Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Biochem Pharmacol ; 163: 169-177, 2019 05.
Article in English | MEDLINE | ID: mdl-30772269

ABSTRACT

Adenylyl cyclases are key points for the integration of stimulatory and inhibitory G protein-coupled receptor (GPCR) signals. Adenylyl cyclase type 5 (AC5) is highly expressed in striatal medium spiny neurons (MSNs), and is known to play an important role in mediating striatal dopaminergic signaling. Dopaminergic signaling from the D1 expressing MSNs of the direct pathway, as well as the D2 expressing MSNs of the indirect pathway both function through the regulation of AC5 activity, controlling the production of the 2nd messenger cAMP, and subsequently the downstream effectors. Here, we used a newly developed cell line that used Crispr-Cas9 to eliminate the predominant adenylyl cyclase isoforms to more accurately characterize a series of AC5 gain-of-function mutations which have been identified in ADCY5-related dyskinesias. Our results demonstrate that these AC5 mutants exhibit enhanced activity to Gαs-mediated stimulation in both cell and membrane-based assays. We further show that the increased cAMP response at the membrane effectively translates into increased downstream gene transcription in a neuronal model. Subsequent analysis of inhibitory pathways show that the AC5 mutants exhibit significantly reduced inhibition following D2 dopamine receptor activation. Finally, we demonstrate that an adenylyl cyclase "P-site" inhibitor, SQ22536 may represent an effective future therapeutic mechanism by preferentially inhibiting the overactive AC5 gain-of-function mutants.


Subject(s)
Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Dyskinesias/genetics , Dyskinesias/metabolism , Gain of Function Mutation/physiology , Genetic Variation/physiology , Adenylyl Cyclase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Gene Knockdown Techniques , HEK293 Cells , Humans , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism
2.
Hum Genet ; 137(10): 807-815, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30276537

ABSTRACT

Hundreds of genes have been implicated in autism spectrum disorders (ASDs). In genetically heterogeneous conditions, large families with multiple affected individuals provide strong evidence implicating a rare variant, and replication of the same variant in multiple families is unusual. We previously published linkage analyses and follow-up exome sequencing in seven large families with ASDs, implicating 14 rare exome variants. These included rs200195897, which was transmitted to four affected individuals in one family. We attempted replication of those variants in the MSSNG database. MSSNG is a unique resource for replication of ASD risk loci, containing whole genome sequence (WGS) on thousands of individuals diagnosed with ASDs and family members. For each exome variant, we obtained all carriers and their relatives in MSSNG, using a TDT test to quantify evidence for transmission and association. We replicated the transmission of rs200195897 to four affected individuals in three additional families. rs200195897 was also present in three singleton affected individuals, and no unaffected individuals other than transmitting parents. We identified two additional rare variants (rs566472488 and rs185038034) transmitted with rs200195897 on 1p36.33. Sanger sequencing confirmed the presence of these variants in the original family segregating rs200195897. To our knowledge, this is the first example of a rare haplotype being transmitted with ASD in multiple families. The candidate risk variants include a missense mutation in SAMD11, an intronic variant in NOC2L, and a regulatory region variant close to both genes. NOC2L is a transcription repressor, and several genes involved in transcription regulation have been previously associated with ASDs.


Subject(s)
Autism Spectrum Disorder/genetics , Eye Proteins/genetics , Genetic Loci , Haplotypes , Mutation, Missense , Polymorphism, Genetic , Repressor Proteins/genetics , Female , Humans , Male , Risk Factors
3.
Mol Psychiatry ; 23(2): 231-239, 2018 02.
Article in English | MEDLINE | ID: mdl-27956748

ABSTRACT

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder often accompanied by intellectual disability, language impairment and medical co-morbidities. The heritability of autism is high and multiple genes have been implicated as causal. However, most of these genes have been identified in de novo cases. To further the understanding of familial autism, we performed whole-exome sequencing on five families in which second- and third-degree relatives were affected. By focusing on novel and protein-altering variants, we identified a small set of candidate genes. Among these, a novel private missense C1143F variant in the second intracellular loop of the voltage-gated sodium channel NaV1.7, encoded by the SCN9A gene, was identified in one family. Through electrophysiological analysis, we show that NaV1.7C1143F exhibits partial loss-of-function effects, resulting in slower recovery from inactivation and decreased excitability in cultured cortical neurons. Furthermore, for the same intracellular loop of NaV1.7, we found an excess of rare variants in a case-control variant-burden study. Functional analysis of one of these variants, M932L/V991L, also demonstrated reduced firing in cortical neurons. However, although this variant is rare in Caucasians, it is frequent in Latino population, suggesting that genetic background can alter its effects on phenotype. Although the involvement of the SCN1A and SCN2A genes encoding NaV1.1 and NaV1.2 channels in de novo ASD has previously been demonstrated, our study indicates the involvement of inherited SCN9A variants and partial loss-of-function of NaV1.7 channels in the etiology of rare familial ASD.


Subject(s)
Autistic Disorder/genetics , NAV1.7 Voltage-Gated Sodium Channel/genetics , Autism Spectrum Disorder/genetics , Case-Control Studies , Family , Female , Humans , Intellectual Disability/genetics , Male , Mutation , Mutation, Missense/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Neurons/physiology , Phenotype , Sodium Channels/genetics , Exome Sequencing
4.
Am J Med Genet B Neuropsychiatr Genet ; 156B(3): 303-11, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21438139

ABSTRACT

Linkage to 7q has been the most robust genetic finding in familial autism. A previous scan of multiplex families with autism spectrum disorders found a linkage signal of genome-wide significance at D7S530 on 7q32. We searched a candidate imprinted region at this location for genetic variants in families with positive linkage scores. Using exon resequencing, we identified three rare potentially pathogenic variants in the TSGA14 gene, which encodes a centrosomal protein. Two variants were missense mutations (c.664C>G; p.P206A and c.766T>G; p.C240G) that changed conserved residues in the same protein domain; the third variant (c.192+5G>A) altered splicing, which resulted in a protein with an internal deletion of 16 residues and a G33D substitution. These rare TSGA14 variants are enriched in the affected subjects (6/348 patients versus 2/670 controls, Fisher's exact two tailed P = 0.022). This is the first report of a possible link of a gene with a centrosomal function with familial autism.


Subject(s)
Child Development Disorders, Pervasive/genetics , Mutation/genetics , Proteins/genetics , Alleles , Amino Acid Sequence , Case-Control Studies , Child , Child Development Disorders, Pervasive/ethnology , Chromosomes, Human, Pair 7/genetics , Family , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Linkage Disequilibrium/genetics , Male , Molecular Sequence Data , Pedigree , Proteins/chemistry , RNA Splicing/genetics , White People/genetics
5.
Neurology ; 65(5): 741-4, 2005 Sep 13.
Article in English | MEDLINE | ID: mdl-16157909

ABSTRACT

Referral-based studies indicate that a mutation (G2019S) in exon 41 of the LRRK2 gene might be a common cause of Parkinson disease (PD). The authors sequenced leucine-rich repeat kinase 2 (LRRK2) exons 31, 35, and 41 in 371 consecutively recruited patients with PD and found mutations in six (1.6%) subjects, including two heterozygous for new putative pathogenic variants (R1441H, IVS31 + 3A-->G). These data confirm the important contribution of LRRK2 to PD susceptibility in a clinic-based population.


Subject(s)
Genetic Predisposition to Disease/genetics , Mutation/genetics , Parkinson Disease/genetics , Protein Serine-Threonine Kinases/genetics , Aged , DNA Mutational Analysis , Exons/genetics , Family Health , Female , Gene Frequency/genetics , Genetic Testing , Genotype , Heterozygote , Homozygote , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Middle Aged , Parkinson Disease/metabolism , Pedigree
6.
Neurology ; 64(7): 1258-60, 2005 Apr 12.
Article in English | MEDLINE | ID: mdl-15824357

ABSTRACT

Spinocerebellar ataxia 14 (SCA14) is associated with missense mutations in the protein kinase C gamma gene (PRKCG), rather than a nucleotide repeat expansion. In this large-scale study of PRKCG in patients with ataxia, two new missense mutations, an in-frame deletion, and a possible splice site mutation were found and can now be added to the four previously described missense mutations. The genotype/phenotype correlations in these families are described.


Subject(s)
Genetic Predisposition to Disease/genetics , Mutation/genetics , Protein Kinase C/genetics , Spinocerebellar Ataxias/enzymology , Spinocerebellar Ataxias/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , DNA Mutational Analysis , Female , Gene Deletion , Genetic Testing , Genotype , Humans , Male , Middle Aged , Mutation, Missense/genetics , Phenotype , Protein Kinase C/chemistry , Protein Structure, Tertiary/genetics , RNA Splice Sites/genetics , Spinocerebellar Ataxias/physiopathology
7.
Mol Psychiatry ; 10(7): 699-711, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15753956

ABSTRACT

Dyslexia is a common and complex developmental disorder manifested by unexpected difficulty in learning to read. Multiple different measures are used for diagnosis, and may reflect different biological pathways related to the disorder. Impaired phonological decoding (translation of written words without meaning cues into spoken words) is thought to be a core deficit. We present a genome scan of two continuous measures of phonological decoding ability: phonemic decoding efficiency (PDE) and word attack (WA). PDE measures both accuracy and speed of phonological decoding, whereas WA measures accuracy alone. Multipoint variance component linkage analyses (VC) and Markov chain Monte-Carlo (MCMC) multipoint joint linkage and segregation analyses were performed on 108 families. A strong signal was observed on chromosome 2 for PDE using both VC (LOD=2.65) and MCMC methods (intensity ratio (IR)=32.1). The IR is an estimate of the ratio of the posterior to prior probability of linkage in MCMC analysis. The chromosome 2 signal was not seen for WA. More detailed mapping with additional markers provided statistically significant evidence for linkage of PDE to chromosome 2, with VC-LOD=3.0 and IR=59.6 at D2S1399. Parametric analyses of PDE, using a model obtained by complex segregation analysis, provided a multipoint maximum LOD=2.89. The consistency of results from three analytic approaches provides strong evidence for a locus on chromosome 2 that influences speed but not accuracy of phonological decoding.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 2/genetics , Dyslexia/genetics , Adolescent , Adult , Articulation Disorders/genetics , Child , DNA/analysis , Family Health , Genetic Linkage , Humans , Lod Score , Markov Chains , Monte Carlo Method , Pedigree , Phenotype , Quantitative Trait Loci/genetics
8.
Am J Hum Genet ; 67(3): 631-46, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10924405

ABSTRACT

Dyslexia is a common and complex disorder with evidence for a genetic component. Multiple loci (i.e., quantitative-trait loci [QTLs]) are likely to be involved, but the number is unknown. Diagnosis is complicated by the lack of a standard protocol, and many diagnostic measures have been proposed as understanding of the component processes has evolved. One or more genes may, in turn, influence these measures. To date, little work has been done to evaluate the mode of inheritance of individual component-as opposed to composite-phenotypes, beyond family or twin correlation studies that initially demonstrate evidence for a genetic basis of such components. Here we use two approaches to segregation analysis in 102 nuclear families to estimate genetic models for component phenotypes associated with dyslexia: digit span and a nonword-repetition task. Both measures are related to phonological skills, one of the key component processes in dyslexia. We use oligogenic-trait segregation analysis to estimate the number of QTLs contributing to each phenotype, and we use complex segregation analysis to identify the most parsimonious inheritance models. We provide evidence in support of both a major-gene mode of inheritance for the nonword-repetition task, with approximately 2.4 contributing QTLs, and for a genetic basis of digit span, with approximately 1.9 contributing QTLs. Results obtained by reciprocal adjustment of measures suggest that genes contributing to digit span may contribute to the nonword-repetition score but that there are additional QTLs involved in nonword repetition. Our study adds to existing studies of the genetic basis of composite phenotypes related to dyslexia, by providing evidence for major-gene modes of inheritance of these single-measure component phenotypes.


Subject(s)
Chromosome Segregation/genetics , Dyslexia/genetics , Dyslexia/physiopathology , Fingers/physiology , Language , Memory/physiology , Age Factors , Environment , Humans , Intelligence Tests , Language Tests , Models, Genetic , Multifactorial Inheritance/genetics , Nuclear Family , Quantitative Trait, Heritable , Sex Factors , Statistics as Topic
9.
Blood ; 95(7): 2262-8, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10733494

ABSTRACT

X-linked thrombocytopenia with thalassemia (XLTT; Online Mendelian Inheritance in Man [OMIM] accession number 314050) is a rare disorder characterized by thrombocytopenia, platelet dysfunction, splenomegaly, reticulocytosis, and unbalanced hemoglobin chain synthesis. In a 4-generation family, the gene responsible for XLTT was mapped to the X chromosome, short arm, bands 11-12 (band Xp11-12). The maximum lod score possible in this family, 2.39, was obtained for markers DXS8054 and DXS1003, at a recombination fraction of 0. Recombination events observed for XLTT and markers DXS8080 and DXS8023 or DXS991 define a critical region that is less than or equal to 7.65 KcM and contains the gene responsible for the Wiskott-Aldrich syndrome (WAS; OMIM accession number 301000) and its allelic variant X-linked thrombocytopenia (XLT; OMIM accession number 313900). Manifestations of WAS include thrombocytopenia, eczema, and immunodeficiency. In WAS/XLT the platelets are usually small, and bleeding is proportional to the degree of thrombocytopenia. In contrast, in XLTT the platelet morphology is normal, and the bleeding time is disproportionately prolonged. In this study no alteration in the WAS gene was detected by Northern blot or Western blot analysis, flow cytometry, or complimentary DNA dideoxynucleotide fingerprinting or sequencing. As has been reported for WAS and some cases of XLT, almost total inactivation of the XLTT gene-bearing X chromosome was observed in granulocytes and peripheral blood mononuclear cells from 1 asymptomatic obligate carrier. The XLTT carrier previously found to have an elevated alpha:beta hemoglobin chain ratio had a skewed, but not clonal, X-inactivation pattern favoring activity of the abnormal allele. Clinical differences and results of the mutation analyses make it very unlikely that XLTT is another allelic variant of WAS/XLT and strongly suggest that X-linked thrombocytopenia mapping to band Xp11-12 is a genetically heterogeneous disorder.


Subject(s)
Genetic Linkage , Thalassemia/genetics , Thrombocytopenia/genetics , X Chromosome , Blotting, Northern , Blotting, Western , Chromosome Mapping , Dosage Compensation, Genetic , Heterozygote , Humans , Male , Pedigree , Proteins/genetics , Thalassemia/complications , Thrombocytopenia/complications , Wiskott-Aldrich Syndrome Protein
10.
Behav Genet ; 30(5): 385-96, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11235984

ABSTRACT

There is evidence for genetic contributions to reading disability, but the phenotypic heterogeneity associated with the clinical diagnosis may make identification of the underlying genetic basis difficult. In order to elucidate distinct phenotypic features that may be contributing to the genotypic heterogeneity, we assessed the familial aggregation patterns of Verbal IQ and 24 phenotypic measures associated with dyslexia in 102 nuclear families ascertained through probands in grades 1 through 6 who met the criteria for this disorder. Correlations between relatives were computed for all diagnostic phenotypes, using a generalized estimating equation (GEE) approach. GEE is a recently developed semiparametric method for handling correlated data. The method is robust to model misspecification and flexible in adjusting for the subjects' characteristics and pedigree sizes as well as for the ascertainment process, while estimating the correlations between related subjects. The Nonword Memory (NWM) subtest of a prepublication version of the Comprehensive Test of Phonological Processing (CTOPP) and Phonemic Decoding Efficiency (PDE) subtest of a prepublication version of the Test of Word Reading Efficiency (TOWRE) showed correlation patterns in relatives that are strongly supportive of a genetic basis. The Wechsler Scale Digit Span, the Word Attack subtest of the Woodcock Reading Mastery Test--Revised, and the Spelling subtest of the Wide Range Achievement Test--Third Edition had slightly weaker evidence of a genetic basis. Five additional phenotypes (the Spelling subtest of the Wechsler Individual Achievement Test, the Accuracy, Rate, and Comprehension subtests of the Gray Oral Reading Test--Third Edition, and Rapid Automatized Naming of Letters and Numbers) gave suggestive evidence of such a pattern. The results cross-validate in that evidence for a pattern consistent with a genetic basis was obtained for two measures of phonological short-term memory (CTOPP Nonword Memory and WISCIII or WAIS-R Digit Span), for two measures of phonological decoding (WRMT-R Word Attack and TOWRE Phonemic Decoding Efficiency), and for two measures of spelling from dictation (WRAT-3 Spelling and, to a lesser extent, WIAT Spelling). These measures are thus good candidates for more sophisticated segregation analyses that can formulate models for incorporation into linkage analyses.


Subject(s)
Dyslexia/genetics , Phenotype , Adult , Child , Dyslexia/diagnosis , Female , Genetic Testing , Humans , Intelligence/genetics , Male , Middle Aged , Neuropsychological Tests
11.
Hum Mol Genet ; 8(5): 743-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10196363

ABSTRACT

X-linked sideroblastic anemia and ataxia (XLSA/A) is a recessive disorder characterized by an infantile to early childhood onset of non-progressive cerebellar ataxia and mild anemia with hypochromia and microcytosis. A gene encoding an ATP-binding cassette (ABC) transporter was mapped to Xq13, a region previously shown by linkage analysis to harbor the XLSA/A gene. This gene, ABC7, is an ortholog of the yeast ATM1 gene whose product localizes to the mitochondrial inner membrane and is involved in iron homeostasis. The full-length ABC7 cDNA was cloned and the entire coding region screened for mutations in a kindred in which five male members manifested XLSA/A. An I400M variant was identified in a predicted transmembrane segment of the ABC7 gene in patients with XLSA/A. The mutation was shown to segregate with the disease in the family and was not detected in at least 600 chromosomes of general population controls. Introduction of the corresponding mutation into the Saccharomyces cerevisiae ATM1 gene resulted in a partial loss of function of the yeast Atm1 protein. In addition, the human wild-type ABC7 protein was able to complement ATM1 deletion in yeast. These data indicate that ABC7 is the causal gene of XLSA/A and that XLSA/A is a mitochondrial disease caused by a mutation in the nuclear genome.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Anemia, Sideroblastic/genetics , Cerebellar Ataxia/genetics , Mutation , Saccharomyces cerevisiae Proteins , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/metabolism , Amino Acid Sequence , Amino Acid Substitution , Blotting, Northern , Cloning, Molecular , Female , Ferrous Compounds/pharmacology , Fungal Proteins/drug effects , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genetic Complementation Test , Humans , Iron/metabolism , Male , Mitochondria/metabolism , Molecular Sequence Data , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , X Chromosome
12.
J Hand Surg Am ; 24(2): 339-44, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10194020

ABSTRACT

X chromosome inactivation patterns at the androgen receptor locus were evaluated to determine clonality in microdissected lesional tissue and in leukocytes from 2 women with Dupuytren's disease. The tissue from both patients generated a polyclonal pattern of X chromosome inactivation of the human androgen receptor gene. This finding supports a polyclonal reactive process as the underlying etiology for palmar fibromatosis.


Subject(s)
Dosage Compensation, Genetic , Dupuytren Contracture/genetics , Receptors, Androgen/genetics , Female , Genotype , Humans , Leukocytes , Middle Aged , Restriction Mapping
13.
J Orthop Res ; 16(5): 521-30, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9820274

ABSTRACT

A human chondrosarcoma cell line has been established from an aggressive chondrosarcoma. The cells grow in a monolayer culture (doubling time: 2 days) and form aggregates. The aggregates consist of a rim of cells surrounding a hollow core. The cell line exhibits a unique pattern of mRNA expression with several molecules characteristic of the chondrocyte phenotype. Consistent with the chondrocyte phenotype, mRNAs encoding types IX and XI collagens were present along with an abundant expression of mRNAs encoding the core protein of the cartilage proteoglycans biglycan and aggrecan. No expression of mRNAs encoding types I or II fibrillar collagens or the proteoglycan decorin was observed. Sodium dodecyl sulfate-polyacrylamide gel electrophoretic analysis of [35S]sulfate-radiolabeled material confirmed the translation of proteoglycans containing glycosaminoglycan chains. The expression of molecules that contribute to cartilage development and tumorigenesis was examined. The cell line produces abundant mRNA that encodes transforming growth factor-beta1, a member of a family of cartilage and bone inductive proteins. The expression of mRNA encoding two proteins associated specifically with chondrogenesis was detected: Cart-1, a homeobox protein involved in cartilage differentiation, and CD-RAP, a secreted molecule restricted under normal conditions to differentiating chondrocytes and cartilage. Overexpression of p53, a tumor-suppressor gene, was detected. DNA analysis revealed a loss of heterozygosity at the chromosomal locus encoding p53, with the deletion of one p53 allele and the mutation of the remaining allele in both the parent tumor and the cell line. The malignant chondrosarcoma phenotype may be related to the unique gene expression pattern that is characteristic in many ways of differentiating chondroblasts, as well as to the inactivation of the p53 function that could contribute to the proliferative capacity of the cell line. This cell line may serve as a biological model for further investigation of the etiology of human chondrosarcomas and for the synthesis and regulation of cartilage-specific genes.


Subject(s)
Bone Neoplasms , Cell Culture Techniques/methods , Chondrosarcoma , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Adult , Blotting, Northern , Cell Differentiation/physiology , Cell Division/physiology , Collagen/genetics , DNA-Binding Proteins/genetics , Heterozygote , Homeodomain Proteins , Humans , Male , Neoplasm Proteins , Proteins/genetics , Proteoglycans/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology , Tumor Suppressor Protein p53/genetics
14.
Hum Mol Genet ; 7(11): 1779-86, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9736780

ABSTRACT

Autosomal dominant familial spastic paraplegia (FSP) is a genetically heterogeneous neurodegenerative disorder displaying anticipation for which three loci have been mapped to the chromosomal positions 14q11.2-q24.3 (SPG3), 2p21-p24 (SPG4) and 15q11.1 (SPG6). The repeat expansion detection (RED) method has been used to demonstrate expanded CAG repeats in some FSP families that map to SPG4. We analyzed 20 FSP families, including four for which there is evidence for linkage to SPG4, and found that in most cases the repeat expansion detected by RED is due to non-pathogenic expansions of the chromosome 18q21.1 SEF2-1 or 17q21.3 ERDA1 locus. Polymorphic expansions at SEF2-1 and ERDA1 appear frequent and may confound RED studies in the search for genes causing disorders demonstrating anticipation. In six FSP families, however, CAG repeat expansion was detected in a subset of affected and at-risk individuals that did not result from expansion of the SEF2-1 and ERDA1 loci. Overall, 11 of 37 (30%) of the FSP patients with a CAG/CTG repeat expansion are unaccounted for by the SEF2-1 and ERDA1 loci, compared with two of 23 (9%) of the unaffected at-risk individuals and none of 19 controls. In the majority of cases these novel expansions were shorter than those previously reported.


Subject(s)
Genes, Dominant , Spastic Paraplegia, Hereditary/genetics , Trinucleotide Repeat Expansion , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Blotting, Western , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 18 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Female , Genetic Linkage , Humans , Male , Pedigree , TATA-Box Binding Protein , TCF Transcription Factors , Trans-Activators/genetics , Transcription Factor 4 , Transcription Factor 7-Like 2 Protein , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
15.
Am J Hum Genet ; 62(2): 346-54, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9463333

ABSTRACT

Hereditary multiple exostoses (EXT; MIM 133700) is an autosomal dominant bone disorder characterized by the presence of multiple benign cartilage-capped tumors (exostoses). Besides suffering complications caused by the pressure of these exostoses on the surrounding tissues, EXT patients are at an increased risk for malignant chondrosarcoma, which may develop from an exostosis. EXT is genetically heterogeneous, and three loci have been identified so far: EXT1, on chromosome 8q23-q24; EXT2, on 11p11-p12; and EXT3, on the short arm of chromosome 19. The EXT1 and EXT2 genes were cloned recently, and they were shown to be homologous. We have now analyzed the EXT1 and EXT2 genes, in 26 EXT families originating from nine countries, to identify the underlying disease-causing mutation. Of the 26 families, 10 families had an EXT1 mutation, and 10 had an EXT2 mutation. Twelve of these mutations have never been described before. In addition, we have reviewed all EXT1 and EXT2 mutations reported so far, to determine the nature, frequency, and distribution of mutations that cause EXT. From this analysis, we conclude that mutations in either the EXT1 or the EXT2 gene are responsible for the majority of EXT cases. Most of the mutations in EXT1 and EXT2 cause premature termination of the EXT proteins, whereas missense mutations are rare. The development is thus mainly due to loss of function of the EXT genes, consistent with the hypothesis that the EXT genes have a tumor- suppressor function.


Subject(s)
Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 8 , Exostoses, Multiple Hereditary/genetics , Mutation , N-Acetylglucosaminyltransferases , Proteins/genetics , Chromosome Mapping , DNA Primers , Exons , Family , Female , Frameshift Mutation , Genes, Tumor Suppressor , Humans , Introns , Male , Point Mutation , Sequence Deletion
16.
Hum Mutat ; 11(3): 231-9, 1998.
Article in English | MEDLINE | ID: mdl-9521425

ABSTRACT

Hereditary multiple exostoses (EXT) is an autosomal dominant disorder characterized by growth of benign bone tumors. Three chromosomal loci have been implicated in this genetically heterogeneous disease: EXT1 at 8q24, EXT2 at 11p13, and EXT3 on 19p. EXT1 and EXT2 were recently cloned. We evaluated 34 families with EXT to estimate the proportion of disease attributable to EXT1, EXT2, and EXT3 and to investigate the spectrum of EXT1 mutations. Linkage analyses combined with heterogeneity testing provides strong evidence in favor of linkage of disease to both chromosomes 8 and 11, but does not support evidence of linkage to chromosome 19 in this data set. The 11 EXT1 exons were PCR-amplified and sequenced in all 11 isolated cases and in 20 of the 23 familial cases. Twelve different novel EXT1 mutations were detected, including 5 frame-shift deletions or insertions, 1 codon deletion, and 6 single base-pair substitutions distributed across 8 of the exons. Only 2 of the mutations were detected in more than one family. Three mutations affect sites in which alterations were previously reported. Nonchain-terminating missense mutations were identified in codons 280 and 340, both coding for conserved arginine residues. These residues may be crucial to the function of this protein. Although the prevalence of EXT has been estimated to be approximately 1/50,000 individuals, the disease has been reported to occur much more frequently in the Chamorro natives on Guam. Our detection of an EXT1 mutation in one Chamorro subject will allow investigation of a possible founder effect in this population. Combined mutational and heterogeneity analyses in this set of families with multiple exostoses suggest that 66% of our total sample, including 45% of isolated and 77% of familial cases, are attributable to abnormalities in EXT1.


Subject(s)
Exostoses, Multiple Hereditary/genetics , Genetic Heterogeneity , Genetic Linkage , Mutation/genetics , N-Acetylglucosaminyltransferases , Proteins/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 8/genetics , DNA Mutational Analysis , Female , Humans , Male , Pedigree
17.
Hum Genet ; 102(1): 93-7, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9490305

ABSTRACT

Paroxysmal dystonic choreoathetosis (PDC) is a rare neurological disorder characterized by episodes of involuntary movement, involving the extremities and face, which may occur spontaneously or be precipitated by caffeine, alcohol, anxiety, and fatigue. PDC is transmitted as an autosomal dominant trait with incomplete penetrance. A gene implicated in this paroxysmal disorder has been mapped to a 10-15 cM region on chromosome 2q31-36 in two families. We describe a third family with PDC. Two-point linkage analyses with markers linked to the candidate PDC locus were performed. A maximum two-point LOD score of 4.20 at a recombination fraction of zero was obtained for marker D2S120, confirming linkage to the distal portion of chromosome 2q. The anion exchanger gene, SLC2C, maps to this region, but the family was poorly informative for polymorphic markers within and flanking this candidate gene. Haplotype analysis revealed a critical recombination event that confines the PDC gene to a 5-cM region bounded by the markers D2S164 and D2S377. We compared the haplotype in our family with that in another chromosome 2-linked PDC family, but did not detect a region of shared genotypes. However, identifying a third family whose disease maps to the same region and narrowing the critical region will facilitate identification of the 2q-linked PDC gene.


Subject(s)
Anion Transport Proteins , Antiporters , Athetosis/genetics , Chorea/genetics , Chromosome Mapping , Chromosomes, Human, Pair 2/genetics , Dystonia/genetics , Adult , Female , Genetic Linkage , Genetic Markers , Humans , Male , Membrane Proteins/genetics , SLC4A Proteins , Syndrome
18.
Leukemia ; 12(2): 108-16, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9519769

ABSTRACT

In November 1996, word reached the University of Washington that Philip Fialkow and his wife, Helen, had died while trekking in Nepal. Over a 30-year period, Dr Fialkow and his colleagues used the cellular mosaicism resulting from X-chromosome inactivation in females as a marker system to investigate the clonal development of human hematopoietic disorders. This review discusses the impact that these studies have had on our understanding of hematopoietic stem cell relationships and the pathogenesis of human neoplasia in general. To appreciate the special role played by studies on clonality, it is necessary to consider how little was known about the origin of leukemias and myeloproliferative disorders and the limited techniques available for their study in the early to mid 1960s. Dr Fialkow and his coworkers were the first to show that myeloproliferative disorders and acute myelogenous leukemias (AML) are clonal diseases at the time of diagnosis and to elucidate the level of differentiation manifested by the originating cell type. Although the myelodysplastic disorders were found to involve a pluripotent stem cell, heterogeneity was found in the level of stem cell involvement in AML. Evidence was obtained to support a multistep pathogenesis of these diseases as well as a clonal but cytogenetically normal stage in some cases of Ph-positive chronic myelogenous leukemia, AML, acute lymphoblastic leukemia and myelodysplasia.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia/genetics , Leukemia/pathology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Cell Differentiation/physiology , Female , Humans , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology
19.
Am J Hum Genet ; 61(4): 873-81, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9382098

ABSTRACT

The identification of genes responsible for the rare cases of familial leukemia may afford insight into the mechanism underlying the more common sporadic occurrences. Here we test a single family with 11 relevant meioses transmitting autosomal dominant acute myelogenous leukemia (AML) and myelodysplasia for linkage to three potential candidate loci. In a different family with inherited AML, linkage to chromosome 21q22.1-22.2 was recently reported; we exclude linkage to 21q22.1-22.2, demonstrating that familial AML is a heterogeneous disease. After reviewing familial leukemia and observing anticipation in the form of a declining age of onset with each generation, we had proposed 9p21-22 and 16q22 as additional candidate loci. Whereas linkage to 9p21-22 can be excluded, the finding of a maximum two-point LOD score of 2.82 with the microsatellite marker D16S522 at a recombination fraction theta = 0 provides evidence supporting linkage to 16q22. Haplotype analysis reveals a 23.5-cM (17.9-Mb) commonly inherited region among all affected family members extending from D16S451 to D16S289. In order to extract maximum linkage information with missing individuals, incomplete informativeness with individual markers in this interval, and possible deviance from strict autosomal dominant inheritance, we performed nonparametric linkage analysis (NPL) and found a maximum NPL statistic corresponding to a P-value of .00098, close to the maximum conditional probability of linkage expected for a pedigree with this structure. Mutational analysis in this region specifically excludes expansion of the AT-rich minisatellite repeat FRA16B fragile site and the CAG trinucleotide repeat in the E2F-4 transcription factor. The "repeat expansion detection" method, capable of detecting dynamic mutation associated with anticipation, more generally excludes large CAG repeat expansion as a cause of leukemia in this family.


Subject(s)
Chromosomes, Human, Pair 16 , Leukemia, Myeloid, Acute/genetics , Chromosome Mapping , Family , Female , Genes, Dominant , Genetic Linkage , Genetic Markers , Humans , Lod Score , Male , Minisatellite Repeats , Myelodysplastic Syndromes/genetics , Pedigree , Polymerase Chain Reaction/methods , Statistics, Nonparametric , Trinucleotide Repeats
20.
Am J Med Genet ; 74(1): 26-36, 1997 Feb 21.
Article in English | MEDLINE | ID: mdl-9034002

ABSTRACT

Familial spastic paraparesis (SPG) is a clinically and genetically heterogeneous group of disorders. At least three loci have been implicated in autosomal dominant pure SPG and mutations in either of two loci may cause the X-linked form. Although the penetrance is high for all forms by age 60, there is wide variation in clinical characteristics, including age of onset. Two-point and multi-point linkage analyses in nine families provided supportive evidence that the most common form of SPG is linked to chromosome 2 (SPG4). Haplotype analysis localized the critical region to a 6 cM interval between D2S392 and D2S367. By haplotype analysis, the disease in at least one family does not appear to be linked to any of the presently known SPG loci, suggesting that there is at least one additional SPG gene. Evaluation at ages of onset in 11 families gave suggestive evidence for anticipation with mean age of onset in parents (41.3 years) being older than mean age of onset in children (26.9 years; P < 0.005).


Subject(s)
Chromosomes, Human, Pair 2/genetics , Spastic Paraplegia, Hereditary/genetics , Age of Onset , Chromosome Mapping , Female , Genetic Linkage , Genetic Markers/genetics , Haplotypes/genetics , Humans , Male , Pedigree , Recombination, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...