Semin Thromb Hemost 2013; 39(03): 291-305
DOI: 10.1055/s-0033-1334466
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Genetic Loci Associated with Platelet Traits and Platelet Disorders

Natalia Bunimov
1   Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
,
Nola Fuller
1   Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
,
Catherine P. M. Hayward
1   Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
2   Department of Medicine, McMaster University, Hamilton, Ontario, Canada
3   Hamilton Regional Laboratory Medicine Program, Hamilton, Ontario, Canada
› Author Affiliations
Further Information

Address for correspondence

Catherine P. M. Hayward, MD, PhD, FRCP(C)
Department of Pathology and Molecular Medicine, McMaster University
2N29A, 1280 Main Street, West Hamilton, Ontario
L8S 4K1 Canada   

Publication History

Publication Date:
06 March 2013 (online)

 

Abstract

Genetic investigations have led to important advances in our knowledge of genes, proteins, and microRNA that influence circulating platelet counts, platelet size, and function. The application of genome-wide association studies (GWAS) to platelet traits has identified multiple loci with a significant association to platelet number, size, and function in aggregation and granule secretion assays. Moreover, the genes altered by disease-causing mutations have now been identified for several platelet disorders, including X-linked recessive, autosomal dominant, and autosomal recessive platelet disorders. Some mutations that cause inherited platelet disorders involve genes that GWAS have associated to platelet traits. Although disease-causing mutations in many rare and syndromic causes of platelet disorders have now been characterized, the genetic mutations that cause common inherited platelet disorders, and impair platelet aggregation and granule secretion, are largely unknown. This review summarizes current knowledge on the genetic loci that influence platelet traits, including the genes with well-characterized mutations in certain inherited platelet disorders.


#

Platelets play an important role in hemostasis and their function traits are emerging to have important genetic influences. Platelet function is complex: with vascular injury, normal platelets adhere to exposed collagen and to von Willebrand factor bound to collagen.[1] [2] [3] This triggers the generation and secretion of thromboxane A2 (TXA2) and platelet storage granule release.[1] [2] [3] Platelets then undergo further activation, with intracellular signaling triggered by their released TXA2, adenosine diphosphate (ADP), serotonin, and other agonists (such as thrombin) that are generated at the sites of vessel injury.[1] [2] [3] Genetic defects can impair platelet hemostatic function in many ways, from modifying platelet–vessel wall interactions, through changes in the number of circulating platelets, and/or their size, adhesive properties, responses to agonists, intracellular signaling, granule release, and the feedback that signaling and secretion have on platelet activation and prohemostatic function.[1] [2] [3] The purpose of this review is to summarize the current state of knowledge on the genetic loci that influence platelet functions and traits, including the genes that may contain disease-causing mutations in those characterized forms of inherited platelet disorders and other conditions that modify platelets.

Inheritance of Platelet Traits and Platelet Disorders

Megakaryocytes transcribe a huge number of genes, and platelets are estimated to contain more than 1,000 proteins.[4] [5] [6] [7] [8] [9] [10] [11] Twin studies have provided evidence that platelet traits and function are influenced by genetic factors.[12] Studies of families and candidate genes have led to the identification of several genetic loci that are strongly associated with platelet physiologic and pathologic function.[13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] Genome-wide association studies (GWAS) of different populations have expanded the list of genetic loci that show significant associations to platelet traits ([Table 1] summarizes information on the associations with p values ≤ 1 × 10−5).[14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] Although there are uncertainties about the degree to which these loci predict normal or pathological platelet variability, the heritability of platelet traits, including platelet “reactivity” to agonists in function tests, offers an attractive explanation for the significant correlation of platelet responses to different agonists in clinical aggregation and secretion assays, for individuals with and without bleeding problems.[30] [31] [32] [33] [34]

Table 1

Human single nucleotide polymorphisms that have been associated with platelet count, volume, and function as measured by light-transmission platelet aggregation or secretion assays[a]

Trait

Gene

Protein

Population

SNP with lowest p value for locus (associated trait)

Cytoband

Reference

Count

HSPB7

Heat shock 27 kDa protein family, member 7

AU, D

rs1763611

1p36.23

[14]

MPV

LAPTM4A, SDC1

Lysosomal-associated transmembrane protein 4A, syndecan-1

E

rs11686303

2p24.1

[15]

Count

KCNJ3

G protein-activated inward rectifier potassium channel 1

AU, D

rs11682195

2q24.1

[14]

Collagen-induced secretion

MAGI1

Membrane-associated guanylate kinase, WW and PDZ domain containing protein 1

SA

rs1318477

3p14.1

[16]

Count, MPV

ARHGEF3

Rho guanine nucleotide exchange factor 3

AU, D

rs1354034 (count)

3p14.3

[14] [15]

E

rs12485738 (MPV)

Aggregation with collagen/thromboxane

MME

Neprilysin

E

rs1436634

3q25.2

[17]

Count

THPO

Thrombopoietin

J

rs6141

3q27.1

[18]

Count

KCNIP4

Kv channel-interacting protein 4

AU, D

rs13150985

4p15.32

[14]

MPV

UNC5C

Netrin receptor UNC5C

E

rs265013

4q22.3

[15]

Count

CDH10

Cadherin-10

AU, D

rs10043237

5p14.2

[14]

Count

BAK1

Bcl-2 homologous antagonist/killer

J

rs5745568

6p21.31

[18]

Count

PHACTR1

Phosphatase and actin regulator

AU, D

rs12212807

6p24.1

[14]

Count

GMDS

GDP-mannose 4,6 dehydratase

SA

rs4463305

6p25.3

[16]

Count

HBS1L, MYB

HBS1-like protein, transcriptional activator Myb

AU, D

rs9399137

6q23.3

[14] [18]

J

rs7775698

Aggregation with collagen/thromboxane

IPCEF1

Interactor protein for cytohesin exchange factors 1

E

rs1534446

6q25.2

[17]

MPV

PIK3CG

Phosphoinositol-4,5-biphosphaste 3-kinase (g subunit)

E

rs342293

7q22.3

[19]

Aggregation with ristocetin

C8orf86

Uncharacterized protein C8orf86

SA

rs7845393

8p11.22

[16]

Aggregation with ristocetin

FGFR1

Fibroblast growth factor receptor 1

SA

rs7845393

8p11.22

[16]

MPV

C8orf22

Chromosome 8 open reading frame 22

E

rs12056729

8q11.21

[15]

MPV

CPQ, TSPYL5

Carboxypeptidase Q, testis-specific Y-encoded-like protein 5

E

rs1835742

8q22.1

[15]

Aggregation with collagen/thromboxane

GLIS3

Zinc finger protein GLIS3

E

rs10116901

9p24.2

[17]

Count

RCL1

RNA 3′-terminal phosphate cyclase-like protein

E

rs385893

9q24.1

[18]

Count

ABCA1

ATP-binding cassette sub-family A member 1

AU, D

rs11999261

9q31.1

[14]

Aggregation with AA

LPAR1

Lysophosphatidic acid receptor 1

SA

rs4366150

9q31.3

[16]

Aggregation with ristocetin

CACNB2

Voltage-dependent L-type calcium channel subunit β-2

SA

rs6415964

10p12.33

[16]

Aggregation with ristocetin

SLC39A12

Zinc transporter ZIP12

SA

rs6415964

10p12.33

[16]

MPV

PFKP

Platelet phosphofructokinase

E

rs1574318

10p15.2

[15]

Aggregation with ADP

LDHAL6A

L-lactate dehydrogenase A-like 6A

AA

rs11024665

11p15.1

[17]

Aggregation (collagen-induced)

MIR100HG

mir-100-let-7a-2 cluster host gene

F

rs565229

11q24.1

[20]

Count

NFE2, COPZ1

Transcription factor NF-E2 45 kDa subunit, coatomer subunit zeta-1

E

rs10876550

12q13.13

[21]

Aggregation with ADP

ANKS1B

Ankyrin repeat and sterile α motif domain-containing protein 1B

AA

rs17029861

12q23.1

[17]

Count

SH2B3

SH2B adapter protein 3

J

rs739496

12q24.21

[18]

Count, MPV

WDR66

WD repeat-containing protein 66

E

rs7961894 (count, MPV)

12q24.31

[15]

Aggregation with AA

RPP25

Ribonuclease P protein subunit p25

SA

rs1867153

15q24.2

[16]

Count, aggregation with AA

SCAMP5

Secretory carrier-associated membrane protein 5

SA

rs1867153 (aggregation)

15q24.2

[14] [16]

AU, D

rs2289583 (count)

Count

GPIBA

Glycoprotein Ibα

J

rs6065

17pter-p12

[18]

Count, MPV

TAOK1

Serine/threonine-protein kinase TAO1

E

rs2138852

17q11.2

[15]

Count, MPV

TPM4

Tropomyosin 4

E, AA

rs8109288

19p13.12

[21]

Abbreviations: AA, African American; AU, Australian; D, Dutch; E, European; F, Framingham Heart Study population; J, Japanese; MPV, mean platelet volume; SA, South American.


a The chromosome positions and gene(s) closest to the single nucleotide polymorphism are shown, along with the respective proteins encoded by the genes. All associations shown were reported have a p value ≤ 1 × 10−5.


Among the heritable markers associated with platelet traits, some show associations with size, count, and/or function, and some are associated with more than one platelet trait. Some associations do not clearly map to a single gene and/or show an association to multiple loci (see [Table 1]). Some single nucleotide polymorphisms (SNPs) have been associated with a platelet characteristic in multiple populations, consistent with an influence upon different genetic backgrounds[14] [15] [16] [18] [35] (see [Table 1]). Meta-analyses, which increase the power for detecting associations, have found additional associations for platelet traits ([Table 2] summarizes data for associations with p values ≤ 1 × 10−5).

Table 2

Human single nucleotide polymorphisms that have been associated with platelet function through meta-analysis of genome-wide association studies[a]

Trait

Gene

Protein

Population

SNP with lowest p value for locus (associated trait)

Cytoband

Reference

MPV

KIF1B

Kinesin-like protein KIF1B

E

rs17396340

1p36.22

[22]

Count

MFN2

Mitofusin-2

E

rs2336384

1p36.22

[22]

ADP and epinephrine aggregation

PEAR1

Platelet endothelial aggregation receptor 1

E

rs12566888 (epinephrine)

1q23.1

[25]

AA

rs12041331 (ADP)

Count, MPV

DNM3

Dynamin 3

E

rs10914144

1q24.3

[22] [23]

Count, MPV

TMCC2

Transmembrane and coiled-coil domains protein 2

E

rs1668871 (count),

rs1172130 (MPV)

1q32.1

[22] [23]

Count

LOC148824

Uncharacterized miscellaneous RNA gene

E

rs7550918

1q44

[22]

Count

TRIM58

Tripartite motif-containing protein 58

E

rs3811444

1q44

[22]

Count

THADA

Thyroid adenoma-associated protein

E

rs17030845

2p21

[22]

Count, MPV

EHD3

EH domain-containing protein 3

E

rs649729 (MPV), rs625132 (count)

2p21

[22] [23]

Count

GCKR

Glucokinase regulatory protein

E

rs1260326

2p23

[22]

MPV

ANKMY1

Ankyrin repeat and MYND domain-containing protein 1

E

rs4305276

2q37.3

[22]

Count, MPV

ARHGEF3

Rho guanine nucleotide exchange factor 3

E

rs1354034 (count), rs12485738 (MPV)

3p14.3

[22] [23]

Count

SATB1

DNA-binding protein SATB1

E

rs7641175

3p23

[22]

Count

SYN2

Synapsin-2

E

rs7616006

3p25

[22]

Count

PDIA5

Protein disulfide-isomerase A5

E

rs3792366

3q21.1

[22]

MPV

KALRN

Kalirin

E

rs10512627

3q21.1

[22]

Count

THPO

Thrombopoietin

E

rs6141

3q27.1

[22]

MPV

KIAA0232

Uncharacterized protein KIAA0232

E

rs11734132

4p16.1

[22]

Count

HSD17B13

17-β-hydroxysteroid dehydrogenase 13

E

rs7694379

4q22.1

[22]

Count, MPV

F2R

Proteinase-activated receptor 1

E

rs2227831 (MPV),

rs17568628 (count)

5q13.3

[22]

Count, MPV

MEF2C

Myocyte-specific enhancer factor 2C

E

rs700585

5q14.3

[22]

Count

IRF1

Interferon regulatory factor 1

E

rs2070729

5q31.1

[22]

MPV

RNF145

RING finger protein 145

E

rs10076782

5q33.3

[22]

Count

BAK1

Bcl-2 homologous antagonist/killer

E

rs1330066

6p21.31

[22] [23] [24]

AA

rs210134

Count

HLA-DOA

HLA class II histocompatibility antigen, DO α chain

E

rs399604

6p21.32

[22]

Count

HLA-B

HLA class I histocompatibility antigen, B-82 α chain

E

rs3819299

6p22.2

[22]

Count

LRRC16A

Leucine-rich repeat containing 16A

AA, E, HA

rs441460

6p22.2

[22] [24]

Count

HBS1L, MYB

HBS1-like protein, transcriptional activator Myb

E

rs9399137

6q23.3

[22] [24]

AA

rs9494145

Count

CD36

Platelet glycoprotein IV (thrombospondin receptor)

AA, E, HA

rs13236689

7q21.11

[24]

Count, MPV, aggregation with epinephrine

PIK3CG

Phosphoinositol-4,5-biphosphate 3-kinase (g subunit)

E

rs342293 (MPV)

7q22.3

[22] [23] [25]

AA

rs342293 (count)

AA

rs342296 (MPV)

E

rs342275 (count)

E

rs342286 (aggregation)

Count

WASL

Wiskott–Aldrich syndrome-like protein

E

rs4731120

7q31.3

[22]

Aggregation with ADP

SHH

Sonic hedgehog protein

E

rs2363910

7q36.3

[25]

AA

rs6943029

Count

ZFPM2

Zinc finger protein ZFPM2

E

rs6993770

8q23.1

[22]

Count

PLEC1

Plectin

E

rs6995402

8q24.3

[22]

Count

CDKN2A

Cyclin-dependent kinase inhibitor 2A, isoform 4

E

rs3731211

9p21.3

[22]

MPV

DOCK8

Dedicator of cytokinesis protein 8

E

rs10813766

9p24.3

[22]

Count

AK3

GTP:AMP phosphor-transferase, mitochondrial

E

rs409801

9q24.1

[22]

Count

RCL1

RNA 3′-terminal phosphate cyclase-like protein

E

rs13300663

9q24.1

[22]

Count

BRD3

Bromodomain-containing protein 3

E

rs11789898

9q34.2

[22]

Count, MPV, aggregation with epinephrine

JMJD1C

Probable JmjC domain-containing histone demethylation protein 2C

E

rs7075195 (MPV)

10q21.2–10q21.3

[22] [23] [24] [25]

E

rs10761731 (count)

AA

rs7896518 (count)

E

rs10761741 (aggregation)

AA

rs2893923 (aggregation)

Aggregation with epinephrine

ADRA2A

Alpha-2A adrenergic receptor

E

rs4311994

10q25.2

[25]

AA

rs869244

Aggregation with ADP

MRVI1

Protein MRVI1

E

rs7940646

11p15.4

[25]

AA

rs1874445

MPV

BET1L

BET1-like protein

E

rs11602954

11p15.5

[23]

Count, MPV

PSMD13

26S proteasome non-ATPase regulatory subunit 13

E

rs17655730 (MPV), rs505404 (count)

11p15.5

[22]

Count

FEN1

Flap endonuclease 1

E

rs4246215

11q12.2

[22]

Count

BAD

Bcl2 antagonist of cell death

AA, E, HA

rs477895

11q13.1

[24]

Count

CBL

E3 ubiquitin-protein ligase CBL

E

rs4938642

11q23.3

[22]

MPV

MLSTD1

Fatty acyl-CoA reductase 2

E

rs2015599

12p11.22

[22]

Count, MPV

CD9, VWF

CD9 antigen, von Willebrand factor

E

rs1558324 (MPV), rs7342306 (count)

12p13.31

[22]

Count, MPV

PTGES3, BAZ2A

Prostaglandin E synthase 3, bromodomain adjacent to zinc finger domain protein 2A

E

rs2950390 (MPV), rs941207 (count)

12q13.3

[22]

MPV

COPZ1, NFE2, CBX5

Coatomer subunit zeta-1, transcription factor NF-E2 45 kDa subunit, chromobox protein homolog 5

E

rs10876550

12q13.13

[22]

Count

ATXN2

Ataxin 2

E

rs11065987

12q24.1

[23]

Count

PTPN11

Tyrosine-protein phosphatase nonreceptor type 11

E

rs11066301

12q24.1

[22] [23]

Count

RPH3A, PTPN11

Rabphilin-3A, tyrosine-protein phosphatase nonreceptor type 11

E

rs17824620

12q24.1

[22]

Count

ACAD10

Acyl-CoA dehydrogenase family member 10

AA

rs6490294

12q24.12

[24]

Count

SH2B3

SH2B adapter protein 3

E

rs3184504

12q24.12

[22]

Count, MPV

WDR66

WD repeat-containing protein 66

E

rs7961894 (count, MPV)

12q24.31

[22] [23]

Count

ABCC4

Multidrug resistance-associated protein 4

E

rs4148441

13q32

[22]

MPV

GRTP1

Growth hormone-regulated TBC protein 1

E

rs7317038

13q34

[22]

Count

RAD51L1

DNA repair protein RAD51 homolog 2

E

rs8022206

14q24.1

[22]

Count

ITPK1

Inositol-tetrakisphosphate 1-kinase

E

rs8006385

14q31

[22]

Count

C14orf70, DLK1

Putative uncharacterized protein encoded LINC00523, protein delta homolog 1

E

rs7149242

14q32.2

[22]

Count

RCOR1

REST corepressor 1

E

rs11628318

14q32.31

[22]

Count, MPV

C14orf73

Exocyst complex component 3-like protein 4

E

rs2297067 (count), rs944002 (MPV)

14q32.32

[22]

MPV

BRF1

Transcription factor IIIB 90 kDa subunit

E

rs3000073

14q32.33

[22]

Count, MPV

TPM1

Tropomyosin α-1 chain

E

rs11071720 (MPV), rs3809566 (count)

15q22.1

[22] [23]

Count

ANKDD1A

Ankyrin repeat and death domain-containing protein 1A

E

rs1719271

15q22.31

[22]

Count

GPIBA

Glycoprotein Ibα

E

rs6065

17pter-p12

[22]

Count

AKAP10

A-kinase anchor protein 10, mitochondrial

E

rs397969

17p11.1

[22]

Count, MPV

TAOK1

Serine/threonine-protein kinase TAO1

E

rs8076739 (MPV)

17q11.2

[22] [24]

AA

rs11653144 (MPV)

E

rs559972 (count)

Count, MPV

SNORD7, AP2B1

Small nucleolar RNA C/D box 7, AP-2 complex subunit β

E

rs10512472 (count), rs16971217 (MPV)

17q12

[22]

Count

FAM171A2, ITGA2B

Protein FAM171A2, integrin α-IIb

E

rs708382

17q21.31

[22]

Count

CABLES1

CDK5 and ABL1 enzyme substrate 1

E

rs11082304

18q11.2

[22]

MPV

CD226

CD226 antigen

E

rs12969657

18q22.3

[22] [23]

Count, MPV

TPM4

Tropomyosin 4

E, AA, HA

rs8109288 (count, MPV)

19p13.12

[22] [24]

Count

EXOC3L2

Exocyst complex component 3-like protein 2

E

rs17356664

19q13.32

[22]

Aggregation with collagen

GP6

Platelet glycoprotein VI

E, AA

rs1671152

19q13.42

[25]

MPV

SIRPA

Tyrosine-protein phosphatase nonreceptor type substrate 1

E

rs13042885

20p13

[22] [23]

Count, MPV

TUBB1, CTSZ,

SLMO2

Tubulin β-1 chain, cathepsin Z, protein slowmo homolog 2

E

rs4812048 (MPV)

20q13.32

[22] [24]

AA, E, HA

rs151361 (count)

Count

ARVCF

Armadillo repeat protein deleted in velocardiofacial syndrome

E

rs1034566

22q11.21

[22]

Abbreviations: AA, African Americans; ADP, adenosine diphosphate; CRP, collagen-related peptide; E, European; HA, Hispanic American; MPV, mean platelet volume; SNP, single nucleotide polymorphism.


a The chromosome positions and gene(s) closest to SNP are shown, along with the respective proteins encoded by the genes. SNPs with p ≤ 1 × 10−5 for associations are shown.


GWAS have associated some noncoding regions of the genome with platelet traits, which indirectly suggests that transcriptional or posttranscriptional regulatory mechanisms are involved in regulating platelet function.[36] Given that both platelets and megakaryocytes contain unique regulatory microRNA (miRNA),[29] some GWAS have explored if the genes encoding these short RNA sequences are associated with platelet function traits.[16] Although strong associations of platelet traits with genes encoding miRNA have not been established by GWAS, this possibility needs to be tested with larger numbers of subjects.

At present, there is small but important overlap between genetic loci that are mutated in platelet disorders ([Table 3]) and those that are known to influence platelet traits ([Tables 1] and [2]). Among the genes that show associations to platelet traits by GWAS ([Tables 1] and [2])[13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [36] but are not yet implicated as causing platelet function disorders, a number have known or implicated importance to platelet function, production, or other traits, including the genes for the following: thrombopoietin,[37] the g subunit of phosphoinositol-4,5-biphosphate 3-kinase,[38] the α-2A adrenergic receptor,[39] platelet endothelial aggregation receptor 1,[40] dynamin 3,[41] multidrug resistance protein 4,[42] tropomyosin 4,[43] proteinase-activated receptor 1 (the thrombin receptor),[44] the transcriptional activator Myb,[45] the ATP-binding cassette transporter A1 ABCA1,[46] the transcription factor NF-E2,[47] secretory carrier-associated membrane protein 5,[48] von Willebrand factor, the tetraspanin CD9,[49] [50] CD226,[51] myocyte-specific enhancer factor 2C,[52] protein MRVI1,[53] E3 ubiquitin-protein ligase CBL,[54] tyrosine-protein phosphatase nonreceptor type 11,[55] tyrosine-protein phosphatase nonreceptor type substrate 1,[56] and Bcl-2 homologous antagonist/killer.[57] For many of the genes showing association, there is a need to validate the GWAS data by other experimental models, to verify that the candidate genes influence platelet traits, as has been done for supervillin.[28] Once characterized, candidate genes that are verified to influence platelet traits will provide attractive targets for investigations of the causes of unidentified bleeding disorders.

Table 3

Summary of the genetic causes of characterized inherited platelet disorders[a]

Type of defect

Name of disorder or affected protein

Mode of inheritance

Gene(s)

Protein(s)

Locus

Description of defect and reference

Activation

GPVI

Autosomal recessive

GP6

Platelet GPVI

19q13.42

Impaired platelet activation by collagen because of mutations of GPVI, which mediates collagen-induced platelet activation[61] [62]

Activation

P2Y12

Autosomal recessive

P2RY12

P2Y purinoceptor 12

3q25.1

Impaired platelet activation by ADP[81]

Activation

P2X1

Autosomal dominant

P2RX1

P2X purinoceptor 1

17p13.2

Impaired platelet activation by ADP[82]

Activation

Thromboxane A2 receptor

Autosomal dominant

TBXA2R

Thromboxane A2 receptor

19p13.3

Defective function of the platelet receptor for thromboxane A2 [83] [84] [85] [86] [87]

Activation

Prostaglandin G/H synthase deficiency

Unproven as deficiencies have been reported, but not mutations

PTGS1

Prostaglandin G/H synthase 1 (cyclo-oxygenase 1)

9q33.2

Defective platelet function because of impaired production of thromboxane A2 [132]

Activation

Thromboxane synthase deficiency

Autosomal recessive

TBXAS1

Thromboxane-A synthase

7q34

Defective platelet functions because of impaired production of thromboxane A2. Some associated with increased bone density with Ghosal hematodiaphyseal syndrome[133]

Adhesion

Platelet-type VWD

Autosomal dominant

GP1BA

Platelet GPIbα chain

17p13.2

Gain-of-function defect in VWF binding to GPIbIXV, because of a mutation in GPIbα[119]

Adhesion

Bernard–Soulier syndrome

Autosomal recessive, some forms autosomal dominant

GP9

Platelet GPIX,

platelet GPIbα chain, or

platelet GPIbβ chain

3q21.3

Deficiency or functional defect in GPIbIXV[134]

GPIBA

17p13.222

GPIBB

22q11.21

Adhesion

α2β1

Autosomal dominant

ITGA2

Integrin α2 subunit of α2β1

5q11.2

Thrombocytopenia associated with deficiency of the platelet integrin receptor for collagen[80]

Adhesion

GPIV

Autosomal recessive

CD36

Platelet GPIV (thrombospondin receptor)

7q21.11

Deficiency of platelet CD36 affecting thrombospondin binding and associated with metabolic syndrome, atherosclerotic cardiovascular diseases and cardiomyopathy[66]

Aggregation

Glanzmann thrombasthenia

Autosomal recessive

ITGA2B

Integrin, αIIb or

17q21.31

Impaired platelet aggregation because of loss or dysfunction of αIIbβ3, the platelet integrin that binds fibrinogen[63] [79]

ITGB3

integrin β3 subunits of αIIbβ3

17q21.32

Aggregation

Leukocyte adhesion deficiency type III (LAD3)

Autosomal recessive

FERMT3

Fermitin family homolog 3 (kindlin-3)

11q13.1

Defective integrin activation involving platelets and leukocytes, because of defects in kindlin 3[109] [111]

Fibrinolysis

Quebec platelet disorder

Autosomal dominant

PLAU

Urokinase-type plasminogen activator

10q22.2

Gain-of-function defect in fibrinolysis from increased platelet urokinase plasminogen activator[102] [103] [104]

Platelet numbers

Glanzmann thrombasthenia-like syndromes

Autosomal dominant

ITGA2B

Integrin, αIIb or

17q21.31

Macrothrombocytopenia associated with activating mutations in αIIbβ3 [63]

ITGB3

integrin β3 subunits of αIIbβ3

17q21.32

Platelet numbers

Thrombocytopenia associated with absent radii syndrome (TAR)

Autosomal recessive

RBM8A

RNA-binding protein 8A

1q21.1

Thrombocytopenia associated with the absence of radii and the presence of thumbs[116] [135]

Platelet numbers

Thrombocytopenia with or without syndromic features

X-linked recessive

FLNA

Filamin A

Xq28

Thrombocytopenia, with or without periventricular nodular heterotopia or otopalatodigital syndromes, because of defects in filamin A[88]

Platelet numbers

Congenital amegakaryocytic thrombocytopenia

Autosomal recessive

MPL

Thrombopoietin receptor

1p34.2

Thrombocytopenia because of a deficiency of the thrombopoietin receptor[37]

Platelet numbers

Wiskott–Aldrich syndrome

X-linked thrombocytopenia

X-linked recessive

WAS

Wiskott–Aldrich syndrome protein

Xp11.23

Related disorders, associated with thrombocytopenia, small platelets, and often eczema, recurrent infections and immune deficiency[118]

Platelet numbers

MYH9-related disorders

Autosomal dominant

MYH9

Myosin-9

22q12.3

Macrothrombocytopenia, leukocyte inclusions (Döhle-like bodies), with or without deafness, cataracts and nephritis[77] [78]

Platelet numbers

Thrombocytopenia (THC2)

Autosomal dominant

MASTL,

ANKRD26

Serine/threonine-protein kinase great wall or ankyrin repeat domain-containing protein 2

10p12.1

Thrombocytopenia[113] [123]

Platelet numbers

Thrombocytopenia Cargeeg

Autosomal dominant

CYCS

Cytochrome C

7p15.3

Thrombocytopenia from a gain-of-function defect in cytochrome C that increases apoptosis and dysregulates megakaryopoiesis[89]

Platelet numbers

GATA-1

X-linked recessive

GATA-1

Erythroid transcription factor

Xp11.23

Thrombocytopenic platelet disorder, that can be associated with thalassemia, neutropenia and megakaryoblastic leukemia, with or without Down syndrome[92]

Platelet numbers

Macrothrombocytopenia

Autosomal dominant

TUBB1

Tubulin β-1 chain

20q13.32

Thrombocytopenia with giant platelets[64] [65]

Platelet numbers

Congenital amegakaryocytic thrombocytopenia associated with synostosis of the radius and ulna

Autosomal dominant

HOXA11 and possibly other genes

Homeobox protein Hox-A11

7p15.2

Thrombocytopenia associated with bilateral or unilateral proximal synostosis of the radius and ulna.[93]

Platelet numbers aggregation and secretion

Familial platelet disorder with propensity to myeloid malignancy

Autosomal dominant

RUNX1

Runt-related transcription factor 1

21q22.12

Thrombocytopenia associated with impaired platelet function and hereditary predisposition to myelodysplastic syndrome and myeloid leukemia[91] [120]

Platelet numbers and α-granules

Paris-Trousseau-Jacobsen syndrome

Autosomal dominant

Deletion includes FLI1

Friend leukemia integration 1 transcription factor

11q23

Thrombocytopenia, giant platelets and α-granules, mental retardation, cardiac and facial defects[122]

Procoagulant function

Scott syndrome

Autosomal recessive

TMEM16F

Anoctamin-6

(transmembrane protein 16F)

12q12

Impaired expression of procoagulant phospholipids on activated platelets for coagulation[114] [105]

Signaling

Signaling defects involving G-protein pathways

Not well documented

GNAS1

Guanine nucleotide-binding protein G(s) subunit α isoforms XLas or

20q13.32

Defective G-protein coupled signaling[105] [106] [107]

GNAQ

guanine nucleotide-binding protein G(q) subunit α

9q21.2

Signaling

Impaired platelet G-protein signaling

Autosomal dominant

RGS2

Regulator of G-protein signaling 2

1q31.2

Platelets showed reduced sensitivity to Gs stimulation and reduced cAMP production after stimulation of Gs-coupled receptors.

Enlarged, round platelets with abnormal α-granules.[108]

α-granule storage

Gray platelet syndrome

Autosomal recessive

NBEAL2

Neurobeachin-like protein 2

3p21.31

Thrombocytopenia associated with severe α-granule protein deficiency[94] [95] [96] [113]

α-granule storage

ARC Syndrome

Autosomal recessive

VPS33B

Vacuolar protein sorting-associated protein 33B

15q26.1

Arthrogryposis, renal dysfunction, cholestasis associated with platelet α-granule deficiency[97]

δ-granule storage

Hermansky–Pudlak syndrome

Autosomal recessive

HPS1

Defects in Hermansky–Pudlak syndrome proteins 1–6,

AP-3 complex subunit β-1, dysbindin,

Biogenesis of lysosome-related organelles complex 1 subunit 3, or palladin

10q24.2

Dense granule deficiency associated with defects of lysosomes and melanosomes with albinism[60] [98] [99] [100] [101]

AP3B1

5q14.1

HPS3

3q24

HPS4

22q12.1

HPS5

11p15.1

HPS6

10q24.32

DTNBP1

6p22.3

BLOC1S3

19q13.32

PLDN

15q21.1

δ-granule storage

Chédiak−Higashi syndrome

Autosomal recessive

LYST

Lysosomal trafficking regulator

1q42.3

Dense granule deficiency associated with hypopigmentation, neutropenia, inclusion bodies in myeloblasts and promyelocytes, susceptibility to infection and lymphoma[99] [136]

δ-granule storage

Griscelli syndrome

Autosomal recessive

MYO5A

Unconventional myosin-Va,

ras-related protein Rab-27A, or melanophilin

15q21.2

Dense granule deficiency associated with hypopigmentation, immunological defects, lymphohistiocytosis and central nervous system defects[128] [136]

RAB27A

15q21.3

MLPH

2q37.3

Abbreviations: ADP, adenosine diphosphate; cAMP, cyclic adenosine monophosphate; GP, glycoprotein.


a More than one gene or protein is shown if there are multiple causes.


At present, the knowledge on associations has not reached the point where genotyping can be used to predict an individual's platelet “reactivity” in function tests. It is also important to recognize that GWAS provides information on the genetic causes of variability, but this technique is unlikely to identify rare causes of variability and it will not identify the genes or miRNA with important roles in platelet function if the genetic sequence has little or no variability between subjects.


#

Gene Defects in Characterized Hereditary Disorders of Platelet Numbers and/or Function

There has been significant progress in finding the molecular defect of inherited platelet disorders, particularly for rare disorders, including those associated with syndromic features, as summarized in [Table 3] and illustrated in [Fig. 1].[58] [59] [60] Nonetheless, only a few of the genes identified to contain mutations in persons with inherited defects of platelet function overlap the genes that show a significant association to platelet “reactivity” in other subjects ([Tables 1] [2] [3]). Such an overlap is evident in the platelet disorders that are associated with mutations in the genes encoding glycoprotein (GP) VI,[61] [62] platelet GPIbα,[59] [60] integrin αIIb,[59] [60] [63] tubulin β-1 chain,[64] [65] and the thrombospondin receptor.[66] Nonetheless, there may be important associations with a disease that are not yet discovered, as recent prospective cohort studies indicate that most individuals with bleeding problems from suspected inherited platelet function disorders (>90%) and impaired platelet aggregation and/or dense granule release have uncharacterized defects.[33] [67] [68] There are many potential candidate genes for uncharacterized, inherited platelet disorders, given the many genes transcribed by megakaryocytes and the large number of proteins found in platelets.[6] [8] [9] [10] [11] [69] [70] [71] [72] [73] [74]

Zoom Image
Fig. 1 Schematic representation of the genetic mutations that have been identified to cause platelet function disorders. Affected proteins and resulting disorders are indicated on the diagram, at the point along the pathway of platelet function (from platelet birth to adhesion, activation, and aggregation) that is disrupted.

Genetic mutations resulting in characterized inherited platelet disorders have been identified to alter various aspects of platelets, including their circulating numbers and hemostatic function ([Fig. 1]). Perhaps not surprisingly, most of the mutations are in the genes that have well-known, and important roles in regulating platelet numbers and/or function.[59] [60] Some are associated with thrombocytopenias, with or without changes to platelet shape and volume.[75] [76] As a comprehensive review of the diagnosis and management of all characterized inherited platelet disorders is beyond the scope of this review, readers interested in information on specific disorders are encouraged to read the references cited for different conditions.

Mechanistically, the characterized defects are difficult to classify into disorders of number or function as some affect both. The defects involve proteins found in several different compartments within platelets, such as the following: (1) the cytoskeleton (e.g., MYH9-related disorders[77] [78] and β1-tubulin defects[64] [65]); (2) platelet membranes (e.g., the membrane receptor for von Willebrand factor, GPIbIXV, in Bernard–Soulier syndrome and platelet type von Willebrand disease[59] [60]; the fibrinogen receptor αIIbβ3 in Glanzmann thrombasthenia and the thrombocytopenic disorders associated with gain-of-function defects in this receptor[59] [60] [79]; the platelet integrin receptor for collagen, α2β1[80]; the thrombospondin receptor, GPIV[66]; the membrane receptors for agonist stimulation, GPVI,[61] [62] P2Y12,[81] P2X1,[82] the TXA2 receptor,[83] [84] [85] [86] [87] among others; the membrane receptor for thrombopoietin in congenital amegakaryocytic thrombocytopenia[37]); (3) the region of platelets linking membrane receptors and cytoskeletal proteins (e.g., filamin A defects)[88]; (4) mitochondria (e.g., cytochrome C, which influences platelet apoptosis)[89]; (5) enzymes in the cytosol (e.g., thromboxane-A synthase[90]); and (6) the nucleus, in the case of factors that regulate megakaryocyte gene expression, such as RUNX1,[91] GATA-1,[92] and HOXA11[93] (see [Fig. 1] and [Table 3]). Additionally, some disorders are caused by mutations in the genes that affect the biogenesis of α-granules[94] [95] [96] [97] and dense granules.[98] [99] [100] [101] A unique copy number variation mutation, causing overexpression of the α-granule protein urokinase-type plasminogen activator by megakaryocytes in Quebec platelet disorder, leads to plasmin-mediated degradation of other stored α-granule proteins and a gain-of-function defect in clot lysis.[102] [103] [104]

The disorders that alter platelet surface receptors can impair platelet function in adhesion or aggregation, alter platelet interactions with collagen, von Willebrand factor, or other ligands ([Fig. 1] and [Table 3]), or alter the process of platelet activation by ADP, collagen or TXA2, and agonist-induced signaling ([Fig. 1] and [Table 3]). Recently, a mutation of the transmembrane protein16F, a Ca2+-activated chloride channel, was identified as the cause of the defective, agonist-induced scrambling of phospholipids and impaired membrane activation and procoagulant function of Scott syndrome. Platelet signaling, which is important for activation induced by agonists and platelet interactions with adhesive ligands, is impaired by mutations in genes encoding G proteins[105] [106] [107] and in proteins that regulate G-protein signaling.[108] Inside-out integrin activation is impaired by mutation in the gene for kindlin-3, an intracellular protein that interacts with β integrins.[109]

Inherited Platelet Disorders: Current Information on Modes of Inheritance

Among the characterized inherited platelet abnormalities, autosomal recessive platelet disorders represent a rare but important cause of bleeding (prevalence approximately 1:106 or less).[60] [110] Some of these recessive platelet disorders derive from mutations in genes that encode proteins that are important for platelet production (e.g., MPL, the thrombopoietin receptor),[37] [76] adhesion or aggregation (e.g., glycoprotein IbIX in Bernard–Soulier syndrome[59] [60]; αIIbβ3 in Glanzmann thrombasthenia[59] [60]; and kindlin-3 in persons with impaired platelet integrin function and leukocyte adhesion defects[59] [109] [111]), agonist responses (e.g., ADP receptor P2Y12, GPVI, and thromboxane synthase),[61] [62] [81] [90] [112] and granule protein storage (e.g., NBEAL2 in gray platelet syndrome)[94] [95] [96] [113] ([Table 3]). The recessively inherited platelet disorders also include conditions such as Scott syndrome,[114] [115] thrombocytopenia with absent radii syndrome,[116] and syndromic disorders associated with δ-granule deficiency ([Table 3]).[97] [98] [99] [100] [101] [117]

X-linked platelet disorders are uncommon and include thrombocytopenia associated with GATA-1 mutations,[92] Wiskott–Aldrich syndrome and the related condition, X-linked thrombocytopenia,[118] in addition to the syndromic and nonsyndromic thrombocytopenias associated with filamin A defects[59] [88] ([Table 3]).

Autosomal dominant platelet disorders are the most prevalent of inherited platelet disorders and their causes include mutations in diverse genes that are important for fibrinolysis (e.g., PLAU in Quebec platelet disorder),[102] [103] [104] platelet adhesion (e.g., activating mutations of the gene for GPαIIbβ3, and GPIbα, and α2β1 deficiency),[60] [63] [80] [119] agonist response (e.g., P2X1 [82] and TXA2 receptor[83] [84] [87]), the platelet cytoskeleton (e.g., MYH9-related disorders),[77] [78] transcriptional regulation (e.g., RUNX1),[91] [120] or other platelet traits,[80] [121] [122] [123] including apoptotic pathways that influence platelet numbers[89] ([Table 3]). Defects in the platelet function from mutations in the gene encoding the TXA2 receptor have been reported in individuals heterozygous for receptor mutations,[84] [87] although some have been homozygous for mutations.[86]

Most patients with uncharacterized inherited platelet function disorders have “secretion defects” (also called “release” or “activation” defects) that impair platelet function in aggregation and/or dense granule release assays, often with multiple (but not necessarily all) agonists.[33] [34] [67] [68] [124] [125] [126] A comprehensive study of the genetic causes of inherited platelet secretion defects has never been undertaken. Inherited secretion defects include δ-granule deficiency, which can result from characterized, autosomal recessive, syndromic disorders associated with hypopigmentation (e.g., Hermansky–Pudlak syndrome, Chédiak–Higashi syndrome, and Griscelli syndrome)[97] [100] [127] [128] or uncharacterized, nonsyndromic autosomal dominant causes ([79] [80] [ 127] [129] and Hayward, unpublished observations). Impaired platelet secretion has been reported in individuals who are heterozygous for disease-causing P2RY12 mutations (gene for the ADP receptor P2Y12), who have impaired ADP aggregation.[130] However, P2RY12 mutations could be an infrequent cause of hereditary secretion defects as many individuals with secretion defects have normal ADP aggregation responses.[34]


#
#

Summary

In recent years, GWAS have become a powerful tool for identifying new genetic factors involved in human diseases and variability in the general population, including platelet function.[13] GWAS, and meta-analyses of GWAS data, have provided new information on the genes that influence platelet function and traits (refer to [Tables 1] and [2]). Nonetheless, some caution is advised as many of the associated genes have not been tested for influence on platelet traits using other models (e.g., mouse knockouts[28] and zebrafish morpholinos[131]) to validate their importance to platelet function and other platelet traits. It is likely that platelet function is influenced by many factors, including genetic background, ethnicity, gender and environment, and exposures to drugs that inhibit platelet function. Although the characterization of several disorders with an altered platelet phenotype has provided important new insights on the genes that influence platelet traits, the causes of most inherited platelet “secretion defects” still need to be thoughtfully characterized. Technical advances in molecular analysis of gene linkage and genomic sequences (e.g., full genome and exome sequencing) will facilitate the discovery of the disease-causing mutations of inherited platelet function disorders and increase our understanding of the genetic loci that influence platelet physiology and pathology.


#
#

Conflict of Interest

The authors have no conflicts of interest to disclose.

Funding

Catherine Hayward is the recipient of a Heart and Stroke Foundation of Ontario Career Investigator Award. The work was supported by grants from the Canadian Institutes for Health Research (MOP 97942) and the Canadian Hemophilia Society.

  • References

  • 1 Gresele P, Page CP, Fuster V, Vermylen J. Platelets in Thrombotic and Non-thrombotic Disorders. Cambridge, UK: Cambridge University Press; 2002: 1-1124
  • 2 Michelson AD. Platelets. 3rd ed. Philadelphia, PA: Elsevier/Academic Press; 2013. (in press)
  • 3 Marder VJ, Aird WC, Bennett JS, Schulman S, White II GC. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 6th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2012: 1-1592
  • 4 Rowley JW, Oler AJ, Tolley ND , et al. Genome-wide RNA-seq analysis of human and mouse platelet transcriptomes. Blood 2011; 118 (14) e101-e111
  • 5 Gnatenko DV, Dunn JJ, Schwedes J, Bahou WF. Transcript profiling of human platelets using microarray and serial analysis of gene expression (SAGE). Methods Mol Biol 2009; 496: 245-272
  • 6 Senzel L, Gnatenko DV, Bahou WF. The platelet proteome. Curr Opin Hematol 2009; 16 (5) 329-333
  • 7 Bahou WF. Platelet systems biology using integrated genetic and proteomic platforms. Thromb Res 2012; 129 (Suppl. 01) S38-S45
  • 8 García A, Prabhakar S, Brock CJ , et al. Extensive analysis of the human platelet proteome by two-dimensional gel electrophoresis and mass spectrometry. Proteomics 2004; 4 (3) 656-668
  • 9 García A, Prabhakar S, Hughan S , et al. Differential proteome analysis of TRAP-activated platelets: involvement of DOK-2 and phosphorylation of RGS proteins. Blood 2004; 103 (6) 2088-2095
  • 10 García A, Zitzmann N, Watson SP. Analyzing the platelet proteome. Semin Thromb Hemost 2004; 30 (4) 485-489
  • 11 Watson SP, Bahou WF, Fitzgerald D, Ouwehand W, Rao AK, Leavitt AD. ISTH Platelet Physiology Subcommittee. Mapping the platelet proteome: a report of the ISTH Platelet Physiology Subcommittee. J Thromb Haemost 2005; 3 (9) 2098-2101
  • 12 Gaxiola B, Friedl W, Propping P. Epinephrine-induced platelet aggregation. A twin study. Clin Genet 1984; 26 (6) 543-548
  • 13 Kunicki TJ, Williams SA, Nugent DJ. Genetic variants that affect platelet function. Curr Opin Hematol 2012; 19 (5) 371-379
  • 14 Ferreira MA, Hottenga JJ, Warrington NM , et al. Sequence variants in three loci influence monocyte counts and erythrocyte volume. Am J Hum Genet 2009; 85 (5) 745-749
  • 15 Meisinger C, Prokisch H, Gieger C , et al. A genome-wide association study identifies three loci associated with mean platelet volume. Am J Hum Genet 2009; 84 (1) 66-71
  • 16 Guerrero JA, Rivera J, Quiroga T , et al. Novel loci involved in platelet function and platelet count identified by a genome-wide study performed in children. Haematologica 2011; 96 (9) 1335-1343
  • 17 Mathias RA, Kim Y, Sung H , et al. A combined genome-wide linkage and association approach to find susceptibility loci for platelet function phenotypes in European American and African American families with coronary artery disease. BMC Med Genomics 2010; 3: 22
  • 18 Kamatani Y, Matsuda K, Okada Y , et al. Genome-wide association study of hematological and biochemical traits in a Japanese population. Nat Genet 2010; 42 (3) 210-215
  • 19 Soranzo N, Rendon A, Gieger C , et al. A novel variant on chromosome 7q22.3 associated with mean platelet volume, counts, and function. Blood 2009; 113 (16) 3831-3837
  • 20 Yang Q, Kathiresan S, Lin JP, Tofler GH, O'Donnell CJ. Genome-wide association and linkage analyses of hemostatic factors and hematological phenotypes in the Framingham Heart Study. BMC Med Genet 2007; 8 (Suppl. 01) S12
  • 21 Lo KS, Wilson JG, Lange LA , et al. Genetic association analysis highlights new loci that modulate hematological trait variation in Caucasians and African Americans. Hum Genet 2011; 129 (3) 307-317
  • 22 Gieger C, Radhakrishnan A, Cvejic A , et al. New gene functions in megakaryopoiesis and platelet formation. Nature 2011; 480 (7376) 201-208
  • 23 Soranzo N, Spector TD, Mangino M , et al. A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium. Nat Genet 2009; 41 (11) 1182-1190
  • 24 Qayyum R, Snively BM, Ziv E , et al. A meta-analysis and genome-wide association study of platelet count and mean platelet volume in African Americans. PLoS Genet 2012; 8 (3) e1002491
  • 25 Johnson AD, Yanek LR, Chen MH , et al. Genome-wide meta-analyses identifies seven loci associated with platelet aggregation in response to agonists. Nat Genet 2010; 42 (7) 608-613
  • 26 Goodall AH, Burns P, Salles I , et al; Bloodomics Consortium. Transcription profiling in human platelets reveals LRRFIP1 as a novel protein regulating platelet function. Blood 2010; 116 (22) 4646-4656
  • 27 Snoep JD, Gaussem P, Eikenboom JC , et al. The minor allele of GP6 T13254C is associated with decreased platelet activation and a reduced risk of recurrent cardiovascular events and mortality: results from the SMILE-Platelets project. J Thromb Haemost 2010; 8 (11) 2377-2384
  • 28 Edelstein LC, Luna EJ, Gibson IB , et al. Human genome-wide association and mouse knockout approaches identify platelet supervillin as an inhibitor of thrombus formation under shear stress. Circulation 2012; 125 (22) 2762-2771
  • 29 Nagalla S, Shaw C, Kong X , et al. Platelet microRNA-mRNA coexpression profiles correlate with platelet reactivity. Blood 2011; 117 (19) 5189-5197
  • 30 Bray PF, Mathias RA, Faraday N , et al. Heritability of platelet function in families with premature coronary artery disease. J Thromb Haemost 2007; 5 (8) 1617-1623
  • 31 Herrera-Galeano JE, Becker DM, Wilson AF , et al. A novel variant in the platelet endothelial aggregation receptor-1 gene is associated with increased platelet aggregability. Arterioscler Thromb Vasc Biol 2008; 28 (8) 1484-1490
  • 32 Faraday N, Yanek LR, Yang XP , et al. Identification of a specific intronic PEAR1 gene variant associated with greater platelet aggregability and protein expression. Blood 2011; 118 (12) 3367-3375
  • 33 Hayward CP, Pai M, Liu Y , et al. Diagnostic utility of light transmission platelet aggregometry: results from a prospective study of individuals referred for bleeding disorder assessments. J Thromb Haemost 2009; 7 (4) 676-684
  • 34 Pai M, Wang G, Moffat KA , et al. Diagnostic usefulness of a lumi-aggregometer adenosine triphosphate release assay for the assessment of platelet function disorders. Am J Clin Pathol 2011; 136 (3) 350-358
  • 35 Lo KA, Bauchmann MK, Baumann AP , et al. Genome-wide profiling of H3K56 acetylation and transcription factor binding sites in human adipocytes. PLoS ONE 2011; 6 (6) e19778
  • 36 Kunicki TJ, Nugent DJ. The genetics of normal platelet reactivity. Blood 2010; 116 (15) 2627-2634
  • 37 Ballmaier M, Germeshausen M. Congenital amegakaryocytic thrombocytopenia: clinical presentation, diagnosis, and treatment. Semin Thromb Hemost 2011; 37 (6) 673-681
  • 38 Hirsch E, Bosco O, Tropel P , et al. Resistance to thromboembolism in PI3Kgamma-deficient mice. FASEB J 2001; 15 (11) 2019-2021
  • 39 Pozgajová M, Sachs UJ, Hein L, Nieswandt B. Reduced thrombus stability in mice lacking the alpha2A-adrenergic receptor. Blood 2006; 108 (2) 510-514
  • 40 Kauskot A, Di Michele M, Loyen S, Freson K, Verhamme P, Hoylaerts MF. A novel mechanism of sustained platelet αIIbβ3 activation via PEAR1. Blood 2012; 119 (17) 4056-4065
  • 41 Wang W, Gilligan DM, Sun S, Wu X, Reems JA. Distinct functional effects for dynamin 3 during megakaryocytopoiesis. Stem Cells Dev 2011; 20 (12) 2139-2151
  • 42 Jedlitschky G, Cattaneo M, Lubenow LE , et al. Role of MRP4 (ABCC4) in platelet adenine nucleotide-storage: evidence from patients with delta-storage pool deficiencies. Am J Pathol 2010; 176 (3) 1097-1103
  • 43 Côté GP. Structural and functional properties of the non-muscle tropomyosins. Mol Cell Biochem 1983; 57 (2) 127-146
  • 44 Lang NN, Guðmundsdóttir IJ, Newby DE. Vascular PAR-1: activity and antagonism. Cardiovasc Ther 2011; 29 (6) 349-361
  • 45 García P, Berlanga O, Vegiopoulos A, Vyas P, Frampton J. c-Myb and GATA-1 alternate dominant roles during megakaryocyte differentiation. J Thromb Haemost 2011; 9 (8) 1572-1581
  • 46 Schmitz G, Schambeck CM. Molecular defects in the ABCA1 pathway affect platelet function. Pathophysiol Haemost Thromb 2006; 35 (1–2) 166-174
  • 47 Fock EL, Yan F, Pan S, Chong BH. NF-E2-mediated enhancement of megakaryocytic differentiation and platelet production in vitro and in vivo. Exp Hematol 2008; 36 (1) 78-92
  • 48 Castermans D, Volders K, Crepel A , et al. SCAMP5, NBEA and AMISYN: three candidate genes for autism involved in secretion of large dense-core vesicles. Hum Mol Genet 2010; 19 (7) 1368-1378
  • 49 Israels SJ, McMillan-Ward EM. Platelet tetraspanin complexes and their association with lipid rafts. Thromb Haemost 2007; 98 (5) 1081-1087
  • 50 Mangin PH, Kleitz L, Boucheix C, Gachet C, Lanza F. CD9 negatively regulates integrin alphaIIbbeta3 activation and could thus prevent excessive platelet recruitment at sites of vascular injury. J Thromb Haemost 2009; 7 (5) 900-902
  • 51 Kojima H, Kanada H, Shimizu S , et al. CD226 mediates platelet and megakaryocytic cell adhesion to vascular endothelial cells. J Biol Chem 2003; 278 (38) 36748-36753
  • 52 Gekas C, Rhodes KE, Gereige LM , et al. Mef2C is a lineage-restricted target of Scl/Tal1 and regulates megakaryopoiesis and B-cell homeostasis. Blood 2009; 113 (15) 3461-3471
  • 53 Antl M, von Brühl ML, Eiglsperger C , et al. IRAG mediates NO/cGMP-dependent inhibition of platelet aggregation and thrombus formation. Blood 2007; 109 (2) 552-559
  • 54 Buitrago L, Tsygankov A, Sanjay A, Kunapuli SP. Cbl proteins in platelet activation. Platelets 2012; (August) 29
  • 55 Derbent M, Öncel Y, Tokel K , et al. Clinical and hematologic findings in Noonan syndrome patients with PTPN11 gene mutations. Am J Med Genet A 2010; 152A (11) 2768-2774
  • 56 Catani L, Sollazzo D, Ricci F , et al. The CD47 pathway is deregulated in human immune thrombocytopenia. Exp Hematol 2011; 39 (4) 486-494
  • 57 Josefsson EC, James C, Henley KJ , et al. Megakaryocytes possess a functional intrinsic apoptosis pathway that must be restrained to survive and produce platelets. J Exp Med 2011; 208 (10) 2017-2031
  • 58 Hayward CP, Favaloro EJ. Diagnostic evaluation of platelet disorders: the past, the present, and the future. Semin Thromb Hemost 2009; 35 (2) 127-130
  • 59 Nurden A, Nurden P. Advances in our understanding of the molecular basis of disorders of platelet function. J Thromb Haemost 2011; 9 (Suppl. 01) 76-91
  • 60 Nurden AT, Freson K, Seligsohn U. Inherited platelet disorders. Haemophilia 2012; 18 (Suppl. 04) 154-160
  • 61 Dumont B, Lasne D, Rothschild C , et al. Absence of collagen-induced platelet activation caused by compound heterozygous GPVI mutations. Blood 2009; 114 (9) 1900-1903
  • 62 Hermans C, Wittevrongel C, Thys C, Smethurst PA, Van Geet C, Freson K. A compound heterozygous mutation in glycoprotein VI in a patient with a bleeding disorder. J Thromb Haemost 2009; 7 (8) 1356-1363
  • 63 Nurden AT, Pillois X, Fiore M, Heilig R, Nurden P. Glanzmann thrombasthenia-like syndromes associated with Macrothrombocytopenias and mutations in the genes encoding the αIIbβ3 integrin. Semin Thromb Hemost 2011; 37 (6) 698-706
  • 64 Navarro-Núñez L, Teruel R, Antón AI , et al. Rare homozygous status of P43 β1-tubulin polymorphism causes alterations in platelet ultrastructure. Thromb Haemost 2011; 105 (5) 855-863
  • 65 Kunishima S, Kobayashi R, Itoh TJ, Hamaguchi M, Saito H. Mutation of the beta1-tubulin gene associated with congenital macrothrombocytopenia affecting microtubule assembly. Blood 2009; 113 (2) 458-461
  • 66 Hirano K, Kuwasako T, Nakagawa-Toyama Y, Janabi M, Yamashita S, Matsuzawa Y. Pathophysiology of human genetic CD36 deficiency. Trends Cardiovasc Med 2003; 13 (4) 136-141
  • 67 Castilloux JF, Moffat KA, Liu Y, Seecharan J, Pai M, Hayward CP. A prospective cohort study of light transmission platelet aggregometry for bleeding disorders: is testing native platelet-rich plasma non-inferior to testing platelet count adjusted samples?. Thromb Haemost 2011; 106 (4) 675-682
  • 68 Hayward CP, Moffat KA, Castilloux JF , et al. Simultaneous measurement of adenosine triphosphate release and aggregation potentiates human platelet aggregation responses for some subjects, including persons with Quebec platelet disorder. Thromb Haemost 2012; 107 (4) 726-734
  • 69 Gnatenko DV, Perrotta PL, Bahou WF. Proteomic approaches to dissect platelet function: Half the story. Blood 2006; 108 (13) 3983-3991
  • 70 Gnatenko DV, Zhu W, Bahou WF. Multiplexed genetic profiling of human blood platelets using fluorescent microspheres. Thromb Haemost 2008; 100 (5) 929-936
  • 71 O'Neill EE, Brock CJ, von Kriegsheim AF , et al. Towards complete analysis of the platelet proteome. Proteomics 2002; 2 (3) 288-305
  • 72 Perrotta PL, Bahou WF. Proteomics in platelet science. Curr Hematol Rep 2004; 3 (6) 462-469
  • 73 Maynard DM, Heijnen HF, Horne MK, White JG, Gahl WA. Proteomic analysis of platelet alpha-granules using mass spectrometry. J Thromb Haemost 2007; 5 (9) 1945-1955
  • 74 Parguiña AF, Rosa I, García A. Proteomics applied to the study of platelet-related diseases: aiding the discovery of novel platelet biomarkers and drug targets. J Proteomics 2012; 76 (Spec No) 275-286
  • 75 Balduini CL, Pecci A, Norris P. Diagnosis and management of inherited thrombocytopenias. Semin Thromb Hemost 2013; ; doi:http://dx.doi.org/ 10.1055/s-0032-1333540
  • 76 Hayward CP, Bunimov N. Thrombocytopenic platelet disorders. Semin Thromb Hemost 2011; 37 (6) 617-620
  • 77 Althaus K, Greinacher A. MYH9-related platelet disorders. Semin Thromb Hemost 2009; 35 (2) 189-203
  • 78 Balduini CL, Pecci A, Savoia A. Recent advances in the understanding and management of MYH9-related inherited thrombocytopenias. Br J Haematol 2011; 154 (2) 161-174
  • 79 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 (23) 5996-6005
  • 80 Noris P, Guidetti GF, Conti V , et al. Autosomal dominant thrombocytopenias with reduced expression of glycoprotein Ia. Thromb Haemost 2006; 95 (3) 483-489
  • 81 Cattaneo M. The platelet P2Y12 receptor for adenosine diphosphate: congenital and drug-induced defects. Blood 2011; 117 (7) 2102-2112
  • 82 Oury C, Toth-Zsamboki E, Van Geet C , et al. A natural dominant negative P2X1 receptor due to deletion of a single amino acid residue. J Biol Chem 2000; 275 (30) 22611-22614
  • 83 Kamae T, Kiyomizu K, Nakazawa T , et al. Bleeding tendency and impaired platelet function in a patient carrying a heterozygous mutation in the thromboxane A2 receptor. J Thromb Haemost 2011; 9 (5) 1040-1048
  • 84 Mumford AD, Dawood BB, Daly ME , et al. A novel thromboxane A2 receptor D304N variant that abrogates ligand binding in a patient with a bleeding diathesis. Blood 2010; 115 (2) 363-369
  • 85 Fuse I, Higuchi W, Aizawa Y. Pathogenesis of a bleeding disorder characterized by platelet unresponsiveness to thromboxane A2. Semin Thromb Hemost 2000; 26 (1) 43-45
  • 86 Higuchi W, Fuse I, Hattori A, Aizawa Y. Mutations of the platelet thromboxane A2 (TXA2) receptor in patients characterized by the absence of TXA2-induced platelet aggregation despite normal TXA2 binding activity. Thromb Haemost 1999; 82 (5) 1528-1531
  • 87 Hirata T, Kakizuka A, Ushikubi F, Fuse I, Okuma M, Narumiya S. Arg60 to Leu mutation of the human thromboxane A2 receptor in a dominantly inherited bleeding disorder. J Clin Invest 1994; 94 (4) 1662-1667
  • 88 Nurden P, Debili N, Coupry I , et al. Thrombocytopenia resulting from mutations in filamin A can be expressed as an isolated syndrome. Blood 2011; 118 (22) 5928-5937
  • 89 Cramer Bordé E, Ouzegdouh Y, Ledgerwood EC, Morison IM. Congenital thrombocytopenia and cytochrome C mutation: a matter of birth and death. Semin Thromb Hemost 2011; 37 (6) 664-672
  • 90 Geneviève D, Proulle V, Isidor B , et al. Thromboxane synthase mutations in an increased bone density disorder (Ghosal syndrome). Nat Genet 2008; 40 (3) 284-286
  • 91 Michaud J, Wu F, Osato M , et al. In vitro analyses of known and novel RUNX1/AML1 mutations in dominant familial platelet disorder with predisposition to acute myelogenous leukemia: implications for mechanisms of pathogenesis. Blood 2002; 99 (4) 1364-1372
  • 92 Millikan PD, Balamohan SM, Raskind WH, Kacena MA. Inherited thrombocytopenia due to GATA-1 mutations. Semin Thromb Hemost 2011; 37 (6) 682-689
  • 93 Thompson AA, Nguyen LT. Amegakaryocytic thrombocytopenia and radio-ulnar synostosis are associated with HOXA11 mutation. Nat Genet 2000; 26 (4) 397-398
  • 94 Kahr WH, Hinckley J, Li L , et al. Mutations in NBEAL2, encoding a BEACH protein, cause gray platelet syndrome. Nat Genet 2011; 43 (8) 738-740
  • 95 Gunay-Aygun M, Falik-Zaccai TC, Vilboux T , et al. NBEAL2 is mutated in gray platelet syndrome and is required for biogenesis of platelet α-granules. Nat Genet 2011; 43 (8) 732-734
  • 96 Albers CA, Cvejic A, Favier R , et al. Exome sequencing identifies NBEAL2 as the causative gene for gray platelet syndrome. Nat Genet 2011; 43 (8) 735-737
  • 97 Abu-Sa'da O, Barbar M, Al-Harbi N, Taha D. Arthrogryposis, renal tubular acidosis and cholestasis (ARC) syndrome: two new cases and review. Clin Dysmorphol 2005; 14 (4) 191-196
  • 98 Meng R, Wang Y, Yao Y , et al. SLC35D3 delivery from megakaryocyte early endosomes is required for platelet dense granule biogenesis and is differentially defective in Hermansky-Pudlak syndrome models. Blood 2012; 120 (2) 404-414
  • 99 Masliah-Planchon J, Darnige L, Bellucci S. Molecular determinants of platelet delta storage pool deficiencies: an update. Br J Haematol 2013; 160 (1) 5-11
  • 100 Wei ML. Hermansky-Pudlak syndrome: a disease of protein trafficking and organelle function. Pigment Cell Res 2006; 19 (1) 19-42
  • 101 Badolato R, Prandini A, Caracciolo S , et al. Exome sequencing reveals a pallidin mutation in a Hermansky-Pudlak-like primary immunodeficiency syndrome. Blood 2012; 119 (13) 3185-3187
  • 102 Blavignac J, Bunimov N, Rivard GE, Hayward CP. Quebec platelet disorder: update on pathogenesis, diagnosis, and treatment. Semin Thromb Hemost 2011; 37 (6) 713-720
  • 103 Hayward CP, Rivard GE. Quebec platelet disorder. Expert Rev Hematol 2011; 4 (2) 137-141
  • 104 Paterson AD, Rommens JM, Bharaj B , et al. Persons with Quebec platelet disorder have a tandem duplication of PLAU, the urokinase plasminogen activator gene. Blood 2010; 115 (6) 1264-1266
  • 105 Van Geet C, Izzi B, Labarque V, Freson K. Human platelet pathology related to defects in the G-protein signaling cascade. J Thromb Haemost 2009; 7 (Suppl. 01) 282-286
  • 106 Gabbeta J, Vaidyula VR, Dhanasekaran DN, Rao AK. Human platelet Galphaq deficiency is associated with decreased Galphaq gene expression in platelets but not neutrophils. Thromb Haemost 2002; 87 (1) 129-133
  • 107 Freson K, Thys C, Wittevrongel C , et al. Pseudohypoparathyroidism type Ib with disturbed imprinting in the GNAS1 cluster and Gsalpha deficiency in platelets. Hum Mol Genet 2002; 11 (22) 2741-2750
  • 108 Noé L, Di Michele M, Giets E , et al. Platelet Gs hypofunction and abnormal morphology resulting from a heterozygous RGS2 mutation. J Thromb Haemost 2010; 8 (7) 1594-1603
  • 109 Jurk K, Schulz AS, Kehrel BE , et al. Novel integrin-dependent platelet malfunction in siblings with leukocyte adhesion deficiency-III (LAD-III) caused by a point mutation in FERMT3. Thromb Haemost 2010; 103 (5) 1053-1064
  • 110 Hayward CP. Diagnostic evaluation of platelet function disorders. Blood Rev 2011; 25 (4) 169-173
  • 111 Kuijpers TW, van de Vijver E, Weterman MA , et al. LAD-1/variant syndrome is caused by mutations in FERMT3. Blood 2009; 113 (19) 4740-4746
  • 112 Defreyn G, Machin SJ, Carreras LO, Dauden MV, Chamone DA, Vermylen J. Familial bleeding tendency with partial platelet thromboxane synthetase deficiency: reorientation of cyclic endoperoxide metabolism. Br J Haematol 1981; 49 (1) 29-41
  • 113 Di Paola J, Johnson J. Thrombocytopenias due to gray platelet syndrome or THC2 mutations. Semin Thromb Hemost 2011; 37 (6) 690-697
  • 114 Lhermusier T, Chap H, Payrastre B. Platelet membrane phospholipid asymmetry: from the characterization of a scramblase activity to the identification of an essential protein mutated in Scott syndrome. J Thromb Haemost 2011; 9 (10) 1883-1891
  • 115 Weiss HJ. Impaired platelet procoagulant mechanisms in patients with bleeding disorders. Semin Thromb Hemost 2009; 35 (2) 233-241
  • 116 Albers CA, Paul DS, Schulze H , et al. Compound inheritance of a low-frequency regulatory SNP and a rare null mutation in exon-junction complex subunit RBM8A causes TAR syndrome. Nat Genet 2012; 44 (4) 435-439 , S1–S2
  • 117 Huizing M, Parkes JM, Helip-Wooley A, White JG, Gahl WA. Platelet alpha granules in BLOC-2 and BLOC-3 subtypes of Hermansky-Pudlak syndrome. Platelets 2007; 18 (2) 150-157
  • 118 Thrasher AJ. New insights into the biology of Wiskott-Aldrich syndrome (WAS). Hematology (Am Soc Hematol Educ Program) 2009; 1: 132-138
  • 119 Othman M. Platelet-type Von Willebrand disease: three decades in the life of a rare bleeding disorder. Blood Rev 2011; 25 (4) 147-153
  • 120 Owen C. Insights into familial platelet disorder with propensity to myeloid malignancy (FPD/AML). Leuk Res 2010; 34 (2) 141-142
  • 121 Raslova H, Komura E, Le Couédic JP , et al. FLI1 monoallelic expression combined with its hemizygous loss underlies Paris-Trousseau/Jacobsen thrombopenia. J Clin Invest 2004; 114 (1) 77-84
  • 122 Favier R, Jondeau K, Boutard P , et al. Paris-Trousseau syndrome : clinical, hematological, molecular data of ten new cases. Thromb Haemost 2003; 90 (5) 893-897
  • 123 Noris P, Perrotta S, Seri M , et al. Mutations in ANKRD26 are responsible for a frequent form of inherited thrombocytopenia: analysis of 78 patients from 21 families. Blood 2011; 117 (24) 6673-6680
  • 124 Mezzano D, Quiroga T, Pereira J. The level of laboratory testing required for diagnosis or exclusion of a platelet function disorder using platelet aggregation and secretion assays. Semin Thromb Hemost 2009; 35 (2) 242-254
  • 125 Quiroga T, Goycoolea M, Matus V , et al. Diagnosis of mild platelet function disorders. Reliability and usefulness of light transmission platelet aggregation and serotonin secretion assays. Br J Haematol 2009; 147 (5) 729-736
  • 126 Hayward CP, Moffat KA, Liu Y. Laboratory investigations for bleeding disorders. Semin Thromb Hemost 2012; 38 (7) 742-752
  • 127 Gunay-Aygun M, Huizing M, Gahl WA. Molecular defects that affect platelet dense granules. Semin Thromb Hemost 2004; 30 (5) 537-547
  • 128 Sandrock K, Zieger B. Current Strategies in Diagnosis of Inherited Storage Pool Defects. Transfus Med Hemother 2010; 37 (5) 248-258
  • 129 Corral J, González-Conejero R, Pujol-Moix N, Domenech P, Vicente V. Mutation analysis of HPS1, the gene mutated in Hermansky-Pudlak syndrome, in patients with isolated platelet dense-granule deficiency. Haematologica 2004; 89 (3) 325-329
  • 130 Cattaneo M, Lecchi A, Lombardi R, Gachet C, Zighetti ML. Platelets from a patient heterozygous for the defect of P2CYC receptors for ADP have a secretion defect despite normal thromboxane A2 production and normal granule stores: further evidence that some cases of platelet 'primary secretion defect' are heterozygous for a defect of P2CYC receptors. Arterioscler Thromb Vasc Biol 2000; 20 (11) E101-E106
  • 131 Jagadeeswaran P. Zebrafish: a tool to study hemostasis and thrombosis. Curr Opin Hematol 2005; 12 (2) 149-152
  • 132 Dubé JN, Drouin J, Aminian M, Plant MH, Laneuville O. Characterization of a partial prostaglandin endoperoxide H synthase-1 deficiency in a patient with a bleeding disorder. Br J Haematol 2001; 113 (4) 878-885
  • 133 Isidor B, Dagoneau N, Huber C , et al. A gene responsible for Ghosal hemato-diaphyseal dysplasia maps to chromosome 7q33-34. Hum Genet 2007; 121 (2) 269-273
  • 134 Pham A, Wang J. Bernard-Soulier syndrome: an inherited platelet disorder. Arch Pathol Lab Med 2007; 131 (12) 1834-1836
  • 135 Toriello HV. Thrombocytopenia-absent radius syndrome. Semin Thromb Hemost 2011; 37 (6) 707-712
  • 136 Huizing M, Helip-Wooley A, Westbroek W, Gunay-Aygun M, Gahl WA. Disorders of lysosome-related organelle biogenesis: clinical and molecular genetics. Annu Rev Genomics Hum Genet 2008; 9: 359-386

Address for correspondence

Catherine P. M. Hayward, MD, PhD, FRCP(C)
Department of Pathology and Molecular Medicine, McMaster University
2N29A, 1280 Main Street, West Hamilton, Ontario
L8S 4K1 Canada   

  • References

  • 1 Gresele P, Page CP, Fuster V, Vermylen J. Platelets in Thrombotic and Non-thrombotic Disorders. Cambridge, UK: Cambridge University Press; 2002: 1-1124
  • 2 Michelson AD. Platelets. 3rd ed. Philadelphia, PA: Elsevier/Academic Press; 2013. (in press)
  • 3 Marder VJ, Aird WC, Bennett JS, Schulman S, White II GC. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 6th ed. Philadelphia, PA: Lippincott Williams and Wilkins; 2012: 1-1592
  • 4 Rowley JW, Oler AJ, Tolley ND , et al. Genome-wide RNA-seq analysis of human and mouse platelet transcriptomes. Blood 2011; 118 (14) e101-e111
  • 5 Gnatenko DV, Dunn JJ, Schwedes J, Bahou WF. Transcript profiling of human platelets using microarray and serial analysis of gene expression (SAGE). Methods Mol Biol 2009; 496: 245-272
  • 6 Senzel L, Gnatenko DV, Bahou WF. The platelet proteome. Curr Opin Hematol 2009; 16 (5) 329-333
  • 7 Bahou WF. Platelet systems biology using integrated genetic and proteomic platforms. Thromb Res 2012; 129 (Suppl. 01) S38-S45
  • 8 García A, Prabhakar S, Brock CJ , et al. Extensive analysis of the human platelet proteome by two-dimensional gel electrophoresis and mass spectrometry. Proteomics 2004; 4 (3) 656-668
  • 9 García A, Prabhakar S, Hughan S , et al. Differential proteome analysis of TRAP-activated platelets: involvement of DOK-2 and phosphorylation of RGS proteins. Blood 2004; 103 (6) 2088-2095
  • 10 García A, Zitzmann N, Watson SP. Analyzing the platelet proteome. Semin Thromb Hemost 2004; 30 (4) 485-489
  • 11 Watson SP, Bahou WF, Fitzgerald D, Ouwehand W, Rao AK, Leavitt AD. ISTH Platelet Physiology Subcommittee. Mapping the platelet proteome: a report of the ISTH Platelet Physiology Subcommittee. J Thromb Haemost 2005; 3 (9) 2098-2101
  • 12 Gaxiola B, Friedl W, Propping P. Epinephrine-induced platelet aggregation. A twin study. Clin Genet 1984; 26 (6) 543-548
  • 13 Kunicki TJ, Williams SA, Nugent DJ. Genetic variants that affect platelet function. Curr Opin Hematol 2012; 19 (5) 371-379
  • 14 Ferreira MA, Hottenga JJ, Warrington NM , et al. Sequence variants in three loci influence monocyte counts and erythrocyte volume. Am J Hum Genet 2009; 85 (5) 745-749
  • 15 Meisinger C, Prokisch H, Gieger C , et al. A genome-wide association study identifies three loci associated with mean platelet volume. Am J Hum Genet 2009; 84 (1) 66-71
  • 16 Guerrero JA, Rivera J, Quiroga T , et al. Novel loci involved in platelet function and platelet count identified by a genome-wide study performed in children. Haematologica 2011; 96 (9) 1335-1343
  • 17 Mathias RA, Kim Y, Sung H , et al. A combined genome-wide linkage and association approach to find susceptibility loci for platelet function phenotypes in European American and African American families with coronary artery disease. BMC Med Genomics 2010; 3: 22
  • 18 Kamatani Y, Matsuda K, Okada Y , et al. Genome-wide association study of hematological and biochemical traits in a Japanese population. Nat Genet 2010; 42 (3) 210-215
  • 19 Soranzo N, Rendon A, Gieger C , et al. A novel variant on chromosome 7q22.3 associated with mean platelet volume, counts, and function. Blood 2009; 113 (16) 3831-3837
  • 20 Yang Q, Kathiresan S, Lin JP, Tofler GH, O'Donnell CJ. Genome-wide association and linkage analyses of hemostatic factors and hematological phenotypes in the Framingham Heart Study. BMC Med Genet 2007; 8 (Suppl. 01) S12
  • 21 Lo KS, Wilson JG, Lange LA , et al. Genetic association analysis highlights new loci that modulate hematological trait variation in Caucasians and African Americans. Hum Genet 2011; 129 (3) 307-317
  • 22 Gieger C, Radhakrishnan A, Cvejic A , et al. New gene functions in megakaryopoiesis and platelet formation. Nature 2011; 480 (7376) 201-208
  • 23 Soranzo N, Spector TD, Mangino M , et al. A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium. Nat Genet 2009; 41 (11) 1182-1190
  • 24 Qayyum R, Snively BM, Ziv E , et al. A meta-analysis and genome-wide association study of platelet count and mean platelet volume in African Americans. PLoS Genet 2012; 8 (3) e1002491
  • 25 Johnson AD, Yanek LR, Chen MH , et al. Genome-wide meta-analyses identifies seven loci associated with platelet aggregation in response to agonists. Nat Genet 2010; 42 (7) 608-613
  • 26 Goodall AH, Burns P, Salles I , et al; Bloodomics Consortium. Transcription profiling in human platelets reveals LRRFIP1 as a novel protein regulating platelet function. Blood 2010; 116 (22) 4646-4656
  • 27 Snoep JD, Gaussem P, Eikenboom JC , et al. The minor allele of GP6 T13254C is associated with decreased platelet activation and a reduced risk of recurrent cardiovascular events and mortality: results from the SMILE-Platelets project. J Thromb Haemost 2010; 8 (11) 2377-2384
  • 28 Edelstein LC, Luna EJ, Gibson IB , et al. Human genome-wide association and mouse knockout approaches identify platelet supervillin as an inhibitor of thrombus formation under shear stress. Circulation 2012; 125 (22) 2762-2771
  • 29 Nagalla S, Shaw C, Kong X , et al. Platelet microRNA-mRNA coexpression profiles correlate with platelet reactivity. Blood 2011; 117 (19) 5189-5197
  • 30 Bray PF, Mathias RA, Faraday N , et al. Heritability of platelet function in families with premature coronary artery disease. J Thromb Haemost 2007; 5 (8) 1617-1623
  • 31 Herrera-Galeano JE, Becker DM, Wilson AF , et al. A novel variant in the platelet endothelial aggregation receptor-1 gene is associated with increased platelet aggregability. Arterioscler Thromb Vasc Biol 2008; 28 (8) 1484-1490
  • 32 Faraday N, Yanek LR, Yang XP , et al. Identification of a specific intronic PEAR1 gene variant associated with greater platelet aggregability and protein expression. Blood 2011; 118 (12) 3367-3375
  • 33 Hayward CP, Pai M, Liu Y , et al. Diagnostic utility of light transmission platelet aggregometry: results from a prospective study of individuals referred for bleeding disorder assessments. J Thromb Haemost 2009; 7 (4) 676-684
  • 34 Pai M, Wang G, Moffat KA , et al. Diagnostic usefulness of a lumi-aggregometer adenosine triphosphate release assay for the assessment of platelet function disorders. Am J Clin Pathol 2011; 136 (3) 350-358
  • 35 Lo KA, Bauchmann MK, Baumann AP , et al. Genome-wide profiling of H3K56 acetylation and transcription factor binding sites in human adipocytes. PLoS ONE 2011; 6 (6) e19778
  • 36 Kunicki TJ, Nugent DJ. The genetics of normal platelet reactivity. Blood 2010; 116 (15) 2627-2634
  • 37 Ballmaier M, Germeshausen M. Congenital amegakaryocytic thrombocytopenia: clinical presentation, diagnosis, and treatment. Semin Thromb Hemost 2011; 37 (6) 673-681
  • 38 Hirsch E, Bosco O, Tropel P , et al. Resistance to thromboembolism in PI3Kgamma-deficient mice. FASEB J 2001; 15 (11) 2019-2021
  • 39 Pozgajová M, Sachs UJ, Hein L, Nieswandt B. Reduced thrombus stability in mice lacking the alpha2A-adrenergic receptor. Blood 2006; 108 (2) 510-514
  • 40 Kauskot A, Di Michele M, Loyen S, Freson K, Verhamme P, Hoylaerts MF. A novel mechanism of sustained platelet αIIbβ3 activation via PEAR1. Blood 2012; 119 (17) 4056-4065
  • 41 Wang W, Gilligan DM, Sun S, Wu X, Reems JA. Distinct functional effects for dynamin 3 during megakaryocytopoiesis. Stem Cells Dev 2011; 20 (12) 2139-2151
  • 42 Jedlitschky G, Cattaneo M, Lubenow LE , et al. Role of MRP4 (ABCC4) in platelet adenine nucleotide-storage: evidence from patients with delta-storage pool deficiencies. Am J Pathol 2010; 176 (3) 1097-1103
  • 43 Côté GP. Structural and functional properties of the non-muscle tropomyosins. Mol Cell Biochem 1983; 57 (2) 127-146
  • 44 Lang NN, Guðmundsdóttir IJ, Newby DE. Vascular PAR-1: activity and antagonism. Cardiovasc Ther 2011; 29 (6) 349-361
  • 45 García P, Berlanga O, Vegiopoulos A, Vyas P, Frampton J. c-Myb and GATA-1 alternate dominant roles during megakaryocyte differentiation. J Thromb Haemost 2011; 9 (8) 1572-1581
  • 46 Schmitz G, Schambeck CM. Molecular defects in the ABCA1 pathway affect platelet function. Pathophysiol Haemost Thromb 2006; 35 (1–2) 166-174
  • 47 Fock EL, Yan F, Pan S, Chong BH. NF-E2-mediated enhancement of megakaryocytic differentiation and platelet production in vitro and in vivo. Exp Hematol 2008; 36 (1) 78-92
  • 48 Castermans D, Volders K, Crepel A , et al. SCAMP5, NBEA and AMISYN: three candidate genes for autism involved in secretion of large dense-core vesicles. Hum Mol Genet 2010; 19 (7) 1368-1378
  • 49 Israels SJ, McMillan-Ward EM. Platelet tetraspanin complexes and their association with lipid rafts. Thromb Haemost 2007; 98 (5) 1081-1087
  • 50 Mangin PH, Kleitz L, Boucheix C, Gachet C, Lanza F. CD9 negatively regulates integrin alphaIIbbeta3 activation and could thus prevent excessive platelet recruitment at sites of vascular injury. J Thromb Haemost 2009; 7 (5) 900-902
  • 51 Kojima H, Kanada H, Shimizu S , et al. CD226 mediates platelet and megakaryocytic cell adhesion to vascular endothelial cells. J Biol Chem 2003; 278 (38) 36748-36753
  • 52 Gekas C, Rhodes KE, Gereige LM , et al. Mef2C is a lineage-restricted target of Scl/Tal1 and regulates megakaryopoiesis and B-cell homeostasis. Blood 2009; 113 (15) 3461-3471
  • 53 Antl M, von Brühl ML, Eiglsperger C , et al. IRAG mediates NO/cGMP-dependent inhibition of platelet aggregation and thrombus formation. Blood 2007; 109 (2) 552-559
  • 54 Buitrago L, Tsygankov A, Sanjay A, Kunapuli SP. Cbl proteins in platelet activation. Platelets 2012; (August) 29
  • 55 Derbent M, Öncel Y, Tokel K , et al. Clinical and hematologic findings in Noonan syndrome patients with PTPN11 gene mutations. Am J Med Genet A 2010; 152A (11) 2768-2774
  • 56 Catani L, Sollazzo D, Ricci F , et al. The CD47 pathway is deregulated in human immune thrombocytopenia. Exp Hematol 2011; 39 (4) 486-494
  • 57 Josefsson EC, James C, Henley KJ , et al. Megakaryocytes possess a functional intrinsic apoptosis pathway that must be restrained to survive and produce platelets. J Exp Med 2011; 208 (10) 2017-2031
  • 58 Hayward CP, Favaloro EJ. Diagnostic evaluation of platelet disorders: the past, the present, and the future. Semin Thromb Hemost 2009; 35 (2) 127-130
  • 59 Nurden A, Nurden P. Advances in our understanding of the molecular basis of disorders of platelet function. J Thromb Haemost 2011; 9 (Suppl. 01) 76-91
  • 60 Nurden AT, Freson K, Seligsohn U. Inherited platelet disorders. Haemophilia 2012; 18 (Suppl. 04) 154-160
  • 61 Dumont B, Lasne D, Rothschild C , et al. Absence of collagen-induced platelet activation caused by compound heterozygous GPVI mutations. Blood 2009; 114 (9) 1900-1903
  • 62 Hermans C, Wittevrongel C, Thys C, Smethurst PA, Van Geet C, Freson K. A compound heterozygous mutation in glycoprotein VI in a patient with a bleeding disorder. J Thromb Haemost 2009; 7 (8) 1356-1363
  • 63 Nurden AT, Pillois X, Fiore M, Heilig R, Nurden P. Glanzmann thrombasthenia-like syndromes associated with Macrothrombocytopenias and mutations in the genes encoding the αIIbβ3 integrin. Semin Thromb Hemost 2011; 37 (6) 698-706
  • 64 Navarro-Núñez L, Teruel R, Antón AI , et al. Rare homozygous status of P43 β1-tubulin polymorphism causes alterations in platelet ultrastructure. Thromb Haemost 2011; 105 (5) 855-863
  • 65 Kunishima S, Kobayashi R, Itoh TJ, Hamaguchi M, Saito H. Mutation of the beta1-tubulin gene associated with congenital macrothrombocytopenia affecting microtubule assembly. Blood 2009; 113 (2) 458-461
  • 66 Hirano K, Kuwasako T, Nakagawa-Toyama Y, Janabi M, Yamashita S, Matsuzawa Y. Pathophysiology of human genetic CD36 deficiency. Trends Cardiovasc Med 2003; 13 (4) 136-141
  • 67 Castilloux JF, Moffat KA, Liu Y, Seecharan J, Pai M, Hayward CP. A prospective cohort study of light transmission platelet aggregometry for bleeding disorders: is testing native platelet-rich plasma non-inferior to testing platelet count adjusted samples?. Thromb Haemost 2011; 106 (4) 675-682
  • 68 Hayward CP, Moffat KA, Castilloux JF , et al. Simultaneous measurement of adenosine triphosphate release and aggregation potentiates human platelet aggregation responses for some subjects, including persons with Quebec platelet disorder. Thromb Haemost 2012; 107 (4) 726-734
  • 69 Gnatenko DV, Perrotta PL, Bahou WF. Proteomic approaches to dissect platelet function: Half the story. Blood 2006; 108 (13) 3983-3991
  • 70 Gnatenko DV, Zhu W, Bahou WF. Multiplexed genetic profiling of human blood platelets using fluorescent microspheres. Thromb Haemost 2008; 100 (5) 929-936
  • 71 O'Neill EE, Brock CJ, von Kriegsheim AF , et al. Towards complete analysis of the platelet proteome. Proteomics 2002; 2 (3) 288-305
  • 72 Perrotta PL, Bahou WF. Proteomics in platelet science. Curr Hematol Rep 2004; 3 (6) 462-469
  • 73 Maynard DM, Heijnen HF, Horne MK, White JG, Gahl WA. Proteomic analysis of platelet alpha-granules using mass spectrometry. J Thromb Haemost 2007; 5 (9) 1945-1955
  • 74 Parguiña AF, Rosa I, García A. Proteomics applied to the study of platelet-related diseases: aiding the discovery of novel platelet biomarkers and drug targets. J Proteomics 2012; 76 (Spec No) 275-286
  • 75 Balduini CL, Pecci A, Norris P. Diagnosis and management of inherited thrombocytopenias. Semin Thromb Hemost 2013; ; doi:http://dx.doi.org/ 10.1055/s-0032-1333540
  • 76 Hayward CP, Bunimov N. Thrombocytopenic platelet disorders. Semin Thromb Hemost 2011; 37 (6) 617-620
  • 77 Althaus K, Greinacher A. MYH9-related platelet disorders. Semin Thromb Hemost 2009; 35 (2) 189-203
  • 78 Balduini CL, Pecci A, Savoia A. Recent advances in the understanding and management of MYH9-related inherited thrombocytopenias. Br J Haematol 2011; 154 (2) 161-174
  • 79 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 (23) 5996-6005
  • 80 Noris P, Guidetti GF, Conti V , et al. Autosomal dominant thrombocytopenias with reduced expression of glycoprotein Ia. Thromb Haemost 2006; 95 (3) 483-489
  • 81 Cattaneo M. The platelet P2Y12 receptor for adenosine diphosphate: congenital and drug-induced defects. Blood 2011; 117 (7) 2102-2112
  • 82 Oury C, Toth-Zsamboki E, Van Geet C , et al. A natural dominant negative P2X1 receptor due to deletion of a single amino acid residue. J Biol Chem 2000; 275 (30) 22611-22614
  • 83 Kamae T, Kiyomizu K, Nakazawa T , et al. Bleeding tendency and impaired platelet function in a patient carrying a heterozygous mutation in the thromboxane A2 receptor. J Thromb Haemost 2011; 9 (5) 1040-1048
  • 84 Mumford AD, Dawood BB, Daly ME , et al. A novel thromboxane A2 receptor D304N variant that abrogates ligand binding in a patient with a bleeding diathesis. Blood 2010; 115 (2) 363-369
  • 85 Fuse I, Higuchi W, Aizawa Y. Pathogenesis of a bleeding disorder characterized by platelet unresponsiveness to thromboxane A2. Semin Thromb Hemost 2000; 26 (1) 43-45
  • 86 Higuchi W, Fuse I, Hattori A, Aizawa Y. Mutations of the platelet thromboxane A2 (TXA2) receptor in patients characterized by the absence of TXA2-induced platelet aggregation despite normal TXA2 binding activity. Thromb Haemost 1999; 82 (5) 1528-1531
  • 87 Hirata T, Kakizuka A, Ushikubi F, Fuse I, Okuma M, Narumiya S. Arg60 to Leu mutation of the human thromboxane A2 receptor in a dominantly inherited bleeding disorder. J Clin Invest 1994; 94 (4) 1662-1667
  • 88 Nurden P, Debili N, Coupry I , et al. Thrombocytopenia resulting from mutations in filamin A can be expressed as an isolated syndrome. Blood 2011; 118 (22) 5928-5937
  • 89 Cramer Bordé E, Ouzegdouh Y, Ledgerwood EC, Morison IM. Congenital thrombocytopenia and cytochrome C mutation: a matter of birth and death. Semin Thromb Hemost 2011; 37 (6) 664-672
  • 90 Geneviève D, Proulle V, Isidor B , et al. Thromboxane synthase mutations in an increased bone density disorder (Ghosal syndrome). Nat Genet 2008; 40 (3) 284-286
  • 91 Michaud J, Wu F, Osato M , et al. In vitro analyses of known and novel RUNX1/AML1 mutations in dominant familial platelet disorder with predisposition to acute myelogenous leukemia: implications for mechanisms of pathogenesis. Blood 2002; 99 (4) 1364-1372
  • 92 Millikan PD, Balamohan SM, Raskind WH, Kacena MA. Inherited thrombocytopenia due to GATA-1 mutations. Semin Thromb Hemost 2011; 37 (6) 682-689
  • 93 Thompson AA, Nguyen LT. Amegakaryocytic thrombocytopenia and radio-ulnar synostosis are associated with HOXA11 mutation. Nat Genet 2000; 26 (4) 397-398
  • 94 Kahr WH, Hinckley J, Li L , et al. Mutations in NBEAL2, encoding a BEACH protein, cause gray platelet syndrome. Nat Genet 2011; 43 (8) 738-740
  • 95 Gunay-Aygun M, Falik-Zaccai TC, Vilboux T , et al. NBEAL2 is mutated in gray platelet syndrome and is required for biogenesis of platelet α-granules. Nat Genet 2011; 43 (8) 732-734
  • 96 Albers CA, Cvejic A, Favier R , et al. Exome sequencing identifies NBEAL2 as the causative gene for gray platelet syndrome. Nat Genet 2011; 43 (8) 735-737
  • 97 Abu-Sa'da O, Barbar M, Al-Harbi N, Taha D. Arthrogryposis, renal tubular acidosis and cholestasis (ARC) syndrome: two new cases and review. Clin Dysmorphol 2005; 14 (4) 191-196
  • 98 Meng R, Wang Y, Yao Y , et al. SLC35D3 delivery from megakaryocyte early endosomes is required for platelet dense granule biogenesis and is differentially defective in Hermansky-Pudlak syndrome models. Blood 2012; 120 (2) 404-414
  • 99 Masliah-Planchon J, Darnige L, Bellucci S. Molecular determinants of platelet delta storage pool deficiencies: an update. Br J Haematol 2013; 160 (1) 5-11
  • 100 Wei ML. Hermansky-Pudlak syndrome: a disease of protein trafficking and organelle function. Pigment Cell Res 2006; 19 (1) 19-42
  • 101 Badolato R, Prandini A, Caracciolo S , et al. Exome sequencing reveals a pallidin mutation in a Hermansky-Pudlak-like primary immunodeficiency syndrome. Blood 2012; 119 (13) 3185-3187
  • 102 Blavignac J, Bunimov N, Rivard GE, Hayward CP. Quebec platelet disorder: update on pathogenesis, diagnosis, and treatment. Semin Thromb Hemost 2011; 37 (6) 713-720
  • 103 Hayward CP, Rivard GE. Quebec platelet disorder. Expert Rev Hematol 2011; 4 (2) 137-141
  • 104 Paterson AD, Rommens JM, Bharaj B , et al. Persons with Quebec platelet disorder have a tandem duplication of PLAU, the urokinase plasminogen activator gene. Blood 2010; 115 (6) 1264-1266
  • 105 Van Geet C, Izzi B, Labarque V, Freson K. Human platelet pathology related to defects in the G-protein signaling cascade. J Thromb Haemost 2009; 7 (Suppl. 01) 282-286
  • 106 Gabbeta J, Vaidyula VR, Dhanasekaran DN, Rao AK. Human platelet Galphaq deficiency is associated with decreased Galphaq gene expression in platelets but not neutrophils. Thromb Haemost 2002; 87 (1) 129-133
  • 107 Freson K, Thys C, Wittevrongel C , et al. Pseudohypoparathyroidism type Ib with disturbed imprinting in the GNAS1 cluster and Gsalpha deficiency in platelets. Hum Mol Genet 2002; 11 (22) 2741-2750
  • 108 Noé L, Di Michele M, Giets E , et al. Platelet Gs hypofunction and abnormal morphology resulting from a heterozygous RGS2 mutation. J Thromb Haemost 2010; 8 (7) 1594-1603
  • 109 Jurk K, Schulz AS, Kehrel BE , et al. Novel integrin-dependent platelet malfunction in siblings with leukocyte adhesion deficiency-III (LAD-III) caused by a point mutation in FERMT3. Thromb Haemost 2010; 103 (5) 1053-1064
  • 110 Hayward CP. Diagnostic evaluation of platelet function disorders. Blood Rev 2011; 25 (4) 169-173
  • 111 Kuijpers TW, van de Vijver E, Weterman MA , et al. LAD-1/variant syndrome is caused by mutations in FERMT3. Blood 2009; 113 (19) 4740-4746
  • 112 Defreyn G, Machin SJ, Carreras LO, Dauden MV, Chamone DA, Vermylen J. Familial bleeding tendency with partial platelet thromboxane synthetase deficiency: reorientation of cyclic endoperoxide metabolism. Br J Haematol 1981; 49 (1) 29-41
  • 113 Di Paola J, Johnson J. Thrombocytopenias due to gray platelet syndrome or THC2 mutations. Semin Thromb Hemost 2011; 37 (6) 690-697
  • 114 Lhermusier T, Chap H, Payrastre B. Platelet membrane phospholipid asymmetry: from the characterization of a scramblase activity to the identification of an essential protein mutated in Scott syndrome. J Thromb Haemost 2011; 9 (10) 1883-1891
  • 115 Weiss HJ. Impaired platelet procoagulant mechanisms in patients with bleeding disorders. Semin Thromb Hemost 2009; 35 (2) 233-241
  • 116 Albers CA, Paul DS, Schulze H , et al. Compound inheritance of a low-frequency regulatory SNP and a rare null mutation in exon-junction complex subunit RBM8A causes TAR syndrome. Nat Genet 2012; 44 (4) 435-439 , S1–S2
  • 117 Huizing M, Parkes JM, Helip-Wooley A, White JG, Gahl WA. Platelet alpha granules in BLOC-2 and BLOC-3 subtypes of Hermansky-Pudlak syndrome. Platelets 2007; 18 (2) 150-157
  • 118 Thrasher AJ. New insights into the biology of Wiskott-Aldrich syndrome (WAS). Hematology (Am Soc Hematol Educ Program) 2009; 1: 132-138
  • 119 Othman M. Platelet-type Von Willebrand disease: three decades in the life of a rare bleeding disorder. Blood Rev 2011; 25 (4) 147-153
  • 120 Owen C. Insights into familial platelet disorder with propensity to myeloid malignancy (FPD/AML). Leuk Res 2010; 34 (2) 141-142
  • 121 Raslova H, Komura E, Le Couédic JP , et al. FLI1 monoallelic expression combined with its hemizygous loss underlies Paris-Trousseau/Jacobsen thrombopenia. J Clin Invest 2004; 114 (1) 77-84
  • 122 Favier R, Jondeau K, Boutard P , et al. Paris-Trousseau syndrome : clinical, hematological, molecular data of ten new cases. Thromb Haemost 2003; 90 (5) 893-897
  • 123 Noris P, Perrotta S, Seri M , et al. Mutations in ANKRD26 are responsible for a frequent form of inherited thrombocytopenia: analysis of 78 patients from 21 families. Blood 2011; 117 (24) 6673-6680
  • 124 Mezzano D, Quiroga T, Pereira J. The level of laboratory testing required for diagnosis or exclusion of a platelet function disorder using platelet aggregation and secretion assays. Semin Thromb Hemost 2009; 35 (2) 242-254
  • 125 Quiroga T, Goycoolea M, Matus V , et al. Diagnosis of mild platelet function disorders. Reliability and usefulness of light transmission platelet aggregation and serotonin secretion assays. Br J Haematol 2009; 147 (5) 729-736
  • 126 Hayward CP, Moffat KA, Liu Y. Laboratory investigations for bleeding disorders. Semin Thromb Hemost 2012; 38 (7) 742-752
  • 127 Gunay-Aygun M, Huizing M, Gahl WA. Molecular defects that affect platelet dense granules. Semin Thromb Hemost 2004; 30 (5) 537-547
  • 128 Sandrock K, Zieger B. Current Strategies in Diagnosis of Inherited Storage Pool Defects. Transfus Med Hemother 2010; 37 (5) 248-258
  • 129 Corral J, González-Conejero R, Pujol-Moix N, Domenech P, Vicente V. Mutation analysis of HPS1, the gene mutated in Hermansky-Pudlak syndrome, in patients with isolated platelet dense-granule deficiency. Haematologica 2004; 89 (3) 325-329
  • 130 Cattaneo M, Lecchi A, Lombardi R, Gachet C, Zighetti ML. Platelets from a patient heterozygous for the defect of P2CYC receptors for ADP have a secretion defect despite normal thromboxane A2 production and normal granule stores: further evidence that some cases of platelet 'primary secretion defect' are heterozygous for a defect of P2CYC receptors. Arterioscler Thromb Vasc Biol 2000; 20 (11) E101-E106
  • 131 Jagadeeswaran P. Zebrafish: a tool to study hemostasis and thrombosis. Curr Opin Hematol 2005; 12 (2) 149-152
  • 132 Dubé JN, Drouin J, Aminian M, Plant MH, Laneuville O. Characterization of a partial prostaglandin endoperoxide H synthase-1 deficiency in a patient with a bleeding disorder. Br J Haematol 2001; 113 (4) 878-885
  • 133 Isidor B, Dagoneau N, Huber C , et al. A gene responsible for Ghosal hemato-diaphyseal dysplasia maps to chromosome 7q33-34. Hum Genet 2007; 121 (2) 269-273
  • 134 Pham A, Wang J. Bernard-Soulier syndrome: an inherited platelet disorder. Arch Pathol Lab Med 2007; 131 (12) 1834-1836
  • 135 Toriello HV. Thrombocytopenia-absent radius syndrome. Semin Thromb Hemost 2011; 37 (6) 707-712
  • 136 Huizing M, Helip-Wooley A, Westbroek W, Gunay-Aygun M, Gahl WA. Disorders of lysosome-related organelle biogenesis: clinical and molecular genetics. Annu Rev Genomics Hum Genet 2008; 9: 359-386

Zoom Image
Fig. 1 Schematic representation of the genetic mutations that have been identified to cause platelet function disorders. Affected proteins and resulting disorders are indicated on the diagram, at the point along the pathway of platelet function (from platelet birth to adhesion, activation, and aggregation) that is disrupted.