Quebec platelet disorder (QPD) is an autosomal dominant disorder with high penetrance that is associated with increased risks for bleeding. The hallmark of QPD is a gain-of-function defect in fibrinolysis due to increased platelet content of urokinase plasminogen activator (uPA) without systemic fibrinolysis. We hypothesized that increased expression of uPA by differentiating QPD megakaryocytes is linked to PLAU. Genetic marker analyses indicated that QPD was significantly linked to a 2-Mb region on chromosome 10q containing PLAU with a maximum multipoint logarithm of the odds (LOD) score of +11 between markers D10S1432 and D10S1136. Analysis of PLAU by sequencing and Southern blotting excluded mutations within PLAU and its known regulatory elements as the cause of QPD. Analyses of uPA mRNA indicated that QPD distinctly increased transcript levels of the linked PLAU allele with megakaryocyte differentiation. These findings implicate a mutation in an uncharacterized cis element near PLAU as the cause of QPD.

Most inherited bleeding disorders result from genetic defects that reduce hemostatic protein expression, secretion, or function.1  Inherited conditions that cause bleeding by increasing gene expression are uncommon and include Quebec platelet disorder (QPD),2,3  an autosomal dominant disorder with high and possibly complete penetrance4  and a prevalence in Quebec of 1:300 000.4  QPD is associated with a unique gain-of-function abnormality in fibrinolysis due to increased platelet stores of urokinase plasminogen activator (uPA) without systemic fibrinolysis or increased uPA in plasma,2,5  urine,6  or CD34+ hematopoietic progenitors.7  QPD increases risks for a number of bleeding symptoms, including delayed-onset bleeding after hemostatic challenges that responds only to fibrinolytic inhibitor therapy.4  Diagnostic tests for QPD include assays for increased platelet uPA and α-granule protein degradation from intraplatelet plasmin generation.2,4 

We considered PLAU, the uPA gene on chromosome 10q24, as a candidate gene for QPD as QPD selectively increases (about 100-fold) PLAU expression during megakaryocyte differentiation.7  In addition, increased PLAU expression in mouse megakaryocytes results in a QPD-like disorder characterized by abnormal bleeding that responds to fibrinolytic inhibitor therapy, stored platelet protein degradation, and accelerated lysis of thrombi.8 PLAU contains 11 exons and 10 introns, and its conserved regulatory elements include a 3′ untranslated region (UTR) that affects mRNA stability (reviewed in Diamandis et al2 ; Figure S1, available on the Blood website; see the Supplemental Materials link at the top of the online article) and a 2.5-kb upstream region that is known to bind transcription factors produced during megakaryopoiesis (reviewed in Kaushansky9 ). These observations led us to investigate PLAU as a candidate gene for QPD.

This study was carried out in accordance with the Helsinki Protocol for research on human subjects and institutional ethics review board approval from Hamilton Health Sciences, McMaster University, and Centre Hôspitalier Universitaire Sainte Justine. Participants were 28 affected and 110 unaffected individuals who were descendants of a common QPD ancestor,4  and 110 healthy unrelated controls. Subjects provided written informed consent; if under 18 years of age, parental consent was obtained. DNA was collected from all subjects for linkage analysis, DNA sequencing, and Southern blotting. PLAU allelic expression was assessed using RNA harvested from platelets, peripheral blood CD34+ cells, and saliva cells of selected individuals (4 with QPD and 5 controls).

A detailed description of materials and methods is provided in Document S1.

The 41 individuals selected for initial genotyping of chromosome 10 microsatellites were predicted to be informative, with estimated average (maximum) logarithm of the odds (LOD) score of +8.4 (+10.9) compared with 10.0 (14.1) for all family members. Genotyping of the 41 subjects (Table 1) indicated significant linkage of GATA121A08 and D10S1432 to QPD (respective LOD scores, +3.6 at θ = 0.05 and +3.3 at θ = 0.1; data not shown). Genotyping of all QPD family members, for these and additional markers (Table 1), confirmed that the region containing PLAU was strongly linked to QPD (LOD scores of +3.0 to +8.4), with a common haplotype in individuals with QPD.

Table 1

Two-point linkage analysis of chromosome 10 microsatellite markers and SNPs

Marker nameRutgers positionBuild 36.1 positionNo. of QPD family members genotypedLOD score at φ (recombination fraction)
0.000.010.050.10.2
D10S1453*  6 615 628 41 −∞ −16.04 −7.74 −4.26 −1.35 
D10S189* 19.62 6 761 879 41 −∞ −4.69 −2.98 −2.11 −1.05 
D10S2325* 31.35 12 832 951 41 −∞ −2.52 0.07 0.93 1.23 
D10S1423* 44.78 19 477 909 41 −∞ −2.98 0.09 1.06 1.46 
D10S1426 57.44 30 535 790 41 −∞ −1.99 1.05 1.94 2.12 
D10S1208* 62.04 35 297 637 41 −∞ −5.2 −0.25 1.33 1.97 
D10S1221*  57 199 892 41 −∞ −10.04 −4.71 -2.6 −0.83 
D10S1225 80.04 64 425 004 41 −∞ −0.92 0.31 0.65 0.66 
GATA121A08 85.32 70 182 554 138 −∞ 4.39 4.55 4.17 3.14 
rs12356915*  73 931 323 138 −2.41 −2.15 −1.14 −0.66 −0.28 
D10S1432 93.7 74 329 402 138 −∞ 4.69 6.65 6.75 5.56 
D10S188 93.7 75 075 749 138 −∞ 2.98 3.23 2.95 2.11 
D10S1737 93.7 75 259 301 138 −∞ 1.06 3.75 4.2 3.42 
QPD-1  75 327 173 138 11.79 11.59 10.71 9.5 6.8 
PLAU  75 340 896       
rs4065 93.7 75 346 470 138 0.76 0.72 0.59 0.45 0.23 
rs1908339  75 513 106 138 2.88 3.0 3.0 2.72 1.97 
D10S535 93.7 76 010 712 138 −∞ 1.64 3.15 3.31 2.69 
rs7899137  76 338 468 138 3.0 2.86 2.58 2.27 1.62 
D10S556 94.38 76 922 909 138 −∞ 1.38 2.33 2.37 1.81 
D10S195* 94.38 77 289 176 138 −∞ −3.38 −1.02 −0.15 0.4 
D10S1136 95.77 78 009 813 138 −∞ 7.83 8.42 7.94 6.17 
D10S1730 96.85 78 600 935 41 −0.4 1.69 2.03 1.86 
D10S2327* 98.55 80 382 014 41 −11.38 −3.88 −1.49 −0.56 0.06 
D10S1419  92 354 576 41 −∞ −1.3 −0.06 0.32 0.43 
D10S677* 114.71 95 954 428 41 −∞ −8.1 −2.33 −0.29 0.98 
D10S1239* 121.96 103 186 224 41 −∞ −3.5 −0.51 0.44 0.87 
D10S1237 133.64 116 109 968 41 −∞ 1.04 2.09 2.22 1.83 
D10S1230* 145.08 122 732 623 41 −∞ −13.29 −6.56 −3.9 −1.59 
D10S1213 150.11 125 396 627 41 0.26 0.24 0.15 0.06 −0.02 
D10S1248* 166.09 130 982 451 41 −∞ −7.09 −3.82 −2.06 −0.53 
D10S212* 174.19 134 299 489 41 −8.49 −2.61 −0.27 0.51 0.85 
Marker nameRutgers positionBuild 36.1 positionNo. of QPD family members genotypedLOD score at φ (recombination fraction)
0.000.010.050.10.2
D10S1453*  6 615 628 41 −∞ −16.04 −7.74 −4.26 −1.35 
D10S189* 19.62 6 761 879 41 −∞ −4.69 −2.98 −2.11 −1.05 
D10S2325* 31.35 12 832 951 41 −∞ −2.52 0.07 0.93 1.23 
D10S1423* 44.78 19 477 909 41 −∞ −2.98 0.09 1.06 1.46 
D10S1426 57.44 30 535 790 41 −∞ −1.99 1.05 1.94 2.12 
D10S1208* 62.04 35 297 637 41 −∞ −5.2 −0.25 1.33 1.97 
D10S1221*  57 199 892 41 −∞ −10.04 −4.71 -2.6 −0.83 
D10S1225 80.04 64 425 004 41 −∞ −0.92 0.31 0.65 0.66 
GATA121A08 85.32 70 182 554 138 −∞ 4.39 4.55 4.17 3.14 
rs12356915*  73 931 323 138 −2.41 −2.15 −1.14 −0.66 −0.28 
D10S1432 93.7 74 329 402 138 −∞ 4.69 6.65 6.75 5.56 
D10S188 93.7 75 075 749 138 −∞ 2.98 3.23 2.95 2.11 
D10S1737 93.7 75 259 301 138 −∞ 1.06 3.75 4.2 3.42 
QPD-1  75 327 173 138 11.79 11.59 10.71 9.5 6.8 
PLAU  75 340 896       
rs4065 93.7 75 346 470 138 0.76 0.72 0.59 0.45 0.23 
rs1908339  75 513 106 138 2.88 3.0 3.0 2.72 1.97 
D10S535 93.7 76 010 712 138 −∞ 1.64 3.15 3.31 2.69 
rs7899137  76 338 468 138 3.0 2.86 2.58 2.27 1.62 
D10S556 94.38 76 922 909 138 −∞ 1.38 2.33 2.37 1.81 
D10S195* 94.38 77 289 176 138 −∞ −3.38 −1.02 −0.15 0.4 
D10S1136 95.77 78 009 813 138 −∞ 7.83 8.42 7.94 6.17 
D10S1730 96.85 78 600 935 41 −0.4 1.69 2.03 1.86 
D10S2327* 98.55 80 382 014 41 −11.38 −3.88 −1.49 −0.56 0.06 
D10S1419  92 354 576 41 −∞ −1.3 −0.06 0.32 0.43 
D10S677* 114.71 95 954 428 41 −∞ −8.1 −2.33 −0.29 0.98 
D10S1239* 121.96 103 186 224 41 −∞ −3.5 −0.51 0.44 0.87 
D10S1237 133.64 116 109 968 41 −∞ 1.04 2.09 2.22 1.83 
D10S1230* 145.08 122 732 623 41 −∞ −13.29 −6.56 −3.9 −1.59 
D10S1213 150.11 125 396 627 41 0.26 0.24 0.15 0.06 −0.02 
D10S1248* 166.09 130 982 451 41 −∞ −7.09 −3.82 −2.06 −0.53 
D10S212* 174.19 134 299 489 41 −8.49 −2.61 −0.27 0.51 0.85 

Markers were evaluated for 41 selected individuals or all 138 QPD family members, as indicated. SNPs are designated by rs numbers, except for the newly identified SNP, QPD-1. PLAU indicates the location of the urokinase plasminogen activator gene. Rutgers map positions are in centimorgans, except for 6 markers whose genetic map positions are unknown. Build 36.1 positions indicate physical distances on chromosome 10.

*

Markers excluding linkage.

Markers giving the best evidence for linkage.

Multipoint linkage analysis with the markers closest to PLAU (Figure 1A) indicated that the most likely site for the disease gene was near PLAU (maximal LOD score of +11). Data for D10S195 excluded part of this region, and defined a 2-Mb region of chromosome 10q as the most likely location for the QPD mutation. This region contains 22 additional genes (Table S1), including 2 transcription factors (HSGT1 and MYST4) that are not expressed by megakaryocytes or known to regulate PLAU.10 

Figure 1

Multipoint linkage and allelic expression analysis of PLAU in QPD. (A) Results of multipoint linkage using 4 microsatellite markers (positions are shown relative to GATA121A08). The genetic location of PLAU between markers D10S1432 and D10S1136 is indicated on the x-axis, which is the region with the highest LOD score for this analysis (+11). (B) Graph of reverse transcriptase–quantitative PCR (RT-qPCR) analyses of PLAU alleles for SNP rs4065 in different tissues and indicates the measured ratios of T/C alleles in saliva cells, CD34+ cells, and platelets in samples from 5 controls and 4 individuals with QPD. T/C ratios were significantly different for QPD compared with control samples for all tissues (P = .016).

Figure 1

Multipoint linkage and allelic expression analysis of PLAU in QPD. (A) Results of multipoint linkage using 4 microsatellite markers (positions are shown relative to GATA121A08). The genetic location of PLAU between markers D10S1432 and D10S1136 is indicated on the x-axis, which is the region with the highest LOD score for this analysis (+11). (B) Graph of reverse transcriptase–quantitative PCR (RT-qPCR) analyses of PLAU alleles for SNP rs4065 in different tissues and indicates the measured ratios of T/C alleles in saliva cells, CD34+ cells, and platelets in samples from 5 controls and 4 individuals with QPD. T/C ratios were significantly different for QPD compared with control samples for all tissues (P = .016).

Close modal

At 13.7 kb upstream of the PLAU transcription start site, we identified a potentially informative marker for QPD (designated QPD-1)—a single nucleotide polymorphism (SNP; T/G; Build 36.1 position: 75 327 173). Genotypes of all 28 affected and 4 of 110 unaffected QPD family members were T/G, whereas other unaffected family members were T/T. Among 105 unrelated French-Canadians, the G allele was uncommon (3 individuals, T/G; 102, T/T). SNP QPD-1 was strongly linked with QPD (LOD score, +11.8; Table 1). This LOD score, and the maximal LOD score from multipoint analyses, were close to the maximum predicted by linkage simulations, excluding the possibility of another locus for QPD in this pedigree.

Southern blotting for a 25.2-kb region of chromosome 10 containing PLAU and its known regulatory elements excluded major alterations from QPD. DNA sequencing excluded the possibility that QPD results from mutations in the exons, introns, and characterized 3′ and 5′ regulatory elements of PLAU, or in the entire 24 kb upstream of the PLAU transcription start site and 2 kb downstream of the 3′ UTR. Complete sequencing of both PLAU alleles for these regions was confirmed, as the overlapping polymerase chain reaction (PCR) products demonstrated SNP heterozygosity in at least 1 QPD subject (Table S2).

To test for cis or trans effects of QPD on PLAU expression, the expressed exon 11 SNP rs4065 (T/C; T allele linked to QPD) was studied in heterozygous individuals. Unlike control platelets (T/C ratio, 1.5), QPD platelets contained an abundance of the T-allele transcript (all ratios greater than 150; P = .016; Figure 1B). In QPD CD34+ cells and saliva cells (which contained normal amounts of uPA mRNA; P = .36), the abundance of the T-allele transcript was more similar to controls (approximately 4-fold and 2-fold increases, respectively, compared with controls; P = .016; Figure 1B). As sequencing excluded mutated PLAU mRNA stability elements, these findings indicated that QPD results from a cis regulatory defect that distinctly increases transcription of the linked PLAU allele during megakaryocyte differentiation.

Genetic disorders with altered tissue-specific patterns of gene transcription appear to be rare.11  We had anticipated that QPD might result from mutations in conserved, 3′ message stability elements or in the conserved, 2.5-kb, 5′ regulatory region of PLAU that binds hematopoietic transcription factors, and other silencers and enhancers (reviewed in Diamandis et al2  and Nagamine et al12 ). As these possibilities were fully excluded, the search for the cis regulatory mutation that causes QPD must now extend beyond PLAU to identify the unique sequence changes that markedly increase transcription of the linked PLAU allele during megakaryocyte differentiation. In some disorders, the causative mutation that alters transcription is quite far (5′ and 3′) from the dysregulated gene,13,14  and it can be in an unrelated, neighboring gene.15,16  Regulatory elements with cis or trans effects have been identified as far as 1 Mb away from the genes transcribed.17-20  Interestingly, the nature of cis mutations that alter gene transcription in other blood disorders (eg, thalassemia) include deletions within locus control regions21  and point mutations that create promoter-like sequences for binding transcription factors.22 

Future identification of the mutation in the linked region of chromosome 10 that increases PLAU transcription in QPD may be helpful to develop improved QPD therapy that targets the cause of uPA overexpression rather than limiting the consequences. The information may also suggest novel ways to up-regulate PLAU expression to protect against arterial and venous thrombosis.8 

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Dr Bhupinder Bharaj and Barry Eng are gratefully acknowledged for technical help.

This work was supported by Heart and Stroke Foundation of Ontario grant 5888 and Career Investigator Award (C.P.M.H.), Bayer Canada (G.E.R.), grants from Genome Canada and Ontario Research Development Challenge Fund (D.E.B.), Ontario Graduate Student Scholarship (M.D.), Canadian Institute of Health Research/Heart and Stroke Foundation of Canada Focus on Stroke Doctoral Research Award (D.K.V.), Canada Research Chairs in Molecular Hemostasis (C.P.M.H.), and the Genetics of Complex Diseases (A.D.P.).

Contribution: M.D. recruited subjects, designed and performed experiments, interpreted results, and wrote the manuscript; A.D.P. participated in study design, data analysis, and writing of the manuscript; J.M.R., D.E.B., J.S.W., and G.E.R. contributed to study design, interpretation of results, and manuscript writing; D.K.V. and J.B. performed experimental work and contributed to manuscript writing; F.D. and G.E.R. recruited subjects; and C.P.M.H. supervised the project, designed experiments, interpreted results, and wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Catherine P. M. Hayward, McMaster University Health Sciences Center, Room 2N30, 1200 Main Street West, Hamilton, Ontario, L8N 3Z5 Canada; e-mail: haywrdc@mcmaster.ca.

1
Dahlback
 
B
Blood coagulation and its regulation by anticoagulant pathways: genetic pathogenesis of bleeding and thrombotic diseases.
J Intern Med
2005
, vol. 
257
 (pg. 
209
-
223
)
2
Diamandis
 
M
Veljkovic
 
DK
Maurer-Spurej
 
E
Rivard
 
GE
Hayward
 
CP
Quebec platelet disorder: features, pathogenesis and treatment.
Blood Coagul Fibrinolysis
2008
, vol. 
19
 (pg. 
109
-
119
)
3
Othman
 
M
Notley
 
C
Lavender
 
FL
, et al. 
Identification and functional characterization of a novel 27-bp deletion in the macroglycopeptide-coding region of the GPIBA gene resulting in platelet-type von Willebrand disease.
Blood
2005
, vol. 
105
 (pg. 
4330
-
4336
)
4
McKay
 
H
Derome
 
F
Haq
 
MA
, et al. 
Bleeding risks associated with inheritance of the Quebec platelet disorder.
Blood
2004
, vol. 
104
 (pg. 
159
-
165
)
5
Kahr
 
WH
Zheng
 
S
Sheth
 
PM
, et al. 
Platelets from patients with the Quebec platelet disorder contain and secrete abnormal amounts of urokinase-type plasminogen activator.
Blood
2001
, vol. 
98
 (pg. 
257
-
265
)
6
Diamandis
 
M
Veljkovic
 
DK
Derome
 
F
Rivard
 
GE
Hayward
 
CP
Evaluation of urokinase plasminogen activator in urine from individuals with Quebec platelet disorder.
Blood Coagul Fibrinolysis
2008
, vol. 
19
 (pg. 
463
-
464
)
7
Veljkovic
 
DK
Rivard
 
GE
Diamandis
 
M
, et al. 
Increased expression of urokinase plasminogen activator in Quebec platelet disorder is linked to megakaryocyte differentiation.
Blood
2009
, vol. 
113
 (pg. 
1535
-
1542
)
8
Kufrin
 
D
Eslin
 
DE
Bdeir
 
K
, et al. 
Antithrombotic thrombocytes: ectopic expression of urokinase-type plasminogen activator in platelets.
Blood
2003
, vol. 
102
 (pg. 
926
-
933
)
9
Kaushansky
 
K
Historical review: megakaryopoiesis and thrombopoiesis.
Blood
2008
, vol. 
111
 (pg. 
981
-
986
)
10
Gnatenko
 
DV
Dunn
 
JJ
McCorkle
 
SR
, et al. 
Transcript profiling of human platelets using microarray and serial analysis of gene expression.
Blood
2003
, vol. 
101
 (pg. 
2285
-
2293
)
11
Johnsen
 
JM
Levy
 
GG
Westrick
 
RJ
Tucker
 
PK
Ginsburg
 
D
The endothelial-specific regulatory mutation, Mvwf1, is a common mouse founder allele.
Mamm Genome
2008
, vol. 
19
 (pg. 
32
-
40
)
12
Nagamine
 
Y
Medcalf
 
RL
Munoz-Canoves
 
P
Transcriptional and posttranscriptional regulation of the plasminogen activator system.
Thromb Haemost
2005
, vol. 
93
 (pg. 
661
-
675
)
13
Cai
 
J
Goodman
 
BK
Patel
 
AS
, et al. 
Increased risk for developmental delay in Saethre-Chotzen syndrome is associated with TWIST deletions: an improved strategy for TWIST mutation screening.
Hum Genet
2003
, vol. 
114
 (pg. 
68
-
76
)
14
de Kok
 
YJ
Vossenaar
 
ER
Cremers
 
CW
, et al. 
Identification of a hot spot for microdeletions in patients with X-linked deafness type 3 (DFN3) 900 kb proximal to the DFN3 gene POU3F4.
Hum Mol Genet
1996
, vol. 
5
 (pg. 
1229
-
1235
)
15
Kleinjan
 
DA
Seawright
 
A
Schedl
 
A
, et al. 
Aniridia-associated translocations, DNase hypersensitivity, sequence comparison and transgenic analysis redefine the functional domain of PAX6.
Hum Mol Genet
2001
, vol. 
10
 (pg. 
2049
-
2059
)
16
Lettice
 
LA
Horikoshi
 
T
Heaney
 
SJ
, et al. 
Disruption of a long-range cis-acting regulator for Shh causes preaxial polydactyly.
Proc Natl Acad Sci U S A
2002
, vol. 
99
 (pg. 
7548
-
7553
)
17
Cook
 
PR
Nongenic transcription, gene regulation and action at a distance.
J Cell Sci
2003
, vol. 
116
 (pg. 
4483
-
4491
)
18
Ho
 
Y
Elefant
 
F
Liebhaber
 
SA
Cooke
 
NE
Locus control region transcription plays an active role in long-range gene activation.
Mol Cell
2006
, vol. 
23
 (pg. 
365
-
375
)
19
Nobrega
 
MA
Ovcharenko
 
I
Afzal
 
V
Rubin
 
EM
Scanning human gene deserts for long-range enhancers.
Science
2003
, vol. 
302
 pg. 
413
 
20
Pfeifer
 
D
Kist
 
R
Dewar
 
K
, et al. 
Campomelic dysplasia translocation breakpoints are scattered over 1 Mb proximal to SOX9: evidence for an extended control region.
Am J Hum Genet
1999
, vol. 
65
 (pg. 
111
-
124
)
21
Kioussis
 
D
Vanin
 
E
deLange
 
T
Flavell
 
RA
Grosveld
 
FG
Beta-globin gene inactivation by DNA translocation in gamma beta-thalassaemia.
Nature
1983
, vol. 
306
 (pg. 
662
-
666
)
22
De
 
GM
Viprakasit
 
V
Hughes
 
JR
, et al. 
A regulatory SNP causes a human genetic disease by creating a new transcriptional promoter.
Science
2006
, vol. 
312
 (pg. 
1215
-
1217
)
Sign in via your Institution