Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 90(7): 3342-54, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26739055

RESUMO

UNLABELLED: The cholesterol synthesis pathway is a ubiquitous cellular biosynthetic pathway that is attenuated therapeutically by statins. Importantly, type I interferon (IFN), a major antiviral mediator, also depresses the cholesterol synthesis pathway. Here we demonstrate that attenuation of cholesterol synthesis decreases gammaherpesvirus replication in primary macrophages in vitro and reactivation from peritoneal exudate cells in vivo. Specifically, the reduced availability of the intermediates required for protein prenylation was responsible for decreased gammaherpesvirus replication in statin-treated primary macrophages. We also demonstrate that statin treatment of a chronically infected host attenuates gammaherpesvirus latency in a route-of-infection-specific manner. Unexpectedly, we found that the antiviral effects of statins are counteracted by type I IFN. Our studies suggest that type I IFN signaling counteracts the antiviral nature of the subdued cholesterol synthesis pathway and offer a novel insight into the utility of statins as antiviral agents. IMPORTANCE: Statins are cholesterol synthesis inhibitors that are therapeutically administered to 12.5% of the U.S. POPULATION: Statins attenuate the replication of diverse viruses in culture; however, this attenuation is not always obvious in an intact animal model. Further, it is not clear whether statins alter parameters of highly prevalent chronic herpesvirus infections. We show that statin treatment attenuated gammaherpesvirus replication in primary immune cells and during chronic infection of an intact host. Further, we demonstrate that type I interferon signaling counteracts the antiviral effects of statins. Considering the fact that type I interferon decreases the activity of the cholesterol synthesis pathway, it is intriguing to speculate that gammaherpesviruses have evolved to usurp the type I interferon pathway to compensate for the decreased cholesterol synthesis activity.


Assuntos
Antivirais/farmacologia , Colesterol/biossíntese , Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Interferon Tipo I/imunologia , Lovastatina/farmacologia , Animais , Células Cultivadas , Gammaherpesvirinae/efeitos dos fármacos , Infecções por Herpesviridae/virologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prenilação de Proteína , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Latência Viral/efeitos dos fármacos , Replicação Viral/genética
2.
Open Biol ; 5(6): 150047, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26063829

RESUMO

RAB18, RAB3GAP1, RAB3GAP2 and TBC1D20 are each mutated in Warburg Micro syndrome, a rare autosomal recessive multisystem disorder. RAB3GAP1 and RAB3GAP2 form a binary 'RAB3GAP' complex that functions as a guanine-nucleotide exchange factor (GEF) for RAB18, whereas TBC1D20 shows modest RAB18 GTPase-activating (GAP) activity in vitro. Here, we show that in the absence of functional RAB3GAP or TBC1D20, the level, localization and dynamics of cellular RAB18 is altered. In cell lines where TBC1D20 is absent from the endoplasmic reticulum (ER), RAB18 becomes more stably ER-associated and less cytosolic than in control cells. These data suggest that RAB18 is a physiological substrate of TBC1D20 and contribute to a model in which a Rab-GAP can be essential for the activity of a target Rab. Together with previous reports, this indicates that Warburg Micro syndrome can be caused directly by loss of RAB18, or indirectly through loss of RAB18 regulators RAB3GAP or TBC1D20.


Assuntos
Anormalidades Múltiplas/etiologia , Anormalidades Múltiplas/patologia , Catarata/congênito , Córnea/anormalidades , Regulação da Expressão Gênica , Hipogonadismo/etiologia , Hipogonadismo/patologia , Deficiência Intelectual/etiologia , Deficiência Intelectual/patologia , Microcefalia/etiologia , Microcefalia/patologia , Atrofia Óptica/etiologia , Atrofia Óptica/patologia , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab1 de Ligação ao GTP/metabolismo , Proteínas rab3 de Ligação ao GTP/metabolismo , Anormalidades Múltiplas/metabolismo , Animais , Western Blotting , Estudos de Casos e Controles , Catarata/etiologia , Catarata/metabolismo , Catarata/patologia , Células Cultivadas , Córnea/metabolismo , Córnea/patologia , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Citometria de Fluxo , Imunofluorescência , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Hidrólise , Hipogonadismo/metabolismo , Deficiência Intelectual/metabolismo , Camundongos , Camundongos Knockout , Microcefalia/metabolismo , Atrofia Óptica/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas rab de Ligação ao GTP/genética , Proteínas rab1 de Ligação ao GTP/genética , Proteínas rab3 de Ligação ao GTP/genética
3.
BMC Genet ; 15: 135, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25476608

RESUMO

BACKGROUND: Loss-of-function mutations in TBC1D20 cause Warburg Micro syndrome 4 (WARBM4), which is an autosomal recessive syndromic disorder characterized by eye, brain, and genital abnormalities. Blind sterile (bs) mice carry a Tbc1d20-null mutation and exhibit cataracts and testicular phenotypes similar to those observed in WARBM4 patients. In addition to TBC1D20, mutations in RAB3GAP1, RAB3GAP2 and RAB18 cause WARBM1-3 respectively. However, regardless of which gene harbors the causative mutation, all individuals affected with WARBM exhibit indistinguishable clinical presentations. In contrast, bs, Rab3gap1 (-/-) , and Rab18 (-/-) mice exhibit distinct phenotypes; this phenotypic variability of WARBM mice was previously attributed to potential compensatory mechanisms. Rab3gap1 (-/-) and Rab18 (-/-) mice were genetically engineered using standard approaches, whereas the Tbc1d20 mutation in the bs mice arose spontaneously. There is the possibility that another unidentified mutation within the bs linkage disequilibrium may be contributing to the bs phenotypes and thus contributing to the phenotypic variability in WARBM mice. The goal of this study was to establish the phenotypic consequences in mice caused by the disruption of the Tbc1d20 gene. RESULTS: The zinc finger nuclease (ZFN) mediated genomic editing generated a Tbc1d20 c.[418_426del] deletion encoding a putative TBC1D20-ZFN protein with an in-frame p.[H140_Y143del] deletion within the highly conserved TBC domain. The evaluation of Tbc1d20 (ZFN/ZFN) eyes identified severe cataracts and thickened pupillary sphincter muscle. Tbc1d20 (ZFN/ZFN) males are infertile and the analysis of the seminiferous tubules identified disrupted acrosomal development. The compound heterozygote Tbc1d20 (ZFN/bs) mice, generated from an allelic bs/+ X Tbc1d20 (ZFN/+) cross, exhibited cataracts and aberrant acrosomal development indicating a failure to complement. CONCLUSIONS: Our findings show that the disruption of Tbc1d20 in mice results in cataracts and aberrant acrosomal formation, thus establishing bs and Tbc1d20 (ZFN/ZFN) as allelic variants. Although the WARBM molecular disease etiology remains unclear, both the bs and Tbc1d20 (ZFN/ZFN) mice are excellent model organisms for future studies to establish TBC1D20-mediated molecular and cellular functions.


Assuntos
Catarata/genética , Endodesoxirribonucleases/genética , Testículo/anormalidades , Proteínas rab1 de Ligação ao GTP/genética , Acrossomo/fisiologia , Animais , Sequência de Bases , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Estudos de Associação Genética , Engenharia Genética , Células HeLa , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Análise de Sequência de DNA , Dedos de Zinco , Proteínas rab1 de Ligação ao GTP/metabolismo
4.
Am J Hum Genet ; 93(6): 1001-14, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24239381

RESUMO

blind sterile (bs) is a spontaneous autosomal-recessive mouse mutation discovered more than 30 years ago. Phenotypically, bs mice exhibit nuclear cataracts and male infertility; genetic analyses assigned the bs locus to mouse chromosome 2. In this study, we first positionally cloned the bs locus and identified a putative causative mutation in the Tbc1d20 gene. Functional analysis established the mouse TBC1D20 protein as a GTPase-activating protein (GAP) for RAB1 and RAB2, and bs as a TBC1D20 loss-of-function mutation. Evaluation of bs mouse embryonic fibroblasts (mEFs) identified enlarged Golgi morphology and aberrant lipid droplet (LD) formation. Based on the function of TBC1D20 as a RABGAP and the bs cataract and testicular phenotypes, we hypothesized that mutations in TBC1D20 may contribute to Warburg micro syndrome (WARBM); WARBM constitutes a spectrum of disorders characterized by eye, brain, and endocrine abnormalities caused by mutations in RAB3GAP1, RAB3GAP2, and RAB18. Sequence analysis of a cohort of 77 families affected by WARBM identified five distinct TBC1D20 loss-of-function mutations, thereby establishing these mutations as causative of WARBM. Evaluation of human fibroblasts deficient in TBC1D20 function identified aberrant LDs similar to those identified in the bs mEFs. Additionally, our results show that human fibroblasts deficient in RAB18 and RAB3GAP1 function also exhibit aberrant LD formation. These findings collectively indicate that a defect in LD formation/metabolism may be a common cellular abnormality associated with WARBM, although it remains unclear whether abnormalities in LD metabolism are contributing to WARBM disease pathology.


Assuntos
Anormalidades Múltiplas/genética , Catarata/congênito , Catarata/genética , Córnea/anormalidades , Hipogonadismo/genética , Infertilidade Masculina/genética , Deficiência Intelectual/genética , Microcefalia/genética , Mutação , Atrofia Óptica/genética , Proteínas rab1 de Ligação ao GTP/genética , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/patologia , Catarata/diagnóstico , Catarata/metabolismo , Linhagem Celular , Córnea/metabolismo , Análise Mutacional de DNA , Fácies , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Hipogonadismo/diagnóstico , Hipogonadismo/metabolismo , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/metabolismo , Cristalino/patologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Microcefalia/diagnóstico , Microcefalia/metabolismo , Atrofia Óptica/diagnóstico , Atrofia Óptica/metabolismo , Linhagem , Fenótipo , Alinhamento de Sequência , Testículo/patologia , Proteínas rab1 de Ligação ao GTP/metabolismo
5.
Invest Ophthalmol Vis Sci ; 54(10): 6646-54, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-24045990

RESUMO

PURPOSE: The goal of this study was to functionally evaluate three previously uncharacterized heat shock factor protein 4 (HSF4) mutations (c.595_599delGGGCC, c.1213C>T, c.1327+4A>G) encoding mutant HSF4 proteins (G199EfsX15, R405X, and M419GfsX29) with missing C-terminal ends. These HSF4 mutations were previously identified in families with congenital autosomal recessive cataracts. METHODS: FLAG-tagged recombinant wild type (WT) and mutant HSF4 proteins were analyzed using the protein stability assay, cellular immunofluorescence, Western blotting, electrophoretic mobility shift assay (EMSA), and reporter activation. RESULTS: HSF4 mutant proteins did not differ in the protein turnover rate when compared with WT HSF4. Immunofluorescence revealed that WT and mutant HSF4 proteins were properly trafficked to the nucleus. EMSA analysis revealed that the G199EfsX15 and M419GfsX29 proteins exhibited decreased heat shock element (HSE)-mediated DNA binding, whereas the R405X mutant exhibited increased HSE-mediated DNA binding when compared with WT HSF4. All three HSF4 mutant proteins exhibited abolished HSE-mediated luciferase reporter activation. Detailed evaluation of the C-terminal region identified three novel domains: two activation domains and one repression domain. CONCLUSIONS: The three HSF4 autosomal recessive mutations evaluated here result in a loss of HSF4 function due to a loss of regulatory domains present at the C-terminal end. These findings collectively indicate that the transcriptional activation of HSF4 is mediated by interactions between activator and repressor domains within the C-terminal end.


Assuntos
Catarata/congênito , Proteínas de Ligação a DNA/genética , DNA/genética , Mutação , Fatores de Transcrição/genética , Western Blotting , Catarata/genética , Catarata/metabolismo , Células Cultivadas , Análise Mutacional de DNA , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Genes Recessivos , Fatores de Transcrição de Choque Térmico , Proteínas de Choque Térmico , Humanos , Fatores de Transcrição/metabolismo , Ativação Transcricional
6.
Invest Ophthalmol Vis Sci ; 54(1): 132-40, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23211830

RESUMO

PURPOSE: During mammalian embryonic eyelid closure ADAM17 has been proposed to play a role as a transactivator of epidermal growth factor receptor (EGFR) signaling by shedding membrane bound EGFR ligands. However, ADAM17 also sheds numerous other ligands, thus implicating ADAM17 in additional molecular pathways. The goal of this study was to experimentally establish the role of ADAM17 and determine ADAM17-mediated pathways essential for the embryonic eyelid closure. METHODS: Wild-type (WT) and woe mice, carrying a hypomorphic mutation in Adam17, were evaluated using H&E and scanning electron microscopy. Expressions of ADAM17, EGFR, and the phosphorylated form EGFR-P were evaluated using immunohistochemistry. BrdU and TUNEL assays were used to evaluate cell proliferation and apoptosis, respectively. In vitro scratch assays of primary cultures were used to evaluate cell migration. Clinical and histologic analyses established if the hypermorphic Egfr(Dsk5) allele can rescue the woe embryonic eyelid closure. RESULTS: woe mice exhibited a failure to develop the leading edge of the eyelid and consequently failure of the embryonic eyelid closure. Expression of ADAM17 was identified in the eyelid epithelium in the cells of the leading edge. ADAM17 is essential for epithelial cell migration, but does not play a role in proliferation and apoptosis. EGFR was expressed in both WT and woe eyelid epithelium, but the phosphorylated EGFR-P form was detected only in WT. The Egfr(Dsk5) allele rescued woe eyelid closure defects, but also rescued woe anterior segment defects and the absence of meibomian glands. CONCLUSIONS: We provide in vivo genetic evidence that the role of ADAM17 during embryonic eyelid closure is to transactivate EGFR signaling.


Assuntos
Proteínas ADAM/metabolismo , Receptores ErbB/metabolismo , Pálpebras/embriologia , Pálpebras/metabolismo , Transdução de Sinais/fisiologia , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Morte Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células , Pálpebras/anormalidades , Pálpebras/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genótipo , Ligantes , Masculino , Glândulas Tarsais/citologia , Glândulas Tarsais/embriologia , Glândulas Tarsais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fenótipo , Gravidez , Cultura Primária de Células
7.
BMC Genet ; 13: 76, 2012 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-22928477

RESUMO

BACKGROUND: Waved with open eyelids 2 (woe2) is a novel autosomal recessive mouse mutation that arose spontaneously in our animal facility. Upon initial evaluation, mutant mice exhibited eyelids open at birth (EOB) and wavy fur phenotypes. The goals of this study were to phenotypically characterize the woe2 mice and to identify the gene harboring the mutation responsible for the woe2 phenotype. RESULTS: Histological analysis of woe2 embryos identified the failure of embryonic eyelid closure. Clinical and histological analysis of woe2 adult eyes identified severe corneal opacities, abnormalities of the anterior segment of the eye, and the absence of meibomian glands. Abnormalities in the fur texture and the absence of meibomian glands prompted us to evaluate other epidermal appendages: skin, teeth, and nails--as well as lacrimal, mammary, salivary, sebaceous and sweat glands. No obvious morphological differences between WT and woe2 mice were identified in these tissues. However, the analysis of woe2 identified cardiac abnormalities. Positional cloning of the woe2 locus identified a 1308 bp deletion in the Ppp1r13l gene. The deletion resulted in an aberrant Ppp1r13l(Δexon9-11) transcript that lacks exons 9, 10 and 11 resulting in a premature stop and a loss of 223 amino acids from the C-terminal end of the putative mutant PPP1R13L protein. Immunohistological analysis during eye development identified expression of PPP1R13L in the palpebral epidermis, palpebral and bulbar conjunctiva, corneal epithelium and meibomian glands. CONCLUSIONS: The woe2 mouse harbors a novel deletion within the Ppp1r13l gene, likely resulting in a complete loss of PPP1R13L function. Results from this study provide evidence that PPP1R13L has an essential role in embryonic eyelid closure as well in development of meibomian glands and the anterior segment of the eye. The woe2 mice are a useful model for investigation of the role of PPP1R13L, especially during ocular and eyelid development.


Assuntos
Anormalidades do Olho/genética , Pálpebras , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos/genética , Mutação , Proteínas Repressoras/genética , Animais , Pálpebras/embriologia , Glândulas Tarsais/anormalidades , Proteína Fosfatase 1
8.
Mamm Genome ; 22(11-12): 661-73, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21858719

RESUMO

Lens opacity 13 (lop13) is a spontaneous, autosomal recessive mouse mutant that exhibits nuclear cataracts. Histological analysis revealed swollen lens fiber cells and the presence of bladder cells within the lens cortex, as well as morgagnian globules and liquefied material at the lens posterior. At 3 months of age, in addition to cataracts, lop13 mice also develop persistent skin wounds. Linkage analysis assigned the lop13 locus to a 1.1-Mb region on mouse Chr 15, encompassing 19 candidate genes. Sequence analysis identified a C3112T mutation in exon 18 of Sterol Regulatory Element Binding-Transcription Factor 2 (Srebf2) resulting in the R1038C substitution of a highly conserved arginine within the Srebf2 regulatory domain. Srebf2 belongs to a family of membrane-bound basic helix-loop-helix leucine zipper transcription factors that control the expression of genes involved in the biosynthesis and uptake of cholesterol and fatty acids. The lack of complementation observed in Srebf2 ( lop13/GT ) compound heterozygotes carrying the Srebf2 gene trapped allele (Srebf2 ( GT )) provides genetic evidence that the identified C3112T substitution in Srebf2 is responsible for the lop13 phenotype. Gas chromatography analysis identified lower levels of cholesterol in the lop13 brain, liver, and lens when compared to wild-type mice. These findings suggest that lop13 is a hypomorphic mutation in Srebf2. As such, the lop13 mouse presents an invaluable in vivo model for studying the contribution of Srebf2 and cholesterol to maintaining the homeostasis of the lens and skin.


Assuntos
Catarata/genética , Catarata/patologia , Pele/patologia , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Animais , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Catarata/metabolismo , Colesterol/metabolismo , Feminino , Genótipo , Cristalino/metabolismo , Cristalino/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Análise de Sequência de DNA
9.
Per Med ; 5(4): 377-385, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19266054

RESUMO

Glaucoma is the second leading cause of blindness worldwide. The primary glaucoma risk factor is elevated intraocular pressure. Topical beta-blockers are affordable and widely used to lower intraocular pressure. Genetic variability has been postulated to contribute to interpersonal differences in efficacy and safety of topical beta-blockers. This review summarizes clinically significant polymorphisms that have been identified in the beta-adrenergic receptors (ADRB1, ADRB2 and ADRB3). The implications of polymorphisms in CYP2D6 are also discussed. Although the candidate-gene approach has facilitated significant progress in our understanding of the genetic basis of glaucoma treatment response, most drug responses involve a large number of genes, each containing multiple polymorphisms. Genome-wide association studies may yield a more comprehensive set of polymorphisms associated with glaucoma outcomes. An understanding of the genetic mechanisms associated with variability in individual responses to topical beta-blockers may advance individualized treatment at a lower cost.

10.
Mol Vis ; 13: 431-42, 2007 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-17417604

RESUMO

PURPOSE: Mutations in PAX6 cause human aniridia. The small eye (sey) mouse represents an animal model for aniridia. However, no large animal model currently exists. We cloned and characterized canine PAX6, and evaluated PAX6 for causal associations with inherited aniridia in dogs. METHODS: Canine PAX6 was cloned from a canine retinal cDNA library using primers designed from human and mouse PAX6 consensus sequences. An RH3000 radiation hybrid panel was used to localize PAX6 within the canine genome. Genomic DNA was extracted from whole blood of dogs with inherited aniridia, and association testing was performed using markers on CFA18. Fourteen PAX6 exons were sequenced and scanned for mutations, and a Southern blot was used to test for large deletions. RESULTS: Like the human gene, canine PAX6 has 13 exons and 12 introns, plus an alternatively spliced exon (5a). PAX6 nucleotide and amino acid sequences were highly conserved between dog, human, and mouse. The canine PAX6 cDNA sequence determined in this study spans 2 large gaps present in the current canine genomic sequence. Radiation hybrid mapping placed canine PAX6 on CFA18 in a region with synteny to HSA11p13. Exon-scanning revealed single nucleotide polymorphisms, but no pathological mutations, and Southern blot analysis revealed no differences between normal and affected animals. CONCLUSIONS: Canine PAX6 was cloned and characterized, and results provide sequence information for gaps in the current canine genome sequence. Canine PAX6 nucleotide and amino acid sequences, as well as gene organization and map location, were highly homologous with that of the human gene. PAX6 was evaluated in dogs with an inherited form of aniridia, and sequence analysis indicated no pathological mutations in the coding regions or splice sites of aniridia-affected dogs, and Southern blot analysis showed no large deletions.


Assuntos
Aniridia/veterinária , Clonagem Molecular , Modelos Animais de Doenças , Doenças do Cão/genética , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Sequência de Aminoácidos , Animais , Aniridia/genética , Sequência de Bases , Sequência Conservada , DNA Complementar , Cães , Éxons , Genoma , Fator de Transcrição PAX6 , Linhagem , Mapeamento de Híbridos Radioativos , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico
11.
Genomics ; 88(5): 541-50, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16859891

RESUMO

Canine progressive rod-cone degeneration (prcd) is a retinal disease previously mapped to a broad, gene-rich centromeric region of canine chromosome 9. As allelic disorders are present in multiple breeds, we used linkage disequilibrium (LD) to narrow the approximately 6.4-Mb interval candidate region. Multiple dog breeds, each representing genetically isolated populations, were typed for SNPs and other polymorphisms identified from BACs. The candidate region was initially localized to a 1.5-Mb zero recombination interval between growth factor receptor-bound protein 2 (GRB2) and SEC14-like 1 (SEC14L). A fine-scale haplotype of the region was developed, which reduced the LD interval to 106 kb and identified a conserved haplotype of 98 polymorphisms present in all prcd-affected chromosomes from 14 different dog breeds. The findings strongly suggest that a common ancestor transmitted the prcd disease allele to many of the modern dog breeds and demonstrate the power of the LD approach in the canine model.


Assuntos
Mapeamento Cromossômico/veterinária , Doenças do Cão/genética , Cães/genética , Degeneração Retiniana/veterinária , Animais , Sequência de Bases , Cruzamento , Cromossomos Artificiais Bacterianos/genética , DNA Complementar/genética , Doenças do Cão/patologia , Feminino , Predisposição Genética para Doença , Genética Populacional , Genômica , Haplótipos , Desequilíbrio de Ligação , Masculino , Filogenia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Especificidade da Espécie
12.
Mol Vis ; 12: 588-96, 2006 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-16760895

RESUMO

PURPOSE: To facilitate the molecular characterization of naturally occurring cataracts in dogs by providing the radiation hybrid location of 21 cataract-associated genes along with their closely associated polymorphic markers. These can be used for segregation testing of the candidate genes in canine cataract pedigrees. METHODS: Twenty-one genes with known mutations causing hereditary cataracts in man and/or mouse were selected and mapped to canine chromosomes using a canine:hamster radiation hybrid RH5000 panel. Each cataract gene ortholog was mapped in relation to over 3,000 markers including microsatellites, ESTs, genes, and BAC clones. The resulting independently determined RH-map locations were compared with the corresponding gene locations from the draft sequence of the canine genome. RESULTS: Twenty-one cataract orthologs were mapped to canine chromosomes. The genetic locations and nearest polymorphic markers were determined for 20 of these orthologs. In addition, the resulting cataract gene locations, as determined experimentally by this study, were compared with those determined by the canine genome project. All genes mapped within or near chromosomal locations with previously established homology to the corresponding human gene locations based on canine:human chromosomal synteny. CONCLUSIONS: The location of selected cataract gene orthologs in the dog, along with their nearest polymorphic markers, serves as a resource for association and linkage testing in canine pedigrees segregating inherited cataracts. The recent development of canine genomic resources make canine models a practical and valuable resource for the study of human hereditary cataracts. Canine models can serve as large animal models intermediate between mouse and man for both gene discovery and the development of novel cataract therapies.


Assuntos
Cães/genética , Mapeamento de Híbridos Radioativos , Animais , Catarata/genética , Linhagem Celular , Cricetinae , Ligação Genética , Marcadores Genéticos , Escore Lod , Polimorfismo Genético , Sitios de Sequências Rotuladas
13.
Hum Mol Genet ; 11(16): 1823-33, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12140185

RESUMO

Cone degeneration (cd ) is an autosomal recessive canine disease that occurs naturally in the Alaskan Malamute and German Shorthaired Pointer breeds. It is phenotypically similar to human achromatopsia, a heterogeneous autosomal recessive disorder associated with three distinct loci. Both the canine disease and its human counterparts are characterized by day-blindness and absence of retinal cone function in adults. We report linkage of the canine cd locus to marker C29.002 on canine chromosome 29 at recombination fraction theta = 0.0 with a maximum LOD score of 24.68 in a series of informative outbred pedigrees derived from cd-affected Alaskan Malamutes. Conserved gene order between CFA29 and the long arm of human chromosome 8 argued for homology between the cd locus and the human achromatopsia locus, ACHM3, at 8q21-22. The canine homolog of the cyclic nucleotide-gated channel beta-subunit gene (CNGB3), responsible for the human ACHM3 disease phenotype, was mapped within the zero-recombination interval for the cd locus. A deletion removing all exons of canine CNGB3 was identified in cd-affected Alaskan Malamute-derived dogs. A missense mutation in exon 6 (D262N, nucleotide 784) within a conserved region of the same gene was detected in German Shorthaired Pointers affected with an allelic disorder. Identification of these canine disorders as homologs of human ACHM3 underscores the power of recent developments in canine genomics, and provides a valuable system for exploring disease mechanisms and evaluating potential therapeutic measures in disorders of cone photoreceptors.


Assuntos
Defeitos da Visão Cromática/genética , Doenças do Cão/genética , Canais Iônicos , Mutação , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Doenças Retinianas/veterinária , Animais , Sequência de Bases , Mapeamento Cromossômico , Canais de Cátion Regulados por Nucleotídeos Cíclicos , DNA/genética , Cães , Feminino , Ligação Genética , Humanos , Hibridização in Situ Fluorescente , Masculino , Linhagem , Doenças Retinianas/genética , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...