531 results on '"Trembath, Richard C."'
Search Results
502. BMPR2 mutations have short lifetime expectancy in primary pulmonary hypertension.
- Author
-
Sankelo M, Flanagan JA, Machado R, Harrison R, Rudarakanchana N, Morrell N, Dixon M, Halme M, Puolijoki H, Kere J, Elomaa O, Kupari M, Räisänen-Sokolowski A, Trembath RC, and Laitinen T
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Family Health, Female, Heterozygote, Humans, Male, Middle Aged, Mutation, Pedigree, Signal Transduction, Bone Morphogenetic Protein Receptors, Type II genetics, Genetic Predisposition to Disease, Hypertension, Pulmonary genetics, Hypertension, Pulmonary mortality, Longevity
- Abstract
In a nationwide study, we identified a total of 59 patients diagnosed with primary pulmonary hypertension (PPH) in Finland between the years 1987 and 1999. These data support a minimum estimate for a PPH population prevalence of 5.8 cases/million with an incidence of 0.2-1.3 cases/million/year. The male-to-female ratio among the patients was 1:4, while 7% (4/59) of the PPH probands had a known family history of the disorder. Familial or sporadic PPH showed no geographic clustering to any region of Finland. Sequencing of the coding regions and exon-intron boundaries of the bone morphogenetic protein receptor type 2 (BMPR2) identified heterozygous BMPR2 mutations in 12% (3/26) of the sporadic and 33% (1/3) of the familial patients. All four mutations were different, and two of those have been previously reported in other populations. Pathogenic defects in BMPR2 include a novel missense mutation (c.2696G>C encoding R899P), located within the receptor intracellular cytoplasmic domain whose function has been poorly characterized. Our analysis demonstrates that this mutant, while localizing to the cell surface, does not impact on SMAD-mediated (mothers against decapentaplegic homolog) intracellular signaling, but leads to constitutive activation of the p38(MAPK) pathway. The absence of a founder mutation in a genetically homogeneous population, such as the Finns, suggests that all identified BMPR2 mutations have to be rather young while the ancestral (if any) mutations have been lost either due to repetitive genetic bottlenecks or due to significant negative selection. Hum Mutat 26(2), 1-6, 2005. (c) 2005 Wiley-Liss, Inc.
- Published
- 2005
- Full Text
- View/download PDF
503. Dysfunctional Smad signaling contributes to abnormal smooth muscle cell proliferation in familial pulmonary arterial hypertension.
- Author
-
Yang X, Long L, Southwood M, Rudarakanchana N, Upton PD, Jeffery TK, Atkinson C, Chen H, Trembath RC, and Morrell NW
- Subjects
- Apoptosis drug effects, Bone Morphogenetic Protein 4, Bone Morphogenetic Protein Receptors, Type II, Bone Morphogenetic Proteins pharmacology, Cell Proliferation drug effects, Cell Survival drug effects, Cells, Cultured, Humans, Mitogen-Activated Protein Kinase 1 physiology, Mitogen-Activated Protein Kinase 3 physiology, Mutation, Protein Serine-Threonine Kinases genetics, Pulmonary Artery drug effects, Smad Proteins, Smad1 Protein, p38 Mitogen-Activated Protein Kinases physiology, DNA-Binding Proteins physiology, Hypertension, Pulmonary genetics, Hypertension, Pulmonary pathology, Muscle, Smooth, Vascular pathology, Pulmonary Artery pathology, Signal Transduction physiology, Trans-Activators physiology
- Abstract
Mutations in the bone morphogenetic protein type II receptor gene (BMPR2) are the major genetic cause of familial pulmonary arterial hypertension (FPAH). Although smooth muscle cell proliferation contributes to the vascular remodeling observed in PAH, the role of BMPs in this process and the impact of BMPR2 mutation remains unclear. Studies involving normal human pulmonary artery smooth muscle cells (PASMCs) suggest site-specific responses to BMPs. Thus, BMP-4 inhibited proliferation of PASMCs isolated from proximal pulmonary arteries, but stimulated proliferation of PASMCs from peripheral arteries, and conferred protection from apoptosis. These differences were not caused by differential activation of BMP signaling pathways because exogenous BMP-4 led to phosphorylation of Smad1, p38(MAPK), and ERK1/2 in both cell types. However, the proproliferative effect of BMP-4 on peripheral PASMCs was found to be p38MAPK/ERK-dependent. Conversely, overexpression of dominant-negative Smad1 converted the response to BMP-4 in proximal PASMCs from inhibitory to proliferative. Furthermore, we confirmed that proximal PASMCs harboring kinase domain mutations in BMPR2 are deficient in Smad signaling and are unresponsive to the growth suppressive effect of BMP-4. Moreover, we show that the pulmonary vasculature of patients with familial and idiopathic PAH are deficient in the activated form of Smad1. We conclude that defective Smad signaling and unopposed p38(MAPK)/ERK signaling, as a consequence of mutation in BMPR2, underlie the abnormal vascular cell proliferation observed in familial PAH.
- Published
- 2005
- Full Text
- View/download PDF
504. Genetic association analysis using data from triads and unrelated subjects.
- Author
-
Epstein MP, Veal CD, Trembath RC, Barker JN, Li C, and Satten GA
- Subjects
- Algorithms, Gene Frequency, Genetic Linkage, Humans, Likelihood Functions, Parents, Polymorphism, Single Nucleotide, Psoriasis genetics, Case-Control Studies, Models, Genetic, Models, Statistical, Research Design
- Abstract
The selection of an appropriate control sample for use in association mapping requires serious deliberation. Unrelated controls are generally easy to collect, but the resulting analyses are susceptible to spurious association arising from population stratification. Parental controls are popular, since triads comprising a case and two parents can be used in analyses that are robust to this stratification. However, parental controls are often expensive and difficult to collect. In some situations, studies may have both parental and unrelated controls available for analysis. For example, a candidate-gene study may analyze triads but may have an additional sample of unrelated controls for examination of background linkage disequilibrium in genomic regions. Also, studies may collect a sample of triads to confirm results initially found using a traditional case-control study. Initial association studies also may collect each type of control, to provide insurance against the weaknesses of the other type. In these situations, resulting samples will consist of some triads, some unrelated controls, and, possibly, some unrelated cases. Rather than analyze the triads and unrelated subjects separately, we present a likelihood-based approach for combining their information in a single combined association analysis. Our approach allows for joint analysis of data from both triad and case-control study designs. Simulations indicate that our proposed approach is more powerful than association tests that are based on each separate sample. Our approach also allows for flexible modeling and estimation of allele effects, as well as for missing parental data. We illustrate the usefulness of our approach using SNP data from a candidate-gene study of psoriasis.
- Published
- 2005
- Full Text
- View/download PDF
505. Sequence changes in predicted promoter elements of STK11/LKB1 are unlikely to contribute to Peutz-Jeghers syndrome.
- Author
-
Hearle NC, Tomlinson I, Lim W, Murday V, Swarbrick E, Lim G, Phillips R, Lee P, O'Donohue J, Trembath RC, Morrison PJ, Norman A, Taylor R, Hodgson S, Lucassen A, and Houlston RS
- Subjects
- AMP-Activated Protein Kinase Kinases, Adolescent, Base Sequence, Binding Sites, Computational Biology methods, Conserved Sequence, DNA genetics, DNA Mutational Analysis, Gene Deletion, Germ-Line Mutation, Humans, Models, Genetic, Molecular Sequence Data, Mutation, Phylogeny, Protein Binding, Sequence Analysis, DNA, Peutz-Jeghers Syndrome genetics, Promoter Regions, Genetic, Protein Serine-Threonine Kinases genetics
- Abstract
Background: Germline mutations or large-scale deletions in the coding region and splice sites of STK11/LKB1 do not account for all cases of Peutz-Jeghers syndrome (PJS). It is conceivable that, on the basis of data from other diseases, inherited variation in promoter elements of STK11/LKB1 may cause PJS., Results: Phylogenetic foot printing and transcription factor binding site prediction of sequence 5' to the coding sequence of STK11/LKB1 was performed to identify non-coding sequences of DNA indicative of regulatory elements. A series of 33 PJS cases in whom no mutation in STK11/LKB1 could be identified were screened for sequence changes in the putative promoter defined by nucleotides -1090 to -1472. Two novel sequence changes were identified, but were found to be present in healthy individuals., Conclusion: These findings indicate that promoter sequence changes are unlikely to contribute to PJS.
- Published
- 2005
- Full Text
- View/download PDF
506. Mutations of the catalytic subunit of RAB3GAP cause Warburg Micro syndrome.
- Author
-
Aligianis IA, Johnson CA, Gissen P, Chen D, Hampshire D, Hoffmann K, Maina EN, Morgan NV, Tee L, Morton J, Ainsworth JR, Horn D, Rosser E, Cole TR, Stolte-Dijkstra I, Fieggen K, Clayton-Smith J, Mégarbané A, Shield JP, Newbury-Ecob R, Dobyns WB, Graham JM Jr, Kjaer KW, Warburg M, Bond J, Trembath RC, Harris LW, Takai Y, Mundlos S, Tannahill D, Woods CG, and Maher ER
- Subjects
- Catalytic Domain, Central Nervous System abnormalities, Eye Abnormalities pathology, Genitalia abnormalities, Humans, Molecular Sequence Data, Syndrome, rab GTP-Binding Proteins genetics, Mutation, rab GTP-Binding Proteins metabolism
- Abstract
Warburg Micro syndrome (WARBM1) is a severe autosomal recessive disorder characterized by developmental abnormalities of the eye and central nervous system and by microgenitalia. We identified homozygous inactivating mutations in RAB3GAP, encoding RAB3 GTPase activating protein, a key regulator of the Rab3 pathway implicated in exocytic release of neurotransmitters and hormones, in 12 families with Micro syndrome. We hypothesize that the underlying pathogenesis of Micro syndrome is a failure of exocytic release of ocular and neurodevelopmental trophic factors.
- Published
- 2005
- Full Text
- View/download PDF
507. Investigation of second genetic hits at the BMPR2 locus as a modulator of disease progression in familial pulmonary arterial hypertension.
- Author
-
Machado RD, James V, Southwood M, Harrison RE, Atkinson C, Stewart S, Morrell NW, Trembath RC, and Aldred MA
- Subjects
- Adult, Alleles, Bone Morphogenetic Protein Receptors, Type II, DNA Mutational Analysis, Disease Progression, Germ-Line Mutation, Humans, Hypertension, Pulmonary pathology, Loss of Heterozygosity, Lung surgery, Microdissection, Microsatellite Repeats, Mutation, Missense, Genetic Predisposition to Disease, Hypertension, Pulmonary genetics, Mutation, Protein Serine-Threonine Kinases genetics
- Abstract
Background: Primary pulmonary arterial hypertension (PAH) is a potentially devastating condition resulting from occlusion of the pulmonary arterioles by the formation of vascular lesions. Heterozygous mutations in the gene encoding the bone morphogenetic protein receptor type II (BMPR2) have been identified in both familial (FPAH) and idiopathic PAH. Mutant alleles are typically of low penetrance, indicating that other factors are required for the onset of PAH. Previous reports have suggested that the characteristic plexiform lesions in affected lungs are akin to neoplasia, showing monoclonal expansion and microsatellite instability. We hypothesized that in patients with germline mutations, BMPR2 might behave as a classic tumor suppressor gene, with somatic loss of the wild-type allele contributing to disease progression., Methods and Results: To test this hypothesis, plexiform and concentric vascular lesions were serially microdissected from lung explant tissue derived from 7 FPAH cases. DNA was analyzed for loss of heterozygosity at BMPR2 and for microsatellite instability (MSI) at 5 loci. MSI was detected in 1 of 37 lesions at a single locus, BAT-26, whereas heterozygosity at BMPR2 was retained at all informative loci. We also describe a FPAH patient carrying biallelic constitutional missense mutations of BMPR2 who manifested disease at a stage and manner similar to heterozygous patients., Conclusions: Taken together, these data demonstrate that MSI is uncommon in FPAH and suggest that somatic loss of the remaining wild-type BMPR2 allele in heterozygous mutation carriers likely does not play a significant role in modulating the onset or progression of FPAH.
- Published
- 2005
- Full Text
- View/download PDF
508. Transforming growth factor-beta receptor mutations and pulmonary arterial hypertension in childhood.
- Author
-
Harrison RE, Berger R, Haworth SG, Tulloh R, Mache CJ, Morrell NW, Aldred MA, and Trembath RC
- Subjects
- Activin Receptors, Type I physiology, Activin Receptors, Type II, Amino Acid Motifs genetics, Amino Acid Substitution, Antigens, CD, Bone Morphogenetic Protein Receptors, Type II, Child, Child, Preschool, Codon, Nonsense, DNA Mutational Analysis, Endoglin, Exons genetics, Female, Genetic Predisposition to Disease, Genotype, Germ-Line Mutation, Heart Defects, Congenital genetics, Humans, Hypertension, Pulmonary etiology, Infant, Infant, Newborn, Male, Mutation, Missense, Protein Serine-Threonine Kinases physiology, RNA Splicing, Receptors, Cell Surface, Receptors, Transforming Growth Factor beta physiology, Sequence Deletion, Telangiectasia, Hereditary Hemorrhagic complications, Telangiectasia, Hereditary Hemorrhagic genetics, Vascular Cell Adhesion Molecule-1 physiology, Activin Receptors, Type I genetics, Hypertension, Pulmonary genetics, Protein Serine-Threonine Kinases genetics, Receptors, Transforming Growth Factor beta genetics, Signal Transduction physiology, Transforming Growth Factor beta physiology, Vascular Cell Adhesion Molecule-1 genetics
- Abstract
Background: Pulmonary arterial hypertension (PAH) is a potentially fatal vasculopathy that can develop at any age. Adult-onset disease has previously been associated with mutations in BMPR2 and ALK-1. Presentation in early life may be associated with congenital heart disease but frequently is idiopathic., Methods and Results: We performed mutation analysis in genes encoding receptor members of the transforming growth factor-beta cell-signaling pathway in 18 children (age at presentation <6 years) with PAH. Sixteen children were initially diagnosed with idiopathic PAH and 2 with PAH in association with congenital heart defects. Germ-line mutations were observed in 4 patients (22%) (age at disease onset, 1 month to 6 years), all of whom presented with idiopathic PAH. The BMPR2 mutations (n=2, 11%) included a partial gene deletion and a nonsense mutation, both arising de novo in the proband. Importantly, a missense mutation of ALK-1 and a branch-site mutation of endoglin were also detected. Presenting clinical features or progression of pulmonary hypertension did not distinguish between patients with mutations in the different genes or between those without mutations., Conclusions: The cause of PAH presenting in childhood is heterogeneous in nature, with genetic defects of transforming growth factor-beta receptors playing a critical role.
- Published
- 2005
- Full Text
- View/download PDF
509. BMP4 inhibits proliferation and promotes myocyte differentiation of lung fibroblasts via Smad1 and JNK pathways.
- Author
-
Jeffery TK, Upton PD, Trembath RC, and Morrell NW
- Subjects
- Activin Receptors, Type I, Bone Morphogenetic Protein Receptors, Bone Morphogenetic Protein Receptors, Type II, Cell Cycle drug effects, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Division drug effects, Cell Division physiology, Cell Line, DNA biosynthesis, Humans, Intracellular Membranes metabolism, Ligands, Receptors, Growth Factor metabolism, Recombinant Proteins pharmacology, Signal Transduction drug effects, Signal Transduction physiology, Smad Proteins, Smad1 Protein, DNA-Binding Proteins metabolism, Fibroblasts cytology, JNK Mitogen-Activated Protein Kinases metabolism, Lung cytology, Myocytes, Smooth Muscle cytology, Protein Serine-Threonine Kinases physiology, Proteins pharmacology, Trans-Activators metabolism
- Abstract
Fibroblast proliferation, differentiation, and migration contribute to the characteristic pulmonary vascular remodeling seen in primary pulmonary hypertension (PPH). The identification of mutations in the bone morphogenetic protein type II receptor (BMPRII) in PPH have led us to question what role BMPRII and its ligands play in pulmonary vascular remodeling. Thus, to further understand the functional significance of BMPRII in the pulmonary vasculature, we examined the expression of TGF-beta superfamily receptors in human fetal lung fibroblasts (HFL) and investigated the role of BMP4 on cell cycle regulation, fibroblast proliferation, and differentiation. Furthermore, signaling pathways involved in these processes were examined. HFL expressed BMPRI and BMPRII mRNA and demonstrated specific I(125)-BMP4 binding sites. BMP4 inhibited [(3)H]thymidine incorporation and proliferation of HFL; protein expression was increased for the cell cycle inhibitor p21 and reduced for the positive regulators cyclin D and cdk2 by BMP4. BMP4 induced differentiation of HFL into a smooth muscle cell phenotype since protein expression of alpha-smooth muscle actin and smooth muscle myosin was increased. Furthermore, p38(MAPK), ERK1/2, JNK, and Smad1 were phosphorylated by BMP4. Using specific MAPK inhibitors, a dominant negative Smad1 construct, and Smad1 siRNA, we found that the antiproliferative and prodifferentiation effects of BMP4 were Smad1 dependent with JNK also contributing to differentiation. Because failure of Smad phosphorylation is a major feature of BMPRII mutations, these results imply that BMPRII mutations may promote the expansion of fibroblasts resistant to the antiproliferative, prodifferentiation effects of BMPs and suggest a mechanism for the vascular obliteration seen in familial PPH.
- Published
- 2005
- Full Text
- View/download PDF
510. The major psoriasis susceptibility locus PSORS1 is not a risk factor for late-onset psoriasis.
- Author
-
Allen MH, Ameen H, Veal C, Evans J, Ramrakha-Jones VS, Marsland AM, Burden AD, Griffiths CE, Trembath RC, and Barker JN
- Subjects
- Adult, Age of Onset, Aged, Aged, 80 and over, Female, Genetic Predisposition to Disease, Genotype, Humans, Male, Middle Aged, Risk Factors, Chromosomes, Human, Pair 6, Psoriasis epidemiology, Psoriasis genetics
- Abstract
PSORS1 is the major susceptibility locus for psoriasis vulgaris (PV) and lies within an approximately 200 kb segment of the major histocompatibility complex on chromosome 6p21.3. Alleles of candidate genes in this region including human leukocyte antigen (HLA)-C, alpha-helical coiled coil rod (HCR), and corneodesmosin (CDSN) show association with early-onset PV. Late-onset psoriasis (LOP) is defined as a disease with onset after 40 y of age and is typically sporadic. We assessed the role of PSORS1 in genetic susceptibility to LOP. Genotyping for HLA-C alleles and seven single nucleotide polymorphisms (SNP) within the genes HCR and CDSN was performed in LOP (n=145) and normal controls (n=309). Statistical analysis of allelic frequencies included calculation of odds ratio and chi2 comparisons. LOP demonstrated only a weak association to PSORS1 alleles HLA-Cw*6 (p=0.037), CDSN*5 (p=0.041), HCR*WC (p=0.013), and HCR SNP +325 (p=0.038). Patients with age of onset for psoriasis of 50 y or above provided no evidence of association with any of these alleles. These data suggest that the study cohort may include a number of subjects who harbor PSORS1 predisposition to early-onset psoriasis and yet do not present with disease by the age of 40 y. Thus this study demonstrates that PSORS1 is not a major inherited risk factor in the pathogenesis of LOP. These data suggest that the exclusion of LOP subjects from case-control studies will aid further delineation of the PSORS1 locus. Future genome-wide studies will be required to identify loci conferring risk for late-onset disease.
- Published
- 2005
- Full Text
- View/download PDF
511. A synonymous SNP of the corneodesmosin gene leads to increased mRNA stability and demonstrates association with psoriasis across diverse ethnic groups.
- Author
-
Capon F, Allen MH, Ameen M, Burden AD, Tillman D, Barker JN, and Trembath RC
- Subjects
- Animals, COS Cells, Case-Control Studies, Chlorocebus aethiops, Consensus Sequence genetics, Female, Haplotypes genetics, Humans, Intercellular Signaling Peptides and Proteins, Male, Mutagenesis, Site-Directed genetics, Mutation genetics, RNA, Messenger analysis, RNA-Binding Proteins metabolism, Serpins metabolism, Transfection, Glycoproteins genetics, Polymorphism, Single Nucleotide, Psoriasis ethnology, Psoriasis genetics, RNA Stability genetics, RNA, Messenger metabolism
- Abstract
Psoriasis is a chronic skin disorder with multifactorial aetiology. Genome-wide scans have provided unambiguous evidence for a major disease susceptibility locus on chromosome 6p21 (PSORS1). A minimal PSORS1 interval has been defined which encompasses three genes (HLA-C, HCR and CDSN) carrying psoriasis-associated SNPs. On the basis of this genetic evidence, we have undertaken an assessment of CDSN allele functional impact. A comparison of CDSN intragenic haplotypes showed that SNPs exclusive to disease-associated chromosomes are located in regions implicated in the stabilization of RNA transcripts. As CDSN is over-expressed in psoriatic lesions, we hypothesised that disease-associated intragenic SNPs may alter the rate of its mRNA decay. Here, we demonstrate that mRNAs transcribed from a CDSN risk haplotype present a 2-fold increase in stability, compared with those transcribed from a neutral haplotype (t-test P=0.004). Site-directed mutagenesis revealed that a single synonymous SNP (CDSN*971T) accounts for the observed increase in RNA stability. CDSN*971T maps to a RNA stability motif and UV cross-linking analysis demonstrated that the SNP affects the transcript affinity for a 39 kDa RNA binding protein. Association analyses show that haplotypes bearing CDSN*971T confer psoriasis susceptibility in a wide range of ethnic groups. These results demonstrate the effect of synonymous variation upon allele specific gene expression, a finding of relevance to future studies of the pathogenesis of common and complex traits.
- Published
- 2004
- Full Text
- View/download PDF
512. Low prevalence of MYOC mutations in UK primary open-angle glaucoma patients limits the utility of genetic testing.
- Author
-
Aldred MA, Baumber L, Hill A, Schwalbe EC, Goh K, Karwatowski W, and Trembath RC
- Subjects
- Adult, Aged, Case-Control Studies, Cytoskeletal Proteins, DNA Mutational Analysis, Family Health, Female, Genetic Predisposition to Disease, Glaucoma, Open-Angle diagnosis, Humans, Male, Middle Aged, Mutation, Prevalence, United Kingdom epidemiology, Eye Proteins genetics, Genetic Testing, Glaucoma, Open-Angle genetics, Glycoproteins genetics
- Abstract
Primary open angle glaucoma (POAG) affects 1% of people over age 40. Early detection and treatment can prevent blindness, but the disease is often asymptomatic until a late stage. Positive family history is an important risk factor and previous studies indicate that approximately 5% of POAG results from mutations in the myocilin ( MYOC) gene, raising the possibility of identifying individuals genetically predisposed to glaucoma. We collected DNA samples from 426 unselected UK POAG patients and analyzed them for MYOC mutations. The Q368X mutation was found in six patients (1.4%). No other mutations were identified, suggesting that amongst patients unselected for family history, the prevalence of MYOC mutations in the UK is lower than in other populations. Genetic and glaucoma screening was offered to first-degree relatives of these six probands (group 1) and of age/sex-matched mutation-negative controls (group 2). Of 11 group-1 relatives, three carried Q368X, one of whom already had glaucoma. Notably, of the 13 relatives in both groups who were mutation negative, one was already being treated for ocular hypertension. We therefore caution against changing glaucoma surveillance regimens in such individuals and suggest that routine untargeted genetic testing for MYOC mutations in patients with POAG would be of limited value until additional significant genetic risk factors are identified.
- Published
- 2004
- Full Text
- View/download PDF
513. An update on the genetics of psoriasis.
- Author
-
Capon F, Trembath RC, and Barker JN
- Subjects
- Female, Humans, Linkage Disequilibrium, Male, Molecular Biology, Prevalence, Psoriasis epidemiology, Risk Assessment, Sensitivity and Specificity, Chromosomes, Human, Pair 6, Genetic Predisposition to Disease, HLA-C Antigens genetics, Polymorphism, Genetic, Psoriasis genetics
- Abstract
Psoriasis is a complex inflammatory disorder whose pathogenesis is likely to require the contribution of several genes and environmental triggers. Despite the difficulties posed by the study of multifactorial conditions, significant progress has been achieved in relation to the molecular genetic basis of psoriasis. It has long been recognized that the major histocompatibility complex (MHC) region on chromosome 6p21 harbors the main determinant conferring psoriasis susceptibility. The identification of non-MHC susceptibility regions across the genome has been hindered by the likely occurrence of genetic heterogeneity. Nonetheless, evidence for the assignment of a number of non-MHC loci has been achieved through studies, including the collaborative analysis of large patient cohorts, and also through the observation of overlap between psoriasis and atopic dermatitis susceptibility regions.
- Published
- 2004
- Full Text
- View/download PDF
514. Genetic basis of pulmonary arterial hypertension: current understanding and future directions.
- Author
-
Newman JH, Trembath RC, Morse JA, Grunig E, Loyd JE, Adnot S, Coccolo F, Ventura C, Phillips JA 3rd, Knowles JA, Janssen B, Eickelberg O, Eddahibi S, Herve P, Nichols WC, and Elliott G
- Subjects
- Animals, Bone Morphogenetic Protein Receptors, Type II, Forecasting, Genetic Predisposition to Disease epidemiology, Genetic Predisposition to Disease genetics, Genetic Testing trends, Humans, Hypertension, Pulmonary diagnosis, Hypertension, Pulmonary etiology, Mutation genetics, Protein Serine-Threonine Kinases genetics, Risk Factors, Hypertension, Pulmonary genetics, Pulmonary Artery pathology
- Abstract
Mutations in two receptors of the transforming growth factor-beta family have recently been shown to be present in the majority of cases of inherited (familial) pulmonary arterial hypertension (PAH). Study of the biology of these receptors, bone morphogenetic protein receptor type-2 (BMPR2), and activin-like kinase type-1 (ALK-1) will certainly reveal pathogenic mechanisms of disease. Exonic mutations in BMPR2 are found in about 50% of patients with familial PAH, and ALK1 mutations are found in a minority of patients with hereditary hemorrhagic telangiectasia and co-existent PAH. Because familial PAH is highly linked to chromosome 2q33, it is likely that the remaining 50% of family cases without exonic mutations have either intronic BMPR2 abnormalities or alterations in the promoter or regulatory genes. Also, only about 10% of patients with "sporadic" idiopathic PAH have identifiable BMPR2 mutations. Mutations in BMPR2 confer a 15% to 20% chance of developing PAH in a carrier's lifetime. Thus, there must be gene-gene or gene-environment interactions that either enhance or prevent the development of the vascular disease in persons carrying a mutation, and there must be other patterns of susceptibility based on genetic makeup. To elucidate the genetic basis of PAH further, investigations are needed, including genome scanning for major and minor genes, analysis of genetic profiles of patients for candidate genes likely to modify risk for disease (e.g., serotonin transporter alleles, nitric oxide-synthases), proteomics, transgenic mice, and altered signal transduction. Advances in genetic testing, presymptomatic screening, and biomarkers should permit early detection of disease in those at risk of PAH and allow trials of preventive therapy in carriers.
- Published
- 2004
- Full Text
- View/download PDF
515. Meta-analysis of genome-wide studies of psoriasis susceptibility reveals linkage to chromosomes 6p21 and 4q28-q31 in Caucasian and Chinese Hans population.
- Author
-
Sagoo GS, Tazi-Ahnini R, Barker JW, Elder JT, Nair RP, Samuelsson L, Traupe H, Trembath RC, Robinson DA, and Iles MM
- Subjects
- Asian People genetics, Chromosomes, Human, Pair 4, Chromosomes, Human, Pair 6, Genetic Predisposition to Disease, Humans, White People genetics, Genetic Linkage, Genome, Human, Psoriasis genetics
- Abstract
Ten genome-wide scans have been conducted over the past few years in the search for psoriasis susceptibility genes, but only one potential susceptibility region has been consistently replicated. A meta-analysis using the genome-search meta-analysis method was undertaken combining the results of six of these psoriasis genome-wide studies. The results of this analysis revealed linkage to the major histocompatibility complex on chromosome 6p21 that includes the PSORS1 locus. In addition, linkage was also recorded to a region on chromosome 4q28-q31 previously identified only in a Chinese Hans population. Both these regions were statistically significant even after correction for multiple testing. A possible reason for the erratic replication of findings could be the large effect of the PSORS1 locus (6p21) masking the effect of other loci involved in psoriasis. To overcome this problem, we suggest that future studies condition on the effect of the PSORS1 locus.
- Published
- 2004
- Full Text
- View/download PDF
516. Mutations in VPS33B, encoding a regulator of SNARE-dependent membrane fusion, cause arthrogryposis-renal dysfunction-cholestasis (ARC) syndrome.
- Author
-
Gissen P, Johnson CA, Morgan NV, Stapelbroek JM, Forshew T, Cooper WN, McKiernan PJ, Klomp LW, Morris AA, Wraith JE, McClean P, Lynch SA, Thompson RJ, Lo B, Quarrell OW, Di Rocco M, Trembath RC, Mandel H, Wali S, Karet FE, Knisely AS, Houwen RH, Kelly DA, and Maher ER
- Subjects
- Blotting, Western, Cell Line, Chromosomes, Human, Pair 15, Electrophoresis, Polyacrylamide Gel, Female, Humans, Male, Membrane Fusion genetics, Membrane Proteins chemistry, Plasmids, Proteins chemistry, SNARE Proteins, Syndrome, Arthrogryposis genetics, Cholestasis genetics, Kidney Diseases genetics, Membrane Fusion physiology, Membrane Proteins genetics, Membrane Proteins physiology, Mutation, Proteins genetics, Vesicular Transport Proteins
- Abstract
ARC syndrome (OMIM 208085) is an autosomal recessive multisystem disorder characterized by neurogenic arthrogryposis multiplex congenita, renal tubular dysfunction and neonatal cholestasis with bile duct hypoplasia and low gamma glutamyl transpeptidase (gGT) activity. Platelet dysfunction is common. Affected infants do not thrive and usually die in the first year of life. To elucidate the molecular basis of ARC, we mapped the disease to a 7-cM interval on 15q26.1 and then identified germline mutations in the gene VPS33B in 14 kindreds with ARC. VPS33B encodes a homolog of the class C yeast vacuolar protein sorting gene, Vps33, that contains a Sec1-like domain important in the regulation of vesicle-to-target SNARE complex formation and subsequent membrane fusion.
- Published
- 2004
- Full Text
- View/download PDF
517. Identification of a fourth locus (EVR4) for familial exudative vitreoretinopathy (FEVR).
- Author
-
Toomes C, Downey LM, Bottomley HM, Scott S, Woodruff G, Trembath RC, and Inglehearn CF
- Subjects
- Chromosome Mapping, Exudates and Transudates, Female, Frizzled Receptors, Genes, Dominant genetics, Genetic Markers, Genotype, Haplotypes, Humans, Male, Pedigree, Proteins genetics, Receptors, Cell Surface, Receptors, G-Protein-Coupled, Chromosomes, Human, Pair 11 genetics, Eye Diseases, Hereditary genetics, Genetic Linkage genetics, Retinal Diseases genetics, Retinal Vessels pathology, Vitreous Body pathology
- Abstract
Purpose: Familial exudative vitreoretinopathy (FEVR) is a genetically heterogeneous inherited blinding disorder of the retinal vascular system. To date three loci have been mapped: EVR1 on chromosome 11q, EVR2 on chromosome Xp, and EVR3 on chromosome 11p. The gene underlying EVR3 remains unidentified whilst the EVR2 gene, which encodes the Norrie disease protein (NDP), was identified over a decade ago. More recently, FZD4, the gene that encodes the Wnt receptor Frizzled-4, was identified as the mutated gene at the EVR1 locus. The purpose of this study was to screen FZD4 in a large family previously proven to be linked to the EVR1 locus., Methods: PCR products were generated using genomic DNA from affected family members with primers designed to amplify the coding sequence of FZD4. The PCR products were screened for mutations by direct sequencing. Genotyping was performed in all available family members using fluorescently labeled microsatellite markers from chromosome 11q., Results: Sequencing of the EVR1 gene, FZD4, in this family identified no mutation. To investigate this family further we performed high-resolution genotyping with markers spanning chromosome 11q. Haplotype analysis excluded FZD4 as the mutated gene in this family and identified a candidate region approximately 10 cM centromeric to EVR1. This new FEVR locus is flanked by markers D11S1368 (centromeric) and D11S937 (telomeric) and spans approximately 15 cM., Conclusions: High-resolution genotyping and haplotype analysis excluded FZD4 as the defective gene in a family previously linked to the EVR1 locus. The results indicate that the gene mutated in this family lies centromeric to the EVR1 gene, FZD4, and is also genetically distinct from the EVR3 locus. This new locus has been designated EVR4 and is the fourth FEVR locus to be described.
- Published
- 2004
518. Pendred syndrome and DFNB4-mutation screening of SLC26A4 by denaturing high-performance liquid chromatography and the identification of eleven novel mutations.
- Author
-
Prasad S, Kölln KA, Cucci RA, Trembath RC, Van Camp G, and Smith RJ
- Subjects
- Biological Transport, Chromatography, High Pressure Liquid, DNA Primers, Diagnosis, Differential, Humans, Polymorphism, Single-Stranded Conformational, Sensitivity and Specificity, Sequence Analysis, DNA, Sulfate Transporters, Sulfates, Syndrome, Carrier Proteins genetics, DNA Mutational Analysis, Genetic Testing, Goiter diagnosis, Goiter genetics, Hearing Loss, Sensorineural diagnosis, Hearing Loss, Sensorineural genetics, Membrane Transport Proteins, Polymorphism, Genetic
- Abstract
Mutations in SLC26A4 cause Pendred syndrome, an autosomal-recessive disorder characterized by sensorineural deafness and goiter, and DFNB4, a type of autosomal recessive nonsyndromic deafness in which, by definition, affected persons do not have thyromegaly. The clinical diagnosis of these two conditions is difficult, making mutation screening of SLC26A4 a valuable test. Although screening can be accomplished in a variety of ways, all techniques are not equally accurate, timely or cost effective. We found single-strand conformational polymorphism analysis (SSCP) to be 63% effective in detecting mutations a panel of different SLC26A4 allele variants when compared to data from direct sequencing. Because direct sequencing can be time consuming and expensive, especially for a gene with 21 exons, we studied DHPLC as an alternative screening method. We found DHPLC as accurate and reliable as direct sequencing but to be more rapid and cost effective. In addition, we report 11 novel disease-causing allele variants of SLC26A4., (Copyright 2003 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
519. Functional interaction between BMPR-II and Tctex-1, a light chain of Dynein, is isoform-specific and disrupted by mutations underlying primary pulmonary hypertension.
- Author
-
Machado RD, Rudarakanchana N, Atkinson C, Flanagan JA, Harrison R, Morrell NW, and Trembath RC
- Subjects
- Bone Morphogenetic Protein Receptors, Type II, HeLa Cells, Humans, Hypertension, Pulmonary metabolism, Lung metabolism, Lung ultrastructure, Models, Biological, Phosphorylation, Plasmids, Protein Isoforms, Protein Serine-Threonine Kinases genetics, Two-Hybrid System Techniques, t-Complex Genome Region, Dyneins metabolism, Hypertension, Pulmonary genetics, Microtubule-Associated Proteins metabolism, Mutation, Nuclear Proteins metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Diverse heterozygous mutations of bone morphogenetic receptor type II (BMPR-II) underlie the inherited form of the vascular disorder primary pulmonary hypertension (PPH). As yet, the molecular detail of how such defects contribute to the pathogenesis of PPH remains unclear. BMPR-II is a member of the transforming growth factor-beta cell signalling superfamily. Ligand binding induces cell surface receptor complex formation and activates a cascade of phosphorylation events of intracellular intermediaries termed Smads, which initiate transcriptional regulation. Some 30% of PPH-causing mutations localize to exon 12, which may be spliced out forming an isoform depleted of the unusually long BMPR-II cytoplasmic tail. To further elucidate the consequences of BMPR2 mutation, we sought to characterize aspects of the cytoplasmic domain function by seeking intracellular binding partners. We now report that Tctex-1, a light chain of the motor complex dynein, interacts with the cytoplasmic domain of BMPR-II and demonstrate that Tctex-1 is phosphorylated by BMPR-II, a function disrupted by PPH disease causing mutations within exon 12. Finally we show that BMPR-II and Tctex-1 co-localize to endothelium and smooth muscle within the media of pulmonary arterioles, key sites of vascular remodelling in PPH. Taken together, these data demonstrate a discrete function for the cytoplasmic domain of BMPR-II and justify further investigation of whether the interaction with and phosphorylation of Tctex-1 contributes to the pathogenesis of PPH.
- Published
- 2003
- Full Text
- View/download PDF
520. Insights into the genetic and molecular basis of primary pulmonary hypertension.
- Author
-
Trembath RC and Harrison R
- Subjects
- Bone Morphogenetic Protein Receptors, Type II, Genetic Testing, Humans, Hypertension, Pulmonary diagnosis, Mutation, Protein Serine-Threonine Kinases genetics, Telangiectasis genetics, Transforming Growth Factor beta genetics, Hypertension, Pulmonary genetics
- Abstract
The pathogenesis of primary pulmonary hypertension (PPH) remains poorly understood. Molecular genetic studies have identified that mutations within the gene BMPR2 on the long arm of chromosome 2 underlie familial PPH. This review explores the significance of the PPH gene identification and examines additional genetic determinants, emphasizing the immediate implications for assessment and management of patients and their relatives.
- Published
- 2003
- Full Text
- View/download PDF
521. An early-onset autosomal dominant macular dystrophy (MCDR3) resembling North Carolina macular dystrophy maps to chromosome 5.
- Author
-
Michaelides M, Johnson S, Tekriwal AK, Holder GE, Bellmann C, Kinning E, Woodruff G, Trembath RC, Hunt DM, and Moore AT
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Chromosome Mapping, DNA analysis, Female, Fluorescein Angiography, Genes, Dominant, Genotype, Humans, Lod Score, Macular Degeneration diagnosis, Male, Microsatellite Repeats, Middle Aged, North Carolina, Pedigree, Polymerase Chain Reaction, Chromosomes, Human, Pair 5 genetics, Genetic Linkage, Macular Degeneration genetics
- Abstract
Purpose: To characterize the phenotype of an autosomal dominant macular dystrophy and identify the chromosomal locus., Methods: Thirteen members of a four-generation, nonconsanguineous British family were examined clinically and also underwent automated perimetry, fundus fluorescein angiography, and fundus autofluorescence imaging. After informed consent was obtained, blood samples were taken for DNA extraction, and genetic linkage analysis was performed., Results: The retinal changes have an early age of onset and are confined to the macular region. The macular abnormalities vary from mild retinal pigment epithelium (RPE) pigmentary change to atrophy. Drusen-like deposits are present to various degrees and are characteristic of the phenotype. Subretinal neovascular membrane (SRNVM) is an established complication. Genetic linkage analysis established linkage to chromosome 5, region p13.1-p15.33 with a maximum LOD score of 3.61 at a recombination fraction of 0.00 for marker D5S630. The locus for this autosomal dominant macular dystrophy lies between flanking markers D5S1981 and D5S2031., Conclusions: A novel locus has been identified for early-onset autosomal dominant macular dystrophy on chromosome 5.
- Published
- 2003
- Full Text
- View/download PDF
522. Missense mutations in the homeodomain of HOXD13 are associated with brachydactyly types D and E.
- Author
-
Johnson D, Kan SH, Oldridge M, Trembath RC, Roche P, Esnouf RM, Giele H, and Wilkie AO
- Subjects
- Amino Acid Sequence, Arm abnormalities, Base Sequence, Binding Sites, DNA Primers, Female, Homeodomain Proteins chemistry, Homeodomain Proteins metabolism, Humans, Leg abnormalities, Male, Molecular Sequence Data, Pedigree, Polymerase Chain Reaction, Sequence Alignment, Sequence Homology, Amino Acid, Fingers abnormalities, Homeodomain Proteins genetics, Mutation, Missense, Synostosis classification, Synostosis genetics, Transcription Factors
- Abstract
HOXD13, the most 5' gene of the HOXD cluster, encodes a homeodomain transcription factor with important functions in limb patterning and growth. Heterozygous mutations of human HOXD13, encoding polyalanine expansions or frameshifts, are believed to act by dominant negative or haploinsufficiency mechanisms and are predominantly associated with synpolydactyly phenotypes. Here, we describe two mutations of HOXD13 (923C-->G encoding Ser308Cys and 940A-->C encoding Ile314Leu) that cause missense substitutions within the homeodomain. Both are associated with distinctive limb phenotypes in which brachydactyly of specific metacarpals, metatarsals, and phalangeal bones is the most constant feature, exhibiting overlap with brachydactyly types D and E. We investigated the binding of synthetic mutant proteins to double-stranded DNA targets in vitro. No consistent differences were found for the Ser308Cys mutation compared with the wild type, but the Ile314Leu mutation (which resides at the 47th position of the homeodomain) exhibited increased affinity for a target containing the core recognition sequence 5'-TTAC-3' but decreased affinity for a 5'-TTAT-3' target. Molecular modeling of the Ile314Leu mutation indicates that this mixed gain and loss of affinity may be accounted for by the relative positions of methyl groups in the amino acid side chain and target base.
- Published
- 2003
- Full Text
- View/download PDF
523. Genetic analysis of PSORS1 distinguishes guttate psoriasis and palmoplantar pustulosis.
- Author
-
Asumalahti K, Ameen M, Suomela S, Hagforsen E, Michaëlsson G, Evans J, Munro M, Veal C, Allen M, Leman J, David Burden A, Kirby B, Connolly M, Griffiths CE, Trembath RC, Kere J, Saarialho-Kere U, and Barker JN
- Subjects
- Diagnosis, Differential, Gene Frequency, Genetic Predisposition to Disease epidemiology, Glycoproteins genetics, HLA-C Antigens genetics, Haplotypes, Heterozygote, Humans, Immunohistochemistry, Intercellular Signaling Peptides and Proteins, Intracellular Signaling Peptides and Proteins, Proteins genetics, Psoriasis epidemiology, Risk Factors, Psoriasis diagnosis, Psoriasis genetics
- Abstract
The PSORS1 locus in the major histocompatibility complex region is the major genetic determinant for psoriasis vulgaris. Within the PSORS1 region reside at least three potential candidate genes for psoriasis susceptibility. Specific allelic variants of the genes HLA-Cw*6, HCR*WWCC, and CDSN*5 are strongly associated with psoriasis vulgaris and are in strong linkage disequilibrium with each other. We have genotyped the three psoriasis vulgaris susceptibility alleles of the PSORS1 locus in two clinical variants of psoriasis (guttate psoriasis and palmoplantar pustulosis) to study whether PSORS1 is also involved in the pathogenesis of these variants. We also asked whether these two clinical subgroups could help us to distinguish the causative gene within the high-risk PSORS1 haplotype. The association of guttate psoriasis with the three PSORS1 susceptibility alleles was similar and even stronger than seen with psoriasis vulgaris. Palmoplantar pustulosis, however, did not show association with any of the three candidate genes at this locus. Finally, no correlation with the age of onset for disease was observed. Our results show conclusively that psoriasis vulgaris and guttate psoriasis have a similar genetic basis for their association to PSORS1, whereas palmoplantar pustulosis appears to be a distinct disorder.
- Published
- 2003
- Full Text
- View/download PDF
524. Mutations in a novel gene Dymeclin (FLJ20071) are responsible for Dyggve-Melchior-Clausen syndrome.
- Author
-
El Ghouzzi V, Dagoneau N, Kinning E, Thauvin-Robinet C, Chemaitilly W, Prost-Squarcioni C, Al-Gazali LI, Verloes A, Le Merrer M, Munnich A, Trembath RC, and Cormier-Daire V
- Subjects
- Amino Acid Sequence, Base Sequence, DNA Mutational Analysis, Female, Genetic Linkage, Humans, Intracellular Signaling Peptides and Proteins, Male, Molecular Sequence Data, Mutation, Pedigree, Proteins metabolism, Sequence Analysis, DNA, Skin pathology, Skin ultrastructure, Abnormalities, Multiple genetics, Intellectual Disability genetics, Proteins genetics
- Abstract
Dyggve-Melchior-Clausen syndrome (DMC) is a rare autosomal-recessive disorder, the gene for which maps to chromosome 18q21.1. DMC is characterized by the association of a spondylo-epi-metaphyseal dysplasia and mental retardation. Electron microscopic study of cutaneous cells of an affected child showed dilated rough endoplasmic reticulum, enlarged and aberrant vacuoles and numerous vesicles. As the etiology of the disorder is unknown, we have used a positional cloning strategy to identify the DMC gene. We detected seven deleterious mutations within a gene predicted from a human transcript (FLJ20071) in 10 DMC families. The mutations were nonsense mutations (R194X, R204X, L219X, Q483X), splice site or frameshift mutations (K626N+92aa to stop). The DMC gene transcript is widely distributed but appears abundant in chondrocytes and fetal brain. The predicted protein product of the DMC gene yields little insight into its likely function, showing no significant homology to any known protein family. However, the carboxy terminal end comprises a cluster of dileucine motifs, highly conserved across species. We conclude that DMC syndrome is consequent upon loss of function of a gene that we propose to name Dymeclin, which may have a role in process of intracellular digestion of proteins.
- Published
- 2003
- Full Text
- View/download PDF
525. Constitutional deletion of chromosome 20q in two patients affected with albright hereditary osteodystrophy.
- Author
-
Aldred MA, Aftimos S, Hall C, Waters KS, Thakker RV, Trembath RC, and Brueton L
- Subjects
- Adolescent, Child, Chromosome Banding, DNA chemistry, DNA genetics, DNA Mutational Analysis, Female, Fibrous Dysplasia, Polyostotic pathology, GTP-Binding Protein alpha Subunits, Gs genetics, Gene Deletion, Humans, Karyotyping, Male, Microsatellite Repeats, Polymorphism, Genetic, Chromosome Deletion, Chromosomes, Human, Pair 20 genetics, Fibrous Dysplasia, Polyostotic genetics
- Abstract
Albright hereditary osteodystrophy (AHO) results from heterozygous inactivation of G(s)alpha, encoded by the GNAS1 locus on the distal long arm of chromosome 20. This autosomal dominant condition is characterized by short stature, obesity, shortening of the metacarpals and metatarsals, and variable mental retardation and may also include end-organ resistance to multiple hormones. Small insertions and deletions or point mutations of GNAS1 are found in approximately 80% of patients with AHO. The remainder may be accounted for by larger genomic rearrangements, but none have been reported to date. We now describe two patients with constitutional 20q deletions and features of AHO. Such deletions are rare in the published literature and have not previously been associated with AHO. Molecular genetic analysis confirmed complete deletion of GNAS1 in both patients. Parental origin could be determined in both cases and provides further support for the parent-of-origin effect on the biochemical status of patients with AHO., (Copyright 2002 Wiley-Liss, Inc.)
- Published
- 2002
- Full Text
- View/download PDF
526. A novel locus for Meckel-Gruber syndrome, MKS3, maps to chromosome 8q24.
- Author
-
Morgan NV, Gissen P, Sharif SM, Baumber L, Sutherland J, Kelly DA, Aminu K, Bennett CP, Woods CG, Mueller RF, Trembath RC, Maher ER, and Johnson CA
- Subjects
- Chromosome Mapping, Female, Genes, Recessive, Genetic Linkage, Humans, Male, Membrane Proteins, Neural Tube Defects genetics, Pedigree, Syndrome, Abnormalities, Multiple genetics, Chromosomes, Human, Pair 8, Proteins genetics
- Abstract
Meckel-Gruber syndrome (MKS), the most common monogenic cause of neural tube defects, is an autosomal recessive disorder characterised by a combination of renal cysts and variably associated features, including developmental anomalies of the central nervous system (typically encephalcoele), hepatic ductal dysplasia and cysts, and polydactyly. Locus heterogeneity has been demonstrated by the mapping of the MKS1locus to 17q21-24 in Finnish kindreds, and of MKS2 to 11q13 in North African-Middle Eastern cohorts. In the present study, we have investigated the genetic basis of MKS in eight consanguineous kindreds, originating from the Indian sub-continent, that do not show linkage to either MKS1 or MKS2. We report the localisation of a third MKS locus ( MKS3) to chromosome 8q24 in this cohort by a genome-wide linkage search using autozygosity mapping. We identified a 26-cM region of autozygosity between D8S586 and D8S1108 with a maximum cumulative two-point LOD score at D8S1179 ( Z(max)=3.04 at theta=0.06). A heterogeneity test provided evidence of one unlinked family. Exclusion of this family from multipoint analysis maximised the cumulative multipoint LOD score at locus D8S1128 ( Z(max)=5.65). Furthermore, a heterozygous SNP in DDEF1, a putative candidate gene, suggested that MKS3 mapped within a 15-cM interval. Comparison of the clinical features of MKS3-linked cases with reports of MKS1- and MKS2-linked kindreds suggests that polydactyly (and possibly encephalocele) appear less common in MKS3-linked families.
- Published
- 2002
- Full Text
- View/download PDF
527. Family-based analysis using a dense single-nucleotide polymorphism-based map defines genetic variation at PSORS1, the major psoriasis-susceptibility locus.
- Author
-
Veal CD, Capon F, Allen MH, Heath EK, Evans JC, Jones A, Patel S, Burden D, Tillman D, Barker JN, and Trembath RC
- Subjects
- Case-Control Studies, Haplotypes genetics, Humans, Linkage Disequilibrium, Molecular Sequence Data, Chromosome Mapping, Chromosomes, Human, Pair 6 genetics, Genetic Predisposition to Disease, Genetic Variation genetics, Polymorphism, Single Nucleotide genetics, Psoriasis genetics
- Abstract
Psoriasis is a common skin disorder of multifactorial origin. Genomewide scans for disease susceptibility have repeatedly demonstrated the existence of a major locus, PSORS1 (psoriasis susceptibility 1), contained within the major histocompatibility complex (MHC), on chromosome 6p21. Subsequent refinement studies have highlighted linkage disequilibrium (LD) with psoriasis, along a 150-kb segment that includes at least three candidate genes (encoding human leukocyte antigen-C [HLA-C], alpha-helix-coiled-coil-rod homologue, and corneodesmosin), each of which has been shown to harbor disease-associated alleles. However, the boundaries of the minimal PSORS1 region remain poorly defined. Moreover, interpretations of allelic association with psoriasis are compounded by limited insight of LD conservation within MHC class I interval. To address these issues, we have pursued a high-resolution genetic characterization of the PSORS1 locus. We resequenced genomic segments along a 220-kb region at chromosome 6p21 and identified a total of 119 high-frequency SNPs. Using 59 SNPs (18 coding and 41 noncoding SNPs) whose position was representative of the overall marker distribution, we genotyped a data set of 171 independently ascertained parent-affected offspring trios. Family-based association analysis of this cohort highlighted two SNPs (n.7 and n.9) respectively lying 7 and 4 kb proximal to HLA-C. These markers generated highly significant evidence of disease association (P<10-9), several orders of magnitude greater than the observed significance displayed by any other SNP that has previously been associated with disease susceptibility. This observation was replicated in a Gujarati Indian case/control data set. Haplotype-based analysis detected overtransmission of a cluster of chromosomes, which probably originated by ancestral mutation of a common disease-bearing haplotype. The only markers exclusive to the overtransmitted chromosomes are SNPs n.7 and n.9, which define a 10-kb PSORS1 core risk haplotype. These data demonstrate the power of SNP haplotype-based association analyses and provide high-resolution dissection of genetic variation across the PSORS1 interval, the major susceptibility locus for psoriasis.
- Published
- 2002
- Full Text
- View/download PDF
528. Functional analysis of bone morphogenetic protein type II receptor mutations underlying primary pulmonary hypertension.
- Author
-
Rudarakanchana N, Flanagan JA, Chen H, Upton PD, Machado R, Patel D, Trembath RC, and Morrell NW
- Subjects
- Animals, Bone Morphogenetic Protein 2, Bone Morphogenetic Protein Receptors, Type II, Bone Morphogenetic Proteins metabolism, Cell Line, Cells, Cultured, Genes, Reporter, HeLa Cells, Humans, Hypertension, Pulmonary metabolism, Ligands, Mice, Mitogen-Activated Protein Kinases, Mutation, Protein Serine-Threonine Kinases metabolism, Protein Structure, Tertiary, Protein Transport physiology, Recombinant Proteins metabolism, p38 Mitogen-Activated Protein Kinases, Hypertension, Pulmonary genetics, Protein Serine-Threonine Kinases genetics, Transforming Growth Factor beta
- Abstract
A wide range of mutations in the type II receptor for bone morphogenetic protein (BMPR-II) have been shown to underlie primary pulmonary hypertension. To determine the mechanism of altered BMPR-II function, we employed transient transfection studies in cell lines and primary cultures of pulmonary vascular smooth muscle cells using green fluorescent protein (GFP)-tagged wild-type and mutant BMPR2 constructs and confocal microscopy to localize receptors. Substitution of cysteine residues in the ligand binding or kinase domain prevented trafficking of BMPR-II to the cell surface, and reduced binding of (125)I-BMP4. In addition, transfection of cysteine-substituted BMPR-II markedly reduced basal and BMP4-stimulated transcriptional activity of a BMP/Smad responsive luciferase reporter gene (3GC2wt-Lux), compared with wild-type BMPR-II, suggesting a dominant-negative effect of these mutants on Smad signalling. In contrast, BMPR-II containing non-cysteine substitutions in the kinase domain were localized to the cell membrane, although these also suppressed the activity of 3GC2wt-Lux. Interestingly, BMPR-II mutations within the cytoplasmic tail trafficked to the cell surface, but retained the ability to activate 3GC2wt-Lux. Transfection of mutant, but not wild-type, constructs into a mouse epithelial cell line (NMuMG cells) led to activation of p38(MAPK) and increased serum-induced proliferation compared with the wild-type receptor, which was partly p38(MAPK)-dependent. We conclude that mutations in BMPR-II heterogeneously inhibit BMP/Smad-mediated signalling by diverse molecular mechanisms. However, all mutants studied demonstrate a gain of function involving upregulation of p38(MAPK)-dependent proproliferative pathways.
- Published
- 2002
- Full Text
- View/download PDF
529. Primary pulmonary hypertension is associated with reduced pulmonary vascular expression of type II bone morphogenetic protein receptor.
- Author
-
Atkinson C, Stewart S, Upton PD, Machado R, Thomson JR, Trembath RC, and Morrell NW
- Subjects
- Activin Receptors, Type I biosynthesis, Adult, Biomarkers analysis, Bone Morphogenetic Protein Receptors, Type II, DNA Mutational Analysis, Endothelium, Vascular pathology, Female, Heterozygote, Humans, Hypertension, Pulmonary genetics, Hypertension, Pulmonary pathology, Immunohistochemistry, In Situ Hybridization, Lung pathology, Male, Middle Aged, Mutation, Organ Specificity, Protein Serine-Threonine Kinases genetics, Pulmonary Circulation genetics, RNA, Messenger analysis, RNA, Messenger biosynthesis, Receptor, Transforming Growth Factor-beta Type I, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta biosynthesis, Endothelium, Vascular metabolism, Hypertension, Pulmonary metabolism, Lung blood supply, Lung metabolism, Protein Serine-Threonine Kinases biosynthesis
- Abstract
Background: Mutations in the type II receptor for bone morphogenetic protein (BMPR-II), a receptor member of the transforming growth factor-beta (TGF-beta) superfamily, underlie many familial and sporadic cases of primary pulmonary hypertension (PPH)., Methods and Results: Because the sites of expression of BMPR-II in the normal and hypertensive lung are unknown, we studied the cellular localization of BMPR-II and the related type I and II receptors for TGF-beta by immunohistochemistry in lung sections from patients undergoing heart-lung transplantation for PPH (n=11, including 3 familial cases) or secondary pulmonary hypertension (n=6) and from unused donor lungs (n=4). In situ hybridization was performed for BMPR-II mRNA. Patients were screened for the presence of mutations in BMPR2. In normal lungs, BMPR-II expression was prominent on vascular endothelium, with minimal expression in airway and arterial smooth muscle. In pulmonary hypertension cases, the intensity of BMPR-II immunostaining varied between lesions but involved endothelial and myofibroblast components. Image analysis confirmed that expression of BMPR-II was markedly reduced in the peripheral lung of PPH patients, especially in those harboring heterozygous BMPR2 mutations. A less marked reduction was also observed in patients with secondary pulmonary hypertension. In contrast, there was no difference in level of staining for TGF-betaRII or the endothelial marker CD31., Conclusions: The cellular localization of BMPR-II is consistent with a role in the formation of pulmonary vascular lesions in PPH, and reduced BMPR-II expression may contribute to the process of vascular obliteration in severe pulmonary hypertension.
- Published
- 2002
- Full Text
- View/download PDF
530. Mutations of the PDS gene, encoding pendrin, are associated with protein mislocalization and loss of iodide efflux: implications for thyroid dysfunction in Pendred syndrome.
- Author
-
Taylor JP, Metcalfe RA, Watson PF, Weetman AP, and Trembath RC
- Subjects
- Biological Transport genetics, Biological Transport physiology, Cell Line, Goiter genetics, Goiter physiopathology, HeLa Cells, Hearing Loss, Sensorineural genetics, Hearing Loss, Sensorineural physiopathology, Humans, Sulfate Transporters, Syndrome, Thyroid Gland physiopathology, Tissue Distribution, Carrier Proteins genetics, Carrier Proteins metabolism, Iodides metabolism, Membrane Transport Proteins, Mutation physiology
- Abstract
Pendred syndrome (PDS) is an autosomal recessive disorder characterized by deafness and goiter. Phenotypic heterogeneity is observed in affected individuals, and thyroid dysfunction is particularly variable. The syndrome is caused by mutations in the PDS (SLC26A4) gene, encoding an anion transporter pendrin, which localizes to the apical membrane of thyroid follicular cells. PDS is thought to enable efflux iodide into the follicle lumen. More than 50 diseases causing mutations of PDS have been reported. Here we have investigated the effect of nine PDS missense mutations on pendrin localization and iodide transport with the view to understanding their functional impact. As demonstrated by transient expression of green fluorescent protein-tagged pendrin mutant constructs in mammalian cell lines, appropriate trafficking to the plasma membrane was observed for only two mutants. The remaining PDS mutants appear to be retained within the endoplasmic reticulum following transfection. Iodide efflux assays were performed using human embryonic kidney 293 cells transfected with mutant pendrin and cotransfected with sodium iodide transporter to provide a mechanism of iodide uptake. The results indicated loss of pendrin iodide transport for all mislocalizing mutations. However, PDS mutants are associated with variable thyroid dysfunction in affected subjects. We concluded that additional genetic and/or environmental factors influence the thyroid activity in Pendred syndrome.
- Published
- 2002
- Full Text
- View/download PDF
531. A novel interaction between lamin A and SREBP1: implications for partial lipodystrophy and other laminopathies.
- Author
-
Lloyd DJ, Trembath RC, and Shackleton S
- Subjects
- 3T3 Cells, Animals, CCAAT-Enhancer-Binding Proteins genetics, DNA-Binding Proteins genetics, Fibroblasts, Gene Expression Regulation, HeLa Cells, Humans, Lamin Type A, Lamins, Lipodystrophy genetics, Mice, Nuclear Proteins genetics, Sterol Regulatory Element Binding Protein 1, Two-Hybrid System Techniques, CCAAT-Enhancer-Binding Proteins metabolism, DNA-Binding Proteins metabolism, Lipodystrophy metabolism, Nuclear Proteins metabolism, Transcription Factors
- Abstract
The gene encoding nuclear lamins A and C is mutated in at least three inherited disorders. Two of these, Emery-Dreifuss muscular dystrophy (EDMD-AD) and a form of dilated cardiomyopathy (CMD1A), involve muscle defects, and the other, familial partial lipodystrophy (FPLD), involves loss of subcutaneous adipose tissue. Mutations causing FPLD, in contrast to those causing muscle disorders, are tightly clustered within the C-terminal domain of lamin A/C. We investigated the expression and subcellular localization of FPLD lamin A mutants and found no abnormalities. We therefore set out to identify proteins interacting with the C-terminal domain of lamin A by screening a mouse 3T3-L1 adipocyte library in a yeast two-hybrid interaction screen. Using this approach, the adipocyte differentiation factor, sterol response element binding protein 1 (SREBP1) was identified as a novel lamin A interactor. In vitro glutathione S-transferase pull-down and in vivo co-immunoprecipitation studies confirmed an interaction between lamin A and both SREBP1a and 1c. A binding site for lamin A was identified in the N-terminal transcription factor domain of SREBP1, between residues 227 and 487. The binding of lamin A to SREBP1 was noticeably reduced by FPLD mutations. Interestingly, one EDMD-AD mutation also interfered with the interaction between lamin A and SREBP1. Whilst the physiological relevance of this interaction has yet to be elucidated, these data raise the intriguing possibility that fat loss seen in laminopathies may be caused, at least in part, by reduced binding of the adipocyte differentiation factor SREBP1 to lamin A.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.