Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Neurobiol Dis ; 112: 85-90, 2018 04.
Article in English | MEDLINE | ID: mdl-29369793

ABSTRACT

Glucocerebrosidase (GCase, deficient in Gaucher disease) enzymatic activity measured in dried blood spots of Parkinson's Disease (PD) cases is within healthy range but reduced compared to controls. It is not known whether activities of additional lysosomal enzymes are reduced in dried blood spots in PD. To test whether reduction in lysosomal enzymatic activity in PD is specific to GCase, we measured GCase, acid sphingomyelinase (deficient in Niemann-Pick disease types A and B), alpha galactosidase A (deficient in Fabry), acid alpha-glucosidase (deficient in Pompe) and galactosylceramidase (deficient in Krabbe) enzymatic activities in dried blood spots of PD patients (n = 648) and controls (n = 317) recruited from Columbia University. Full sequencing of glucocerebrosidase (GBA) and the LRRK2 G2019S mutation was performed. Enzymatic activities were compared between PD cases and controls using t-test and regression models adjusted for age, gender, and GBA and LRRK2 G2019S mutation status. Alpha galactosidase A activity was lower in PD cases compared to controls both when only non-carriers were included (excluding all GBA and LRRK2 G2019S carriers and PD cases with age-at-onset below 40) [2.85 µmol/l/h versus 3.12 µmol/l/h, p = 0.018; after controlling for batch effect, p = 0.006 (468 PD cases and 296 controls)], and when including the entire cohort (2.89 µmol/l/h versus 3.10 µmol/l/h, p = 0.040; after controlling for batch effect, p = 0.011). Because the alpha galactosidase A gene is X-linked, we stratified the analyses by sex. Among women who were non-carriers of GBA and LRRK2 G2019S mutations (PD, n = 155; control, n = 194), alpha galactosidase A activity was lower in PD compared to controls (2.77 µmol/l/h versus 3.10 µmol/l/h, p = 0.044; after controlling for a batch effect, p = 0.001). The enzymatic activity of acid sphingomyelinase, acid alpha-glucosidase and galactosylceramidase was not significantly different between PD and controls. In non-carriers, most lysosomal enzyme activities were correlated, with the strongest association in GCase, acid alpha-glucosidase, and alpha galactosidase A (Pearson correlation coefficient between 0.382 and 0.532). In a regression model with all five enzymes among non-carriers (adjusted for sex and age), higher alpha galactosidase A activity was associated with lower odds of PD status (OR = 0.54; 95% CI:0.31-0.95; p = 0.032). When LRRK2 G2019S PD carriers (n = 37) were compared to non-carriers with PD, carriers had higher GCase, acid sphingomyelinase and alpha galactosidase A activity. We conclude that alpha galactosidase A may have a potential independent role in PD, in addition to GCase.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Parkinson Disease/enzymology , Parkinson Disease/genetics , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism , Aged , Cohort Studies , Enzyme Activation/physiology , Female , Humans , Male , Middle Aged , Parkinson Disease/diagnosis
2.
Neurology ; 73(4): 279-86, 2009 Jul 28.
Article in English | MEDLINE | ID: mdl-19636047

ABSTRACT

OBJECTIVE: Mutations in both alleles of parkin have been shown to result in Parkinson disease (PD). However, it is unclear whether haploinsufficiency (presence of a mutation in only 1 of the 2 parkin alleles) increases the risk for PD. METHODS: We performed comprehensive dosage and sequence analysis of all 12 exons of parkin in a sample of 520 independent patients with familial PD and 263 controls. We evaluated whether presence of a single parkin mutation, either a sequence (point mutation or small insertion/deletion) or dosage (whole exon deletion or duplication) mutation, was found at increased frequency in cases as compared with controls. We then compared the clinical characteristics of cases with 0, 1, or 2 parkin mutations. RESULTS: We identified 55 independent patients with PD with at least 1 parkin mutation and 9 controls with a single sequence mutation. Cases and controls had a similar frequency of single sequence mutations (3.1% vs 3.4%, p = 0.83); however, the cases had a significantly higher rate of dosage mutations (2.6% vs 0%, p = 0.009). Cases with a single dosage mutation were more likely to have an earlier age at onset (50% with onset at < or =45 years) compared with those with no parkin mutations (10%, p = 0.00002); this was not true for cases with only a single sequence mutation (25% with onset at < or =45 years, p = 0.06). CONCLUSIONS: Parkin haploinsufficiency, specifically for a dosage mutation rather than a point mutation or small insertion/deletion, is a risk factor for familial PD and may be associated with earlier age at onset.


Subject(s)
Gene Dosage/genetics , Genetic Predisposition to Disease/genetics , Mutation/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Ubiquitin-Protein Ligases/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Base Sequence/genetics , DNA Mutational Analysis , Female , Gene Deletion , Gene Frequency/genetics , Genetic Markers/genetics , Genetic Testing , Genotype , Haplotypes/genetics , Humans , Male , Middle Aged , Parkinson Disease/physiopathology , Point Mutation/genetics , Risk Factors , Young Adult
3.
Neurology ; 72(22): 1886-92, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19279319

ABSTRACT

OBJECTIVE: A recent study reported that mutations in a gene on chromosome 2q36-37, GIGYF2, result in Parkinson disease (PD). We have previously reported linkage to this chromosomal region in a sample of multiplex PD families, with the strongest evidence of linkage obtained using the subset of the sample having the strongest family history of disease and meeting the strictest diagnostic criteria. We have tested whether mutations in GIGYF2 may account for the previously observed linkage finding. METHODS: We sequenced the GIGYF2 coding region in 96 unrelated patients with PD used in our original study that contributed to the chromosome 2q36-37 linkage signal. Subsequently, we genotyped the entire sample of 566 multiplex PD kindreds as well as 1,447 controls to test whether variants in GIGYF2 are causative or increase susceptibility for PD. RESULTS: We detected three novel variants as well as one of the previously reported seven variants in a total of five multiple PD families; however, there was no consistent evidence that these variants segregated with PD in these families. We also did not find a significant increase in risk for PD among those inheriting variants in GIGYF2 (p = 0.28). CONCLUSIONS: We believe that variation in a gene other than GIGYF2 accounts for the previously reported linkage finding on chromosome 2q36-37.


Subject(s)
Carrier Proteins/genetics , Genetic Linkage/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Mutation/genetics , Parkinson Disease/genetics , Aged , Base Sequence/genetics , Chromosome Mapping/methods , Chromosomes, Human, Pair 2/genetics , DNA Mutational Analysis , Female , Genetic Testing , Genotype , Humans , Male , Middle Aged , Open Reading Frames/genetics , Pedigree , Polymorphism, Single Nucleotide/genetics
4.
Neurology ; 72(4): 310-6, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-18987351

ABSTRACT

OBJECTIVE: To characterize sequence variation within the glucocerebrosidase (GBA) gene in a select subset of our sample of patients with familial Parkinson disease (PD) and then to test in our full sample whether these sequence variants increased the risk for PD and were associated with an earlier onset of disease. METHODS: We performed a comprehensive study of all GBA exons in one patient with PD from each of 96 PD families, selected based on the family-specific lod scores at the GBA locus. Identified GBA variants were subsequently screened in all 1325 PD cases from 566 multiplex PD families and in 359 controls. RESULTS: Nine different GBA variants, five previously reported, were identified in 21 of the 96 PD cases sequenced. Screening for these variants in the full sample identified 161 variant carriers (12.2%) in 99 different PD families. An unbiased estimate of the frequency of the five previously reported GBA variants in the familial PD sample was 12.6% and in the control sample was 5.3% (odds ratio 2.6; 95% confidence interval 1.5-4.4). Presence of a GBA variant was associated with an earlier age at onset (p = 0.0001). On average, those patients carrying a GBA variant had onset with PD 6.04 years earlier than those without a GBA variant. CONCLUSIONS: This study suggests that GBA is a susceptibility gene for familial Parkinson disease (PD) and patients with GBA variants have an earlier age at onset than patients with PD without GBA variants.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Glucosylceramidase/genetics , Mutation/genetics , Parkinson Disease/epidemiology , Parkinson Disease/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Female , Genetic Variation/genetics , Humans , Male , Middle Aged , Parkinson Disease/enzymology
5.
Neurology ; 70(16 Pt 2): 1456-60, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18337586

ABSTRACT

OBJECTIVE: Leucine-rich repeat kinase 2 (LRRK2) mutations are the most common cause of Parkinson disease (PD). Several dominantly inherited pathogenic substitutions have been identified in different domains of the Lrrk2 protein. Herein, we characterize the clinical and genetic features associated with Lrrk2 p.R1441C. METHODS: We identified 33 affected and 15 unaffected LRRK2 c.4321C>T (p.R1441C) mutation carriers through an international consortium originating from three continents. The age-specific cumulative incidence of PD was calculated by Kaplan-Meier analysis. RESULTS: The clinical presentation of Lrrk2 p.R1441C carriers was similar to sporadic PD and Lrrk2 p.G2019S parkinsonism. The mean age at onset for parkinsonism was 60 years, range 30-79 years; fewer than 20% of the patients had symptoms before the age 50 years, while by 75 years >90% of them had developed symptoms. Haplotype analysis suggests four independent founders for the p.R1441C mutation. CONCLUSIONS: The distribution in age at onset and clinical features in Lrrk2 p.R1441C patients are similar to idiopathic and Lrrk2 p.G2019S parkinsonism. Several independent founders of the p.R1441C substitution suggest this site is prone to recurrent mutagenesis.


Subject(s)
Amino Acid Substitution/genetics , Parkinsonian Disorders/genetics , Parkinsonian Disorders/physiopathology , Protein Serine-Threonine Kinases/genetics , Adult , Aged , Aged, 80 and over , Arginine/genetics , Cysteine/genetics , DNA Mutational Analysis , Female , Glycine/genetics , Haplotypes/genetics , Humans , Internationality , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Middle Aged , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Serine/genetics
6.
Neurology ; 69(18): 1737-44, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17804834

ABSTRACT

BACKGROUND: Pathogenic mutations in the leucine-rich repeat kinase 2 gene (LRRK2) have been found to cause typical, later-onset Parkinson disease (PD). Although G2019S is the most common mutation, other mutations have also been reported. It is critical to catalog the types of mutations found in LRRK2 that can cause PD, so as to provide insight regarding disease susceptibility and potential novel treatments. METHODS: We performed a comprehensive study of all 51 exons of the LRRK2 gene in one PD patient from each of 88 multiplex PD families who had the highest family-specific multipoint lod score at the LRRK2 locus from a cohort of 430 PD families without the G2019S mutation. RESULTS: Five families (5.7%) harbored what seem to be novel, pathogenic mutations (L1795F, I1192V, E10K, E334K, Q1111H). Three of these apparent mutations were in known, functional domains of the LRRK2 protein, where other studies have also identified disease producing mutations. However, two of the novel variants were found in the N-terminal region of LRRK2, where no pathogenic substitutions have yet been reported. Similar to previous studies, all subjects with an LRRK2 mutation had classic symptoms of PD and typical, later age at onset. CONCLUSIONS: We have identified five novel variants in LRRK2, with two of these in the N-terminal region of LRRK2, where no pathogenic substitutions have been previously reported. If confirmed to be causative, these mutations would broaden the potential mechanisms whereby mutations in LRRK2 result in Parkinson disease.


Subject(s)
DNA Mutational Analysis , Genetic Linkage , Parkinson Disease/genetics , Point Mutation , Protein Serine-Threonine Kinases/genetics , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Exons , Female , Genetic Predisposition to Disease , Genetic Testing , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Lod Score , Male , Microsatellite Repeats , Middle Aged , Parkinson Disease/physiopathology , Pedigree , Polymorphism, Genetic
7.
Neurology ; 65(12): 1936-40, 2005 Dec 27.
Article in English | MEDLINE | ID: mdl-16380616

ABSTRACT

BACKGROUND: Spheroid body myopathy (SBM) is a rare, autosomal dominant, neuromuscular disorder, which has only been previously reported in a single large kindred. Identification of the mutated gene in this disorder may provide insight regarding abnormal neuromuscular function. METHODS: The authors completed a detailed clinical evaluation on an extensive kindred diagnosed with SBM. Genome-wide linkage analysis was performed to localize the disease gene to a specific chromosomal region. Further marker genotyping and screening of a positional, functional candidate gene were completed to detect the disease-causing mutation. Pathologic analysis of muscle biopsy was performed on three individuals. Biochemical studies were performed on one muscle biopsy specimen from an affected individual. RESULTS: Linkage to chromosome 5q23-5q31 was detected with a lod score of 2.9. Genotyping of additional markers in a larger sample of family members produced a maximum lod score of 6.1 and narrowed the critical interval to 12.2 cM. Screening of the candidate gene titin immunoglobulin domain protein (TTID, also known as MYOT) detected a cytosine-to-thymine mutation in exon 2 of all clinically affected family members. Similar pathologic changes were present in all muscle biopsy specimens. Immunohistologic and biochemical analysis revealed that the TTID protein, also known as myotilin, is a component of the insoluble protein aggregate. CONCLUSIONS: A novel mutation in the TTID gene results in the clinical and pathologic phenotype termed "spheroid body myopathy." Mutations in this gene also cause limb-girdle muscular dystrophy 1A and are associated with myofibrillar myopathy.


Subject(s)
Cytoskeletal Proteins/genetics , Genetic Predisposition to Disease/genetics , Inclusion Bodies/genetics , Muscle Proteins/genetics , Muscle, Skeletal/physiopathology , Muscular Diseases/genetics , Mutation/genetics , Adult , Aged , Aged, 80 and over , Chromosome Disorders/genetics , Chromosome Mapping , Chromosomes, Human, Pair 5/genetics , Connectin , DNA Mutational Analysis , Exons/genetics , Female , Genes, Dominant/genetics , Genetic Markers/genetics , Genetic Testing , Humans , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Male , Microfilament Proteins , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Pedigree , Point Mutation/genetics
9.
Neurology ; 62(9): 1616-8, 2004 May 11.
Article in English | MEDLINE | ID: mdl-15136695

ABSTRACT

A genome screen to identify genes influencing the age at Parkinson disease (PD) onset was completed using 276 families without parkin mutations. Significant evidence of linkage to chromosome 2p near the PARK3 locus (logarithm of odds [lod] = 4.8) was observed. Evidence of linkage was also detected to chromosomes 1q (lod = 3.0) and 8q (lod = 2.6). These data suggest that the genes influencing age at PD onset likely differ from those that contribute to PD susceptibility.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 2/genetics , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/genetics , Adolescent , Adult , Age of Onset , Aged , Chromosome Mapping , Family , Female , Genotype , Germany/ethnology , Humans , Lod Score , Male , Middle Aged , Mutation , Parkinson Disease/ethnology , United States/ethnology , White People/genetics
10.
Neurology ; 60(5): 796-801, 2003 Mar 11.
Article in English | MEDLINE | ID: mdl-12629236

ABSTRACT

BACKGROUND: The vast majority of the parkin mutations previously identified have been found in individuals with juvenile or early onset PD. Previous screening of later onset PD cohorts has not identified substantial numbers of parkin mutations. METHODS: Families with at least two siblings with PD were ascertained to identify genes contributing to PD susceptibility. Screening of the parkin gene, by both quantitative PCR and exon sequencing, was performed in those families with either early onset PD (age onset < or =50 years) or positive lod score with a marker in intron 7 of the parkin gene. RESULTS: A total of 25 different mutations in the parkin gene were identified in 103 individuals from 47 families. Mutations were found in both parkin alleles in 41 of the individuals, whereas a single mutation in only one of the two parkin alleles was observed in 62 individuals. Thirty-five of the subjects (34%) with a parkin mutation had an age at onset of 60 years or above with 30 of these 35 (86%) having a detectable mutation on only one parkin allele. Few significant clinical differences were observed among the individuals with two, one, or no mutated copies of the parkin gene. CONCLUSION: Mutations in the parkin gene occur among individuals with PD with an older age at onset (> or =60 years) who have a positive family history of the disease. In addition, the clinical findings of parkin-positive individuals are remarkably similar to those without mutations.


Subject(s)
Mutation , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/genetics , Age of Onset , Aged , Genetic Testing , Heterozygote , Humans , Middle Aged , North America/epidemiology , Parkinson Disease/epidemiology
12.
Nature ; 413(6855): 488-94, 2001 Oct 04.
Article in English | MEDLINE | ID: mdl-11586351

ABSTRACT

Thrombotic thrombocytopenic purpura (TTP) is a life-threatening systemic illness of abrupt onset and unknown cause. Proteolysis of the blood-clotting protein von Willebrand factor (VWF) observed in normal plasma is decreased in TTP patients. However, the identity of the responsible protease and its role in the pathophysiology of TTP remain unknown. We performed genome-wide linkage analysis in four pedigrees of humans with congenital TTP and mapped the responsible genetic locus to chromosome 9q34. A predicted gene in the identified interval corresponds to a segment of a much larger transcript, identifying a new member of the ADAMTS family of zinc metalloproteinase genes (ADAMTS13). Analysis of patients' genomic DNA identified 12 mutations in the ADAMTS13 gene, accounting for 14 of the 15 disease alleles studied. We show that deficiency of ADAMTS13 is the molecular mechanism responsible for TTP, and suggest that physiologic proteolysis of VWF and/or other ADAMTS13 substrates is required for normal vascular homeostasis.


Subject(s)
Metalloendopeptidases/genetics , Mutation , Purpura, Thrombotic Thrombocytopenic/genetics , von Willebrand Factor/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 9 , DNA Mutational Analysis , Female , Humans , Male , Metalloendopeptidases/blood , Metalloendopeptidases/physiology , Molecular Sequence Data , Multigene Family , Pedigree , Physical Chromosome Mapping , Purpura, Thrombotic Thrombocytopenic/congenital , Purpura, Thrombotic Thrombocytopenic/enzymology
13.
N Engl J Med ; 345(5): 325-34, 2001 Aug 02.
Article in English | MEDLINE | ID: mdl-11484689

ABSTRACT

BACKGROUND: Most patients with familial primary pulmonary hypertension have defects in the gene for bone morphogenetic protein receptor II (BMPR2), a member of the transforming growth factor beta (TGF-beta) superfamily of receptors. Because patients with hereditary hemorrhagic telangiectasia may have lung disease that is indistinguishable from primary pulmonary hypertension, we investigated the genetic basis of lung disease in these patients. METHODS: We evaluated members of five kindreds plus one individual patient with hereditary hemorrhagic telangiectasia and identified 10 cases of pulmonary hypertension. In the two largest families, we used microsatellite markers to test for linkage to genes encoding TGF-beta-receptor proteins, including endoglin and activin-receptor-like kinase 1 (ALK1), and BMPR2. In subjects with hereditary hemorrhagic telangiectasia and pulmonary hypertension, we also scanned ALK1 and BMPR2 for mutations. RESULTS: We identified suggestive linkage of pulmonary hypertension with hereditary hemorrhagic telangiectasia on chromosome 12q13, a region that includes ALK1. We identified amino acid changes in activin-receptor-like kinase 1 that were inherited in subjects who had a disorder with clinical and histologic features indistinguishable from those of primary pulmonary hypertension. Immunohistochemical analysis in four subjects and one control showed pulmonary vascular endothelial expression of activin-receptor-like kinase 1 in normal and diseased pulmonary arteries. CONCLUSIONS: Pulmonary hypertension in association with hereditary hemorrhagic telangiectasia can involve mutations in ALK1. These mutations are associated with diverse effects, including the vascular dilatation characteristic of hereditary hemorrhagic telangiectasia and the occlusion of small pulmonary arteries that is typical of primary pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Activin Receptors , Adult , Bone Morphogenetic Protein Receptors, Type II , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 12 , Female , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/pathology , Lung/pathology , Male , Microsatellite Repeats , Middle Aged , Mutation, Missense , Pedigree , Signal Transduction , Telangiectasia, Hereditary Hemorrhagic/complications , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
14.
Am J Hum Genet ; 68(1): 92-102, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11115378

ABSTRACT

Primary pulmonary hypertension (PPH) is a potentially lethal disorder, because the elevation of the pulmonary arterial pressure may result in right-heart failure. Histologically, the disorder is characterized by proliferation of pulmonary-artery smooth muscle and endothelial cells, by intimal hyperplasia, and by in situ thrombus formation. Heterozygous mutations within the bone morphogenetic protein type II receptor (BMPR-II) gene (BMPR2), of the transforming growth factor beta (TGF-beta) cell-signaling superfamily, have been identified in familial and sporadic cases of PPH. We report the molecular spectrum of BMPR2 mutations in 47 additional families with PPH and in three patients with sporadic PPH. Among the cohort of patients, we have identified 22 novel mutations, including 4 partial deletions, distributed throughout the BMPR2 gene. The majority (58%) of mutations are predicted to lead to a premature termination codon. We have also investigated the functional impact and genotype-phenotype relationships, to elucidate the mechanisms contributing to pathogenesis of this important vascular disease. In vitro expression analysis demonstrated loss of BMPR-II function for a number of the identified mutations. These data support the suggestion that haploinsufficiency represents the common molecular mechanism in PPH. Marked variability of the age at onset of disease was observed both within and between families. Taken together, these studies illustrate the considerable heterogeneity of BMPR2 mutations that cause PPH, and they strongly suggest that additional factors, genetic and/or environmental, may be required for the development of the clinical phenotype.


Subject(s)
Genes, Dominant/genetics , Hypertension, Pulmonary/genetics , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Adolescent , Adult , Age of Onset , Base Sequence , Bone Morphogenetic Protein Receptors, Type II , Cells, Cultured , Child , Child, Preschool , Codon, Terminator/genetics , DNA Mutational Analysis , Exons/genetics , Female , Fluorescence , Gene Dosage , Haplotypes/genetics , Humans , Hypertension, Pulmonary/epidemiology , Infant , Introns/genetics , Male , Middle Aged , Models, Genetic , Pedigree , Polymorphism, Genetic/genetics , RNA Splice Sites/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Sequence Deletion/genetics
15.
J Med Genet ; 37(10): 741-5, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11015450

ABSTRACT

BACKGROUND: Primary pulmonary hypertension (PPH), resulting from occlusion of small pulmonary arteries, is a devastating condition. Mutations of the bone morphogenetic protein receptor type II gene (BMPR2), a component of the transforming growth factor beta (TGF-beta) family which plays a key role in cell growth, have recently been identified as causing familial PPH. We have searched for BMPR2 gene mutations in sporadic PPH patients to determine whether the same genetic defect underlies the more common form of the disorder. METHODS: We investigated 50 unrelated patients, with a clinical diagnosis of PPH and no identifiable family history of pulmonary hypertension, by direct sequencing of the entire coding region and intron/exon boundaries of the BMPR2 gene. DNA from available parent pairs (n=5) was used to assess the occurrence of spontaneous (de novo) mutations contributing to sporadic PPH. RESULTS: We found a total of 11 different heterozygous germline mutations of the BMPR2 gene in 13 of the 50 PPH patients studied, including missense (n=3), nonsense (n=3), and frameshift (n=5) mutations each predicted to alter the cell signalling response to specific ligands. Parental analysis showed three occurrences of paternal transmission and two of de novo mutation of the BMPR2 gene in sporadic PPH. CONCLUSION: The sporadic form of PPH is associated with germline mutations of the gene encoding the receptor protein BMPR-II in at least 26% of cases. A molecular classification of PPH, based upon the presence or absence of BMPR2 mutations, has important implications for patient management and screening of relatives.


Subject(s)
Germ-Line Mutation/genetics , Hypertension, Pulmonary/genetics , Multigene Family , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/chemistry , Adolescent , Adult , Age of Onset , Bone Morphogenetic Protein Receptors, Type II , Child , Codon/genetics , DNA Mutational Analysis , Exons/genetics , Female , Genetic Testing , Heterozygote , Humans , Hypertension, Pulmonary/epidemiology , Introns/genetics , Male , Middle Aged , Pedigree , Protein Serine-Threonine Kinases/physiology , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction
16.
Nat Genet ; 26(1): 81-4, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973254

ABSTRACT

Primary pulmonary hypertension (PPH), characterized by obstruction of pre-capillary pulmonary arteries, leads to sustained elevation of pulmonary arterial pressure (mean >25 mm Hg at rest or >30 mm Hg during exercise). The aetiology is unknown, but the histological features reveal proliferation of endothelial and smooth muscle cells with vascular remodelling (Fig. 1). More than one affected relative has been identified in at least 6% of cases (familial PPH, MIM 178600). Familial PPH (FPPH) segregates as an autosomal dominant disorder with reduced penetrance and has been mapped to a locus designated PPH1 on 2q33, with no evidence of heterogeneity. We now show that FPPH is caused by mutations in BMPR2, encoding a TGF-beta type II receptor (BMPR-II). Members of the TGF-beta superfamily transduce signals by binding to heteromeric complexes of type I and II receptors, which activates serine/threonine kinases, leading to transcriptional regulation by phosphorylated Smads. By comparison with in vitro studies, identified defects of BMPR-II in FPPH are predicted to disrupt ligand binding, kinase activity and heteromeric dimer formation. Our data demonstrate the molecular basis of FPPH and underscore the importance in vivo of the TGF-beta signalling pathway in the maintenance of blood vessel integrity.


Subject(s)
Germ-Line Mutation , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Protein Serine-Threonine Kinases/genetics , Amino Acid Sequence , Bone Morphogenetic Protein Receptors, Type II , Chromosomes, Human, Pair 2/genetics , Cloning, Molecular , DNA, Complementary/metabolism , Endothelium, Vascular/metabolism , Exons , Family Health , Female , Genes, Dominant , Genetic Linkage , Genetic Markers , Humans , Hypertension, Pulmonary/diagnostic imaging , Introns , Ligands , Lung/blood supply , Lung/diagnostic imaging , Male , Molecular Sequence Data , Muscle, Smooth/metabolism , Pedigree , Protein Isoforms , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Radiography , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/chemistry , Receptors, Transforming Growth Factor beta/genetics , Recombination, Genetic , Restriction Mapping , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Signal Transduction/genetics
17.
Genomics ; 68(2): 220-8, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10964520

ABSTRACT

Primary pulmonary hypertension (PPH), an often fatal disorder, is characterized by sustained elevation of pulmonary artery pressure of unknown cause. In its familial form (FPPH), the disorder segregates as an autosomal dominant and displays markedly reduced penetrance. A gene for FPPH was previously localized to a 25-cM interval on the long arm of chromosome 2 (2q31-q33). We now report a complete yeast artificial chromosome (YAC) and bacterial artificial chromosome (BAC)/P1 artificial chromosome contig (PAC), assembled by STS content mapping, across a newly identified minimum nonrecombinant interval containing the gene designated PPH1. The physical map has served to establish polymorphic marker order unequivocally, enabling the establishment of detailed haplotypes for the region. Together with the identification of novel recombination events in affected individuals from six newly ascertained kindreds, these data have allowed the significant reduction of the minimum PPH1 critical interval to a 4.8-cM region. The region, flanked by the polymorphic markers D2S115 (centromeric) and D2S1384 (telomeric), corresponds to a minimum physical distance of 5.8 Mb at 2q33. Numerous expressed sequence tags and known genes were placed on the YAC/BAC contig spanning the PPH1 gene critical region.


Subject(s)
Genetic Predisposition to Disease/genetics , Hypertension, Pulmonary/genetics , Physical Chromosome Mapping , Bacteriophage P1/genetics , Chromosomes, Artificial, Yeast/genetics , Chromosomes, Bacterial/genetics , Chromosomes, Human, Pair 2/genetics , Contig Mapping , DNA/genetics , Expressed Sequence Tags , Family Health , Female , Genetic Linkage , Genotype , Haplotypes , Humans , Male , Microsatellite Repeats , Pedigree , Sequence Tagged Sites , Transcription, Genetic
18.
J Biol Chem ; 274(46): 32539-42, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10551804

ABSTRACT

The endoplasmic reticulum-Golgi intermediate compartment (ERGIC) is the site of segregation of secretory proteins for anterograde transport, via packaging into COPII-coated transport vesicles. ERGIC-53 is a homo-hexameric transmembrane lectin localized to the ERGIC that exhibits mannose-selective properties in vitro. Null mutations in ERGIC-53 were recently shown to be responsible for the autosomal recessive bleeding disorder, combined deficiency of coagulation factors V and VIII. We have studied the effect of defective ER to Golgi cycling by ERGIC-53 on the secretion of factors V and VIII. The secretion efficiency of factor V and factor VIII was studied in a tetracycline-inducible HeLa cell line overexpressing a wild-type ERGIC-53 or a cytosolic tail mutant of ERGIC-53 (KKAA) that is unable to exit the ER due to mutation of two COOH-terminal phenylalanine residues to alanines. The results show that efficient trafficking of factors V and VIII requires a functional ERGIC-53 cycling pathway and that this trafficking is dependent on post-translational modification of a specific cluster of asparagine (N)-linked oligosaccharides to a fully glucose-trimmed, mannose9 structure.


Subject(s)
Endoplasmic Reticulum/metabolism , Factor VIII/metabolism , Factor V/metabolism , Golgi Apparatus/genetics , Mannose-Binding Lectins , Mannose/metabolism , Membrane Proteins/metabolism , Biological Transport , Glycosylation , Golgi Apparatus/chemistry , HeLa Cells , Humans , Protein Conformation , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
19.
Int J Clin Lab Res ; 29(1): 1-7, 1999.
Article in English | MEDLINE | ID: mdl-10356656

ABSTRACT

von Willebrand disease (VWD) is a clinically heterogeneous bleeding disorder that reflects a wide array of defects. Quantitative subtypes of the disorder, including types 1 and 3 VWD, result in bleeding due to reduced levels of circulating von Willebrand factor (VWF) protein. Qualitative subtypes, defined as type 2 VWD, act through altered VWF function. A range of molecular defects are responsible for many of these subtypes, including missense, nonsense, splicing, insertion, and deletion mutations, resulting in either dominant or recessive inheritance. While many mutations correspond to selected variants, the basis for variation in expression and the imperfect correlations between genotype and phenotype remain to be understood.


Subject(s)
Frameshift Mutation , Mutation, Missense , von Willebrand Diseases/genetics , von Willebrand Factor/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...