Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Biomaterials ; 90: 40-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26986855

RESUMO

X-linked juvenile retinoschisis (XLRS) is a retinal degenerative disorder caused by mutations in the RS1 gene encoding a protein termed retinoschisin. The disease is an excellent candidate for gene replacement therapy as the majority of mutations have been shown to lead to a complete deficiency of the secreted protein in the retinal structures. In this work, we have studied the ability of non-viral vectors based on solid lipid nanoparticles (SLN) to induce the expression of retinoschisin in photoreceptors (PR) after intravitreal administration to Rs1h-deficient mice. We designed two vectors prepared with SLN, protamine, and dextran (DX) or hyaluronic acid (HA), bearing a plasmid containing the human RS1 gene under the control of the murin opsin promoter (mOPS). In vitro, the nanocarriers were able to induce the expression of retinoschisin in a PR cell line. After injection into the murine vitreous, the formulation prepared with HA induced a higher transfection level in PR than the formulation prepared with DX. Moreover, the level of retinoschisin in the inner nuclear layer (INL), where bipolar cells are located, was also higher. Two weeks after vitreal administration into Rs1h-deficient mice, both formulations showed significant improvement of the retinal structure by inducing a decrease of cavities and PR loss, and an increase of retinal and outer nuclear layer (ONL) thickness. HA-SLN resulted in a significant higher increase in the thickness of both retina and ONL, which can be explained by the higher transfection level of PR. In conclusion, we have shown the structural improvement of the retina of Rs1h-deficient mice with PR specific expression of the RS1 gene driven by the specific promoter mOPS, after successful delivery via SLN-based non-viral vectors.


Assuntos
Moléculas de Adesão Celular/genética , Proteínas do Olho/genética , Nanopartículas/química , Retina/patologia , Retinosquise/genética , Retinosquise/terapia , Animais , Deleção de Genes , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Ácido Hialurônico/química , Camundongos , Camundongos Endogâmicos C57BL , Retina/metabolismo , Retina/ultraestrutura , Retinosquise/patologia
2.
J Control Release ; 217: 273-83, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26400864

RESUMO

X-linked juvenile retinoschisis (XLRS), which results from mutations in the gene RS1 that encodes the protein retinoschisin, is a retinal degenerative disease affecting between 1/5000 and 1/25,000 people worldwide. Currently, there is no cure for this disease and the treatment is based on the application of low-vision aids. The aim of the present work was the in vitro and in vivo evaluation of two different non-viral vectors based on solid lipid nanoparticles (SLNs), protamine and two anionic polysaccharides, hyaluronic acid (HA) or dextran (DX), for the treatment of XLRS. First, the vectors containing a plasmid which encodes both the reporter green fluorescent protein (GFP) and the therapeutic protein retinoschisin, under the control of CMV promoters, were characterized in vitro. Then, the vectors were subretinally or intravitreally administrated to C57BL/6 wild type mice. One week later, GFP was detected in all treated mice and in all retinal layers except in the Outer Nuclear Layer (ONL) and the Inner Nuclear Layer (INL), regardless of the administration route and the vector employed. Finally, two weeks after subretinal or intravitreal injection to Rs1h-deficient mice, GFP and retinoschisin expression was detected in all retinal layers, except in the ONL, which was maintained for at least two months after subretinal administration. The structural analysis of the treated Rs1h-deficient eyes showed a partial recovery of the retina related to the production of retinoschisin. This work shows for the first time a successful RS1 gene transfer to Rs1h-deficient animals using non-viral nanocarriers, with promising results that point to non-viral gene therapy as a feasible future therapeutic tool for retinal disorders.


Assuntos
Moléculas de Adesão Celular/genética , Proteínas do Olho/genética , Terapia Genética/métodos , Retinosquise/terapia , Animais , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , DNA/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lipídeos/administração & dosagem , Lipídeos/química , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nanopartículas/administração & dosagem , Nanopartículas/química , Retina/metabolismo
3.
Klin Monbl Augenheilkd ; 232(3): 250-8, 2015 Mar.
Artigo em Alemão | MEDLINE | ID: mdl-25803554

RESUMO

An early diagnosis of inherited retinal or optic nerve disorders is often delayed due to unspecific clinical signs, multiple clinical manifestations and striking genetic heterogeneity of the underlying molecular defects. This study represents a retrospective analysis of findings in 4,021 patients with inherited retinal or optic nerve disorders seen between 1986 and 2014 (1,171 with follow-up). In addition to the basic ophthalmological examination, electrophysiological tests (ERG, n = 2,088, since 1986; EOG, n = 381, since 1986; VEP n = 595, since 1986; mfERG, n = 819, since 1998) and non-invasive retinal imaging (fundus autofluorescence (FAF, n = 1,784, since 2002), near-infrared autofluorescence (NIA, n = 1,091, since 2006), spectral domain OCT (SD-OCT, n = 848, since 2008) and three-wavelengths multicolour spectral reflection imaging (MC, n = 366, since 2013) were performed at least once. Molecular DNA testing was done in 383 patients between 2006 and 2014. Based on these data an efficient diagnostic strategy is suggested: 1) inclusion of inherited retinal and optic nerve disorders into the differential diagnosis of visual loss or visual field defects with undefined causes; 2) non-invasive retinal imaging; 3) electrophysiological tests; 4) DNA testing to confirm the initial clinical diagnosis; 5) examination in specialised centres, therapy and follow-up. In recent years, the spectrum of diagnostic techniques has continuously expanded. Importantly, non-invasive retinal imaging has become the primary diagnostic tool and DNA testing based on state-of-the-art high throughput techniques increases the identification of associated gene mutations. In conclusion, a structured process in the diagnostic procedure of inherited retinal and optic nerve disorders greatly reduces a diagnostic delay, enables an earlier counselling and therapy and avoids further unnecessary diagnostic tests.


Assuntos
Técnicas de Diagnóstico Oftalmológico , Testes Genéticos/métodos , Doenças do Nervo Óptico/diagnóstico , Doenças do Nervo Óptico/genética , Doenças Retinianas/diagnóstico , Doenças Retinianas/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Criança , Pré-Escolar , Feminino , Predisposição Genética para Doença/genética , Alemanha , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Patologia Molecular/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Adulto Jovem
4.
J Orofac Orthop ; 75(3): 226-39, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24825834

RESUMO

BACKGROUND: Primary failure of eruption (PFE) may be associated with pathogenic mutations in the PTHR1 gene. It has numerous manifestations and is characterized by severe posterior open bite. However, there are also phenotypically similar types of eruption anomalies not associated with a known pathogenic PTHR1 mutation. The purpose of this study was to evaluate whether a distinction can be made between PTHR1-mutation carriers and noncarriers based on clinical and radiological findings. PATIENTS AND METHODS: A total of 36 patients with suspected PFE diagnoses were included and analyzed in accordance with specific clinical and radiographic criteria. In addition, all patients underwent Sanger DNA sequencing analysis of all coding sequences (and the immediate flanking intronic sequences) of the PTHR1 gene. RESULTS: Of these patients, 23 exhibited a heterozygous pathogenic mutation in the PTHR1 gene (PTHR1-mutation carriers), while molecular genetic analysis revealed nosequence alteration in the other 13 patients (non-PTHR1-mutation carriers). Relevant family histories were obtained from 5 patients in the carrier group; hence, this group included a total of 13 familial and 10 simplex cases. The group of noncarriers revealed no relevant family histories. All patients in the carrier group met six of the clinical and radiographic criteria explored in this study: (1) posterior teeth more often affected; (2) eruption disturbance of an anterior tooth in association with additional posterior-teeth involvement; (3) affected teeth resorbing the alveolar bone located coronal to them; (4) involvement of both deciduous and permanent teeth; (5) impaired vertical alveolar-process growth; and (6) severe subsequent finding of posterior open bite. None of the analyzed criteria were, by contrast, met by all patients in the noncarrier group. All patients in the carrier group could be assigned to one of three classifications indicating the extent of eruption disturbance, whereas 4 of the 13 noncarriers presented none of these three patterns. The clinical and radiographic criteria employed in this study would have correctly identified 10 of the 13 PFE patients in the noncarrier group as possessing no detectable PTHR1 mutation. CONCLUSION: The evaluation of clinical and radiographic characteristics can heighten the specificity of ruling out suspected PTHR1 involvement in PFE patients. A hereditary element of PTHR1-associated PFE is clearly identifiable. More studies with more patients are needed to optimize the sensitivity of this preliminary approach on the differential identification of PTHR1-mutation carriers versus noncarriers by multivariate analysis.


Assuntos
Predisposição Genética para Doença/genética , Técnicas de Diagnóstico Molecular/métodos , Radiografia Dentária/métodos , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Doenças Dentárias/diagnóstico por imagem , Doenças Dentárias/genética , Adolescente , Criança , Diagnóstico Diferencial , Feminino , Marcadores Genéticos/genética , Humanos , Masculino , Mutação/genética , Variações Dependentes do Observador , Polimorfismo de Nucleotídeo Único/genética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Adulto Jovem
7.
Cell Prolif ; 43(4): 372-7, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20590662

RESUMO

OBJECTIVES: In a previous study, we have reported the existence of neural crest-derived stem cell-like cells originating from the corneal limbus of juvenile mice (termed murine corneal cells, MCCs). To yield a sufficient number of MCCs, for example, for cell-therapy approaches, here we have investigated MCCs' ability for extensive proliferation, and we have evaluated their stem cell qualities and genetic stability after large-scale culture. MATERIALS AND METHODS: MCCs were established from corneal limbal tissue of juvenile mice. To determine their cell proliferation and self-renewing potential, MTT tests and an estimation of colony forming unit efficiency were carried out. Multipotency of cell differentiation was examined by applying adipogenic and osteogenic differentiation protocols. Moreover, karyotyping was performed and expression of stem cell markers and cell cycle-associated genes was analysed. RESULTS: MCCs, as primary cells, could be cultured for more than 60 passages. We observed increased cell proliferation and high number of colony forming units (CFUs) after extensive culture. Interestingly, there were no changes in expression of MCC markers. Furthermore, cell differentiation potentials remained comparable with MCCs at early passages. However, karyotyping revealed numeric chromosomal aberrations at higher passages. Moreover, tumour suppressor genes such as p16 and p21 were found to be down-regulated after large-scale cell culture. CONCLUSIONS: MCCs immortalize spontaneously after extensive cell culture, but still demonstrate stem cell-like qualities.


Assuntos
Limbo da Córnea/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Técnicas de Cultura de Células , Diferenciação Celular/genética , Proliferação de Células , Aberrações Cromossômicas , Córnea , Cariotipagem , Limbo da Córnea/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/metabolismo , Neurônios/citologia , Neurônios/metabolismo
8.
Klin Monbl Augenheilkd ; 226(12): 999-1011, 2009 Dec.
Artigo em Alemão | MEDLINE | ID: mdl-19757352

RESUMO

BACKGROUND: Hereditary retinal dystrophies comprise a heterogeneous group of inherited retinal disorders with variable clinical presentation and multiple associated genes. Clinical diagnosis and differential diagnosis are difficult. The purpose of the current paper is to provide guidelines for an effective diagnostic approach. METHODS: A literature search was carried out and our own data on clinical (n = 3200) and molecular genetic (n = 4050) diagnosis of patients with retinal dystrophies were evaluated. RESULTS: For an early diagnosis it is of importance to include inherited retinal dystrophies in the differential diagnosis of unexplained visual disturbances. The most important clinical test is the full-field electroretinogram (ERG), which allows detection or exclusion of generalised retinal dystrophies. If the full-field ERG is normal, a multifocal ERG will distinguish macular dystrophies. Fundus autofluorescence, near-infrared autofluorescence and high resolution optical coherence tomography improve the early diagnosis because morphological alterations can be detected prior to their ophthalmoscopic visibility. In addition, these non-invasive imaging techniques reveal new phenomena which are important for the differential diagnosis and follow-up of retinal dystrophies as well as for an improved understanding of their pathogenesis. Routine molecular genetic diagnosis is available for an increasing number of retinal dystrophies. A succinct clinical diagnosis is a prerequisite to allow selection of the gene(s) to be analysed. If genetic testing is indicated, a human geneticist should be involved for counselling of the patient and possibly further family members and initiation of the necessary steps for DNA testing. CONCLUSION: The combination of electrophysiological testing, retinal imaging and molecular genetic analysis allows a differentiated diagnosis of inherited retinal dystrophies and an individual counselling of patients. If inherited retinal dystrophies are suspected, a detailed examination in a retinal centre specialised on inherited retinal dystrophies is recommended.


Assuntos
Eletrorretinografia/tendências , Testes Genéticos/tendências , Degeneração Retiniana/diagnóstico , Degeneração Retiniana/genética , Retinoscopia/tendências , Medicina Baseada em Evidências , Predisposição Genética para Doença/genética , Humanos
10.
Eye (Lond) ; 23(12): 2238-44, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19169232

RESUMO

AIMS: Little is known about the role of genetic variants in the early stages of age-related macular degeneration (AMD). We aimed to investigate how genetic variations within five well-defined genes relate to AMD severity. METHODS: We analysed SNPs in the genes for complement factor H (CFH), age-related maculopathy susceptibility (ARMS2), HtrA serine peptidase 1 (HtrA1), complement factor B (CFB), and complement component 2 (C2)in 183 controls and 730 patients with increasing severity of AMD from the Muenster aging and retina study (MARS). Severity scoring was based on the Rotterdam classification of fundus photographs. RESULTS: Compared with controls, patients with very early AMD showed a significantly increased minor allele frequency (MAF) only for CFH-rs1061170. With increasing severity of AMD, SNPs in CFH-rs1061170,as well as ARMS2-rs10490924, became consistently more common (P<0.001). Likewise, HtrA1-rs11200638 was less clearly associated with AMD severity, whereas C2-rs9332739 and CFB-rs641153 showed no relation. Multifactorial models confirmed CFH and ARMS2 as major determinants of AMD severity, whereas addition of HtrA1, C2 and CFB did not improve model prediction. In the models, age did not contribute to very early but to all more severe AMD stages, whereas smoking history had a significant impact only for late AMD. CONCLUSION: Our findings indicate that the CFH gene is involved in the onset of AMD, whereas both, the CFH and ARMS2 genes, and more weakly, the HtrA1 gene, appear to account for the advancement of AMD. The results for SNPs in the C2 and CFB genes were inconclusive. Genetic factors dominated in their impact over age and smoking history.


Assuntos
Proteínas do Sistema Complemento/genética , Degeneração Macular/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas/genética , Serina Endopeptidases/genética , Idoso , Alelos , Complemento C2/genética , Fator B do Complemento/genética , Fator H do Complemento/genética , Feminino , Fundo de Olho , Frequência do Gene , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Modelos Logísticos , Degeneração Macular/fisiopatologia , Masculino , Retina/patologia , Análise de Sequência de DNA , Índice de Gravidade de Doença
11.
Eye (Lond) ; 23(6): 1349-59, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18791550

RESUMO

AIMS: To compare melanin-related near-infrared fundus autofluorescence (FAF; NIA, excitation 787 nm, emission >800 nm) with lipofuscin-related FAF (excitation 488 nm, emission >500 nm) in retinitis pigmentosa (RP). METHODS: Thirty-three consecutive RP patients with different modes of inheritance were diagnosed clinically, with full-field ERG, and if possible with molecular genetic methods. FAF and NIA imaging were performed with a confocal scanning laser ophthalmoscope (Heidelberg Retina Angiograph 2). RESULTS: Rings of increased FAF were present within an area of preserved retinal pigment epithelium (RPE) at the posterior pole (31/33). Rings of increased NIA were located in the same region as rings of increased FAF. In contrast to FAF, NIA showed a precipitous decline of NIA peripheral to the ring. In larger areas of preserved NIA (11/31), pericentral and foveal NIA were of similar intensity with an area of lower NIA in between. In smaller areas of preserved NIA (20/31), NIA was homogeneous from the perifovea to the fovea. In one patient without a ring of increased FAF, NIA distribution was normal. In the remaining patient with severely advanced RP, no residual RPE as well as no FAF and NIA were detectable. CONCLUSION: Characteristic features for FAF and NIA alterations in a heterogeneous group of RP patients indicate a common pathway of RPE degeneration. Patterns of NIA and FAF indicate different pathophysiologic processes involving melanin and lipofuscin. Combined NIA and FAF imaging will provide further insight into the pathogenesis of RP and non-invasive monitoring of future therapeutic interventions.


Assuntos
Técnicas de Diagnóstico Oftalmológico , Fluorescência , Lipofuscina/análise , Melaninas/análise , Epitélio Pigmentado da Retina/patologia , Retinose Pigmentar/diagnóstico , Adolescente , Adulto , Idoso , Biomarcadores/análise , Criança , Feminino , Fundo de Olho , Humanos , Masculino , Pessoa de Meia-Idade , Retinose Pigmentar/patologia , Adulto Jovem
12.
Cytogenet Genome Res ; 115(1): 35-44, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16974082

RESUMO

Early schisis cavities in the retinal bipolar cell layer accompanied by progressive loss of cone and rod photoreceptor cells are the hallmark of the retinoschisin-deficient (Rs1h(-/Y)) murine retina. With this study we aimed at elucidating the molecular events underlying the photoreceptor cell death in this established murine model of X-linked juvenile retinoschisis. We show that photoreceptor degeneration in the Rs1h(-/Y) mouse is due to apoptotic events peaking around postnatal day 18. Cell death is accompanied by increased expression of initiator and inflammatory caspases but not by downstream effector caspases. The strong induction of caspase-1 (Casp1) prompted us to explore its involvement in the apoptotic process. We therefore generated double knock-out mice deficient for both retinoschisin and Casp1. No direct influence of the Casp1 genotype on apoptosis could be identified although striking differences in the overall number of resident microglia were observed independent of the Rs1h genotype.


Assuntos
Apoptose , Caspase 1/fisiologia , Caspases/fisiologia , Moléculas de Adesão Celular/genética , Proteínas do Olho/genética , Animais , Caspase 1/genética , Caspases/genética , Contagem de Células , Regulação Enzimológica da Expressão Gênica , Genótipo , Camundongos , Microglia , Células Fotorreceptoras de Vertebrados , Retinosquise/genética , Retinosquise/patologia
13.
Ophthalmologe ; 102(11): 1029-35, 2005 Nov.
Artigo em Alemão | MEDLINE | ID: mdl-16170519

RESUMO

Age-related macular degeneration is a complex genetic disorder. Recent data suggest that the additive genetic risk for late-stage disease is more than two-thirds. Comprehensive genetic studies (candidate gene approaches, linkage and association studies) have been performed in recent years to identity the genetic risk factors at the molecular lavel. Very recently, a significant risk allele, Y402H, has been discovered in the complement factor H (CFH) gene. The relative risk of developing AMD has been estimated between 2.4-4.6 for heterozygotes and 3.3-7.4 for homozygotes. This polymorphism accounts for approximately 20-50% of the overall risk of developing AMD. In this review the results from molecular genetic studies in AMD are summarized, with a special emphasis on the recent data obtained for the CFH gene.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Degeneração Macular/sangue , Degeneração Macular/epidemiologia , Medição de Risco/métodos , Fator H do Complemento/genética , Análise Mutacional de DNA/métodos , França/epidemiologia , Predisposição Genética para Doença/epidemiologia , Testes Genéticos/métodos , Humanos , Incidência , Degeneração Macular/genética , Polimorfismo Genético , Fatores de Risco
14.
Ophthalmologe ; 102(2): 116-21, 2005 Feb.
Artigo em Alemão | MEDLINE | ID: mdl-15627199

RESUMO

Best's vitelliform macular dystrophy (Best's disease) is an autosomal dominant disease of the central retina and is caused by mutations in the VMD2 gene located on the long arm of chromosome 11. VMD2 encodes bestrophin, a transmembrane protein with putative Ca(2+)-dependent chloride channel activity at the basolateral portion of the retinal pigment epithelium. The N-terminal half of bestrophin reveals high sequence homology to three bestrophin-like proteins in humans but also to protein sequences from evolutionarily distant organisms. Most of the known VMD2 mutations are located within this presumably important functional part of the protein and cause amino acid substitutions and small in-frame deletions of single amino acid residues. The pathogenicity of VMD2 mutations is likely based on a dominant negative effect possibly by oligomerization of normal and mutated bestrophin molecules to form a defective ion channel. Missense mutations in VMD2 were also shown to be associated with vitreoretinochoroidopathy and ocular developmental abnormalities. In this case, the pathogenic sequence changes influence the peptide sequences but simultaneously alter the regulation of mRNA splicing and maturation. Different disease mechanisms may therefore be responsible for the distinct phenotypes associated with VMD2 mutations.


Assuntos
Cromossomos Humanos Par 11 , Distrofias Hereditárias da Córnea/genética , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Predisposição Genética para Doença/genética , Degeneração Macular/metabolismo , Bestrofinas , Biomarcadores/metabolismo , Canais de Cloreto/metabolismo , Distrofias Hereditárias da Córnea/diagnóstico , Distrofias Hereditárias da Córnea/metabolismo , Proteínas do Olho/química , Humanos , Degeneração Macular/diagnóstico , Degeneração Macular/genética , Polimorfismo Genético , Doenças Retinianas/diagnóstico , Doenças Retinianas/genética , Doenças Retinianas/metabolismo , Fatores de Risco , Relação Estrutura-Atividade
15.
Cytogenet Genome Res ; 108(1-3): 229-33, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15545735

RESUMO

The gene encoding claudin-1 (CLDN1) has been mapped to human chromosome 3 (HSA3; 3q28-->q29) using a radiation hybrid panel. Employing fluorescence in situ hybridization (FISH) we here show that a human P1-derived artificial chromosome (PAC) containing CLDN1 detects the orthologous sites in chromosomes of the great apes, chimpanzee, gorilla, and orangutan. Furthermore, the chromosomal position of CLDN1 was determined in mouse chromosomes by FISH. The position of fluorescent signals is confined to a single chromosomal site in both great apes and mouse and in each case maps to the chromosomal region that has conserved synteny with HSA3 (PTR2q28, GGO2q28, PPY2q38 and MMU16B1). Using a gene-specific probe our results are consistent with reports of the striking similarity of great ape and human genomes as illustrated previously by chromosome painting.


Assuntos
Mapeamento Cromossômico/métodos , Gorilla gorilla/genética , Proteínas de Membrana/genética , Pan troglodytes/genética , Pongo pygmaeus/genética , Animais , Cromossomos Humanos Par 3/genética , Cromossomos de Mamíferos/genética , Claudina-1 , Humanos , Linfócitos/química , Linfócitos/metabolismo , Metáfase/genética , Camundongos , Sintenia/genética
16.
Hum Pathol ; 35(10): 1260-5, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15492994

RESUMO

PTEN: and beta-catenin mutations constitute the predominant genetic alterations in endometrioid carcinomas of the endometrium. PTEN encodes a dual-specificity phosphatase with lipid phosphatase and protein tyrosine phosphatase activities that regulate both apoptosis and interactions with the extracellular matrix. Recent studies have associated PTEN mutations with tumorigenesis of prostate carcinoma via the Wnt signaling pathway, leading to nuclear beta-catenin accumulation. To elucidate the potential interaction of PTEN and beta-catenin in endometrial cancer, we performed mutation analyses of the entire PTEN gene and of exon 3 of the beta-catenin gene that is most frequently targeted by mutations. A total of 82 endometrial carcinomas comprising 62 type I endometrioid carcinomas and 20 type II high-grade carcinomas were investigated. In addition in a subset of 22 carcinomas, the intracellular beta-catenin distribution was analyzed by immunohistochemistry. Overall, 20 (24.4%) of 82 tumors revealed mutations in the PTEN gene, and 16 (19.5%) of 82, in the beta-catenin gene. Six tumors (7.3%) showed mutations in both the PTEN and beta-catenin gene. Mutations were mainly detected in endometrioid carcinomas of the endometrium. As expected, a striking nuclear accumulation of beta-catenin could be shown in tumors with beta-catenin mutations. In the vast majority of tumors with PTEN mutations, a regular staining pattern of the cytoplasmic and membranous compartments was found. We therefore conclude that, in contrast to prostate cancer, mutations in the PTEN gene seem not to affect cellular distribution of the beta-catenin protein in endometrial carcinomas.


Assuntos
Carcinoma Endometrioide/genética , Carcinoma Endometrioide/metabolismo , Proteínas do Citoesqueleto/metabolismo , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Mutação , Monoéster Fosfórico Hidrolases/genética , Transativadores/metabolismo , Proteínas Supressoras de Tumor/genética , Carcinoma Endometrioide/patologia , Núcleo Celular/metabolismo , Análise Mutacional de DNA , Neoplasias do Endométrio/patologia , Feminino , Humanos , Imuno-Histoquímica , PTEN Fosfo-Hidrolase , beta Catenina
17.
Cytogenet Genome Res ; 106(1): 74-81, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15218245

RESUMO

Retina and retinal pigment epithelium (RPE) cells are of neuroectodermal origin with highly specialized functions in light perception. Identification and characterization of genes differentially expressed in these cells will greatly aid our understanding of their functional roles in retinal biology. As a source enriched for gene transcripts from the retina/RPE, we generated a human retina and a bovine RPE cDNA library applying the PCR-based technique of suppression subtractive hybridization (SSH). Sequencing of 1,080 retina and 2,350 RPE SSH clones resulted in the identification of 321 and 343 non-redundant human transcripts, respectively. Of these, only 27 genes were in common between the two cDNA libraries. One transcript expressed exclusively in retina and RPE is the novel gene C4orf11 which is comprised of four exons on chromosome 4q21.2. We report the full-length cloning of two isoforms of C4orf11, 919 bp and 857 bp in length, both of which contain four identical open reading frames (ORFs). While ORFs 1 to 3 show no homologies to known proteins or protein domains, ORF4 reveals 50% sequence identity to RPE-spondin, a hypothetical protein on 8q13.3 with unknown function. We demonstrate that both the retina and the RPE SSH cDNA libraries are excellent resources for identifying known and novel genes exclusively or abundantly expressed in the retina/RPE complex. In combination with other approaches such as microarray analysis or serial analysis of gene expression (SAGE), the availability of highly sensitive and specific SSH cDNA libraries will facilitate the comprehensive description of the retina/RPE transcriptome.


Assuntos
Expressão Gênica , Epitélio Pigmentado Ocular/metabolismo , Retina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Cromossomos Humanos Par 4 , Éxons , Componentes do Gene , Biblioteca Gênica , Humanos , Dados de Sequência Molecular , Hibridização de Ácido Nucleico/métodos , Especificidade de Órgãos/genética , RNA Mensageiro/análise
18.
Cytogenet Genome Res ; 105(1): 107-14, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15218265

RESUMO

Mutations in the human vitelliform macular dystrophy type 2 (VMD2) gene are known to cause autosomal dominant Best macular dystrophy (BMD), a degenerative disorder of the central retina. VMD2, together with VMD2L1, VMD2L2 and VMD2L3, belong to a closely related gene family characterized by several transmembrane (TM) spanning helical domains and an invariant arginine, phenylalanine and proline (RFP) tripeptide motif, thus termed VMD2 RFP-TM. The four genes are thought to encode a novel family of anion channels. We now report the cloning and characterization of the murine orthologs by combining biocomputational analyses and molecular genetic approaches. While the murine Vmd2, Vmd2l1 and Vmd2l3 genes are functional, murine Vmd2l2p was found to be a non-transcribed pseudogene. Expression profiling of the murine Vmd2 RFP-TM family members revealed tissue-restricted expression with predominant transcription of Vmd2 in testis, of Vmd2l1 in colon and of Vmd2l3 in heart. Differential splicing was observed for Vmd2l3 in a number of tissues (e.g. in brain, retina/RPE, kidney) although the functional importance of the splice variants remains to be determined.


Assuntos
Proteínas do Olho/genética , Processamento Alternativo , Animais , Sequência de Bases , Bestrofinas , Canais de Cloreto , Clonagem de Organismos , DNA Complementar , Proteínas de Ligação a DNA/genética , Bases de Dados Genéticas , Etiquetas de Sequências Expressas , Perfilação da Expressão Gênica , Humanos , Canais Iônicos , Camundongos , Dados de Sequência Molecular , Família Multigênica , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Ubiquitina-Proteína Ligases
20.
Br J Ophthalmol ; 85(8): 969-75, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11466257

RESUMO

BACKGROUND: A long term project was initiated to identify and to characterise genes that are expressed exclusively or preferentially in the retina as candidates for a genetic susceptibility to age related macular degeneration (AMD). A transcript represented by a cluster of five human expressed sequence tags (ESTs) derived exclusively from retinal cDNA libraries was identified. METHODS: Northern blot and RT-PCR analyses confirmed preferential retinal expression of the gene, which encodes a G protein coupled receptor, GPR75. Following isolation of the full length cDNA and determination of the genomic organisation, the coding sequence of GPR75 was screened for mutations in 535 AMD patients and 252 controls from Germany, the United States, and Italy. Employed methods included single stranded conformational polymorphism (SSCP) analysis, denaturing high performance liquid chromatography (DHPLC), and direct sequencing. RESULTS: Nine different sequence variations were identified in patients and control individuals. Three of these (-30A>C, 150G>A, and 346G>A) likely represent polymorphic variants. Each of six alterations (-4G>A, N78K, P99L, S108T, T135P, and Q234X) were found once in single AMD patients and were considered variants that could affect the protein function and potentially cause retinal pathology. CONCLUSION: The presence of six potential pathogenic variants in a cohort of 535 AMD patients alone does not provide statistically significant evidence for the association of sequence variation in GPR75 with genetic predisposition to AMD. However, a possible connection between the variants and age related retinal pathology cannot be discarded. Functional studies are needed to clarify the role of GPR75 in retinal physiology.


Assuntos
Etiquetas de Sequências Expressas , Degeneração Macular/genética , Receptores de Superfície Celular/genética , Retina/metabolismo , Adulto , Distribuição Binomial , Northern Blotting , Estudos de Casos e Controles , Cromatografia Líquida de Alta Pressão , Biblioteca Gênica , Predisposição Genética para Doença , Humanos , Degeneração Macular/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação/genética , Polimorfismo Conformacional de Fita Simples , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA