Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Genomics ; 103(2-3): 169-76, 2014.
Article in English | MEDLINE | ID: mdl-24603341

ABSTRACT

Exome and whole-genome analyses powered by next-generation sequencing (NGS) have become invaluable tools in identifying causal mutations responsible for Mendelian disorders. Given that individual exomes contain several thousand single nucleotide variants and insertions/deletions, it remains a challenge to analyze large numbers of variants from multiple exomes to identify causal alleles associated with inherited conditions. To this end, we have developed user-friendly software that analyzes variant calls from multiple individuals to facilitate identification of causal mutations. The software, termed exomeSuite, filters for putative causative variants of monogenic diseases inherited in one of three forms: dominant, recessive caused by a homozygous variant, or recessive caused by two compound heterozygous variants. In addition, exomeSuite can perform homozygosity mapping and analyze the variant data of multiple unrelated individuals. Here we demonstrate that filtering of variants with exomeSuite reduces datasets to a fraction of a percent of their original size. To the best of our knowledge this is the first freely available software developed to analyze variant data from multiple individuals that rapidly assimilates and filters large data sets based on pattern of inheritance.


Subject(s)
Datasets as Topic , Exome , Genetic Diseases, Inborn/genetics , INDEL Mutation , Polymorphism, Single Nucleotide , Software , Alleles , DNA Mutational Analysis/methods , Female , Genome-Wide Association Study/methods , Heterozygote , Homozygote , Humans , Male , Pedigree
2.
Doc Ophthalmol ; 125(3): 229-33, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22903242

ABSTRACT

PURPOSE: To establish the normal range of values for rod-isolated b-wave amplitudes in achromatopsia and cone dystrophies. METHODS: We reviewed charts of 112 patients with various types of cone dystrophy, and compared their standardized electroretinographic rod b-wave amplitudes with age-matched normal controls. Twenty-six patients had known mutations in achromatopsia and cone dystrophy genes, while 53 were characterized by their inheritance pattern since they had yet to have their gene identified. Visual acuity information and scotomata were documented. RESULTS: We found that patients with achromatopsia and cone dystrophy had rod b-wave amplitudes that were significantly lower than age-matched controls, but found no evidence of rod amplitude progression nor loss of peripheral visual fields in the study group. CONCLUSIONS: We found that cone dystrophy patients of all types had depressed rod-isolated ERGs across the board. If typical diagnostic criteria are used, these patients might be considered to have "abnormal" rod-isolated electroretinographic values, and might be called "cone-rod dystrophy", even though the waveforms are stable for years. Patients with cone-rod dysfunction patterns on ERG can be better understood by also performing kinetic (Goldmann) visual fields, which will help to distinguish cone dystrophies from progressive cone-rod dystrophies by central scotomata size and progression over time in many forms of cone-rod dystrophy.


Subject(s)
Color Vision Defects/physiopathology , Electroretinography , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Adult , Aged , Color Vision Defects/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Female , Humans , Male , Middle Aged , Potassium Channels, Voltage-Gated/genetics , Retrospective Studies , Scotoma/physiopathology , Visual Acuity/physiology , Visual Field Tests , Visual Fields/physiology
4.
Eye (Lond) ; 24(5): 764-74, 2010 May.
Article in English | MEDLINE | ID: mdl-19893586

ABSTRACT

PURPOSE: To document the progression of disease in male and female members of a previously described family with X-linked dominant retinitis pigmentosa (RP) caused by a de novo insertion after nucleotide 173 in exon ORF15 of RPGR. METHODS: The clinical records of 19 members of family UTAD054 were reviewed. Their evaluations consisted of confirmation of family history, standardised electroretinograms (ERGs), Goldmann visual fields, and periodic ophthalmological examinations over a 23-year period. RESULTS: Male members of family UTAD054 had non-recordable to barely recordable ERGs from early childhood. The males showed contracted central fields and developed more severe retinopathy than the females. The female members showed a disease onset delayed to teenage years, recordable but diminishing photopic and scotopic ERG amplitudes in a cone-rod pattern, progressive loss and often asymmetric visual fields, and diffuse atrophic retinopathy with fewer pigment deposits compared with males. CONCLUSIONS: This insertion mutation in the RPGR exon ORF15 is associated with a RP phenotype that severely affects males early and females by 30 years of age, and is highly penetrant in female members. Families with dominant-acting RPGR mutations may be mistaken to have an autosomal mode of inheritance resulting in an incorrect prediction of recurrence risk and prognosis. Broader recognition of X-linked RP forms with dominant inheritance is necessary to facilitate appropriate counselling of these patients.


Subject(s)
Eye Proteins/genetics , Genetic Diseases, X-Linked/genetics , Mutagenesis, Insertional/genetics , Retinitis Pigmentosa/genetics , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Dark Adaptation/physiology , Disease Progression , Electroretinography , Exons/genetics , Female , Follow-Up Studies , Genetic Diseases, X-Linked/physiopathology , Humans , Infant , Male , Middle Aged , Refractive Errors , Retinitis Pigmentosa/physiopathology , Sensory Thresholds , Visual Acuity , Visual Fields/physiology , Young Adult
5.
Vision Res ; 47(5): 624-33, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17267005

ABSTRACT

We report the chromosomal localization, mutant gene identification, ophthalmic appearance, histology, and functional analysis of two new hereditary mouse models of retinal degeneration not having the Pde6brd1("r", "rd", or "rodless") mutation. One strain harbors an autosomal recessive mutation that maps to mouse chromosome 5. Sequence analysis showed that the retinal degeneration is caused by a missense point mutation in exon 13 of the beta-subunit of the rod cGMP phosphodiesterase (beta-PDE) gene (Pde6b). The gene symbol for this strain was set as Pde6brd10, abbreviated rd10 hereafter. Mice homozygous for the rd10 mutation showed histological changes at postnatal day 16 (P16) of age and sclerotic retinal vessels at four weeks of age, consistent with retinal degeneration. Retinal sections were highly positive for TUNEL and activated caspase-3 immunoreactivity, specifically in the outer nuclear layer (ONL). ERGs were never normal, but rod and cone ERG a- and b-waves were easily measured at P18 and steadily declined over 90% by two months of age. Protein extracts from rd10 retinas were positive for beta-PDE immunoreactivity starting at about the same time as wild-type (P10), though signal averaged less than 40% of wild-type. Interestingly, rearing rd10 mice in total darkness delayed degeneration for at least a week, after which morphological and functional loss progressed irregularly. With the second strain, a complementation test with rd1 mice revealed that the retinal degeneration phenotype observed represents a possible new allele of Pde6b. Sequencing demonstrated a missense point mutation in exon 16 of the beta-subunit of rod phosphodiesterase gene, different from the point mutations in rd1 and rd10. The gene symbol for this strain was set as Pde6bnmf137, abbreviated nmf137 hereafter. Mice homozygous for this mutation showed retinal degeneration with a mottled retina and white retinal vessels at three weeks of age. The exon 13 missense mutation (rd10) is the first known occurrence of a second mutant allele spontaneously arising in the Pde6b gene in mice and may provide a model for studying the pathogenesis of autosomal recessive retinitis pigmentosa (arRP) in humans. It may also provide a better model for experimental pharmaceutical-based therapy for RP because of its later onset and milder retinal degeneration than rd1 and nmf137.


Subject(s)
Mutation, Missense , Phosphoric Diester Hydrolases/genetics , Retinal Degeneration/genetics , Retinal Rod Photoreceptor Cells/enzymology , Animals , Apoptosis , Base Sequence , Cyclic Nucleotide Phosphodiesterases, Type 6 , Dark Adaptation , Disease Models, Animal , Electroretinography , Eye Proteins/genetics , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phenotype , Phosphoric Diester Hydrolases/metabolism , Retinal Degeneration/enzymology , Retinal Degeneration/pathology
6.
Vision Res ; 46(22): 3926-34, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16814838

ABSTRACT

One eye of rd12 mice received a sub-retinal injection of a vector carrying normal human RPE65 cDNA at post-natal day 18, and at 6- and 13-months of age. Electroretinograms (ERGs) and visual-evoked potentials (VEPs) were recorded to luminance, and to spatially and temporally modulated stimuli to assess the consequences of delayed treatment on visual pathway function. Early treatment resulted in better overall retinal rescue and better rescue of cone-mediated function. VEPs to low temporal frequency luminance modulation were well preserved at all but the oldest treatment age and corresponded to predictions based on the amount of retinal rescue. In contrast, VEPs to high frequency spatially and temporally modulated stimuli were impaired even at the earliest age. These results provide further support that early treatment in human LCA will have the most hope for optimal visual performance.


Subject(s)
Blindness/genetics , Genetic Therapy/methods , Retina/physiopathology , Visual Cortex/physiopathology , Animals , Blindness/congenital , Blindness/physiopathology , Dark Adaptation , Disease Models, Animal , Electroretinography/methods , Evoked Potentials, Visual/physiology , Mice , Mice, Mutant Strains , Rhodopsin/metabolism , Time Factors , Vision Tests/methods , Visual Acuity/physiology
7.
Vis Neurosci ; 22(5): 587-93, 2005.
Article in English | MEDLINE | ID: mdl-16332269

ABSTRACT

The Jackson Laboratory, having the world's largest collection of mouse mutant stocks and genetically diverse inbred strains, is an ideal place to discover genetically determined eye variations and disorders. In this paper, we list and describe mouse models for ocular research available from Mouse Eye Mutant Resource at The Jackson Laboratory. While screening mouse strains and stocks at The Jackson Laboratory (TJL) for genetic mouse models of human ocular disorders, we have identified numerous spontaneous or naturally occurring mutants. We characterized these mutants using serial indirect ophthalmoscopy, fundus photography, electroretinography (ERG) and histology, and performed genetic analysis including linkage studies and gene identification. Utilizing ophthalmoscopy, electroretinography, and histology, to date we have discovered 109 new disorders affecting all aspects of the eye including the lid, cornea, iris, lens, and retina, resulting in corneal disorders, glaucoma, cataracts, and retinal degenerations. The number of known serious or disabling eye diseases in humans is large and affects millions of people each year. Yet research on these diseases frequently is limited by the obvious restrictions on studying pathophysiologic processes in the human eye. Likewise, many human ocular diseases are genetic in origin, but appropriate families often are not readily available for genetic studies. Mouse models of inherited ocular disease provide powerful tools for rapid genetic analysis, characterization, and gene identification. Because of the great similarity among mammalian genomes, these findings in mice have direct relevance to the homologous human conditions.


Subject(s)
Eye Diseases/genetics , Mice, Mutant Strains/genetics , Animals , Cataract/genetics , Cataract/pathology , Chromosomes/metabolism , Chromosomes/ultrastructure , Disease Models, Animal , Electroretinography , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Eye Diseases/pathology , Eye Diseases/physiopathology , Glaucoma/genetics , Glaucoma/pathology , Mice , Ophthalmoscopy , Retinal Diseases/genetics , Retinal Diseases/pathology
8.
Am J Hum Genet ; 71(2): 262-75, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12075507

ABSTRACT

Usher syndrome type I is characterized by congenital hearing loss, retinitis pigmentosa (RP), and variable vestibular areflexia. Usher syndrome type ID, one of seven Usher syndrome type I genetic localizations, have been mapped to a chromosomal interval that overlaps with a nonsyndromic-deafness localization, DFNB12. Mutations in CDH23, a gene that encodes a putative cell-adhesion protein with multiple cadherin-like domains, are responsible for both Usher syndrome and DFNB12 nonsyndromic deafness. Specific CDH23 mutational defects have been identified that differentiate these two phenotypes. Only missense mutations of CDH23 have been observed in families with nonsyndromic deafness, whereas nonsense, frameshift, splice-site, and missense mutations have been identified in families with Usher syndrome. In the present study, a panel of 69 probands with Usher syndrome and 38 probands with recessive nonsyndromic deafness were screened for the presence of mutations in the entire coding region of CDH23, by heteroduplex, single-strand conformation polymorphism, and direct sequence analyses. A total of 36 different CDH23 mutations were detected in 45 families; 33 of these mutations were novel, including 18 missense, 3 nonsense, 5 splicing defects, 5 microdeletions, and 2 insertions. A total of seven mutations were common to more than one family. Numerous exonic and intronic polymorphisms also were detected. Results of ophthalmologic examinations of the patients with nonsyndromic deafness have found asymptomatic RP-like manifestations, indicating that missense mutations may have a subtle effect in the retina. Furthermore, patients with mutations in CDH23 display a wide range of hearing loss and RP phenotypes, differing in severity, age at onset, type, and the presence or absence of vestibular areflexia.


Subject(s)
Cadherins/genetics , Deafness/genetics , Mutation , Retinitis Pigmentosa/genetics , Adolescent , Adult , Aged , Amino Acid Sequence , Cadherin Related Proteins , Child , Child, Preschool , DNA Mutational Analysis , Female , Genetic Heterogeneity , Humans , Infant , Male , Molecular Sequence Data , Phenotype , Sequence Alignment , Syndrome , Vestibular Function Tests
9.
Br J Ophthalmol ; 86(6): 658-62, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12034689

ABSTRACT

BACKGROUND/AIMS: Few studies have reported on the change in visual acuity (VA) in patients with choroideraemia. In order to determine the degree and rate of VA impairment associated with this disease, the central VA was analysed in a large group of patients with choroideraemia. METHODS: The authors completed a retrospective, cross sectional review of 115 patients with choroideraemia from three tertiary care centres. A longitudinal analysis was performed on 45 of these patients who met the inclusion criteria of at least three visits over a minimum period of 4.5 years. Multiple linear regression analysis was used to explore the 5 year rate of VA change while controlling for initial VA and initial age. Multiple logistic regression was also used to investigate VA impairment. RESULTS: In the cross sectional group (n = 115), 84% (87/103) of patients under the age of 60 had a VA of 20/40 or better while 33% (4/12) of patients 60 years of age or older had a VA of 20/200 or worse at their most recent visit. The majority of the patients (93%) in the longitudinal subgroup of 45 patients had a VA of 20/30 or better at their initial visit. The mean 5 year rate of VA change was 0.09 logMAR equivalent (approximately one line on the Lighthouse chart). CONCLUSION: In this cohort of patients with choroideraemia, there was typically a slow rate of VA loss and the prognosis for central VA retention was, as a group, favourable until the seventh decade.


Subject(s)
Choroideremia/physiopathology , Vision Disorders/etiology , Visual Acuity , Adolescent , Adult , Aged , Child , Choroideremia/complications , Cross-Sectional Studies , Disease Progression , Humans , Linear Models , Longitudinal Studies , Middle Aged , Prognosis , Retrospective Studies , Vision Disorders/physiopathology
10.
Vision Res ; 42(4): 517-25, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11853768

ABSTRACT

The Jackson Laboratory, having the world's largest collection of mouse mutant stocks and genetically diverse inbred strains, is an ideal place to look for genetically determined eye variations and disorders. Through ophthalmoscopy, electroretinography and histology, we have discovered disorders affecting all aspects of the eye including the lid, cornea, iris, lens and retina, resulting in corneal disorders, cataracts, glaucoma and retinal degenerations. Mouse models of retinal degeneration have been investigated for many years in the hope of understanding the causes of photoreceptor cell death. Sixteen naturally occurring mouse mutants that manifest degeneration of photoreceptors in the retina with preservation of all other retinal cell types have been found: retinal degeneration (formerly rd, identical with rodless retina, r, now Pde6b(rd1)); Purkinje cell degeneration (pcd); nervous (nr); retinal degeneration slow (rds, now Prph(Rd2)); retinal degeneration 3 (rd3); motor neuron degeneration (mnd); retinal degeneration 4 (Rd4); retinal degeneration 5 (rd5, now tub); vitiligo (vit, now Mitf(mi-vit)); retinal degeneration 6 (rd6); retinal degeneration 7 (rd7, now Nr2e3(rd7)); neuronal ceroid lipofuscinosis (nclf); retinal degeneration 8 (rd8); retinal degeneration 9 (Rd9); retinal degeneration 10 (rd10, now Pde6b(rd10)); and cone photoreceptor function loss (cpfl1). In this report, we first review the genotypes and phenotypes of these mutants and second, list the mouse strains that carry each mutation. We will also provide detailed information about the cpfl1 mutation. The phenotypic characteristics of cpfl1 mice are similar to those observed in patients with complete achromatopsia (ACHM2, OMIM 216900) and the cpfl1 mutation is the first naturally-arising mutation in mice to cause cone-specific photoreceptor function loss. cpfl1 mice may provide a model for congenital achromatopsia in humans.


Subject(s)
Apoptosis , Mice, Mutant Strains , Models, Animal , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Animals , Electroretinography , Fundus Oculi , Mice , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Rhodopsin/metabolism
11.
Can J Ophthalmol ; 36(6): 315-22, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11714117

ABSTRACT

BACKGROUND: In experimental models of several forms of human retinitis pigmentosa (RP) the dystrophy begins in the neonatal period, during a "critical period" in which photoreceptors are sensitive to hypoxia. We performed a study to test whether perinatal stress is associated with human RP, particularly in simplex (nonfamilial) cases. METHODS: Two surveys were carried out in 1999. In one, Australians with RP were surveyed for information on whether they had experienced stress at birth and whether any members of their family had RP. In the other, the diagnostic type and inheritance patterns of a group of patients with RP seen at a university-affiliated eye institute in Los Angeles between 1997 and 1999 were established as part of their clinical assessment. In neither cohort was the RP part of a syndrome. RESULTS: After entry criteria were applied, there were 293 cases (of a total of 446 replies) available for analysis from the Australian survey and 119 cases (after exclusion of 229 cases with incomplete data) from the US survey. A total of 52.2% and 53.8% of the cases respectively were simplex. Perinatal stress was reported by about 15% of the respondents with familial RP (15.0% in the Australian cohort and 14.5% in the US cohort), compared with 30% of those with simplex RP (27.4% and 29.7% respectively), a significant difference (p < 0.05). In the Australian cohort four forms of stress--cyanosis, difficult presentation, prematurity and a perinatal period of intensive care--were reported more than twice as often by respondents in the simplex group than those in the familial group. For only one factor, cyanosis, was the difference between the two groups significant (chi2 test, p = 0.01). In the US cohort no single form of stress was significantly related to simplex RP. INTERPRETATION: Our findings support the hypothesis that perinatal stress is associated with simplex RP in a minority of cases. Larger cohorts need to be studied to test whether perinatal stress can interact with predisposing genes in the genesis of some forms of RP.


Subject(s)
Pregnancy Complications , Prenatal Exposure Delayed Effects , Retinitis Pigmentosa/etiology , Stress, Physiological/complications , Australia/epidemiology , Cohort Studies , Embryonic and Fetal Development , Female , Health Surveys , Humans , Infant, Newborn , Pregnancy , Retinitis Pigmentosa/epidemiology , Retinitis Pigmentosa/physiopathology , Stress, Physiological/epidemiology , Stress, Physiological/physiopathology , Surveys and Questionnaires , United States/epidemiology
12.
Am J Hum Genet ; 69(1): 198-203, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11389483

ABSTRACT

Mutations in the crumbs homologue 1 (CRB1) gene cause a specific form of retinitis pigmentosa (RP) that is designated "RP12" and is characterized by a preserved para-arteriolar retinal pigment epithelium (PPRPE) and by severe loss of vision at age <20 years. Because of the early onset of disease in patients who have RP with PPRPE, we considered CRB1 to be a good candidate gene for Leber congenital amaurosis (LCA). Mutations were detected in 7 (13%) of 52 patients with LCA from the Netherlands, Germany, and the United States. In addition, CRB1 mutations were detected in five of nine patients who had RP with Coats-like exudative vasculopathy, a relatively rare complication of RP that may progress to partial or total retinal detachment. Given that four of five patients had developed the complication in one eye and that not all siblings with RP have the complication, CRB1 mutations should be considered an important risk factor for the Coats-like reaction, although its development may require additional genetic or environmental factors. Although no clear-cut genotype-phenotype correlation could be established, patients with LCA, which is the most severe retinal dystrophy, carry null alleles more frequently than do patients with RP. Our findings suggest that CRB1 mutations are a frequent cause of LCA and are strongly associated with the development of Coats-like exudative vasculopathy in patients with RP.


Subject(s)
Blindness/genetics , Mutation/genetics , Optic Atrophies, Hereditary/genetics , Optic Atrophies, Hereditary/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology , Adult , Age of Onset , Blindness/pathology , Child , DNA Mutational Analysis , Female , Genes, Recessive/genetics , Genotype , Humans , Infant , Male , Pedigree , Phenotype
13.
Invest Ophthalmol Vis Sci ; 42(6): 1179-89, 2001 May.
Article in English | MEDLINE | ID: mdl-11328725

ABSTRACT

PURPOSE: To assess the allelic variation of the ATP-binding transporter protein (ABCA4). METHODS: A combination of single-strand conformation polymorphism (SSCP) and automated DNA sequencing was used to systematically screen this gene for sequence variations in 374 unrelated probands with a clinical diagnosis of Stargardt disease, 182 patients with age-related macular degeneration (AMD), and 96 normal subjects. RESULTS: There was no significant difference in the proportion of any single variant or class of variant between the control and AMD groups. In contrast, truncating variants, amino acid substitutions, synonymous codon changes, and intronic variants were significantly enriched in patients with Stargardt disease when compared with their presence in subjects without Stargardt disease (Kruskal-Wallis P < 0.0001 for each variant group). Overall, there were 2480 instances of 213 different variants in the ABCA4 gene, including 589 instances of 97 amino acid substitutions, and 45 instances of 33 truncating variants. CONCLUSIONS: Of the 97 amino acid substitutions, 11 occurred at a frequency that made them unlikely to be high-penetrance recessive disease-causing variants (HPRDCV). After accounting for variants in cis, one or more changes that were compatible with HPRDCV were found on 35% of all Stargardt-associated alleles overall. The nucleotide diversity of the ABCA4 coding region, a collective measure of the number and prevalence of polymorphic sites in a region of DNA, was found to be 1.28, a value that is 9 to 400 times greater than that of two other macular disease genes that were examined in a similar fashion (VMD2 and EFEMP1).


Subject(s)
ATP-Binding Cassette Transporters/genetics , Alleles , Genetic Variation , Macular Degeneration/genetics , Adult , Humans , Linkage Disequilibrium , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational , Sequence Analysis, DNA
14.
Invest Ophthalmol Vis Sci ; 42(6): 1319-27, 2001 May.
Article in English | MEDLINE | ID: mdl-11328746

ABSTRACT

PURPOSE: To define the phenotypic expression of a deletion in the gene encoding the transcription factor CRX in a large, seven-generation, white family. METHODS: Fourteen affected individuals, all heterozygous for the Leu146del12 mutation in the cone-rod homeobox gene (CRX), and four nonaffected relatives from the same family were examined with visual function tests, and 10 underwent bone mineral density (BMD) measurement. RESULTS: The ability of the mutated CRX protein to transactivate rhodopsin promoter was decreased by approximately 25%, and its ability to react synergistically with neural retinal leucine zipper (NRL) was reduced by more than 30%. The affected members of the family had an autosomal dominant ocular condition most closely resembling Leber congenital amaurosis (LCA) with severe visual impairment at an early age. Depending on age, affected members showed varying degrees of significant visual acuity loss, elevated dark-adaptation thresholds, significantly reduced cone and rod electroretinogram (ERG) amplitudes, and progressive constriction of the visual fields, in most cases leading to complete blindness. Six affected members had reduced levels of BMD in the spine and the hip (osteopenia). Four affected female members who were receiving long-term hormonal replacement therapy (HRT) demonstrated normal values of BMD. CONCLUSIONS: This large deletion of the CRX gene is associated with a severe form of autosomal dominant retinal degeneration. Affected members not receiving HRT showed reduced BMD (osteopenia). This phenotype may reflect the abnormal influence of mutant CRX on both retinal and pineal development.


Subject(s)
Base Sequence , Bone Diseases, Metabolic/genetics , Homeodomain Proteins/genetics , Retinal Degeneration/genetics , Sequence Deletion , Trans-Activators/genetics , Adult , Aged , Bone Density , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/pathology , Child , Child, Preschool , DNA Mutational Analysis , Electroretinography , Estrogen Replacement Therapy , Female , Genes, Dominant , Humans , Male , Middle Aged , Mutation , Pedigree , Phenotype , Retinal Degeneration/pathology , Vision Disorders/genetics , Visual Acuity
15.
BMC Genet ; 2: 1, 2001.
Article in English | MEDLINE | ID: mdl-11178107

ABSTRACT

BACKGROUND: Glaucoma is a common disease but its molecular etiology is poorly understood. It involves retinal ganglion cell death and optic nerve damage that is often associated with elevated intraocular pressure. Identifying genes that modify glaucoma associated phenotypes is likely to provide insights to mechanisms of glaucoma. We previously reported glaucoma in DBA/2J mice caused by recessive alleles at two loci, isa and ipd, that cause iris stromal atrophy and iris pigment dispersion, respectively. A approach for identifying modifier genes is to study the effects of specific mutations in different mouse strains. When the phenotypic effect of a mutation is modified upon its introduction into a new strain, crosses between the parental strains can be used to identify modifier genes. The purpose of this study was to determine if the effects of the DBA/2J derived isa and ipd loci are modified in strain AKXD-28/Ty. RESULTS: AKXD-28/Ty mice develop glaucoma characterized by intraocular pressure elevation, retinal ganglion loss, and optic nerve excavation. In AKXD-28/Ty, isa causes an iris stromal atrophy phenotype as in DBA/2J. However, the iris pigment dispersion phenotype associated with ipd in DBA/2J does not occur in AKXD-28/Ty. Additionally, a greater severity and speed of retinal and optic nerve damage following intraocular pressure elevation in AKXD-28/Ty compared to DBA/2J mice suggests that AKXD-28/Ty is more susceptible to pressure-induced cell death. CONCLUSIONS: The consequences of the ipd and isa mutations are modified in the AKXD-28/Ty background. These strains provide a resource for the identification of modifier genes that modulate pigment dispersion and susceptibility to pressure-induced cell death.


Subject(s)
Genetic Predisposition to Disease , Glaucoma/genetics , Glaucoma/pathology , Animals , Atrophy , Female , Glaucoma/diagnosis , Iris/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Mutation , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Phenotype , Pigment Epithelium of Eye/pathology , Retinal Diseases/genetics , Retinal Diseases/pathology , Sex Factors , Species Specificity
16.
Hum Mutat ; 17(1): 42-51, 2001.
Article in English | MEDLINE | ID: mdl-11139241

ABSTRACT

Inherited retinopathies are a genetically and phenotypically heterogeneous group of diseases affecting approximately one in 2000 individuals worldwide. For the past 10 years, the Laboratory for Molecular Diagnosis of Inherited Eye Diseases (LMDIED) at the University of Texas-Houston Health Science Center has screened subjects ascertained in the United States and Canada for mutations in genes causing dominant and recessive autosomal retinopathies. A combination of single strand conformational analysis (SSCA) and direct sequencing of five genes (rhodopsin, peripherin/RDS, RP1, CRX, and AIPL1) identified the disease-causing mutation in approximately one-third of subjects with autosomal dominant retinitis pigmentosa (adRP) or with autosomal dominant cone-rod dystrophy (adCORD). In addition, the causative mutation was identified in 15% of subjects with Leber congenital amaurosis (LCA). Overall, we report identification of the causative mutation in 105 of 506 (21%) of unrelated subjects (probands) tested; we report five previously unreported mutations in rhodopsin, two in peripherin/RDS, and one previously unreported mutation in the cone-rod homeobox gene, CRX. Based on this large survey, the prevalence of disease-causing mutations in each of these genes within specific disease categories is estimated. These data are useful in estimating the frequency of specific mutations and in selecting individuals and families for mutation-specific studies.


Subject(s)
Membrane Glycoproteins , Mutation , Retinitis Pigmentosa/epidemiology , Retinitis Pigmentosa/genetics , Amino Acid Substitution/genetics , Animals , Arginine/genetics , Cysteine/genetics , Genetic Variation , Glutamine/genetics , Homeodomain Proteins/genetics , Humans , Intermediate Filament Proteins/genetics , Leucine/genetics , Nerve Tissue Proteins/genetics , Optic Atrophies, Hereditary/genetics , Peripherins , Prevalence , Proline/genetics , Retinal Degeneration/genetics , Retinal Diseases/epidemiology , Retinal Diseases/genetics , Rhodopsin/genetics , Trans-Activators/genetics , Tyrosine/genetics
17.
Proc Natl Acad Sci U S A ; 97(26): 14461-6, 2000 Dec 19.
Article in English | MEDLINE | ID: mdl-11106380

ABSTRACT

We report a method for introducing mtDNA mutations into the mouse female germ line by means of embryonic stem (ES) cell cybrids. Mitochondria were recovered from the brain of a NZB mouse by fusion of synaptosomes to a mtDNA-deficient (rho degrees ) cell line. These cybrids were enucleated and the cytoplasts were electrofused to rhodamine-6G (R-6G)-treated female ES cells. The resulting ES cell cybrids permitted transmission of the NZB mtDNAs through the mouse maternal lineage for three generations. Similarly, mtDNAs from a partially respiratory-deficient chloramphenicol-resistant (CAP(R)) cell line also were introduced into female chimeric mice and were transmitted to the progeny. CAP(R) chimeric mice developed a variety of ocular abnormalities, including congenital cataracts, decreased retinal function, and hamaratomas of the optic nerve. The germ-line transmission of the CAP(R) mutation resulted in animals with growth retardation, myopathy, dilated cardiomyopathy, and perinatal or in utero lethality. Skeletal and heart muscle mitochondria of the CAP(R) mice were enlarged and atypical with inclusions. This mouse ES cell-cybrid approach now provides the means to generate a wide variety of mouse models of mitochondrial disease.


Subject(s)
DNA, Mitochondrial , Genomic Imprinting , Stem Cells , Animals , Brain/pathology , Cell Line , Chimera , Chloramphenicol/pharmacology , Drug Resistance , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Myocardium/pathology , Ovum , Pedigree , Phenotype , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology
18.
Arch Ophthalmol ; 118(11): 1525-33, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11074809

ABSTRACT

PURPOSE: To investigate whether antirecoverin antibodies are present in patients with retinitis pigmentosa (RP). Recoverin, a retinal protein, has been implicated as a cause of cancer-associated retinopathy (CAR), which manifests as an RP-like retinal degeneration. The rationale is that the ocular findings in CAR syndrome are similar to those found in many forms of RP, and since 40% of patients with RP have no family history, some patients may have an underlying autoimmune process causing or contributing to their retinopathy. METHODS: Serum samples from 521 patients diagnosed with RP were screened for antiretinal proteins activity by Western blot analysis. Fifty-one patients had antibody reactivity against retinal proteins in the range of 23 to 26 kd and underwent dot-blot analysis for antirecoverin antibody, checking IgG and IgM antibodies. Enzyme-linked immunosorbent assay (ELISA) was performed to evaluate the titer of antirecoverin antibodies in patients with positive results on dot-blot analysis. Lymphocyte proliferation assays using recoverin were performed on 26 samples. RESULTS: Ten patients were found to have antirecoverin antibody and/or cellular immunoreactivity. Eight patients had positive dot-blot testing: 6 patients had both IgG and IgM antirecoverin activity, and 1 patient each had IgG or IgM activity. In these 8 patients, numerous other antiretinal protein antibodies were present. Three patients had positive recoverin-mediated lymphocyte proliferation, and all patients were positive for antirecoverin antibodies on ELISA testing. CONCLUSIONS: Antirecoverin immunoreactivity was found in 10 patients without systemic malignancy but with clinical findings consistent with RP. These results suggest that there are other immunogenic mechanisms occurring in the formation of antirecoverin antibodies in addition to the putative tumor-mediated mechanisms. This survey suggests that there may be rare cases of CAR-like syndrome in the category of simplex RP, or that some patients with RP also have antirecoverin antibodies that may be exacerbating their underlying disease. Arch Ophthalmol. 2000;118:1525-1533


Subject(s)
Antigens, Neoplasm/immunology , Autoantibodies/blood , Autoimmune Diseases/immunology , Calcium-Binding Proteins/immunology , Eye Proteins/immunology , Lipoproteins , Nerve Tissue Proteins , Retinitis Pigmentosa/immunology , Adult , Autoantigens/immunology , Autoimmune Diseases/pathology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Fluorescein Angiography , Fundus Oculi , Hippocalcin , Humans , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Lymphocyte Activation/immunology , Male , Middle Aged , Recoverin , Retinitis Pigmentosa/pathology , Visual Acuity
19.
Ophthalmology ; 107(12): 2256-66, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11097607

ABSTRACT

PURPOSE: To clarify the pathogenesis of late-onset retinal degeneration (L-ORD), an autosomal dominant disorder characterized by thick deposits of lipid-rich material between the retinal pigment epithelium (RPE) and Bruch's membrane. STUDY DESIGN: Comparative clinicopathologic case report and case series. TISSUES: Eyes of an 82-year-old L-ORD eye donor and an age-matched control. SUBJECTS: Five descendants of the eye donor and his affected sister. METHODS: The eyes were processed for histopathologic examination, including electron microscopy and immunohistochemistry. Family members were examined clinically and with retinal function tests. RESULTS: The L-ORD eye had sub-RPE deposits that were positive for lipid, including esterified and unesterified cholesterol. The deposits were thinnest in the macula, which retained the highest percentage of photoreceptors. In the periphery, RPE thinning and photoreceptor loss correlated with thickness of the sub-RPE deposits. The eye donor was asymptomatic until his late 50s, when he developed problems with adapting to darkness. At age 68, the eye donor had normal acuity but a midperipheral scotoma and subnormal electroretinograms (ERGs); visual loss was progressive. The five descendants (at the time of examination ages 44-58) of the eye donor and his affected sister, who were at 50/50 risk of inheriting L-ORD, had normal ERGs, but four showed defects in dark adaptation. The dark adaptation abnormalities had a distribution similar to the thickness of the sub-RPE deposits in the eye donor, with slow kinetics in the midperiphery and normal kinetics centrally. CONCLUSIONS: The L-ORD donor eye differed from a previous case in the regional distribution of sub-RPE deposits and photoreceptors. In the next generation of this L-ORD family, the first expression of disease, abnormal dark adaptation, mirrored the regional distribution of the deposits in the donor eye. The fine structure and staining characteristics of the sub-RPE deposits in L-ORD resemble those in age-related macular degeneration and Sorsby fundus dystrophy.


Subject(s)
Eye Diseases, Hereditary/pathology , Photoreceptor Cells, Vertebrate/pathology , Pigment Epithelium of Eye/pathology , Retina/physiology , Retinal Degeneration/pathology , Adult , Aged , Aged, 80 and over , Apolipoproteins/metabolism , Bruch Membrane/pathology , Dark Adaptation , Electroretinography , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/physiopathology , Female , Filipin/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Pedigree , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/ultrastructure , Pigment Epithelium of Eye/metabolism , Pigment Epithelium of Eye/ultrastructure , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Tissue Donors , Visual Acuity , Visual Fields
20.
Am J Hum Genet ; 67(6): 1569-74, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11060213

ABSTRACT

Usher syndrome type I is an autosomal recessive disorder marked by hearing loss, vestibular areflexia, and retinitis pigmentosa. Six Usher I genetic subtypes at loci USH1A-USH1F have been reported. The MYO7A gene is responsible for USH1B, the most common subtype. In our analysis, 151 families with Usher I were screened by linkage and mutation analysis. MYO7A mutations were identified in 64 families with Usher I. Of the remaining 87 families, who were negative for MYO7A mutations, 54 were informative for linkage analysis and were screened with the remaining USH1 loci markers. Results of linkage and heterogeneity analyses showed no evidence of Usher types Ia or Ie. However, one maximum LOD score was observed lying within the USH1D region. Two lesser peak LOD scores were observed outside and between the putative regions for USH1D and USH1F, on chromosome 10. A HOMOG chi(2)((1)) plot shows evidence of heterogeneity across the USH1D, USH1F, and intervening regions. These results provide conclusive evidence that the second-most-common subtype of Usher I is due to genes on chromosome 10, and they confirm the existence of one Usher I gene in the previously defined USH1D region, as well as providing evidence for a second, and possibly a third, gene in the 10p/q region.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 10/genetics , Deafness/genetics , Genetic Heterogeneity , Retinitis Pigmentosa/genetics , Chromosome Mapping , Consanguinity , DNA Mutational Analysis , Genes, Recessive/genetics , Humans , Lod Score , Mutation/genetics , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...