Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
J Med Genet ; 43(7): 576-81, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16371502

ABSTRACT

INTRODUCTION: The majority of hearing loss in children can be accounted for by genetic causes. Non-syndromic hearing loss accounts for 80% of genetic hearing loss in children, with mutations in DFNB1/GJB2 being by far the most common cause. Among the second tier genetic causes of hearing loss in children are mutations in the DFNB9/OTOF gene. METHODS: In total, 65 recessive non-syndromic hearing loss families were screened by genotyping for association with the DFNB9/OTOF gene. Families with genotypes consistent with linkage or uninformative for linkage to this gene region were further screened for mutations in the 48 known coding exons of otoferlin. RESULTS: Eight OTOF pathological variants were discovered in six families. Of these, Q829X was found in two families. We also noted 23 other coding variant, believed to have no pathology. A previously published missense allele I515T was found in the heterozygous state in an individual who was observed to be temperature sensitive for the auditory neuropathy phenotype. CONCLUSIONS: Mutations in OTOF cause both profound hearing loss and a type of hearing loss where otoacoustic emissions are spared called auditory neuropathy.


Subject(s)
Connexins/genetics , Hearing Loss/genetics , Membrane Proteins/genetics , Mutation , Child , Chromosome Mapping , Connexin 26 , Family , Female , Genetic Variation , Genotype , Humans , Male
4.
Surg Endosc ; 16(3): 450-2, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11928026

ABSTRACT

BACKGROUND: The risk factors for gallstone disease are well known, but they have not been updated to take the development of better ultrasound technology and the advent of laparoscopic surgery into consideration. METHODS: We compared two groups of patients who underwent ultrasound-one group (n = 100) who underwent cholecystectomy after ultrasound revealed the presence of gallstones and a control group (n = 107) in whom no gallstones were shown on ultrasound. RESULTS: Seven patients in the control group refused to participate in the study; otherwise, the groups are sequential. Age in the surgery group was 51 years (+/- 16) vs 50 (+/- 16) for the control group. The percentage of female patients was 59% and 52%, respectively (p = ns). Body mass index was 32 (+/- 8) and 28 (+/- 6), respectively (p = 0.013). Parity > 2 was 0.49% and 0.37%, respectively (p = 0.000001). The number who breast-fed at least one child was 17 (24%) and eight (12%), respectively (p = 0.03). Oral contraceptive use was 37 (52%) and 17 (22%), respectively (p = 0.0005). Primary relatives who had had gallbladder surgery was 0.68 (+/- 1) and 0.35 (+/- 0.6), respectively (p = 0.02). CONCLUSION: Body mass index, breast-feeding, oral contraceptives, parity > 2, and family history were found to be risk factors for gallstone disease. Age and female sex were not, probably due to selection bias.


Subject(s)
Cholelithiasis/etiology , Body Mass Index , Case-Control Studies , Cholecystectomy/statistics & numerical data , Cholelithiasis/diagnostic imaging , Cholelithiasis/surgery , Contraceptives, Oral/administration & dosage , Family , Female , Humans , Male , Middle Aged , Parity , Risk Factors , Ultrasonography
5.
Genome Res ; 11(10): 1716-24, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11591648

ABSTRACT

Haplotype analysis of disease chromosomes can help identify probable historical recombination events and localize disease mutations. Most available analyses use only marginal and pairwise allele frequency information. We have developed a Bayesian framework that utilizes full haplotype information to overcome various complications such as multiple founders, unphased chromosomes, data contamination, and incomplete marker data. A stochastic model is used to describe the dependence structure among several variables characterizing the observed haplotypes, for example, the ancestral haplotypes and their ages, mutation rate, recombination events, and the location of the disease mutation. An efficient Markov chain Monte Carlo algorithm was developed for computing the estimates of the quantities of interest. The method is shown to perform well in both real data sets (cystic fibrosis data and Friedreich ataxia data) and simulated data sets. The program that implements the proposed method, BLADE, as well as the two real datasets, can be obtained from http://www.fas.harvard.edu/~junliu/TechRept/01folder/diseq_prog.tar.gz.


Subject(s)
Chromosome Mapping/statistics & numerical data , Cystic Fibrosis/genetics , Friedreich Ataxia/genetics , Haplotypes/genetics , Linkage Disequilibrium/genetics , Bayes Theorem , Computer Simulation , Humans , Models, Statistical
6.
J Mol Evol ; 52(3): 232-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11428460

ABSTRACT

Friedreich ataxia is an autosomal recessive neurodegenerative disorder associated with a GAA repeat expansion in the first intron of the gene (FRDA) encoding a novel, highly conserved, 210 amino acid protein known as frataxin. Normal variation in repeat size was determined by analysis of more than 600 DNA samples from seven human populations. This analysis showed that the most frequent allele had nine GAA repeats, and no alleles with fewer than five GAA repeats were found. The European and Syrian populations had the highest percentage of alleles with 10 or more GAA repeats, while the Papua New Guinea population did not have any alleles carrying more than 10 GAA repeats. The distributions of repeat sizes in the European, Syrian, and African American populations were significantly different from those in the Asian and Papua New Guinea populations (p < 0.001). The GAA repeat size was also determined in five nonhuman primates. Samples from 10 chimpanzees, 3 orangutans, 1 gorilla, 1 rhesus macaque, 1 mangabey, and 1 tamarin were analyzed. Among those primates belonging to the Pongidae family, the chimpanzees were found to carry three or four GAA repeats, the orangutans had four or five GAA repeats, and the gorilla carried three GAA repeats. In primates belonging to the Cercopithecidae family, three GAA repeats were found in the mangabey and two in the rhesus macaque. However, an AluY subfamily member inserted in the poly(A) tract preceding the GAA repeat region in the rhesus macaque, making the amplified sequence approximately 300 bp longer. The GAA repeat was also found in the tamarin, suggesting that it arose at least 40 million years ago and remained relatively small throughout the majority of primate evolution, with a punctuated expansion in the human genome.


Subject(s)
Friedreich Ataxia/genetics , Genetic Variation , Phylogeny , Trinucleotide Repeats/genetics , Alleles , Animals , Base Sequence , Cercocebus atys/genetics , Evolution, Molecular , Hominidae/genetics , Humans , Macaca mulatta/genetics , Molecular Sequence Data , Primates/genetics , Saguinus/genetics , Sequence Alignment
7.
Am J Ophthalmol ; 131(3): 355-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11239869

ABSTRACT

PURPOSE: To search for patients with Usher syndrome type IC among those with Usher syndrome type I who reside in New England. METHODS: Genotype analysis of microsatellite markers closely linked to the USH1C locus was done using the polymerase chain reaction. We compared the haplotype of our patients who were homozygous in the USH1C region with the haplotypes found in previously reported USH1C Acadian families who reside in southwestern Louisiana and from a single family residing in Lebanon. RESULTS: Of 46 unrelated cases of Usher syndrome type I residing in New England, two were homozygous at genetic markers in the USH1C region. Of these, one carried the Acadian USH1C haplotype and had Acadian ancestors (that is, from Nova Scotia) who did not participate in the 1755 migration of Acadians to Louisiana. The second family had a haplotype that proved to be the same as that of a family with USH1C residing in Lebanon. Each of the two families had haplotypes distinct from the other. CONCLUSION: This is the first report that some patients residing in New England have Usher syndrome type IC. Patients with Usher syndrome type IC can have the Acadian haplotype or the Lebanese haplotype compatible with the idea that at least two independently arising pathogenic mutations have occurred in the yet-to-be identified USH1C gene.


Subject(s)
Carrier Proteins/genetics , Deafness/genetics , Haplotypes , Retinitis Pigmentosa/genetics , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Cytoskeletal Proteins , DNA Mutational Analysis , Deafness/classification , Deafness/congenital , Deafness/ethnology , Female , Genetic Linkage/genetics , Humans , Male , Microsatellite Repeats , New England/epidemiology , Pedigree , Retinitis Pigmentosa/classification , Retinitis Pigmentosa/ethnology , Syndrome
8.
Nat Genet ; 26(1): 51-5, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973247

ABSTRACT

Usher syndrome type 1 (USH1) is an autosomal recessive sensory defect involving congenital profound sensorineural deafness, vestibular dysfunction and blindness (due to progressive retinitis pigmentosa)1. Six different USH1 loci have been reported. So far, only MYO7A (USH1B), encoding myosin VIIA, has been identified as a gene whose mutation causes the disease. Here, we report a gene underlying USH1C (MIM 276904), a USH1 subtype described in a population of Acadian descendants from Louisiana and in a Lebanese family. We identified this gene (USH1C), encoding a PDZ-domain-containing protein, harmonin, in a subtracted mouse cDNA library derived from inner ear sensory areas. In patients we found a splice-site mutation, a frameshift mutation and the expansion of an intronic variable number of tandem repeat (VNTR). We showed that, in the mouse inner ear, only the sensory hair cells express harmonin. The inner ear Ush1c transcripts predicted several harmonin isoforms, some containing an additional coiled-coil domain and a proline- and serine-rich region. As several of these transcripts were absent from the eye, we propose that USH1C also underlies the DFNB18 form of isolated deafness.


Subject(s)
Carrier Proteins/genetics , Frameshift Mutation , Hair Cells, Auditory, Inner/pathology , Hearing Loss, Sensorineural/genetics , Mutation , Retinal Degeneration/genetics , Adaptor Proteins, Signal Transducing , Alleles , Animals , Base Sequence , Blotting, Northern , Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Cell Cycle Proteins , Cytoskeletal Proteins , DNA Mutational Analysis , DNA, Complementary/metabolism , Exons , Family Health , Gene Deletion , Gene Library , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Vestibular/metabolism , Heterozygote , Humans , Immunohistochemistry , Introns , Mice , Minisatellite Repeats/genetics , Models, Genetic , Molecular Sequence Data , Pedigree , Protein Isoforms , Protein Structure, Tertiary , RNA Splicing/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Tissue Distribution , Transcription, Genetic
9.
Hum Hered ; 50(2): 126-32, 2000.
Article in English | MEDLINE | ID: mdl-10799971

ABSTRACT

Linkage analyses of simulated quantitative trait data were performed using the Haseman-Elston (H-E) sib pair regression test to investigate the effects of inaccurate allele frequency estimates on the type I error rates of this test. Computer simulations generating a quantitative trait in nuclear families were performed using GASP [1]. Assuming no linkage, several data sets were simulated; they differed in marker allele numbers and frequencies, number of sib pairs and number of sibships. Each set of simulated data was analyzed using (1) all parental marker data, (2) half of the parental marker data, and (3) no parental marker data, using both correct and incorrect allele frequencies in the latter 2 cases. The H-E sib pair linkage method was found to be robust to misspecification of marker allele frequencies regardless of the number of alleles.


Subject(s)
Chromosome Mapping/standards , Gene Frequency , Computer Simulation , Heterozygote , Humans , Quantitative Trait, Heritable , Research Design , Software
10.
Gene ; 246(1-2): 265-74, 2000 Apr 04.
Article in English | MEDLINE | ID: mdl-10767548

ABSTRACT

The combined DFNB7-DFNB11 deafness locus maps to chromosome 9q13-q21 between markers D9S1806 and D9S769. We have determined the cDNA sequence and genomic structure of a novel gene, TMEM2, that maps to this interval and is expressed in the cochlea. The mouse orthologue of this gene (Tmem2) maps to the murine dn (deafness) locus on mouse chromosome 19. Screens for transmembrane helices reveal the presence of at least one putative transmembrane domain in the TMEM2 protein. To determine whether mutations in TMEM2 cause hearing loss at the DFNB7-DFNB11 locus, we screened the coding region of this gene in DFNB7-DFNB11 affected families by direct sequencing. All DNA variants that segregated with the deafness and changed the predicted amino acid sequence of TMEM2 were common polymorphisms, as demonstrated by allele-specific amplification of pooled control DNA. Northern blot analysis showed no difference in transcript size or expression level of Tmem2 in dn/dn and control mice. The intragenic polymorphisms in TMEM2 represent a novel centromeric boundary for the DFNB7-DFNB11 interval.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Deafness/genetics , Genes/genetics , Membrane Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Blotting, Northern , Chromosome Mapping , Chromosomes/genetics , Cochlea/embryology , Cochlea/metabolism , Contig Mapping , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , DNA, Complementary/chemistry , DNA, Complementary/genetics , Exons , Family Health , Female , Gene Expression , Gene Expression Regulation, Developmental , Humans , Introns , Male , Mice , Molecular Sequence Data , Mutation , Pedigree , Polymorphism, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Tissue Distribution
11.
Genome Res ; 9(1): 7-16, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9927480

ABSTRACT

Hearing impairment is clinically and genetically heterogeneous. There are >400 disorders in which hearing impairment is a characteristic of the syndrome, and family studies demonstrate that there are at least 30 autosomal loci for nonsyndromic hearing impairment. The genes that have been identified encode diaphanous (HDIA1), alpha-tectorin (TECTA), the transcription factor POU4F3, connexin 26 (GJB2), and two unconventional myosins (MYO7A and MYO15), and four novel proteins (PDS, COCH, DFNA5, DFNB9). The same clinical phenotype in hearing-impaired individuals, even those within the same family, can result from mutations in different genes. Conversely, mutations in the same gene can result in a variety of clinical phenotypes with different modes of inheritance. For example, mutations in the gene encoding MYO7A cause Usher syndrome type IB, autosomal-recessive nonsyndromic hearing impairment (DFNB2), and autosomal-dominant nonsyndromic hearing impairment (DFNA11). Additionally, the mouse ortholog of the MYO7A gene is the shaker-1 gene. Mouse models such as shaker-1 have facilitated the identification of genes that cause hearing impairment in humans. The availability of high-resolution maps of the human and mouse genomes and new technologies for gene identification are advancing molecular understanding of hearing impairment and the complex mechanisms of the auditory system.


Subject(s)
Hearing Disorders/genetics , Animals , Connexin 26 , Connexins , Disease Models, Animal , Genome, Human , Hearing Disorders/history , History, 16th Century , History, 17th Century , History, 19th Century , History, 20th Century , Humans
12.
Am J Med Genet ; 89(3): 158-66, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10704190

ABSTRACT

Mutations in the gene (MYO7A) encoding myosin-VIIa, a member of the large superfamily of myosin motor proteins that move on cytoplasmic actin filaments, and in the USH2A gene, which encodes a novel protein resembling an extracellular matrix protein or a cell adhesion molecule, both cause Usher syndrome (USH), a clinically heterogeneous autosomal recessive disorder comprising hearing and visual impairment. Patients with USH1 have severe to profound congenital hearing impairment, vestibular dysfunction, and retinal degeneration beginning in childhood, while those with USH2 have moderate to severe hearing impairment, normal vestibular function, and later onset of retinal degeneration. USH3 is characterized by progressive hearing loss and variable age of onset of retinal degeneration. The phenotype resulting from MYO7A and USH2A mutations is variable. While most MYO7A mutations cause USH1, some cause nonsyndromic hearing impairment, and one USH3 phenotype has been described. USH2A mutations cause atypical USH as well as USH2. MYO7A is on chromosome region 11q13 and USH2A is on 1q41. Seven other USH genes have been mapped but have not yet been identified. USH1A, USH1C, USH1D, USH1E, and USH1F have been assigned to chromosome bands 14q32, 11p15.1, 10q, 21q21, and 10, respectively, while USH2B is on 5q, and USH3 is at 3q21-q25. Myosin VIIa mutations also result in the shaker-1 (sh1) mouse, providing a model for functional studies. One possibility is that myosin-VIIa is required for linking stereocilia in the sensory hair bundle; another is that it may be needed for membrane trafficking. The ongoing studies of myosin-VIIa, the USH2A protein, and the yet to be identified proteins encoded by the other USH genes will advance understanding of the Usher syndromes and contribute to the development of effective therapies. Am. J. Med. Genet. (Semin. Med. Genet.) 89:158-166, 1999.


Subject(s)
Deafness , Animals , Chromosome Mapping , Deafness/diagnosis , Deafness/genetics , Deafness/physiopathology , Humans , Mice , Mutation , Syndrome
13.
Gene ; 215(2): 461-9, 1998 Jul 30.
Article in English | MEDLINE | ID: mdl-9758550

ABSTRACT

The DFNB7/11 locus for autosomal recessive non-syndromic hearing loss (ARNSHL) has been mapped to an approx. 1.5 Mb interval on human chromosome 9q13-q21. We have determined the cDNA sequence and genomic structure of a novel cochlear-expressed gene, ZNF216, that maps to the DFNB7/11 interval. The mouse orthologue of this gene maps to the murine dn (deafness) locus on mouse chromosome 19. The ZNF216 gene is highly conserved between human and mouse, and contains two regions that show homology to the putative zinc linger domains of other proteins. To determine it mutations in ZNF216 might be the cause of hearing loss at the DFNB7/11 locus, we screened the coding region of this gene in DFNB7/11 families by direct sequencing. No potential disease-causing mutations were found. In addition, Northern blot analysis showed no difference in ZNF216 transcript size or abundance between dn and control mice. These data Suggest that the ZNF216 gene is unlikely to be responsible for hearing loss at the DFNB7/11 and dn loci.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 19 , Cochlea/metabolism , Hearing Loss/genetics , Proteins/genetics , Algorithms , Amino Acid Sequence , Animals , Base Sequence , Chromosomes, Artificial, Yeast , DNA Mutational Analysis , DNA-Binding Proteins , Exons , Fetus , Genes, Recessive , Human Genome Project , Humans , Introns , Mice , Molecular Sequence Data , Protein Biosynthesis , Proteins/chemistry , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Zinc Fingers
14.
Biochim Biophys Acta ; 1407(3): 257-62, 1998 Sep 30.
Article in English | MEDLINE | ID: mdl-9748617

ABSTRACT

Recombination data for the mouse deafness locus (dn) on chromosome 19 are consistent with the presence of an inversion for which one of the breakpoints is between D19Mit14 and D19Mit96, a distance of less than 226 kb. Fluorescence in situ hybridization studies using a bacterial artificial chromosome on interphase (G1) nuclei provide additional support for the presence of an inversion. The dn gene is probably the orthologue of the human DFNB7/DFNB11 gene on chromosome 9.


Subject(s)
Chromosome Inversion , Deafness/genetics , Genes , Animals , Fluorescein-5-isothiocyanate , Genotype , In Situ Hybridization, Fluorescence , Inbreeding , Mice , Polymerase Chain Reaction , Rhodamines
15.
Biochim Biophys Acta ; 1407(1): 84-91, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9639681

ABSTRACT

Usher syndrome type 1C (USH1C) occurs in a small population of Acadian descendants from southwestern Louisiana. Linkage and linkage disequilibrium analyses localize USH1C to chromosome 11p between markers D11S1397 and D11S1888, an interval of less than 680 kb. Here, we refine the USH1C linkage to a region less than 400 kb, between genetic markers D11S1397 and D11S1890. Using 17 genetic markers from this interval, we have isolated a contiguous set of 60 bacterial artificial chromosomes (BACs) that span the USH1C critical region. Exon trapping of BAC clones from this region resulted in the recovery of an exon of the nuclear EF-hand acidic (NEFA) gene. However, DNA sequence analysis of the NEFA cDNA from lymphocytes of affected individuals provided no evidence of mutation, making structural mutations in the NEFA protein unlikely as the cellular cause of Acadian Usher syndrome.


Subject(s)
Chromosome Mapping , Chromosomes, Human, Pair 11 , DNA-Binding Proteins/genetics , Hearing Loss, Sensorineural/genetics , Retinitis Pigmentosa/genetics , Bacteriophage P1/genetics , Calcium-Binding Proteins , Canada/ethnology , Chromosomes, Artificial, Yeast , Cloning, Molecular , France/ethnology , Hearing Loss, Sensorineural/classification , Hearing Loss, Sensorineural/epidemiology , Humans , Louisiana/epidemiology , Microsatellite Repeats , Nerve Tissue Proteins , Nucleobindins , Retinitis Pigmentosa/classification , Retinitis Pigmentosa/epidemiology , Sequence Analysis, DNA , Syndrome
16.
Hear Res ; 117(1-2): 24-30, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9557975

ABSTRACT

The f2/f1 frequency ratio of 1.3 in combination with stimulus levels of L1/L2 = 50/60 and 50/50 dB SPL produced a higher level of distortion product otoacoustic emissions (DPOAE) in the heterozygous (+/dn) mice than in the homozygous (+/+) mice. These results suggest that the dn gene carriers have a unique cochlear trait which may be related to the dn gene locus and expressed via a frequency- and intensity-dependent DPOAE function.


Subject(s)
Heterozygote , Homozygote , Otoacoustic Emissions, Spontaneous/genetics , Acoustic Stimulation , Animals , Deafness/genetics , Disease Models, Animal , Female , Male , Mice , Phenotype
17.
Am J Med Genet ; 74(5): 488-93, 1997 Sep 19.
Article in English | MEDLINE | ID: mdl-9342197

ABSTRACT

The CAG repeat tract at the autosomal dominant spinocerebellar ataxia type 1 (SCA1) locus was analyzed in SCA1 families and French-Acadian, African-American, Caucasian, Greenland Inuit, and Thai populations. The normal alleles had 9-37 repeats, whereas disease alleles contained 44-64 repeats. The CAG repeat tract contained one or two CAT interruptions in 44 of 47 normal human chromosomes and in all five chimpanzees examined. In contrast, no CAT interruptions were found in Old World monkeys or expanded human alleles. The number and positions of CAT interruptions may be important in stabilizing CAG repeat tracts in normal chromosomes. At least five codons occupy the region corresponding to the polyglutamine tract at the SCA1 locus in mice, rats, and other rodents. They comprise three or four CCN (coding for proline) in addition to one or two CAG repeats.


Subject(s)
Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Spinocerebellar Degenerations/genetics , Trinucleotide Repeats/genetics , Amino Acid Sequence , Animals , Ataxin-1 , Ataxins , Base Sequence , Cercocebus , Gerbillinae , Guinea Pigs , Humans , Macaca , Mice , Molecular Sequence Data , Pan troglodytes , Peromyscus , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sciuridae , Spinocerebellar Degenerations/etiology
18.
Ann Neurol ; 41(5): 675-82, 1997 May.
Article in English | MEDLINE | ID: mdl-9153531

ABSTRACT

We studied genotype-phenotype correlations in a group of 100 patients with typical Friedreich ataxia (FRDA), and in three groups of patients with atypical clinical presentations, including 44 Acadian FRDA, 8 late-onset FRDA (LOFA), and 6 FRDA with retained reflexes (FARR). All patients, except 3 with typical FRDA, carried two copies of the FRDA-associated GAA triplet repeat expansion. Overall, the phenotypic spectrum of FRDA appeared to be wider than defined by the currently used diagnostic criteria. Our study indicated the existence of several sources of variability in FRDA. Patients with larger GAA expansions tended to have earlier onset and were more likely to show additional manifestations of the disease. Mitotic instability of the expanded GAA repeats may partially account for the limited degree of correlation between expansion sizes as determined in lymphocytes and clinical parameters. Some clinical variants associated with specific FRDA haplotypes, such as Acadian FRDA and FARR, turned out to be unrelated to expansion sizes. No polymorphism in the frataxin coding sequence could be associated with these clinical variants.


Subject(s)
Friedreich Ataxia/classification , Friedreich Ataxia/genetics , Trinucleotide Repeats , Adolescent , Adult , Age of Onset , Child , DNA/analysis , Disease Progression , Friedreich Ataxia/epidemiology , Genotype , Humans , Phenotype
19.
Hum Mutat ; 9(5): 402-8, 1997.
Article in English | MEDLINE | ID: mdl-9143918

ABSTRACT

A large Costa Rican kindred has been identified with 15 males affected with congenital blindness, progressive bearing loss, and venous insufficiency. Due to ophthalmological and audio-otological findings, including bilateral retinal dysplasia and detachment, progressive bilateral sensorineural hearing loss, and an X-linked pattern of inheritance, a tentative diagnosis of Norrie disease was considered. However, venous insufficiency is a clinical finding not reportedly associated with Norrie disease. Genetic linkage analysis using microsatellite repeat markers demonstrated linkage to Xp11.23-11.4 (z = 2.723 at theta = 0.0). A candidate gene approach using the Norrie disease gene (NDP), which maps to Xp11.3, revealed a point mutation in the third exon resulting in substitution of phenylalanine for leucine at position 61. The precise function of the gene product, norrin, has yet to be elucidated; however, it has been postulated to be involved in the regulation of neural cell differentiation and proliferation, although hypotheses have been considered for its role in vascular development in the eye. The finding of a mutation in NDP in association with peripheral vascular disease may provide valuable insight into the potential role of this gene in cellular processes.


Subject(s)
Blindness/genetics , Deafness/genetics , Intellectual Disability/genetics , Peripheral Vascular Diseases/genetics , Costa Rica , Genetic Linkage , Humans , Karyotyping , Male , Phenotype , X Chromosome
20.
Hear Res ; 98(1-2): 18-21, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8880177

ABSTRACT

Distortion product otoacoustic emissions (DPOE) were obtained from five different hearing mouse groups: CBA/J, MOLF/Rk, ct (homozygous normal mice of the curly-tail stock), and the F1 hybrid offspring of the matings CBA/J x dn/dn and MOLF/Rk x dn/dn (dn/dn mice are the curly-tail stock with recessive deafness). The DPOE patterns of the CBA/J and ct strains were similar to each other and different from that of the MOLF/Rk. The two sets of F1 hybrid mice, (CBA/J x dn/dn)F1 and (MOLF/Rk x dn/dn)F1, were found to have significantly larger DPOE amplitudes than their hearing parent strains, MOLF/Rk and CBA/J, respectively. In addition, the DPOE amplitudes were greater for the offspring of the MOLF/Rk x dn/dn cross than for those of the CBA/J x dn/dn cross, even though they were lower for MOLF/Rk than for CBA/J. The distinct features of DPOE patterns among these five groups suggest that DPOE testing can be used for auditory phenotyping.


Subject(s)
Deafness/genetics , Hearing/genetics , Hybrid Vigor/genetics , Otoacoustic Emissions, Spontaneous , Animals , Female , Mice , Mice, Inbred Strains , Phenotype , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL