GPVI levels and the collagen response was associated with loss of her nephrotic syndrome. An unusual case reported by Sanchez-Guiu et al [118] concerned an atypical plateletreacting IgM class cold agglutinin in a woman with a lifelong history of mild bleeding and chronic moderate thrombocytopenia. A unique diagnostic feature was pseudothrombocytopenia that was temperature-dependent but EDTA-independent. Her IgM bound GPVI and aggregated control platelets. Her own platelets had elevated bound IgM and showed signs of activation

ACCEPTED MANUSCRIPT (increased TXA2 production and surface P-selectin). Low GPVI expression and increased levels of sGPVI in her plasma was suggestive of shedding. The patient had prolonged closure times in the PFA-100 with both cartridges with platelet plugs forming more quickly when the test was performed immediately after the blood was taken (due to the spontaneous agglutination). Platelet function testing was abandoned for this reason. Platelet activation was

T

blocked by a GPVI-Fc fusion protein and by inhibitors of the tyrosine kinase Src family

SC RI P

and of Syk . Her IgM agglutinin has persisted over a 5-yr period. No abnormalities were seen on sequencing of the GP6 coding sequences. No other family member was affected. Finally in this Section, Rabbolini et al [119] described a middle-aged woman with a

NU

long-standing history of ITP and easy bruising. Severe thrombocytopenia improved with immunosuppressive therapy but a relapse required treatment with IVIgG given with antibiotics.

MA

Tests performed prior to hip surgery revealed a specifically defective collagen response and severe platelet deficiencies of GPVI. Next generation sequencing failed to reveal potential

ED

disease-causing mutations. Her serum aggregated control platelets; a response prevented by

PT

prior cleavage (and loss) of GPVI or by blocking the platelet FcRIIA receptor. An important feature as seen with other cases is that bleeding continued despite a platelet count well above

CE

the 30x109 /L threshold normally considered to merit special precautions in ITP. Quite remarkably, this section deals with 10 case reports describing patients with

AC

acquired GPVI deficiency due mainly to the presence of anti-GPVI antibodies - and all are women (Table 2). It will be interesting to see if new case reports continue this trend. This section also proves that anti-GPVI antibodies can give rise to ITP and I agree with Rabbolini et al [119] that they may well be an under-recognized cause. Also, the continued occurrence of bleeding in some patients after platelet count (and often GPVI expression) improves on treatment suggests that some antibodies may be function blocking although this has rarely been

ACCEPTED MANUSCRIPT proven. While antibodies clearly provoke sGPVI shedding for some internalization and intracellular processing of antibody-GPVI complexes is a possible alternative pathway.

8. GPVI and megakaryocytopoiesis In a companion paper in Blood Reviews [120] I have assessed the roles of autoantibodies to

T

αIIbβ3 in acquired GT. Some marked differences can be observed between antibodies to GPVI

SC RI P

and those directed against αIIbβ3. First, while autoantibodies to GPVI may induce receptor shedding those to αIIbβ3 do not. Secondly αIIbβ3 appears early in MK differentiation (being present in stem cells) and autoantibodies to the integrin can directly influence proplatelet

NU

production resulting in altered platelet biogenesis [discussed in 120]. In contrast, the expression of GPVI increases late in MK maturation and while the interaction of both GPVI and α2β1 with

MA

collagen combine to negatively regulate proplatelet production, this is part of a natural process to prevent premature platelet release in the bone marrow [121,122]. So far, inherited diseases of

ED

GPVI do not lead to platelet size increases contrasting also with acquired antibodies to GPIbIX-V and the inherited loss of GPIb-IX-V in BSS [65,123]. Still to be resolved, however, is the

PT

reason why platelet GPVI levels in mice return to normal much later than the platelet count

CE

after antibody-induced GPVI loss [71]. Does this have a parallel in human acquired GPVI loss?

AC

9. Bleeding syndromes associated with acquired defects in GPVI-dependent intracellular signaling pathways

Rare reports also describe patients for whom the molecular basis for loss of GPVI function is intra-cellualr. Bellucci et al [124] described defective collagen- induced platelet activation associated with bleeding in two patients with malignant hemopathies (cases 11 and 12, Table 2). A man with myelodysplasia, anemia and low white blood cell numbers suddenly developed a bleeding syndrome. His platelet count was normal but his bleeding time was very long. He

ACCEPTED MANUSCRIPT gradually developed severe aplasia and severe thrombocytopenia with an eventual transformation to myelomonoblastic leukemia from which he died. An elderly woman with eventually fatal chronic lymphocytic leukemia started bleeding after treatment with heparin for a pulmonary embolism. Her PFA-100 closure time was prolonged for collagen/epinephrine. Both patients had a severely impaired platelet response to collagen, CRP and Cvx whereas that

T

to other agonists including ADP, TRAP, AA and ristocetin was normal or only partially

SC RI P

decreased. GPVI was fully expressed in the platelets of both patients and bound Cvx yet neither Cvx nor specific activating MoAbs to GPVI induced secretion or Ca2+-mobilization. Perfusiondependent platelet adhesion and thrombus formation on collagen were both highly impaired.

NU

Inhibitory antibodies were not present in the plasma of the patients and a signaling defect proposed but not resolved. Best et al [51] also showed that as well as a low α2β1 content some

MA

patients with myeloproliferative syndromes also had decreased GPVI function. The case histories in this review conclude with a report that illustrates the difficulties of

ED

working up a suspected defect of GPVI [125]. A woman with a lifelong experience of mild bleeding gave birth to 2 children without excessive bleeding. There was no family history of

PT

bleeding and platelet counts were normal. When first tested, her platelets failed to aggregate

CE

with collagen; but the response later improved and while remaining delayed and low to Cvx was normal to all other agonists. Collagen- induced release from alpha-granules and dense-

AC

granules was variable. Anti-platelet antibodies were not detected in her serum or on her platelets and plasma levels of sGPVI matched those of controls. The PFA-100 closure time was not prolonged for either cartridge. Sequencing of the GP6 gene showed no disease-causing mutations. A rare heterozygous c.1038C>T variant in the 3’untranslated region had no effect on GPVI mRNA expression and GPVI was normally expressed in her platelets. The fact that collagen- induced platelet responses improved over time suggests an acquired defect (perhaps drug-induced) but the molecular cause remains an enigma.

ACCEPTED MANUSCRIPT

10. Platelets and GPVI in inflammation and other non-hemostatic events By adhering to foreign surfaces and by secreting metabolites and many biologically active proteins platelets promote not only tissue repair and wound healing but are vital for inflammation and innate immunity (reviewed in [3]). Clearly, as a collagen and fibrin receptor,

T

GPVI is an important player in these processes. In addition, the structurally related ITAM

SC RI P

receptor CLEC-2 acting through podoplanin expressed on vascular cells also exerts a major and multifaceted role in inflammation and is key for the development of the vascular and lymphatic systems [6,126,127]. While altered development of the lymphatic system is seen with CLEC-2

NU

deficiency [128]; ductus arteriosus is not a feature of GPVI deficiency in mice or man. During inflammation, platelets not only facilitate leukocyte infiltration at endothelial cell junctions

MA

through expression of P-selectin they also assure hemostasis by sealing breaks in the endothelium (reviewed in [129]). While deficiencies of GPVI or CLEC-2 in mice do not

ED

compromise wound healing; their double deficiency actually enhanced it probably by allowing more bleeding into wound sites by severely interfering with hemostasis in mice [130,131].

PT

How deficiencies of GPVI in man influence inflammation is an important area for future

CE

research.

Pioneering studies by Goerge et al [132], showed how thrombocytopenia promoted

AC

intra-alveolar bleeding and respiratory distress during endotoxin-induced lung inflammation. They also showed bleeding from venules during the cutaneous Arthus reaction where loss of vascular integrity is linked to immune complex deposition. Studies in mice showed that single platelets seal neutrophil- induced vascular breaches via GPVI during immune-complexmediated dermatitis [133]. The absence of GPVI (and a fall in platelet count) favored neutrophil- induced inflammatory bleeding. Another mouse model showed that platelet recruitment to the inflamed glomerulus also occurs in part by a GPVI-mediated mechanism

ACCEPTED MANUSCRIPT with P-selectin crucial [134]. Gros et al [133] speculated such interactions might involve EMMPRIN, a counter-receptor for GPVI expressed by leukocytes although other proteins potentially exposed in the vessel wall and able to bind GPVI include laminin, Fn and Vn (Fig. 1); fibrin is another possibility as are histones if DNA nets are present. A pathological role for GPVI and the platelet-collagen interaction in the release of biologically active microparticles

T

(MPs) from platelets was shown in a mouse model of rheumatoid arthritis suggesting GPVI as

SC RI P

a therapeutic target in this condition [135]. However, later studies by the same group showed that rather than MPs, lipid derivatives are the active platelet-released components [136,137]. Platelets are necessary for host defense during bacterial infection and limit bacterial

NU

growth and dissemination [138,139]. They also help prevent hemorrhage during sepsis and limit cytokine responses. A role for GPVI is illustrated by increased bacterial growth in the

MA

lungs and in distant body sites when GPVI-/- mice (or mice depleted of GPVI by antibodyinduced shedding) were infected with klebsiella pneumonia bacteria to induce pneumonia-

ED

induced sepsis [140]. Fewer platelets and platelet-leukocyte aggregates were observed in the bronchoalveolar space. GPVI on platelets reacting with its ligands collagen, fibrin and histones

PT

at sites of inflammation leads to up-regulated leukocyte function; in parallel changes in

CE

cytokine levels suggest that GPVI has an anti-inflammatory effect. The net effect will depend on the site and the nature of the inflammation. For example, in mouse models of cutaneous

AC

inflammation, GPVI has a pro-inflammatory effect. its absence leading to decreased platelet interplay with macrophages and increased anti-inflammatory markers and therefore a lower vasculo-protective effect [141]. On a wider scale, elevated sGPVI levels in many acute and inflammatory conditions including patients with severe burns, inflammatory bowel and kidney disease, elective cardiac surgery, trauma, acute brain injury or prolonged ventilation suggest ongoing GPVI loss and this was particularly so in sepsis [81]. For further information in this field I invite the reader to consult an excellent recent review by Boulaftali et al [142].

ACCEPTED MANUSCRIPT Clearly the role of GPVI is complex with GPVI loss favoring bleeding at some inflamed sites particularly when thrombocytopenia is also a factor. But much research is needed in this field and it is already apparent that the role of GPVI (and platelets) may vary considerably with respect to the site and cause of different clinical inflammatory states. The influence of administered therapeutics on bleeding (for example, anti-inflammatatory drugs) and to combat

SC RI P

T

thrombocytopenia (for example, in sepsis) must also be taken into account.

11. Thrombosis and the potential therapeutic use of anti-GPVI agents Platelet receptor polymorphisms emerged as risk factors for arterial thrombotic disease and this

NU

has included those influencing α2β1 and GPVI expression (see above Section 2). More importantly, GPVI has become a novel pharmacological target for drugs effective in thrombotic

MA

therapy but with preserved hemostatic risk (reviewed in [143]). a) Collagen receptor polymorphisms as thrombotic risk factors. Early studies suggested

ED

that variations in collagen receptor density helped determine risk in ischemic or cardiac disease and in thrombo-inflammatory conditions such as stroke (reviewed in [144]). For example, the

PT

dimorphic α2β1 C807/T873 allele associated with high α2β1 density was linked to myocardial

CE

infarction or stroke in younger patients, findings that were not confirmed in Japan suggesting race variations [145-147]. For GPVI, early studies suggested that high expression with

AC

polymorphic variation at the GP6 locus gave increased risk of coronary artery disease and myocardial infarction but the study groups were small [49,148-151]. Interestingly, a recent study associated the GPVI “a” allele with a two-fold higher level of platelet MPs release and with increased sepsis severity in a pediatric population [152]. Hopefully, the use of modern genomics will put such studies on a different level and enable a more personalized risk assessment than at present [153]. So far, diagnostic or treatment changing data has not been obtained.

ACCEPTED MANUSCRIPT b) Sticky platelet syndrome. The so-called “sticky platelet syndrome” refers to a loosely defined group of patients with increased thrombotic tendency usually defined as an increased sensitivity of platelets to aggregate with low doses of ADP and/or epinephrine (reviewed in [154]). The syndrome is claimed to have an autosomal dominant trait linked to variants in a wide range of genes. The Kubisz group [154] have linked polymorphisms in the GP6 gene to

T

its pathogenicity describing over-expression of GP6 haplotypes; however, much more stringent

SC RI P

evidence is required to prove the existence of sticky platelet syndrome as a distinct entity and to establish a molecular basis for the role of GPVI. One of the associated symptoms is fetal loss (and fetal growth retardation) and perhaps the most convincing finding is the association found

NU

by Skerenova et al. [155] between fetal loss and the presence of the low frequency GP6 “b” allele (PEAN). A reduced GPVI signaling was a suggested cause.

MA

c) Direct targeting of platelet GPVI in arterial thrombosis and stroke. The results obtained for genetically modified mice or mice with antibody-induced GPVI depletion showed

ED

that loss of GPVI occurred without major disruption of hemostasis (Sections 4 and 5). Most patients with inherited or acquired GPVI deficiency have mild bleeding while no thrombo-

PT

embolic events have been reported (Table 1). It is therefore not surprising that research into

CE

GPVI as a potential target for safe antithrombotic therapy including stroke has attracted much attention. Collagen fibrils have multiple partners during platelet adhesion and thrombus

AC

formation following atherosclerotic plaque rupture [156-158], and the continued functioning of other platelet membrane receptors including GPIbα, α2β1 and αIIbβ3 may limit bleeding. Diverging concepts for abrogating GPVI-dependent platelet signaling have emerged, three of which are now briefly mentioned. The first is the intravenous infusion of a recombinant soluble dimeric GPVI-Fc fusion protein (Revacept) blocking GPVI-reactive sites in collagen exposed in atherosclerotic plaques. Animal studies showed that Revacept both reduces thrombus formation and improves vascular dysfunction [159,160]. It is about to enter a phase 2 trial in

ACCEPTED MANUSCRIPT patients undergoing elective percutaneous coronary intervention [161]. A second approach is to therapeutically induce GPVI shedding [74]. This, however, remains largely unproven despite increases in our knowledge of how intracellular mechanisms can regulate sGPVI release, as for example through the production of reactive oxygen species [162]. Large problems remain however, for example, with regard to controlling the specificity, extent and duration of GPVI

T

shedding. The third and currently the most promising approach is by direct pharmacological

SC RI P

inhibition of GPVI using an antibody or with peptides derived from snake venom proteins [163-167]. Fab fragments of a MoAb to GPVI, ACT017, have been recently shown to have excellent safety and tolerability profiles in a phase 1 study in man while blocking collagen-

NU

induced platelet aggregation in a dose-dependent manner [168]. As well as arterial thrombosis and stroke, GPVI inhibitors have potential to prevent cardiac ischemia perfusion injury

MA

[158,169]. The results of phase 2 and possibly phase 3 clinical trials with these new drugs will be anxiously awaited. Whether patients with deficiencies of GPVI are protected against

ED

cardiovascular disease is an interesting question to answer in coming years as is the question of whether collagen receptor polymorphisms (see Section 2) will prove useful in predicting those

PT

patients most susceptible to benefit from these drugs.

CE

d) Soluble fibrin as a regulator of GPVI. Interestingly, difference in the way that GPVI interacts with collagen and fibrin or Fg provides the opportunity to developing inhibitors

AC

specific for each ligand [39-44]. Interestingly, soluble fibrin formed by restricted thrombin generation acts as a GPVI antagonist sterically blocking access to its ligands and the functional response [170]. Thrombin generation in response to trauma, cancer and disseminated intravascular coagulation (DIC) and plasmin generation may lead to circulating fibrin monomers or soluble protofibrils [171]. Soluble fibrin monomers can be maintained for several hours and render platelets non-responsive to collagen, a finding that would extend to donor platelets in the event of transfusion. According to Lee et al [170] 1% conversion of circulating

ACCEPTED MANUSCRIPT Fg would lead to 90 nM soluble fibrin – enough to overwhelm platelet GPVI and lead to loss of function of endogenous or transfused platelets. However, Onselaer et al [41] reached more conservative conclusions. Whether the soluble fibrin simply blocks GPVI or additionally desensitizes its signaling mechanism requires further research. The interaction between fibrin and/or Fg with GPVI explains early observations where mice deficient in GPVI failed to form

T

an occlusive thrombus in a ferric chloride injury model, even though the onset of thrombosis

SC RI P

was unaltered [172].

e) Deep vein thrombosis. An exciting new development is the concept that GPVI may mediate, at least in part, platelet involvement in deep vein thrombosis. Using a microfluidic

NU

model mimicking the low stasis conditions seen in human venous valves, Loyau et al [173] showed that tissue factor-dependent fibrin formation was followed by platelet accumulation

MA

that was blocked by a Fab fragment to GPVI or by D-dimer. Platelet accumulation occurred in a red cell-dependent manner and platelet activation with PS expression favored thrombin

ED

generation and thrombus growth. This opens the possibility of using anti-GPVI blockers in combination with tissue-type plasminogen activator in venous thrombosis. Whether patients

PT

lacking GPVI are protected against deep vein thrombosis is unknown.

CE

e) GPVI and cancer. Platelets and platelet-derived MPs exert important roles in cancer dissemination and metastasis and GPVI is one of the receptors involved [174,175]. This opens

AC

a particularly exciting field for anti-GPVI drugs, particularly as blocking GPVI in mouse models increases bleeding within newly formed tumors increasing the efficacy of chemotherapy [176].

12. Conclusions and future directions The data in this review mostly confirm the often-cited tendency of a low hemorrhagic tendency for patients with inherited deficiencies of GPVI and only moderately prolonged bleeding times.

ACCEPTED MANUSCRIPT However, the number of patients with disease-causing GP6 variants remains very low and there is already evidence for two subjects of excessive trauma-related bleeding (Table 1). Dominant are the families living in rural areas of Chile with a common 2bp insertion followed by a stop codon suggestive of a founder effect [64]. A much larger cohort of patients of wider geographical distribution is required before definite conclusions can be made on a modest

T

bleeding severity. Notwithstanding, the studies on GP6 knockout mouse models also point in

SC RI P

this direction. Acquired loss of GPVI function due to metalloproteinase- induced shedding is more common and again in most case reports results in little bleeding. However, acquired GPVI loss is often associated with thrombocytopenia that on occasion is severe; anti-GPVI

NU

antibodies clearly are a cause of ITP [119]. Thus platelet count is another possible factor influencing phenotype, although guidelines for ITP suggest that platelet counts >50 x 109 /L are

MA

unlikely alone to cause major bleeding [177]. However blocking GPVI function with antibodies acting as inhibitors would amplify loss of the GPVI/collagen interaction. Acquired GPVI

ED

deficiency may also be underestimated as testing for antibody against GPVI in ITP is not generalized while in some subjects their presence may be asymptomatic as indeed was the case

PT

for a Chilean subject with inherited GPVI deficiency (Table 1). In rare patients antibody to

CE

GPVI was only shown after elution from the patients’ platelets. While these may be of low titer but high affinity this has yet to be proven. GPVI shedding itself could result in rapid antibody

AC

clearance from plasma by immunoadsorption. Another explanation for negative testing is endocytosis of occupied GPVI with bound antibodies transported to intra-cellular sites. Striking is the high proportion of women featured in all case reports for GPVI deficiency and in acquired causes only women were identified. While this could suggest the presence of unrecognized sex-linked factors in controlling bleeding or antibody production, why should this be a unique feature of GPVI?

ACCEPTED MANUSCRIPT Perhaps the most important finding seen for GPVI in recent years is that it in part mediates platelet interaction with fibrin and receptor-bound Fg. Thus not only does GPVI help bring about platelet binding to the vessel wall, it also intervenes in thrombus build up. Historically, αIIbβ3 was thought to be the platelet receptor for fibrin; however, GT platelets that lack this integrin bind to fibrin polymers even though they are not activated as a result

T

(reviewed in [120]). Similarly, while GT platelets attach to collagen or exposed

SC RI P

subendothelium, they do not spread [178] and this is despite the normal presence of α2β1 and GPVI. Thus intracellular signaling in response to collagen requires receptor inter-play and is quite complex and it is interesting to note that GT platelets lacking αIIbβ3 fail to retract a clot

NU

whereas platelets deficient in GPVI retain clot retraction. Further research is needed here. The emergence of agents blocking GPVI or GPVI signaling for anti-thrombotic therapy

MA

is highly encouraging. Nonetheless, the complexity of the platelet/collagen interactions and of their signaling pathways will necessarily require the most careful evaluation of the situations

ED

where drugs to GPVI are used. Not to be forgotten are the possible consequences of their longterm use on platelet production in the marrow for GPVI has been shown to regulate platelet

PT

biogenesis by mediating the MK interaction with collagen [121,122]. High trauma situations

CE

including childbirth still need to be evaluated for these drugs, as does their use in combination with specialized medications in cardiovascular disease, inflammation and major illnesses such

AC

as sepsis and rheumatoid arthritis. Studies on their potential for wider use in cancer are highly promising but in their infancy and offer much scope for future use as an adjunct to chemotherapy. Finally, a major requirement is a rapid and more adept clinical test to assess the clinical risk of bleeding after GPVI loss or therapeutic blockade. Results obtained with the PFA-100 were highly variable for patients with inherited and acquired GPVI loss although the collagen/epinephrine cartridge was generally more sensitive, perhaps better showing the

ACCEPTED MANUSCRIPT importance of collagen-induced ADP release from platelets. Use of computer-derived analysis of platelet adhesion and thrombus formation under shear or under static conditions on collagen and other surfaces would be a great improvement. Potential procedures have been reviewed by

T

Brouns et al [179]

-

SC RI P

Practice points

Patients with inherited or acquired collagen or receptor function mostly have conserved hemostasis with little bleeding although trauma and some inflammatory

NU

states may represent an exception. The use of anti-GPVI drugs in antithrombotic therapy is promising but their long-term use has yet to be tested. The detection and characterization of anti-GPVI antibody is vital in acquired disorders

MA

-

and testing should take into account low-titre antibodies that may be in large part

-

ED

adsorbed onto the platelets.

Autoantibodies to GPVI may induce sGPVI release through metalloproteinase-

-

AC

Research agenda

CE

PT

induced shedding and testing for the soluble form is recommended.

Improvements in identifying autoantibodies to GPVI with rapid and easy to perform tests to identify both epitope targets and inhibitor activity. Large studies should investigate any possible sex linkage.

-

Studies to increase our knowledge of the active sites on GPVI whose loss leads to defective function, to better understand the role of dimerization and to design a test that better measures the effect of GPVI loss on bleeding risk.

ACCEPTED MANUSCRIPT -

The application of high-throughput screening to identify platelet, blood cell, plasma and vascular gene variants that will influence bleeding and will help choose the use of anti-GPVI drugs.

-

Inaugurating active international databases to clearly establish the frequency of GPVI deficiencies (inherited or acquired) and to evaluate the effectiveness of different

T

treatments blocking collagen receptor function in a cardiovascular disease or other

SC RI P

context.

Conflicts of Interest

MA

Acknowledgements and Addendum

NU

The author declares no conflict of interest.

The author thanks Paquita Nurden for her help and for her support during the writing of this

ED

review. After this manuscript was submitted, Martine Jandrot-Perrus and her colleagues

PT

published a short review on this subject that gives an alternative view of some of the data that I covered (Jandrot-Perrus M, Hermans C, Mezzano D. Platelet glycoprotein VI genetic and

AC

CE

qualitative defects. Platelets 2019;30:708-713).

ACCEPTED MANUSCRIPT References 1.

Gimbrone MA Jr, Aster RH, Cotran RS, Corkery J, Jandl JH, Folkman J. Preservation of vascular integrity in organs perfused in vitro with a platelet-rich medium. Nature 1969;222:33-36.

2.

Nachman RL, Rafii S. Platelets, petechiae, and preservation of the vessel wall. New

SC RI P

3.

T

Engl J Med 2008;359:1261-1270.

Nurden AT. The biology of the platelet with special reference to inflammation, wound healing and immunity. Front Biosci Landmark 2018;23:726-751. Ruggeri ZM. Platelets in atherothrombosis. Nat Med 2002;8:1227-1234.

5.

Nieswandt B, Pleines I, Bender M. Platelet adhesion and activation mechanisms in

NU

4.

arterial thrombosis and ischaemic stroke. J Thromb Haemost 2011; Suppl 1:92-104. Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune

MA

6.

receptors GPVI and CLEC-2. J Clin Invest 2019;129:12-33. Savage B, Saldivar E, Ruggeri ZM. Initiation of platelet adhesion by arrest onto

ED

7.

8.

PT

fibrinogen or translocation on von Willebrand factor. Cell 1996;84:289-297. Varga-Szabo D, Pleines I, Nieswandt B. Cell adhesion mechanisms in platelets.

9.

CE

Arterioscler Thromb Vasc Biol 2008;28:403-412. Chen H, Locke D, Liu Y, Liu C, Kahn ML. The platelet receptor GPVI mediates

AC

both adhesion and signaling responses to collagen in a receptor density-dependent fashion. J Biol Chem 2002;277:3011-3019. 10.

Kuijpers MJ, Schulte V, Bergmeier W, Lindhout T, Brakebusch C, Offermans S, Fässler R, Heemskerk JW, Nieswandt B. Complementary roles of glycoprotein VI and alpha2beta1 integrin in collagen- induced thrombus formation in flowing whole blood ex vivo. FASEB J 2003;17:685-687.

ACCEPTED MANUSCRIPT 11.

Chen H, Kahn ML. Reciprocal signaling by integrin and nonintegrin receptors during collagen activation of platelets. Mol Cell Biol 2003;23:4764-4777.

12.

Nieswandt B, Watson SP. Platelet-collagen interaction: Is GPVI the central receptor? Blood 2003;102:449-461.

13.

Clemetson JM, Polgar J, Magnenat E, Wells TN, Clemetson KJ. Platelet collagen

T

receptor glycoprotein VI is a member of the immunoglobulin superfamily closely

SC RI P

related to FcalphaR and the natural killer receptors. J Biol Chem 1999;274:2901929024. 14.

Jandrot-Perrus M, Busfield S, Lagrue AH, Xiong X, Debili N, Chickering T, Le

NU

Couedic JP, Dussault B, Fraser C, Vainchenker W, Villeval JL. Cloning, characterization and functional studies of human and mouse glycoprotein VI: a

MA

platelet specific collagen receptor from the immunoglobulin superfamily. Blood 2000;96:1798-1807.

Ezumi Y, Uchiyama T, Takayama H. Molecular cloning, genomic structure,

ED

15.

chromosomal localization, and alternative splice forms of the platelet collagen

Kunicki TJ, Cheli Y, Moroi M, Furihata K. The influence of N-linked glycosylation

CE

16.

PT

receptor glycoprotein VI. Biochem Biophys Res Commun 2000;277:27-36.

on the function of platelet glycoprotein VI. Blood 2005;106:2744-2749. Wang Y, Jobe SM, Ding X, Choo H, Mi R, Ju T, Cummings RD. Platelet biogenesis

AC

17.

and functions require correct protein O-glycosylation. Proc Natl Acad Sci USA 2012;109:16143-16148. 18.

Lecut C, Arocas V, Ulrichts H, Elbaz A, Villeval JL, Lacapère JJ, Deckmyn H, Jandrot-Perrus M, Identification of residues within human glycoprotein VI involved in the binding to collagen: evidence for the existence of distinct binding sites. J Biol Chem 2004;279:52293-52299.

ACCEPTED MANUSCRIPT 19.

Andrews RK, Suzuki-Inoue K, Shen Y, Tulasne D, Watson SP, Berndt MC. Interaction of calmodulin with the cytoplasmic domain of platelet glycoprotein VI. Blood 2002;99:4219-4221.

20.

Locke D, Liu C, Peng X, Chen H, Kahn ML. FcR-independent signaling by the platelet collagen receptor glycoprotein VI. J Biol Chem 2003;278:15441-15448.

T

Stalker TJ, Newman DK, Ma P, Wannemacher KM, Brass LF. Platelet signaling. Handb Exp Pharmacol 2012;210:59-85.

22.

SC RI P

21.

Watson SP, Auger JM, McCarty OJ, Pearce AC. GPVI and integrin alphaIIbbeta3 signaling in platelets. J Thromb Haemost 2005;3:1752-1762.

Van de Walle GR, Schoolmeester A, Iserby BF, Cosemans JM, Heemskerk JW,

NU

23.

Hoylaerts MF, Nurden A, Vanhoorelbeke K, Deckmyn H. Activation of

MA

alphaIIbbeta3 is a sufficient but also an imperative prerequisite for activation of alpha2beta1 on platelets. Blood 2007;109:595-602. Pugh N, Simpson AMC, Smethurst PA, de Groot PG, Raynal N, Farndale RW.

ED

24.

Synergism between platelet collagen receptors defined using receptor-specific

Mori J, Pearce AC, Spalton JC, Griegielska B, Eble JA, Tomlinson MG, Senis YA,

CE

25.

PT

collagen- mimetic peptide substrata in flowing blood. Blood 2010;115:5069-579.

Watson SP. G6b-B inhibits constitutive and agonist-induced signaling by

26.

AC

glycoprotein VI and CLEC-2. J Biol Chem 2008;283:35419-35427. Ming Z, Hu Y, Xiang J, Polewski P, Newman PJ, Newman DK. Lyn and PECAM-1 function as interdependent inhibitors of platelet aggregation. Blood 2011;117:39033906. 27.

Mori J, Wang YJ, Ellison S, Heising S, Neel BG, Tremblay ML, Watson SP, Senis YA. Dominant role of the protein-tyrosine phosphatase CD148 in regulating platelet

ACCEPTED MANUSCRIPT activation relative to tyrosine phosphatase-1B. Arterioscler Thromb Vasc Biol 2012;32:2956-2965. 28.

Arthur JF, Shen Y, Kahn ML, Berndt MC, Andrews RK, Gardiner EE. Ligand binding rapidly induces disulfide-dependent dimerization of glycoprotein VI on platelet plasma membrane. J Biol Chem 2007;282:30434-30441. Jung SM, Tsui K, Soejima K, Nakagaki T, Miura Y, Berndt MC, Gardiner EE,

T

29.

SC RI P

Howes JL, Pugh N, Bihan D, Watson SP, Farndale RW. Constitutive dimerization of glycoprotein VI (GVI) in resting platelets is essential for binding to collagen and activation in flowing blood. J Biol Chem 2012;287:3000-3013. Loyau S, Dumont B, Ollivier V, Boulaftali Y, Feldman L, Ajzenberg N, Jandrot-

NU

30.

Perrus M. Platelet glycoprotein VI dimerization, an active process inducing receptor

MA

competence, is an indicator of platelet reactivity. Arterioscler Thromb Vasc Biol 2012;32:778-785.

Poulter NS, Pollitt AY, Owen DM, Gardiner EE, Andrews RK, Shimzu H, Ishikawa

ED

31.

D, Bihan D, Farndale RW, Moroi M, Watson SP, Jung SM. Clustering of

PT

glycoprotein VI (GPVI) dimers upon adhesion to collagen as a mechanism to

32.

CE

regulate GPVI signaling in platelets. J Thromb Haemost 2017;15:549-564. Inoue O, Suzuki-Inoue K, McCarty OJT, Moroi M, Ruggeri ZM, Kunicki TJ, Ozaki

AC

Y, Watson SP. Laminin stimulates spreading of platelets through integrin α6β1dependent activation of GPVI. Blood 2006;107:1405-1412. 33.

Seizer P, Borst O, Langer HF, Büllmann A, Münch G, Herouy Y, Stellos K, Krämer B, Bigalke B, Büchele B, Bachem MG, Vestweber D, Simmet T, Gawaz M, May AE. EMMPRIN (CD147) is a novel receptor for platelet GPVI and mediates platelet rolling via GPVI-EMMPRIN interaction. Thromb Haemost 2009;101:682-686

ACCEPTED MANUSCRIPT 34.

Bültmann A, Li Z, Wagner S, Peluso M, Schönberger T, Weiss C, Konrad J, Stellos K, Massberg S, Nieswandt B, Gawaz M, Ungerer M, Münch G. Impact of glycoprotein VI and platelet adhesion on atherosclerosis – a possible role of fibronectin. J Mol Cell Cardiol 2010;49:532-542.

35.

Schulz C, von Brühl ML, Barocke V, Cullen P, Mayer K, Okrojek R, Steinhart A,

T

Ahmad Z, Kremmer E, Nieswandt B, Frampton J, Massberg S, Schmidt R.

SC RI P

EMMPRIN (CD147/basigin) mediates platelet-monocyte interactions in vivo and augments monocyte recruitment to the vascular wall. J Thromb Haemost 2011;9:1007-1019.

Schönberger T, Ziegler M, Borst O, Konrad J, Nieswandt B, Massberg S, Ochmann

NU

36.

C, Jürgens T, Seizer P, Langer H, Münch G, Ungerer M, Preissner KT, Elvers M,

MA

Gawaz M. The dimeric platelet collagen receptor GPVI-Fc reduces platelet adhesion to activated endothelium and preserves myocardial functio n after transient ischemia

37.

ED

in mice. Am J Physiol Cell Physiol 2012;303:C757-766. Riba R, Hughes CE, Graham A, Watson SP, Naseem KM. Globular adiponectin

PT

induces platelet activation through the collagen receptor GPVI-Fc receptor chain

38.

CE

complex. J Thromb Haemost 2008;6:1012-1020. Alshehri OM, Montague S, Watson S, Carter P, Sarker N, Manne BK, Miller JL,

AC

Herr AB, Pollitt AY, O’Callaghan CA, Kunapuli S, Arman M, Hughes CE, Watson SP. Activation of glycoprotein VI (GPVI) and C-type lectin- like receptor-2 (CLEC2) underlies platelet activation by diesel exhaust particles and other charged/hydrophobic ligands. Biochem J 2015;468:459-473. 39.

Mammadova-Bach E, Ollivier V, Loyau S, Schaff M, Dumont B, Favier R, Freyburger G, Latger-Cannard V, Nieswandt B, Gachet C, Mangin PH, Jandrot-

ACCEPTED MANUSCRIPT Perrus M. Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood 2015;126:683-691. 40.

Alshehri OM, Hughes CE, Montague S, Watson SK, Frampton J, Bender M, Watson SP. Fibrin activates GPVI in human and mouse platelets. Blood 2015;126:16011608. Onselaer M-B, Hardy AT, Wilson C, Sanchez X, Babar AK, Miller LLC, Watson

T

41.

SC RI P

CN, Watson SK, Bonna A, Philippou H, Herr AB, Mezzano D, Ariëns RAS, Watson SP. Fibrin and D-dimer bind to monomeric GPVI. Blood Adv 2017;1:1495-1504. 42.

Ebrahim M, Jamashi J, Adler K, Megens RTA, M’Bengue Y, Blanchet X, Uhland K,

NU

Ungerer M, Brandl R, Weber C, Elia N, Lorenz R, Münch G, Siess W. Dimeric glycoprotein VI binds to collagen but not to fibrin. Thromb Haemost 2018;118:351-

43.

MA

361.

Induruwa I, Moroi M, Bonna A, Malcor J-D, Howes J-M, Warburton EA, Farndale

ED

RW, Jung SM. Platelet collagen receptor glycoprotein VI-dimer recognizes fibrinogen and fibrin through their D-domains, contributing to platelet adhesion and

Mangin PH, Onselaer MB, Receveur N, Le lay N, Hardy AT, Wilson C, Sanchez X,

CE

44.

PT

activation through thrombus formation. J Thromb Haemost 2018;16:389-404.

Loyau S, Dupuis A, Babar AK, Miller JCL, Philippou H, Hughes CE, Herr AB,

AC

Ariëns RAS, Mezzano D, Jandrot-Perrus M, Gachet C, Watson SP. Immobilized fibrinogen activates human platelets through glycoprotein VI. Haematologica 2018;103:898-907. 45.

Slater A, Perrella G, Onselaer M-B, Martin EM, Gauer JS, Xu R-G, Heemskerk JWM, Ariens RAS, Watson SP. Does fibrin(ogen) bind to monomeric or dimeric GPVI, or not at all? Platelets 2019;30:281-289.

ACCEPTED MANUSCRIPT 46.

Kritzik M, Savage B, Nugent DJ, Santoso S, Ruggeri ZM, Kunicki TJ. Nucleotide polymorphisms in the α2 gene define multiple alleles which are associated with differences in platelet α2β1. Blood 1998;92:2382-2388.

47.

Jacquelin B, Rozenshteyn D, Kanaji S, Koziol JA, Nurden AT, Kunicki TJ.

gene. J Biol Chem 2001;276:23518-23524.

Furihata K, Clemetson KJ, Deguchi H, Kunicki TJ. Variation in human platelet

SC RI P

48.

T

Characterization of inherited differences in transcription of the human integrin α2

glycoprotein VI content modulates glycoprotein VI-specific prothrombinase activity. Arterioscler Thromb Vasc Biol 2001;21:1857-1863.

Croft SA, Samani NJ, Teare MD, Hampton KK, Steeds RP, Channer KS, Daley ME.

NU

49.

Novel platelet membrane glycoprotein VI dimorphism is a risk factor for myocardial

50.

MA

infarction. Circulation 2001;104:1459-1463.

Joutsi-Korhonen L, Smethurst PA, Rankin A, Gray E, Ijsseldijk M, Onley CM,

ED

Watkins NA, Williamson LM, Goodall AH, de Groot PG, Farndale RW, Ouwehand WH. The low- frequency allele of the platelet collagen signaling receptor

PT

glycoprotein VI is associated with reduced functional responses and expression.

51.

CE

Blood 2003;101:4372-4379. Best D, Senis YA, Jarvis GE, Eagleton HJ, Roberts DJ, Saito T, Jung SM, Moroi M,

AC

Harrison P, Green FR, Watson SP. GPVI levels in platelets: relationship to platelet function at high shear. Blood 2003;102:2811-2818. 52.

Watkins NA, O’Connor MN, Rankin A, Jennings N, Wilson E, Harmer IJ, Davies L, Smethurst PA, Dudbridge F, Farndale RW, Ouwehand WH. Definition of novel GP6 polymorphisms and major difference in haplotype frequencies between populations by a combination of in-depth exon re-sequencing and genotyping with tag single nucleotide polymorphisms. J Thromb Haemost 2006;4:1197-1205.

ACCEPTED MANUSCRIPT 53.

Jones CI, Garner SF, Angenent W, Bernard A, Berzuini C, Burns P, Farndale RW, Hogwood J, Rankin A, Stephens JC, Tom BD, Walton J, Dudbridge F, Ouwehand WH and Goodall AH on behalf of the Bloodomics Consortium. Mapping the platelet profile for functional genomic studies and demonstration of the effect size of the GP6 locus. J Thromb Haemost 2007;5:1756-1765. Trifiro E, Williams SA, Cheli Y, Furihata K, Pulcinelli FM, Nugent DJ, Kunicki TJ.

T

54.

SC RI P

The low frequency isoform of glycoprotein VIb attenuates ligand- mediated signal transduction but not receptor expression or ligand binding. Blood 2009;114:18931899.

Kunicki TJ, Federici AB, Salomon DR, Koziol JA, Head SR, Mondala TS, Chismar

NU

55.

JD, Baronciani L, Canciani T, Peake IR. An association of candidate gene haplotypes

2004;104:2359-2367.

Kunicki TJ, Baronciani L, Canciani MT, Gianniello F, Head SR, Mondala TS,

ED

56.

MA

and bleeding severity in von Willebrand disease (VWD) type I pedigrees. Blood

Salamon DR, Federici AB. An association of candidate gene haplotypes and bleeding

PT

severity in von Willebrand disease type 2A, 2B and 2M pedigrees. J Thromb

57.

CE

Haemost 2006;4:137-147. Cheli Y, Jensen D, Marchese P, Habart D, Wiltshire T, Cooke M, Fernandez JA,

AC

Ware J, Ruggeri ZM, Kunicki TJ. The modifier of hemostasis (Mh) locus on chromosome 4 controls in vivo hemostasis of GP6-/- mice. Blood 2008;111:12661273. 58.

Moroi M, Jung SM, Okuma M, Shinmyozu K. A patient with platelets deficient in glycoprotein VI that lack both collagen- induced aggregation and adhesion. J Clin Invest 1989;84:1440-1445.

ACCEPTED MANUSCRIPT 59.

Sugiyama T, Okuma M, Ushikubi F, Sensaki S, Kanaji K, Uchino H. A novel platelet aggregating factor found in a patient with defective collagen- induced platelet aggregation and autoimmune thrombocytopenia. Blood 1987;69:1712-1720.

60.

Arai M, Yamamoto N, Moroi M, Akamatsu N, Fukutake K, Tanoue K. Platelets with 10% of the normal amount of glycoprotein VI have an impaired response to collagen

Goto S, Tamura N, Handa S, Arai M, Kodama K, Takayama H. Involvement of

SC RI P

61.

T

that results in a mild bleeding tendency. Br J Haematol 1995;89:124-130.

glycoprotein VI in platelet thrombus formation on both collagen and von Willebrand factor surfaces under flow conditions. Circulation 2002;106:266-272. Dumont B, Lasne D, Rothschild C, Bouabdelli M, Ollivier V, Oudin C, Ajzenberg N,

NU

62.

Grandchamp B, Jandrot-Perrus M. Absence of collagen-induced platelet activation

63.

MA

caused by compound heterozygous GPVI mutations. Blood 2009;114:1900-1903. Hermans C, Wittevrongel C, Thys C, Smethurst PA, Van Geet C, Freson K. A

ED

compound heterozygous mutation in glycoprotein VI in a patient with a bleeding disorder. J Thromb Haemost 2009;7:1356-1363. Matus V, Valenzuela G, Saez CG, Hidalgo P, Lagos M, Aranda E, Panes O, Pereira

PT

64.

CE

J, Pillois X, Nurden AT, Mezzano D. An adenine insertion in exon 6 of human GP6 generates a truncated protein associated with a bleeding disorder in four Chilean

65.

AC

families. J Thromb Haemost 2013;11:1751-1759. Savoia A, Kunishima S, De Rocco D, Zieger B, Rand ML, Pujol-Moix N, Caliskan U, Tokgoz H, Pecci A, Noris P, Srivastava A, Ward C, Morel-Kopp MC, Alessi MC, Bellucci S, Beurrier P, de Maistre E, Favier R, Hézard N, Hurtaud-Roux MF, LatgerCannard V, Lavenu-Bombled C, Proulle V, Meunier S, Négrier C, Nurden A, Randrianaivo H, Fabris F, Platokouki H, Rosenberg N, HadjKacem B, Heller PG,

ACCEPTED MANUSCRIPT Karimi M, Balduini CL, Pastore A, Lanza F. Spectrum of the mutations in BernardSoulier syndrome. Hum Mutat 2014;35:1033-1045. 66.

Nurden AT, Fiore M, Nurden P, Pillois X. Glanzmann thrombasthenia: a review of ITGA2B and ITGB3 defects with emphasis on variants, phenotypic variability, and mouse models. Blood. 2011;118:5996-6005. Nieswandt B, Brakebusch C, Bergmeier W, Schulte V, Bouvard D, Mokhtari-Nejad

T

67.

SC RI P

R, Lindhout T, Heemskerk JWM, Zirngibl H, Fässler R. Glycoprotein VI but not α2β1 integrin is essential for platelet interaction with collagen. EMBO J 2001;9:2120-2130.

Höltkötter O, Nieswandt B, Smyth N, Müller W, Hafner M, Schulte V, Krieg T,

NU

68.

Eckes B. Integrin α2-deficient mice develop normally, are fertile, but display

MA

partially defective platelet interaction with collagen. J Biol Chem 2002;277:1078910794.

Chen J, Diacovo TG, Grenache DG, Santoro SA, Zutter MM. The α2 integrin

ED

69.

subunit-deficient mouse. A multifaceted phenotype including defects of branching

Nieswandt B, , Bergmeier W, Schulte V, Gessner JE, Zimgibl H. Expression and

CE

70.

PT

morphogenesis and hemostasis. Am J Pathol 2002;161:337-344.

function of the mouse collagen receptor glycoprotein VI is strictly dependent on its

71.

AC

association with the FcRgamma chain. J Biol Chem 2000;275:23998-24002. Nieswandt B, Schulte V, Bergmeier W, Mokhtari-Nejad R, Rackebrandt K, Cazenave JP, Ohlmann P, Gachet C, Zimgibl H. Long-term antithrombotic protection by in vivo depletion of platelet glycoprotein VI in mice. J Exp Med. 2001;193:459-469. 72.

Kato K, Kanaji T, Russell S, Kunicki TJ, Furihata K, Kanaji S, Marchese P, Reininger A, Ruggeri ZM, Ware J. The contribution of glycoprotein VI to stable

ACCEPTED MANUSCRIPT platelet adhesion and thrombus formation illustrated by targeted gene deletion. Blood 2003;102:1701-1707. 73.

Schulte V, Rabie T, Prostredna M, Aktas B, Grüner S, Nieswandt B. Targeting of the collagen-binding site on glycoprotein VI is not essential for in vivo depletion of the receptor. Blood 2003;101:3948-3952. Bender M, Hofmann S, Stegner D, Chalaris A, Bösl M, Braun A, Scheller J, Rose-

T

74.

SC RI P

John S, Nieswandt B. Differentially regulated GPVI ectodomain shedding by multiple platelet-expressed proteinases. Blood 2010;116:3347-3355. 75.

Al-Tamimi M, Mu F-T, Arthur JF, Shen Y, Moroi M, Berndt MC, Andrews RK,

NU

Gardiner EE. Antiglycoprotein VI monoclonal antibodies directly aggregate platelets independently of FcyRIIa and induce GPVI ectodomain shedding. Platelets

76.

MA

2009;20:75-82.

Rabie T, Varga-Szabo D, Bender M, Pozgaj R, Lanza F, Saito T, Watson SP,

ED

Nieswandt B. Diverging signaling events control the pathway of GPVI downregulation in vivo. Blood 2007;110:529-535. Boylan B, Berndt MC, Kahn ML, Newman PJ. Activation-independent, antibody-

PT

77.

CE

mediated removal of GPVI from circulating human platelets: development of a novel NOD/SCID mouse model to evaluate the in vivo effectiveness of anti-human platelet

78.

AC

agents. Blood 2006;108:908-914. Schulte V, Reusch HP, Pozgajova M, Varga-Szabo D, Gachet C, Nieswandt B. Twophase antithrombotic protection after anti-glycoprotein VI treatment in mice. Arterioscler Thromb Vasc Biol 2006;26:1640-1670. 79.

Li H, Lockyer S, Concepcion A, Gong X, Takizawa H, Guertin M, Matsumoto Y, Kambayashi J, Tandon NN, Liu Y. The Fab fragment of a novel anti-GPVI

ACCEPTED MANUSCRIPT monoclonal antibody, OM4, reduces in vivo thrombosis without bleeding risk in rats. Arterioscler Thromb Vasc Biol 2007;27:1199-1205. 80.

Ohlmann P, Hechler B, Ravanat C, Loyau S, Herrenschmidt N, Wanert F, JandrotPerrus M, Gachet C. Ex vivo inhibition of thrombus formation by an antiglycoprotein VI Fab fragment in non-human primates without modification of

Montague SJ, Delierneux C, Lecut C, Layios N, Dinsdale RJ, Lee CS-M, Poulter NS,

SC RI P

81.

T

glycoprotein VI expression. J Thromb Haemost 2008;6:1003-1011.

Andrews RK, Hampson P, Wearn CM, Maes N, Bishop J, Bamford A, Gardiner C, Lee WM, Iqbal T, Moiemen N, Watson SP, Oury C, Harrison P, Gardiner EE.

NU

Soluble GPVI is elevated in injured patients: shedding is mediated by fibrin activation of GPVI. Blood Adv 2018;2:240-251. Andrews RK, Karunakaran D, Gardiner EE, Berndt MC. Platelet receptor

MA

82.

proteolysis: a mechanism for downregulating platelet reactivity. Arterioscler Thromb

83.

ED

Vasc Biol 2007;27:1511-1520.

Gardiner EE, Karunakaran D, Shen Y, Arthur JF, Andrews RK, Berndt MC.

PT

Controlled shedding of platelet glycoprotein (GP)VI and GPIb-IX-V by ADAM

84.

CE

family metalloproteases. J Thromb Haemost 2007;5:1530-1537. Takayama H, Hosaka Y, Nakayama K, Shirakawa K, Naitoh K, Matsusue T,

AC

Shinozaki M, Honda M, Yatagai Y, Kawahara T, Hirose J, Yokohama T, Kurihara M, Furusako S. A novel antiplatelet antibody therapy that induces cAMP-dependent endocytosis of the GPVI/Fc receptor gamma-chain complex. J Clin Invest 2008;118:1785-1795. 85.

Al-Tamimi M, Grigoriadis G, Tran H, Paul E, Servadei P, Berndt MC, Gardiner EE, Andrews RK. Coagulation- induced shedding of platelet glycoprotein VI mediated by factor Xa. Blood 2011;117:3912-3920.

ACCEPTED MANUSCRIPT 86.

Villeneuve J, Block A, Le-Bousse-Kerdilès MC, Lepreux S, Nurden P, Ripoche J, Nurden AT. Tissue inhibitors of matrix metalloproteinases in platelets and megakaryocytes: a novel organization for these secreted proteins. Exp Hematol 2009;37:849-856.

87.

Facey A, Pinar I, Arthur J, Qiao J, Jing J, Mado B, Carberry J, Andrews RK,

88.

SC RI P

activated platelets. Biochemistry 2016;55:1187-1194.

T

Gardiner EE. A-Disintegrin-and Metalloprotease (ADAM) 10 activity on resting and

Noy PJ, Yang Y, Reyat JS, Matthews AL, Charlton AE, Furmstom J, Rogers DA, Rainger GE, Tomlinson MG. TSPANC8 and a Disinitigrin and Metalloprotease 10

NU

(ADAM10) interact via their extracellular regions: Evidence for distinct binding mechanisms for different TspanC8 Proteins. J Biol Chem 2016;291:3145-3157. Al-Tamimi M, Tan CW, Qiao J, Pennings GJ, Javadzadegan A, Yong AS, Arthur JF,

MA

89.

Davis AK, Jing J, Mu FT, Hamilton JR, Jackson SP, Ludwig A, Berndt MC, Ward

ED

CM, Kritharides L, Andrews RK, Gardiner EE. Pathologic shear triggers shedding of vascular receptors: a novel mechanism for down-regulation of platelet glycoprotein

Al-Tamimi M, Gardiner EE, Thom JY, Shen Y, Cooper MN, Hankey GJ, Berndt

CE

90.

PT

VI in stenosed coronary vessels. Blood 2012;119:4311-4320.

MC, Baker RI, Andrews RK. Soluble glycoprotein VI is raised in the plasma of

91.

AC

patients with acute ischemic stroke. Stroke 2011;2:498-500. Lukito P, Wong A, Jing J, Arthur JF, Marasco SF, Murphy DA, Bergin PJ, Shaw JA, Collecutt M, Andrews RK, Gardiner EE, Davis AK. Mechanical circulatory support is associated with loss of platelet receptors glycoprotein Ibalpha and glycoprotein VI. J Thromb Haemost 2016:14:2253-2260.

ACCEPTED MANUSCRIPT 92.

Nieuwenhuis HK, Akkerman JW, Houdijk WP, Sixma JJ. Human platelets showing no response to collagen fail to express surface glycoprotein Ia. Nature 1985;318:470472.

93.

Nieuwenhuis HK, Sakariassen KS, Houdijk WP, Nievelstein PF, Sixma JJ. Deficiency of platelet membrane glycoprotein Ia associated with a decreased platelet

Kehrel B, Balleisen L, Kokott R, Mesters R, Stenzinger W, Clemetson KJ, van de

SC RI P

94.

T

adhesion to subendothelium: a defect in platelet spreading. Blood 1986;68:692-695.

Loo J. Deficiency of intact thrombospondin and membrane glycoprotein Ia in platelets with defective collagen-induced aggregation and spontaneous loss of

95.

NU

disorder. Blood 1988;71:1074-1078.

Handa M, Watanabe K, Kawai Y, Kamata T, Koyama T, Nagai H, Ikeda Y. Platelet

MA

unresponsiveness to collagen: involvement of glycoprotein Ia-IIa (alpha2beta1 integrin) deficiency associated with a myeloproliferative disorder. Thromb Haemost

96.

ED

1995;73:521-528.

Noris P, Guidetti GF, Conti V, Ceresa IF, Di Pumpo M, Pecci A, Torti M, Savoia M,

PT

Balduini CL. Autosomal dominant thrombocytopenias with reduced expression of

Noris P, Perrotta S, Seri M, Pecci A, Gnan C, Loffredo G, Pujol-Moix N, Zecca M, Scognamiglio F, De Rocco D, Punzo F, Melazzini F, Scianguetta S, Casale M,

AC

97.

CE

glycoprotein Ia. Thromb Haemost 2006;95:483-489.

Marconi C, Pippucci T, Amendola G, Notarengela LD, Kiersey C, Civaschi E, Balduini CL, Savoia A. Mutations in ANKRD26 are responsible for a frequent form of inherited thrombocytopenia: analysis of 78 patients from 21 families. Blood 2011;117: 6673-6680.

ACCEPTED MANUSCRIPT 98.

Greenwalt DE, Lipsky RH, Ockenhouse CF, Ikeda H, Tandon NN, Jamieson GA. Mmebrane glycoprotein CD36: A review of its roles in adherence, signal transduction, and transfusion medicine. Blood 1992;80:1105-1115.

99.

Daniel JL, Dangelmaier C, Strouse R, Smith JB. Collagen induces normal signal transduction in platelets deficient in CD36 (platelet glycoprotein IV). Thromb

T

Haemost 1994;71:353-356.

SC RI P

100. Clemetson KJ, Clemetson JM. Platelet collagen receptors. Thromb Haemost 2001;86:189-197.

101. Deckmyn H, Chew SL, Vermylen J. Lack of platelet response to collagen associated

NU

with an autoantibody against glycoprotein Ia: a novel cause of acquired qualitative platelet dysfunction. Thromb Haemost 1990;64:74-79.

MA

102. Beer JH, Rabaglio M, Berchtold P, von Felten A, Clemetson KJ, Tsakiris DA, Kehrel B, Brandenberger S. Autoantibodies against the platelet glycoproteins (GP) IIb/IIIa,

ED

Ia/IIa and IV and partial deficiency of GPIV in a patient with a bleeding disorder and a platelet collagen interaction. Blood 1993;82:820-829.

PT

103. Deckmyn H, Zhang J, Van Houtte E, Vermylen J. Production and nucleotide

CE

sequence of an inhibitory human IgM autoantibody directed against glycoprotein Ia/IIa. Blood 1994;84:1968-1974.

AC

104. Shadle PJ, Ginsberg MH, Plow EF, Barondes SH. Platelet-collagen adhesion: inhibition by a monoclonal antibody that binds glycoprotein IIb. J Cell Biol 1984;99: 2056-260. 105. Takahashi H, Moroi M. Antibody against platelet membrane glycoprotein VI in a patient with systemic lupus erythematosus. Am J Hematol 2001;67:262-267.

ACCEPTED MANUSCRIPT 106. Ichinohe T, Takayama H, Ezumi Y, Yanagi S, Yamamura H, Okuma M. Cyclic AMP-insensitive activation of c-Src and Syk protein-tyrosine kinase through platelet membrane glycoprotein VI. J Biol Chem 1995;270:28029-28036. 107. Ichinohe T, Takayama H, Ezumi Y, Arai M, Yamamoto M, Takahashi H, Okuma M. Collagen-stimulated activation of Syk but not c-Src is severely compromised in

T

human platelets lacking membrane glycoprotein VI. J Biol Chem 1997;272:63-68.

SC RI P

108. Boylan B, Chen H, Rathore V, Paddock C, Salacz M, Friedman KD, Curtis BR, Stapleton M, Newman DK, Kahn ML, Newman PJ. Anti-GPVI-associated ITP: an acquired platelet disorder caused by autoantibody- mediated clearance of the

NU

GPVI/FcR-chain complex from the human platelet surface. Blood 2004;104:13501355.

MA

109. Nurden P, Jandrot-Perrus M, Combrié R, Winkler J, Arocas V, Lecut C, Pasquet JM, Kunicki TJ, Nurden AT. Severe deficiency of GPVI in a patient with gray platelet

ED

syndrome. Blood 2004;104:107-114.

110. Lo RW, Li L, Leung R, Pluthero FG, Kahr WHA. NBEAL2 (neurobeachin- like 2) is

PT

required for retention of cargo proteins by -granules during their production by

CE

megakaryocytes. Arterioscler Thromb Vasc Biol 2018;38:2435-2447. 111. Nurden AT, Nurden P. The gray platelet syndrome: Clinical spectrum of the disease.

AC

Blood Rev 2007;21:21-36. 112. Kojima H, Moroi M, Jung SM, Goto S, Tamura N, Kozuma Y, Suzukawa K, Nagasawa T. Characterization of a patient with glycoprotein (GP) VI autoantibody nor genetic aberration. J Thromb Haemost 2006; 4:2433-2442. 113. Akiyama M, Kashiwagi H, Todo K, Moroi M, Berndt MC, Kojima H, Kanakura Y, Tomiyama Y. Presence of platelet-associated anti-glycoprotein (GP)VI

ACCEPTED MANUSCRIPT autoantibodies and restoration of GPVI expression in patients with GPVI deficiency. J Thromb Haemost 2009;7:1373-1383. 114. Gardiner EE, Al-Tamimi M, Mu FT, Karunakaran D, Thom JY, Moroi M, Andrews RK, Berndt MC, Baker RI. Compromised ITAM-based platelet receptor function in a patient with immune thrombocytopenic purpura. J Thromb Haemost 2008;6:1175-

T

1182.

SC RI P

115. Gardiner EE, Thom JY, Al-Tamimi M, Hughes A, Berndt MC, Andrews RK, Baker RI. Restored platelet function after romiplostim treatment in a patient with immune thrombocytopenic purpura. Br J Haematol 2010;148:625-628.

NU

116. Kanaji S, Kanaji T, Jacquelin B, Chang M, Nugent DJ, Komatsu N, Moroi M, Izuhara K, Kunicki TJ. Thrombopoietin initiates demethylation-based transcription

MA

of GP6 during megakaryocyte differentiation. Blood 2005;105:3888-92. 117. Nurden P, Tandon N, Takizawa H, Couzi L, Morel D, Fiore M, Pillois X, Loyau S,

ED

Jandrot-Perrus M, Nurden AT. An acquired inhibitor to the GPVI platelet collagen receptor in a patient with lupus nephritis. Res Pract Thromb Haemost 2009;7:1541-

PT

1549.

CE

118. Sanchez Guiu IM, Martinez-Martinez I, Martinez C, Navarro-Fernandez J, GarciaCandel F, Ferrer-Marin F, Vicente V, Watson SP, Andrews RK, Gardiner EE,

AC

Lozano ML, Rivera J. An atypical IgM class platelet cold agglutinin induces GPVIdependent aggregation of human platelets. Thromb Haemost 2015;114:313-324. 119. Rabbolini DJ, Gardiner EE, Morel-Kopp MC, Dunkely S, Jahangiri A, Lee CS-M, Stevenson WS, Ward CM. Anti-glycoprotein VI mediated immune thrombocytopenia: An under-recognized and significant entity. Res Pract Thromb Haemost 2017;1:291-295.

ACCEPTED MANUSCRIPT 120. Nurden AT. Acquired Glanzmann thrombasthenia: From antibodies to antiplatelet drugs. Blood Rev, 2019;36:10-22. 121. Sabri S, Jandrot-Perrus M, Bertolino J, Farndale RW, Mas VM, Debili N, Vainchenker W. Differential regulation of actin stress fiber assembly and proplatelet formation by alpha2beta1 integrin and GPVI in human megakaryocytes. Blood 2004;

T

104:3117-3125.

SC RI P

122. Semeniak D, Kulawig R, Stegner D, Meyer I, Schwiebert S, Bosing H, Eckes B, Nieswandt B, Schulze H. Proplatelet formation is inhibited by collagen type I through Syk-dependent collagen signaling. J Cell Sci 2016:129:3473-3484.

NU

123. Nurden P, Nurden AT. Is the mysterious platelet receptor GPV an unsuspected major target for platelet autoantibodies. Haematologica 2019; 104:1103-1105.

MA

124. Bellucci S, Huisse MG, Boval B, Hainaud P, Robert A, Fauvel-Lafève F, JandrotPerrus M. Defective collagen-induced platelet activation in two patients with

ED

malignant haemopathies is related to a defect in the GPVI-coupled signalling pathway. Thromb Haemost 2005;93:130-138.

PT

125. Klümpers V, Müller I, Hellstern P, Schulze TJ, Mannhalter C, Bugert P, Eichler H.

CE

Characterization and diagnostic work-up of a patient with functionally impaired platelet GP6. Hämostaseologie 2018;38:112-120.

AC

126. Watson SP, Herbert JM, Pollitt AY. GPVI and CLEC-2 in hemostasis and vascular integrity. J Thromb Haemost 2010;8:1456-1467. 127. Boulaftali Y, Hess PR, Getz TM, Cholka A, Stolla M, Mackman N, Owens AP 3 rd, Ware J, Kahn ML, Bergmeier W. Platelet ITAM signaling is critical for vascular integrity in inflammation. J Clin Invest 2013;123:908-916. 128. Suzuki-Inoue K, Inoue O, Ding G, Nishimura S, Hokamura K, Eto K, Kashiwagi H, Tomiyama Y, Yatomi Y, Umemura K, Shin Y, Hirashima M, Ozaki Y. Essential in

ACCEPTED MANUSCRIPT vivo roles of the C-type lectin receptor CLEC-2: embryonic/neonatal lethality of CLEC-2 deficient mice by blood/lymphatic misconnections and impaired thrombus formation of CLEC-2-deficient platelets. J Biol Chem 2010;285:24494-29507. 129. Ho-Tin-Noé B. The multifaceted roles of platelets in inflammation and vascular immunity. Platelets 2018; 29:531-532.

T

130. Bender M, May F, Lorenz V, Thielmann I, Hagedorn I, Finney BA, Vögle T, Kremer

SC RI P

K, Braun A, Bösl M, Watson SP, Nieswandt B. Combined in vivo depletion of glycoprotein VI and C-type lectin- like receptor 2 severely compromises hemostasis and abrogates arterial thrombosis in mice. Arterioscler Thromb Vasc Biol

NU

2013;33:926-933.

131. Wichaiyo S, Lax S, Montague SJ, Li Z, Grygielska B, Pike JA, Haining EJ, Brill A,

MA

Watson SP, Rayes J. Platelet glycoprotein VI and C-type lectin-like receptor 2 deficiency accelerates wound healing by impairing vascular integrity in mice.

ED

Haematologica 2019 Feb 7. pli: haematol 2018.208363. doi: 10.3324/haematol.2018.208363. (Epub ahead of print).

PT

132. Goerge T, Ho-Tin-Noé B, Carbo C, Benarafa C, Remold-O’Donnell E, Zhao BQ,

CE

Cifuni SM, Wagner DD. Inflammation induces hemorrhage in thrombocytopenia. Blood 2008;111:4958-4964.

AC

133. Gros A, Syvannarath V, Lamrani L, Ollivier V, Loyau S, Goerge T, Nieswandt B, Jandrot-Perrus M, Ho-Tin-Noé B. Single platelets seal neutrophil- induced vascular breaches via GPVI during immune-complex-mediated inflammation in mice. Blood 2015;126:1017-1026. 134. Devi S, Kuligowski MP, Kwan RY, Westein E, Jackson SP, Kitching AR, Hickey MJ. Platelet recruitment to the inflamed glomerulus occurs via an alphaIIbbeta3/GPVI-dependent pathway. Am J Pathol 2010;177:1131-1142.

ACCEPTED MANUSCRIPT 135. Boilard E, Nigrovic PA, Larabee K, Watts GF, Coblyn JS, Weinblatt ME, Massarotti EM, Remold-O’Donnell E, Farndale RW, Ware J, Lee DM. Platelets amplify inflammation in arthritis via collagen-dependent microparticle production. Science 2010;237:580-583. 136. Boilard E, Larabee K, Shnayder R, Jacobs K, Farndale RW, Ware J, Lee DM.

T

Platelets participate in synovitis via Cox-1-dependent synthesis of prostacyclin

SC RI P

independently of microparticle generation. J. Immunol 2011;186:4361-4366. 137. Duchez AC, Boudreau LH, Naika GS, Rousseau M, Cloutier N, Levesque T, Gaib MH, Boilard E. Respective contribution of cytosolic phospholipase A2alpha and

NU

secreted phospholipase A2 IIA to inflammation and eicosanoid production in arthritis. Prostaglandins Other Lipid Mediat 2019;143:106340. doi:

MA

10.1016/j.prostaglandins.2019.106340 (Epub ahead of print). 138. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD,

ED

Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, Devinney R, Doig CJ, Green FH, Kubes P. Platelet TLR4 activates neutrophil extracellular traps

PT

to ensnare bacteria in septic blood. Nat Med 2007;13:463-469.

CE

139. De Stoppelaar SF, van ‘t Veer C, Claushuis TA, Albersen BJ, Rolsofs JJ, van der Poll T. Thrombocytopenia impairs host defense in gram negative pneumonia derived

AC

sepsis in mice. Blood 2014;124:3781-3790. 140. Claushuis TAM, de Vos AF, Nieswandt B, Boon L, Roelofs JJTH, de Boer OJ, van’t Veer C, van der Poll T. Platelet glycoprotein VI aids in local immunity during pneumonia-derived sepsis caused by gram-negative bacteria. Blood 2018;131:864876. 141. Pierre S, Linke B, Suo J, Tarighi N, Del Turco D, Thomas D, Ferreiros N, Stegner D, Frölich S, Sisignano M, Meyer Dos Santos S, deBruin N, Nüsing RM, Deller T,

ACCEPTED MANUSCRIPT Nieswandt B, Geisslinger G, Scholich K. GPVI and thromboxane receptor on platelets promote proinflammatory macrophage phenotypes during cutaneous inflammation. J Invest Dermatol 2017;137:686-695. 142. Boulaftali Y, Mawhin M-A, Jandrot-Perrus M, Ho-Tin-Noé B. Glycoprotein VI in securing vascular integrity in inflamed vessels. Glycoprotein VI. Res Pract Thromb

T

Haemost 2018;2:228-239.

SC RI P

143. McFadyen JD, Schaff M, Peter K. Current and future antiplatelet therapies: emphasis on preserving hemostasis. Nat Rev Cardiol 2018;15:181-191.

144. Kunicki TJ, Williams SA, Nugent DJ. Genetic variants that affect platelet function.

NU

Curr Opin Hematol 2012;19:371-379.

145. Santoso S, Kunicki TJ, Kroll H, Haberbosch W, Gardemann A. Association of the

MA

platelet glycoprotein Ia C807T gene polymorphism with nonfatal myocardial infarction in younger patients. Blood 1999;93:2449-2453.

ED

146. Carlsson LE, Santoso S, Spitzer C, Kessler C, Greinacher A. The alpha2 gene coding sequence T807/A873 of the platelet collagen receptor integrin alpha2beta1 might be

PT

a genetic risk factor for the development of stroke in younger patients. Blood

CE

1999;93:3583-3586.

147. Morita H, Kurihara H, Imai Y, Sugiyama T, Hamada C, Sakai E, Mori M, Nagai R.

AC

Lack of association between platelet the platelet glycoprotein Ia C807T gene polymorphism and myocardial infarction in Japanese. An approach entailing melting curve analysis with specific fluorescent hybridization probes. Thromb Haemost 2001;85:226-230. 148. Takagi S, Iwai N, Baba S, Mannami T, Ono K, Miyata T, Miyazaki S, Nonogi H, Goto Y. A GPVI polymorphisms is a risk factor for myocardial infarction in Japanese. Atherosclerosis 2002;165:387-398.

ACCEPTED MANUSCRIPT 149. Ollikainen E, Mikkelson J, Perola M, Pentilla A, Karhunen PJ. Platelet membrane collagen receptor glycoprotein VI polymorphism is associated with coronary thrombosis and fatal myocardial infarction in middle-aged men. Atherosclerosis 2004;176:95-99. 150. Samaha FF, Hibard C, Sacks J, Chen H, Varello MA, George T, Kahn ML. Density

T

of platelet collagen receptors glycoprotein VI and alpha2beta1 and prior myocardial

SC RI P

infarction in human subjects, a pilot study. Med Sci Monit 2005;11:CR224-229. 151. Bigalke B, Lindemann S, Ehrlers R, Seizer P, Daub K, Langer H, Schonberger T, Kremmer E, Siegel-Axel D, May AE, Gawaz M. Expression of platelet collagen

NU

receptor glycoprotein VI is associated with acute coronary syndrome. Eur Heart J 2006;27:2165-2169.

MA

152. Asfari A, Dent JA, Corken A, Herington D, Kaliki V, Sra N, Hefley G, Pasala S, Prodhan P, Ware J. Platelet glycoprotein VI haplotypes and the presentation of

ED

paediatric sepsis. Thromb Haemost 2019;119:431-438. 153. Nagalia S, Bray PF. Personalized medicine in thrombosis: back to the future. Blood

PT

2016;127:2665-2671.

CE

154. Kubisz P, Ruiz-Arguelles GJ, Stasko J, Holly P, Ruiz-Delgado GJ. Sticky platlet syndrome: History and future directions. Semin Thromb 2014;40:526-534.

AC

155. Skerenova M, Sokol J, Biringer K, Ivankova J, Stasko J, Kubisz P, Lasabova Z. GP6 haplotype of missense variants is associated with sticky platelet syndrome manifested by fetal loss. Clin Appl Thromb Hemost 2018;24:63-69. 156. Massberg S, Gawaz M, Grüner S, Schulte V, Konrad I, Zohlinhöfer D, Heinzmann U, Nieswandt B. A crucial role of glycoprotein VI for platelet recruitment to the injured artery wall in vivo. J Exp Med 2003;197:41-49.

ACCEPTED MANUSCRIPT 157. Konishi H, Kato Y, Takaya N, Kashiwakura Y, Itoh S, Ra C, Daida H. Platelets activated by collagen through immunoreceptor tyrosine-based activation motif play pivotal role in initiation and generation of neointimal hyperplasia after vascular injury. Circulation 2002;105:912-916. 158. Takaya H, Katch Y, Iwabuchi K, Hayashi I, Konishi H, Itoh S, Okumura K, Ra C,

T

Nagaoka I, Daida H. Platelets activated by collagen through the immunoreceptor

SC RI P

tyrosine based activation motif in the Fc receptor gamma chain play a pivotal role in the development of myocardial ischemia reperfusion injury. J Mol Cell Cardiol 2005;39:856-864.

NU

159. Ungerer M, Rosport K, Bültmann A, Piechatzek R, Uhland K, Schlieper P, Gawaz M, Münch G. Novel antiplatelet drug Revacept (dimeric glycoprotein VI-Fc)

MA

specifically and efficiently inhibited collagen- induced platelet aggregation without affecting general haemostasis in humans. Circulation 2011;123:1891-1899.

ED

160. Ungerer M, Li Z, Baumgartner C, Goebel S, Vogelmann J, Holthoff HP, Gawaz M, Münch G. The GPVI fusion protein Revacept reduces thrombus formation and

PT

improves vascular dysfunction in atherosclerosis without impact on bleeding times.

CE

PLoS One 2013; 8(8):e71193. doi: 10.1371/journal.pone.0071193. ecollection 2013. 161. Schüpke S, Hein-Rothweiler R, Mayer K, Janisch M, Sibbing D, Ndrepepa G, Hilz

AC

R, Laugwitz KL, Berniocher I, Gschwendtner S, Kupka D, Gori T, Zeiher AM, Schunkert H, Massberg S, Kastrati A, ISAR-PLASTER-Trial Investigators. Revacept, a novel inhibitor of platelet adhesion, in patients undergoing elective PCIdesign and rationale of the randomized ISAR-PLASTER trial. Thromb Haemost 2019; Jun 21.doi: 10.10.1055/s-0039-1692423. (Epub ahead of print).

ACCEPTED MANUSCRIPT 162. Qiao J, Arthur JF, Gardiner EE, Andrews RK, Zeng L, Xu K. Regulation of platelet activation and thrombus formation by reactive oxygen species. Redox Biol 2018; 14:126-130. 163. Mangin PH, Tang C, Bourdon C, Loyau S, Freund M, Hechler B, Gachet C, JandrotPerrus M. A humanized glycoprotein VI (GPVI) mouse model to assess the

T

antithrombotic efficacies of anti-GPVI agents. J Pharmacol Exp Ther 2012;341:156-

SC RI P

163.

164. Jiang P, Jandrot-Perrus M. New advances in treating thrombotic disease: GPVI as a platelet drug target. Drug Discov Today 2014;19:1471-1475.

NU

165. Induruwa I, Jung S, Warburton EA. Beyond antiplatelets: The role of glycoprotein VI in ischemic stroke. Int J Stroke 2016; 11:618-625.

MA

166. Chang CH, Chung CH, Tu YS, Tsai CC, Hsu CC, Peng HC, Tseng YJ, Huang TF. Trowaglerix venom polypeptides as a novel antithrombotic agent by targeting

2017;37:1307-1314.

ED

immuno- globulin- like domains in platelet. Arterioscler Thromb Vasc Biol

PT

167. Florian P, Wonerow P, Harder S, Kuczka K, Dubar M, Graff J. Anti-GPVI Fab

CE

SAR264565 effectively blocks GPVI function in ex vivo human platelets under arterial shear in a perfusion chamber. Eur J Clin Pharmacol 2017;73:949-956.

AC

168. Voors-Pette C, Lebozec K, Dogterom P, Jullien L, Billiald P, Ferlan P, Renaud L, Favre-Bulle O, Avenard G, Machacek M, Piétan Y, Jandrot-Perrus M. Safety and tolerability, pharmacokinetics, and pharmacodynamics of ACT017, an antiplatelet GPVI (glycoprotein VI) Fab. Arterioscler Thromb Vasc Biol 2019;39:956-964. 169. Pachel C, Mathes D, Arias-Loza AP, Heitzmann W, Nordbeck P, Deppermann C, Lorenz V, Hofmann U, Nieswandt B, Frantz S. Inhibition of platelet GPVI protects

ACCEPTED MANUSCRIPT against myocardial ischemia-reperfusion injury. Arterioscler Thromb Vasc Biol 2016;36:629-635. 170. Lee MY, Verni CC, Herbig BA, Diamond SL. Soluble fibrin causes an acquired platelet glycoprotein VI signaling defect: implications for coagulopathy. J Thromb Haemost 2017;15:2396-2407.

T

171. Schutte T, Thijs A, Smulders YM. Never ignore extremely elevated D-dimer levels:

SC RI P

they are specific for serious illness. Neth J Med 2016;74:443-448. 172. Bender M, Hagedorn I, Nieswandt B. Genetic and antibody-induced glycoprotein VI deficiency equally protects mice from mechanically and FeCl(3)-induced thrombosis.

NU

J Thromb Haemost 2011;9:1423-1426.

173. Loyau S, Ho-Tin-Noé B, Bourrienne MC, Boulaftali Y, Jandrot-Perrus M.

MA

Microfluidic modeling of thrombolysis. Arterioscler Thromb Vasc Biol 2018;38:2626-2637.

ED

174. Jain S, Russell S, Ware J. Platelet glycoprotein VI facilitates experimental lung metastasis in syngenic mouse models. J Thromb Haemost 2009; 8:1456-1467.

PT

175. Schlesinger M. Role of platelets and platelet receptors in cancer metastasis. J

CE

Hematol Oncol 2018; 11(1):125. doi: 10.1186/s13045-018-0669-2. 176. Volz J, Mammadova-Bach E, Gil-Pulido J, Nandigama R, Remer K, Sorokin L,

AC

Zernecke A, Abrams SL, Ergun S, Henke E, Nieswandt B. Inhibition of platelet glycoprotein VI induces intratumor hemorrhage and increases efficacy of chemotherapy in mice. Blood 2019;133:2696-2706. 177. Cooper N. State of the art - how I manage immune thrombocytopenia. Br J Haematol 2017;177:39-54. 178. Patel D, Väänänen H, Jirouskova M, Hoffmann T, Bodian C, Coller BS. Dynamics of GPIIb-IIIa-mediated platelet-platelet interactions in platelet/adhesion thrombus

ACCEPTED MANUSCRIPT formation on collagen in vitro as revealed by videomicroscopy. Blood 2003;101:929936. 179. Brouns SLN, van Geffen JP, Heemskerk JWM. High-throughput measurement of human platelet aggregation under flow: application in hemostasis and beyond.

AC

CE

PT

ED

MA

NU

SC RI P

T

Platelets 2018;29:662-669.

ACCEPTED MANUSCRIPT Figure Legends

Figure 1. Cartoon showing the purported structure of the GPVI-FcR complex with essential information on its ligands, signaling pathways and its modification in inherited and acquired

SC RI P

T

disease. Note the presence of negative (-ve) and positive (+ve) signaling pathways.

Figure 2. Cartoon showing the organization of the GP6 gene as it would encode the common GPVI type I splice form. Featured are the published disease-causing mutations, those giving rise to compound heterozgous expression are in the same color. *A homozygous mutation

NU

common to 6 unrelated families in Chile. Common high density “a” (SKTQH) and low

AC

CE

PT

ED

variants are fully referenced in the text.

MA

density “b” (PEALN) alleles are also shown. The gene structure and the mutations or gene

ACCEPTED MANUSCRIPT Table 1: Clinical and biological data for cases with inherited defects of GP6

1

2

F

F

Japan

Normal

Moderate thrombocyto penia

Japan

(110140x109 /L)

F

France

Normal

Absent

Specifi c severe reducti on to collage n

10%

Specifi c absenc e with collage n and CRP

<20%; residual GPVI, smeared on WB

ED

3

Specifi c absenc e to collage n

Belgiu m

511

Normal

AC

Sever al cases (M and F)

Chile

Severe reducti on for collage n, Cvx and CRP

Much reduced

Specifi c absenc e for collage n, CRP and Cvx.

No surface GPVI. Residual truncated protein in cytoplas m

PT

F

CE

4

Normal for all patients

GP6 variant

Bleeding symptom s

Family

Refere nce

ND

Mild bleeding (purpura, menorrha gia and epistaxis)

Parents nonsymptomat ic with 50% expression of GPVI

Moroi et al, 1989

Lifelong mild bleeding. Bleeding after surgery required transfusio n

No family history of bleeding

Arai et al, 1995

Comp Het Arg38Cys + c.356_560 dup + fs + stop

Mild lifelong bleeding (ecchymo ses)

Parents heterozygo us for one mutation

Dumon t et al, 2009

Comp Het Ser176Asn + 16bp out-offrame del + stop

Moderate life-long bleeding (ecchymo ses, epistaxis, menorrha gia) + posttrauma bleeding

Parents heterozygo tes and asymptom atic.

Herman s et al, 2009

Homozygo us c.711_712i nsA + fs + stop

Mild bleeding (easy bruising, epistaxis, gum bleeding), one sister asymptom atic

All cases in unrelated families from different parts of Chile. Founder effect suspected.

Matus et al., 2013; Onselae r et al, 2017; Mangin et al, 2018

T

1

Platelet GPVI Platelet count aggregati expressi on on2

SC RI P

Count ry of origin

ND

NU

Gend er

MA

Ca se

Country of residence/origin; 2GPVI expression determined by flow cytometry or western blot (WB); Cvx, convulxin; CRP, collagen-related peptide; ND, not determined; M, male; F, female; ins, insert; fs, frameshift; Comp het, compound heterozygote 1

ACCEPTED MANUSCRIPT Table 2: Clinical and biological data for cases with acquired defects in GPVI and defective platelet/collagen interactions

F

Japan

USA

Bleeding symptoms

Severe (15x109 /L) improved with corticotherapy (140x109 / L)

Specif ic absen ce to collag en

Bleeding late in life (skin bruising, gum bleeding, epistaxis)

Severe (20x109 /L) normalizing after corticotherapy

Specif ic reduct ion to collag en

<10%

Moderate (105135x109 /L)

Specif ic absen ce with collag en and CRP

Reduc ed to 1015% of normal

Striki ng reduct ion to collag en and Cvx

Much reduce d

PT

France

CE

F

Macrothrombo cytopenia (1835x109 /L)

AC

4

Not evaluat ed

5

F

Japan

Moderate (48x109 /L) later normalizing

6

F

Japan

Moderate (80133x109 /L) then

Antibo dy to GPVI

Yes. Platele t activat ing

Refer Clinical condition ence

ITP-like syndrome + Graves disease

T

Platelet GPVI aggrega expres tion sion3

ED

3

F

Japan

Thrombocytop enia (platelet count2 )

SC RI P

2

F

Origin1

Sugiy ama et al, 1987; Ichino he et al, 1995

Yes. Platele t activat ing

ITP + SLE + thyroiditis

Takah ashi & Moroi , 2001; Ichino he et al, 1995

Moderate bleeding (petechia, easy bruising)

Yes

ITP-like syndrome

Boyla n et al, 2004

Lifelong moderate bleeding

Not detect ed

Gray platelet syndrome and suspected myelofibrosis

Nurde n et al, 2004

Initiall y no but later detect ed in platele t eluates

ITP-like syndrome suspected

Kojim a et al, 2006; Akiya ma et al, 2009

Detect ed in platele t

ITP-like syndrome with progressive loss of

Akiya ma et al,

Mild subcutaneo uss bleeding

NU

1

Gen der

MA

Ca se

Specif ic absen ce with collag en, CRP and Cvx

Absent

Mild bleeding (petechia, gum bleeding)

Specif ic absen ce

Absent then progre ssive

Mild bleeding (easy bruising,

ACCEPTED MANUSCRIPT

F

Australia

ITP, romiplostim treatment was curative

Gardi ner et al, 2008; 2010

No severe bleeding

Yes. In plasm a and on platele ts, activat ing when strong

SLE and ITPlike conditions responsive to treatment

Nurde n et al, 2009

Lifelong bleeding

IgM cold aggluti nin

Pseudothrombo cytopenia (5x109 /L in citrated blood, 100x109 /L in EDTA)

Sanch ezGuiu et al, 2015

Severe deficie ncy.

Exagerated bruising

Plasm a antibo dy that aggreg ates contro l platele ts

ITP-like syndrome. Platelet count improved under treatment

Rabbo lini et al, 2017

None. GPVIsignali ng defcet

Myelodysplasi a and fatal myelomonobla stic leukemia

Belluc ci et al, 2004

None.

Chronic

Belluc

1020% of normal

Major deficie ncy, then recove ry

Low

Specif ic absen ce with collag en, Cvx and CRP

Norma l

Purpura in lower limbs

Specif

Norma

Extensive

11

M

France

Severe (10x109 /L) progressing to severe thrombocytope nia

12

F

France

Normal then

2009

Yes. Activa ting, presen t in plasm a and in platele t eluates

Specif ic absen ce with collag en

CE

F

bleeding symptoms

Epistaxis and lower limb purpura; symptoms continued despite corticother apy

Severe ITP when first studied. Moderate (91x109 /L) at time of study

AC

10

eluates but not in plasm a

T

Not able to be assess ed

epistaxis)

SC RI P

Spain

Chronic moderate thrombocytope nia

PT

9

F

France/M orocco

Severe (4x109 /L) normalizing with immunosuppre ssive and corticoid therapy

Specif ic absen ce with collag en and Cvx

Australia

recove ry

MA

8

F

Severe (2x109 /L) normalizing with corticotherapy

Specif ic absen ce with collag en and Cvx

ED

7

with collag en, Cvx

NU

normalizing

ACCEPTED MANUSCRIPT

F

Germany

Normal

bleeding (also heparindependent thrombocyt openia)

Norma l GPVI expres sion

Mild lifelong skin hematomas + menorrhagi a

GPVI signali ng defect

lymphocytic leukemia

ci et al, 2004

T

Specif ic reduct ion with collag en and Cvx, impro ving with time

l

NU

13

ic absen ce with collag en, Cvx and CRP

Norma l GPVI expres sion

SC RI P

progressive thrombocytope nia

Unknown cause. No autoantibody to GPVI

Klüm pers et al, 2018

Country of residence/origin; 2Platelet count at its nadir; 3GPVI expression determined by flow cytometry or western blot (WB); M, male; F, female; Cvx, convulxin; CRP, collagen-related peptide

AC

CE

PT

ED

MA

1

Figure 1

Figure 2