Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Virol ; 88(17): 9673-82, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24920813

RESUMO

UNLABELLED: Human endogenous retrovirus type K (HERV-K) proviruses are scattered throughout the human genome, but as no infectious HERV-K virus has been detected to date, the mechanism by which these viruses replicated and populated the genome remains unresolved. Here, we provide evidence that, in addition to the RNA genomes that canonical retroviruses package, modern HERV-K viruses can contain reverse-transcribed DNA (RT-DNA) genomes. Indeed, reverse transcription of genomic HERV-K RNA into the DNA form is able to occur in three distinct times and locations: (i) in the virus-producing cell prior to viral release, yielding a DNA-containing extracellular virus particle similar to the spumaviruses; (ii) within the extracellular virus particle itself, transitioning from an RNA-containing particle to a DNA-containing particle; and (iii) after entry of the RNA-containing virus into the target cell, similar to canonical retroviruses, such as murine leukemia virus and HIV. Moreover, using a resuscitated HERV-K virus construct, we show that both viruses with RNA genomes and viruses with DNA genomes are capable of infecting target cells. This high level of genomic flexibility historically could have permitted these viruses to replicate in various host cell environments, potentially assisting in their many integration events and resulting in their high prevalence in the human genome. Moreover, the ability of modern HERV-K viruses to proceed through reverse transcription and package RT-DNA genomes suggests a higher level of replication competency than was previously understood, and it may be relevant in HERV-K-associated human diseases. IMPORTANCE: Retroviral elements comprise at least 8% of the human genome. Of all the endogenous retroviruses, HERV-K viruses are the most intact and biologically active. While a modern infectious HERV-K has yet to be found, HERV-K activation has been associated with cancers, autoimmune diseases, and HIV-1 infection. Thus, determining how this virus family became such a prevalent member of our genome and what it is capable of in its current form are of the utmost importance. Here, we provide evidence that HERV-K viruses currently found in the human genome are able to proceed through reverse transcription and historically utilized a life cycle with a surprising degree of genomic flexibility in which both RNA- and DNA-containing viruses were capable of mediating infection.


Assuntos
DNA Viral/genética , DNA Viral/metabolismo , Retrovirus Endógenos/genética , Genoma Viral , Provírus/genética , Linhagem Celular Tumoral , Retrovirus Endógenos/fisiologia , Humanos , RNA Viral/genética , RNA Viral/metabolismo , Transcrição Reversa , Montagem de Vírus
2.
Genome Res ; 23(9): 1505-13, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23657884

RESUMO

Human endogenous retroviruses (HERVs) make up 8% of the human genome. The HERV-K (HML-2) family is the most recent group of these viruses to have inserted into the genome, and we have detected the activation of HERV-K (HML-2) proviruses in the blood of patients with HIV-1 infection. We report that HIV-1 infection activates expression of a novel HERV-K (HML-2) provirus, termed K111, present in multiple copies in the centromeres of chromosomes throughout the human genome yet not annotated in the most recent human genome assembly. Infection with HIV-1 or stimulation with the HIV-1 Tat protein leads to the activation of K111 proviruses. K111 is present as a single copy in the genome of the chimpanzee, yet K111 is not found in the genomes of other primates. Remarkably, K111 proviruses appear in the genomes of the extinct Neanderthal and Denisovan, while modern humans have at least 100 K111 proviruses spread across the centromeres of 15 chromosomes. Our studies suggest that the progenitor K111 integrated before the Homo-Pan divergence and expanded in copy number during the evolution of hominins, perhaps by recombination. The expansion of K111 provides sequence evidence suggesting that recombination between the centromeres of various chromosomes took place during the evolution of humans. K111 proviruses show significant sequence variations in each individual centromere, which may serve as markers in future efforts to annotate human centromere sequences. Further, this work is an example of the potential to discover previously unknown genomic sequences through the analysis of nucleic acids found in the blood of patients.


Assuntos
Retrovirus Endógenos/genética , Genoma Humano , Infecções por HIV/genética , Provírus/genética , Integração Viral , Animais , Centrômero/genética , Centrômero/virologia , Cromossomos Humanos/genética , Cromossomos Humanos/virologia , Evolução Molecular , Hominidae/genética , Hominidae/virologia , Humanos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
3.
Arch Ophthalmol ; 129(1): 23-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21220625

RESUMO

OBJECTIVE: To investigate the etiology of acute exudative polymorphous vitelliform maculopathy (AEPVM) in a patient with metastatic melanoma, undiagnosed at initial examination, by testing for autoimmune mechanisms. METHODS: Serum samples were obtained from a 50-year-old man with AEPVM and metastatic unknown primary melanoma during the acute stage and 3 years later when subretinal fluid had resolved and melanoma was in remission (AEPVM convalescent stage). Western immunoblots using both serum samples against human donor retinal extract and cultured primary human retinal pigment epithelium (RPE) cell extract were performed to identify antiretinal and anti-RPE antibodies. Serum samples from 5 unaffected participants were tested as controls. Protein identification was performed using 2-dimensional gel electrophoresis and mass spectrometry and was then confirmed by blotting against purified protein. RESULTS: Western immunoblots of the patient's serum against human donor retinal extract and RPE cell extract demonstrated several antiretinal antibodies, as well as anti-RPE antibodies against a 26-kDa protein that was identified as peroxiredoxin 3 (PRDX3). Serum reactivity against PRDX3 was greatly decreased in the convalescent-stage serum sample compared with the acute-stage serum sample, while results of retinal extract Western immunoblots remained essentially unchanged. Five separate serum samples from participants without AEPVM had no autoantibodies against PRDX3. CONCLUSIONS: Paraneoplastic autoimmune reaction against RPE, with PRDX3 as the putative antigen, may be a cause of AEPVM. This is the first report to date linking a human RPE disease with anti-RPE antibodies against a heretofore undetermined putative protein. Testing for RPE autoantibodies may be useful in exploring the pathogenesis of other presumed RPE-related diseases.


Assuntos
Autoanticorpos/sangue , Autoantígenos/imunologia , Síndromes Paraneoplásicas/imunologia , Peroxirredoxinas/sangue , Epitélio Pigmentado da Retina/imunologia , Distrofia Macular Viteliforme/imunologia , Doença Aguda , Antineoplásicos Alquilantes/uso terapêutico , Western Blotting , Células Cultivadas , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Eletroforese em Gel Bidimensional , Eletrorretinografia , Exsudatos e Transudatos , Angiofluoresceinografia , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Metástase Linfática , Masculino , Espectrometria de Massas , Melanoma/tratamento farmacológico , Melanoma/imunologia , Melanoma/secundário , Pessoa de Meia-Idade , Neoplasias Primárias Desconhecidas/tratamento farmacológico , Neoplasias Primárias Desconhecidas/patologia , Síndromes Paraneoplásicas/diagnóstico , Peroxirredoxina III , Temozolomida , Tomografia de Coerência Óptica , Distrofia Macular Viteliforme/diagnóstico
4.
Biochim Biophys Acta ; 1811(2): 76-83, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21081177

RESUMO

Fenofibrate, a PPAR-α agonist, lowers triglycerides (TG) and raises high-density lipoproteins (HDL-C) in humans. While fenofibrate is very effective in lowering TG, it does not raise HDL-C in humans to the same extent as seen in human apoAI transgenic (hAI-Tg) mice. We studied the mechanism of this discordance using the following compounds as tools: cholic acid that down-regulates human apoAI, and fenofibrate, that elevates hapoAI and HDL-C in hAI-Tg mice. We hypothesized that additional sequences, including apoCIII and AIV genes on chromosome 11, not present in the hapoAI transgene may be responsible for the dampened effect of fibrates on HDL-C seen in humans. For this, hAI-Tg mice with 11kb DNA segment and hapoAI-CIII-AIV-Tg mice with 33kb DNA segment harboring apoCIII and AIV genes were employed. These mice were treated with fenofibrate and cholic acid. Fenofibrate increased apoAI and HDL-C levels, and HDL size in the apoAI-Tg mice via up-regulation of the hapoAI mRNA and increased activity and mRNA of PLTP, respectively. Consistent with earlier findings, cholic acid showed similar effects of lowering HDL-C, and elevating LDL-C in hAI-Tg mice as well as in the hAI-CIII-AIV-Tg mice. Fenofibrate decreased TG and increased HDL size in hAI-CIII-AIV-Tg mice as well, but surprisingly, did not elevate serum levels of hapoAI or hepatic AI mRNA, suggesting that additional sequences not present in the hapoAI transgene (11kb) may be partly responsible for the dampened effect on HDL-C seen in hAI-CIII-AIV-Tg mice. Since hAI-CIII-AIV-Tg mouse mimics fenofibrate effects seen in humans, this transgenic mouse could serve as a better predictive model for screening HDL-C raising compounds.


Assuntos
Apolipoproteína A-I/sangue , Fenofibrato/metabolismo , Hipolipemiantes/metabolismo , Lipoproteínas HDL/sangue , Animais , Apolipoproteína A-I/genética , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ácido Cólico/metabolismo , Humanos , Lipoproteínas HDL/genética , Camundongos , Camundongos Transgênicos , Triglicerídeos/sangue
5.
Mol Vis ; 16: 1936-48, 2010 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-21031137

RESUMO

PURPOSE: Recoverin has been demonstrated to be one of the main causative antigenic retinal proteins common in many cases of autoimmune retinopathy (AIR). Strategies for producing two different AIR mouse models associated with anti-recoverin antibodies were tested. METHODS: (1) Six-week-old female B6.MRL-Fal(lpr)/J mice (LPR) mice were immunized with recombinant recoverin three times at 2-4 week intervals. (2) Five-month-old Balb/cJ mice were injected with hybridoma cells designed to produce recoverin monoclonal antibodies. Anti-recoverin antibodies were analyzed by immunoblot and enzyme-linked immunosorbent assay (ELISA). Electroretinograms (ERG), histopathologic examination, and flow cytometric analysis were assessed. RESULTS: High anti-recoverin antibody levels were achieved in both models, accompanied by significantly reduced scotopic and photopic responses on the ERGs. Retinal histology showed swollen cell bodies in the inner nuclear layer in recoverin-immunized LPR mice, while photoreceptor and outer nuclear layer swelling was observed in recoverin hybridoma cells injected into balb/cJ mice. Glial fibrillary acidic protein (GFAP) staining detected a marked increase of Müller cells and astrocyte reactive gliosis in both mouse models. Rhodopsin and S-opsin staining was similar to controls, while decreased numbers of bipolar cells were observed in both models. Complement component C1q and C3 deposits increased upon immunohistopathologic retinal staining in both models, while increased numbers of CD4+ and CD68+ cells from retinas were found upon flow cytometric analysis. CONCLUSIONS: These two models had similar pathology in the retina, indicating the retinal antigens to recoverin antibody set off pathologic events that include leukocyte invasion, complement deposition, reactive gliosis in the retina, and selective retinal degeneration of inner nuclear layer neurons. These two AIR mouse models will allow for detailed pathologic investigation and testing of protein antigens associated with human AIR and can be used to test treatments. It is important to note that, since most AIR patients have multiple anti-retinal antibodies, it will be possible to study which antibodies are pathologic and which have no retinal pathologic effects. These models can also serve as an important research resource for studying the pathophysiology of specific retinal proteins by creating autoantibodies, which potentially will give a better understanding of retinal protein interactions.


Assuntos
Doenças Autoimunes/complicações , Doenças Autoimunes/metabolismo , Modelos Animais de Doenças , Recoverina/metabolismo , Doenças Retinianas/complicações , Doenças Retinianas/metabolismo , Animais , Anticorpos/imunologia , Doenças Autoimunes/patologia , Eletrorretinografia , Feminino , Citometria de Fluxo , Humanos , Hibridomas/metabolismo , Imunização , Imuno-Histoquímica , Inflamação/complicações , Inflamação/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Recoverina/imunologia , Recoverina/isolamento & purificação , Retina/patologia , Doenças Retinianas/patologia
6.
Mol Cell Biochem ; 345(1-2): 197-206, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20740305

RESUMO

Cholesterol ester transfer protein (CETP) and apolipoprotein (apo) E are important in peroxisome proliferation activated receptor-α (PPAR-α)-mediated regulation of lipoprotein metabolism. Therefore, popularly used apolipoprotein E knockout mice are not suitable to evaluate PPAR-α agonists. In this study, we aimed to: a) evaluate hamster as a model for insulin resistance, hyperlipidemia and atherosclerosis; and b) investigate the effect of a PPAR-α activator, fenofibrate, in this model. A high fat high cholesterol (HFHC) diet increased serum cholesterol and triglycerides, but inclusion of fenofibrate in the diet decreased cholesterol and proatherogenic lipoproteins, VLDL and LDL, in a time-dependent manner. Concomitantly, serum levels of triglycerides also decreased. These reductions were attributed, in part, to the down-regulation of lipogenic genes and upregulation of lipoprotein lipase. The HFHC diet caused body weight gain and mild insulin resistance, both of which were prevented following the treatments with fenofibrate. Insulin resistance was further investigated in high fructose-fed hamsters. Fenofibrate prevented both hyperinsulinemia and hypertriglyceridemia. The insulin sensitizing activity of fenofibrate appeared to occur via reductions in protein tyrosine phophatase-1B. To determine whether lowering of lipids by fenofibrate treatment contributed to the reduced risks of developing atherosclerosis in hyperlipidemic hamsters, we measured lipid deposition in the aorta. Our results showed that fenofibrate treatment reduced aortic lipid deposition by 70%. These findings suggest that hamster may be an adequate animal model to evaluate the efficacy of lipid lowering, insulin sensitizing and antiatherosclerotic agents. We also show that fenofibrate is an effective antiatherosclerotic agent in hyperlipidemic hamster model.


Assuntos
Aorta/patologia , Aterosclerose/tratamento farmacológico , Fenofibrato/farmacologia , Hiperlipidemias/tratamento farmacológico , Resistência à Insulina , Metabolismo dos Lipídeos/efeitos dos fármacos , Animais , Aterosclerose/prevenção & controle , Colesterol/administração & dosagem , Colesterol/sangue , Cricetinae , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/sangue , Fenofibrato/uso terapêutico , Hipolipemiantes/farmacologia , Mesocricetus , Modelos Animais , PPAR alfa/agonistas , Aumento de Peso
7.
Arch Ophthalmol ; 127(12): 1572-80, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20008709

RESUMO

OBJECTIVES: To study 11 patients with melanoma-associated retinopathy (MAR) to clarify the reliability of various methods of diagnostic testing, to determine the underlying antigenic retinal proteins, and to study the clinical histories and types of associated melanomas. METHODS: Clinical data were obtained from patients with melanoma who developed marked visual problems. Testing included electroretinography, kinetic visual fields, comparative studies of Western blots, and indirect immunohistologic examination to detect antiretinal antibodies, as well as proteomic studies to identify underlying antigenic retinal proteins. RESULTS: Patients with MAR typically have rapid onset of photopsias, scotomata, and loss of central or paracentral vision. Ophthalmoscopy seldom shows significant changes early, but electroretinograms are abnormal. Results of Western blots and immunohistologic examination can show antiretinal antibodies but not always. Most patients (9 of 11) had a strong family history of autoimmune disorders. Any type of melanoma (cutaneous, choroidal, ciliary body, or choroidal nevi) may be associated with this paraneoplastic autoimmune reactivity. MAR may precede or follow the diagnosis of melanoma. Patients with MAR have the same antigenic retinal proteins that have been associated with cancer-associated retinopathy. In addition, 2 new antigenic retinal proteins, aldolase A and aldolase C, were found. CONCLUSIONS: There was a high prevalence of positive family histories of autoimmune disease in patients with MAR. To confirm the disorder, multiple clinical and serum diagnostic techniques (Western blot or indirect immunohistologic examination) are needed. Two newly observed antigenic retinal proteins, aldolase A and aldolase C, are associated with MAR.


Assuntos
Autoanticorpos/sangue , Autoantígenos/análise , Doenças Autoimunes/imunologia , Melanoma/imunologia , Síndromes Paraneoplásicas/imunologia , Doenças Retinianas/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Doenças Autoimunes/etiologia , Western Blotting , Eletroforese em Gel de Poliacrilamida , Eletrorretinografia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Frutose-Bifosfato Aldolase/análise , Humanos , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Síndromes Paraneoplásicas/etiologia , Doenças Retinianas/etiologia , Escotoma/etiologia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Neoplasias Uveais/imunologia , Neoplasias Uveais/patologia , Campos Visuais
8.
Arch Ophthalmol ; 127(4): 390-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19365013

RESUMO

OBJECTIVE: To report the results of treating autoimmune retinopathy (AIR) with immunosuppression therapy. METHODS: Retrospective review of 30 consecutive patients with AIR followed for 3 to 89 months (median, 17 months) who were treated with immunosuppression (systemic or local). Subgroups were cancer-associated retinopathy (CAR), nonparaneoplastic AIR (npAIR), and npAIR with cystoid macular edema (npAIR/CME). Outcome measures were improvement of Snellen visual acuity by at least 2 lines, expansion of the visual field area by more than 25%, and resolution of CME. RESULTS: Overall, 21 of 30 patients (70%) showed improvement. All 6 CAR patients, 7 of 13 (54%) with npAIR, and 8 of 11 (73%) with npAIR/CME showed improvement. Five of 21 patients (24%) had improvement in visual acuity, 15 of 21 (71%) had expansion of visual field area, and 6 of 11 (55%) had resolution of CME. Twenty-six of 30 patients exhibited diffuse retinal atrophy without pigment deposits. An autoimmune family history was common in all the groups: npAIR, 69% (9 of 13); npAIR/CME, 64% (7 of 11); and CAR, 50% (3 of 6). CONCLUSIONS: Long-term treatment with immunosuppression resulted in clinical improvement in all subgroups of AIR. The most responsive subgroup was CAR; the least was npAIR. These results challenge the commonly held belief that AIR is untreatable.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Azatioprina/uso terapêutico , Ciclosporina/uso terapêutico , Imunossupressores/uso terapêutico , Prednisona/uso terapêutico , Doenças Retinianas/tratamento farmacológico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Autoanticorpos/sangue , Autoantígenos/imunologia , Doenças Autoimunes/imunologia , Western Blotting , Criança , Quimioterapia Combinada , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Edema Macular/tratamento farmacológico , Edema Macular/imunologia , Masculino , Pessoa de Meia-Idade , Síndromes Paraneoplásicas/tratamento farmacológico , Síndromes Paraneoplásicas/imunologia , Doenças Retinianas/imunologia , Estudos Retrospectivos , Resultado do Tratamento
9.
Vision Res ; 48(3): 366-76, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17904189

RESUMO

Mutations in the cilia-centrosomal protein Retinitis Pigmentosa GTPase Regulator (RPGR) are a frequent cause of retinal degeneration. The RPGR gene undergoes complex alternative splicing and encodes multiple protein isoforms. To elucidate the function of major RPGR isoforms (RPGR 1-19 and RPGR ORF15), we have generated isoform-specific antibodies and examined their expression and localization in the retina. Using sucrose-gradient centrifugation, immunofluorescence and co-immunoprecipitation methods, we show that RPGR isoforms localize to distinct sub-cellular compartments in mammalian photoreceptors and associate with a number of cilia-centrosomal proteins. The RCC1-like domain of RPGR, which is present in all major RPGR isoforms, is sufficient to target it to the cilia and centrosomes in cultured cells. Our findings indicate that multiple isotypes of RPGR may perform overlapping yet somewhat distinct transport-related functions in photoreceptors.


Assuntos
Proteínas de Transporte/fisiologia , Oftalmopatias Hereditárias/genética , Proteínas do Olho/fisiologia , Doenças Genéticas Ligadas ao Cromossomo X/genética , Retinose Pigmentar/genética , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Bovinos , Ciclo Celular/fisiologia , Células Cultivadas , Centrossomo/metabolismo , Oftalmopatias Hereditárias/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/imunologia , Proteínas do Olho/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Humanos , Camundongos , Células Fotorreceptoras de Vertebrados/metabolismo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Retina/metabolismo , Retinose Pigmentar/metabolismo , Especificidade da Espécie
10.
Proc Natl Acad Sci U S A ; 104(41): 16227-32, 2007 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-17884985

RESUMO

Genetic variants at chromosomes 1q31-32 and 10q26 are strongly associated with susceptibility to age-related macular degeneration (AMD), a common blinding disease of the elderly. We demonstrate, by evaluating 45 tag SNPs spanning HTRA1, PLEKHA1, and predicted gene LOC387715/ARMS2, that rs10490924 SNP alone, or a variant in strong linkage disequilibrium, can explain the bulk of association between the 10q26 chromosomal region and AMD. A previously suggested causal SNP, rs11200638, and other examined SNPs in the region are only indirectly associated with the disease. Contrary to previous reports, we show that rs11200638 SNP has no significant impact on HTRA1 promoter activity in three different cell lines, and HTRA1 mRNA expression exhibits no significant change between control and AMD retinas. However, SNP rs10490924 shows the strongest association with AMD (P = 5.3 x 10(-30)), revealing an estimated relative risk of 2.66 for GT heterozygotes and 7.05 for TT homozygotes. The rs10490924 SNP results in nonsynonymous A69S alteration in the predicted protein LOC387715/ARMS2, which has a highly conserved ortholog in chimpanzee, but not in other vertebrate sequences. We demonstrate that LOC387715/ARMS2 mRNA is detected in the human retina and various cell lines and encodes a 12-kDa protein, which localizes to the mitochondrial outer membrane when expressed in mammalian cells. We propose that rs10490924 represents a major susceptibility variant for AMD at 10q26. A likely biological mechanism is that the A69S change in the LOC387715/ARMS2 protein affects its presumptive function in mitochondria.


Assuntos
Degeneração Macular/genética , Proteínas Mitocondriais/genética , Proteínas/genética , Serina Endopeptidases/genética , Idoso , Sequência de Aminoácidos , Animais , Sequência de Bases , Cromossomos Humanos Par 10/genética , Sequência Conservada , Expressão Gênica , Predisposição Genética para Doença , Variação Genética , Heterozigoto , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Homozigoto , Humanos , Desequilíbrio de Ligação , Dados de Sequência Molecular , Pan troglodytes/genética , Polimorfismo de Nucleotídeo Único , RNA Mensageiro/genética , Retina/metabolismo , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico
11.
Hum Mutat ; 28(11): 1074-83, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17554762

RESUMO

Mutations in the centrosomal-ciliary gene CEP290/NPHP6 are associated with Joubert syndrome and are the most common cause of the childhood recessive blindness known as Leber congenital amaurosis (LCA). An in-frame deletion in Cep290 shows rapid degeneration in the rod-rich mouse retina. To explore the mechanisms of the human retinal disease, we studied CEP290-LCA in patients of different ages (7-48 years) and compared results to Cep290-mutant mice. Unexpectedly, blind CEP290-mutant human retinas retained photoreceptor and inner laminar architecture in the cone-rich central retina, independent of severity of visual loss. Surrounding the cone-rich island was photoreceptor loss and distorted retina, suggesting neural-glial remodeling. The mutant mouse retina at 4-6 weeks of age showed similar features of retinal remodeling, with altered neural and synaptic laminae and Muller glial activation. The visual brain pathways in CEP290-LCA were anatomically intact. Our findings of preserved foveal cones and visual brain anatomy in LCA with CEP290 mutations, despite severe blindness and rapid rod cell death, suggest an opportunity for visual restoration of central vision in this common form of inherited blindness.


Assuntos
Antígenos de Neoplasias/genética , Cegueira/genética , Cegueira/terapia , Encéfalo/fisiopatologia , Mutação , Proteínas de Neoplasias/genética , Células Fotorreceptoras de Vertebrados/fisiologia , Vias Visuais , Adolescente , Adulto , Animais , Cegueira/fisiopatologia , Proteínas de Ciclo Celular , Criança , Proteínas do Citoesqueleto , Feminino , Humanos , Macaca fascicularis , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Mutantes , Pessoa de Meia-Idade
12.
Hum Mol Genet ; 15(11): 1847-57, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16632484

RESUMO

Centrosome- and cilia-associated proteins play crucial roles in establishing polarity and regulating intracellular transport in post-mitotic cells. Using genetic mapping and positional candidate strategy, we have identified an in-frame deletion in a novel centrosomal protein CEP290 (also called NPHP6), leading to early-onset retinal degeneration in a newly identified mouse mutant, rd16. We demonstrate that CEP290 localizes primarily to centrosomes of dividing cells and to the connecting cilium of retinal photoreceptors. We show that, in the retina, CEP290 associates with several microtubule-based transport proteins including RPGR, which is mutated in approximately 15% of patients with retinitis pigmentosa. A truncated CEP290 protein (DeltaCEP290) is detected in the rd16 retina, but in considerably reduced amounts; however, the mutant protein exhibits stronger association with specific RPGR isoform(s). Immunogold labeling studies demonstrate the redistribution of RPGR and of phototransduction proteins in the photoreceptors of rd16 retina. Our findings suggest a critical function for CEP290 in ciliary transport and provide insights into the mechanism of early-onset photoreceptor degeneration.


Assuntos
Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Proteínas de Transporte/metabolismo , Centrossomo/ultraestrutura , Proteínas do Olho/genética , Deleção de Genes , Proteínas Nucleares/genética , Degeneração Retiniana/genética , Animais , Sequência de Bases , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Centrossomo/metabolismo , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Proteínas do Olho/metabolismo , Humanos , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/fisiologia , Ligação Proteica
13.
J Biol Chem ; 280(39): 33580-7, 2005 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16043481

RESUMO

Mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene account for almost 20% of patients with retinitis pigmentosa. Most mutations are detected in alternatively spliced RPGR-ORF15 isoform(s), which are primarily but not exclusively expressed in the retina. We show that, in addition to the axoneme, the RPGR-ORF15 protein is localized to the basal bodies of photoreceptor connecting cilium and to the tip and axoneme of sperm flagella. Mass spectrometric analysis of proteins that were immunoprecipitated from the retinal axoneme-enriched fraction using an anti-ORF15 antibody identified two chromosome-associated proteins, structural maintenance of chromosomes (SMC) 1 and SMC3. Using pulldown assays, we demonstrate that the interaction of RPGR with SMC1 and SMC3 is mediated, at least in part, by the RCC1-like domain of RPGR. This interaction was not observed with phosphorylation-deficient mutants of SMC1. Both SMC1 and SMC3 localized to the cilia of retinal photoreceptors and Madin-Darby canine kidney cells, suggesting a broader physiological relevance of this interaction. Additional immunoprecipitation studies revealed the association of RPGR-ORF15 isoform(s) with the intraflagellar transport polypeptide IFT88 as well as microtubule motor proteins, including KIF3A, p150Glued, and p50-dynamitin. Inhibition of dynein function by overexpressing p50 abrogated the localization of RPGR-ORF15 to basal bodies. Taken together, these results provide novel evidence for the possible involvement of RPGR-ORF15 in microtubule organization and regulation of transport in primary cilia.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas do Olho/metabolismo , Retinose Pigmentar/prevenção & controle , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/ultraestrutura , Proteínas de Ciclo Celular/ultraestrutura , Proteoglicanas de Sulfatos de Condroitina/ultraestrutura , Proteínas Cromossômicas não Histona/ultraestrutura , Proteínas do Olho/genética , Proteínas do Olho/ultraestrutura , Humanos , Camundongos , Camundongos Knockout , Microtúbulos/ultraestrutura , Mutação , Fases de Leitura Aberta , Retinose Pigmentar/metabolismo , Retinose Pigmentar/patologia
14.
Nat Genet ; 37(3): 282-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15723066

RESUMO

Nephronophthisis (NPHP) is the most frequent genetic cause of chronic renal failure in children. Identification of four genes mutated in NPHP subtypes 1-4 (refs. 4-9) has linked the pathogenesis of NPHP to ciliary functions. Ten percent of affected individuals have retinitis pigmentosa, constituting the renal-retinal Senior-Loken syndrome (SLSN). Here we identify, by positional cloning, mutations in an evolutionarily conserved gene, IQCB1 (also called NPHP5), as the most frequent cause of SLSN. IQCB1 encodes an IQ-domain protein, nephrocystin-5. All individuals with IQCB1 mutations have retinitis pigmentosa. Hence, we examined the interaction of nephrocystin-5 with RPGR (retinitis pigmentosa GTPase regulator), which is expressed in photoreceptor cilia and associated with 10-20% of retinitis pigmentosa. We show that nephrocystin-5, RPGR and calmodulin can be coimmunoprecipitated from retinal extracts, and that these proteins localize to connecting cilia of photoreceptors and to primary cilia of renal epithelial cells. Our studies emphasize the central role of ciliary dysfunction in the pathogenesis of SLSN.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Calmodulina/metabolismo , Proteínas do Olho/metabolismo , Mutação , Sequência de Aminoácidos , Northern Blotting , Proteínas de Ligação a Calmodulina/química , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Síndrome , Técnicas do Sistema de Duplo-Híbrido
15.
J Biol Chem ; 279(40): 42211-20, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15292180

RESUMO

Many mammalian retinas are rod-dominant, and hence our knowledge of cone photoreceptor biology is relatively limited. To gain insights into the molecular differences between rods and cones, we compared the gene expression profile of the rod-dominated retina of wild type mouse with that of the cone-only retina of Nrl(-/-) (Neural retina leucine zipper knockout) mouse. Our analysis, using custom microarrays of eye-expressed genes, provided equivalent data using either direct or reference-based experimental designs, confirmed differential expression of rod- and cone-specific genes in the Nrl(-/-) retina and identified novel genes that could serve as candidates for retinopathies or for functional studies. In addition, we detected altered expression of several genes that encode cell signaling or structural proteins. Prompted by these findings, additional real-time PCR analysis revealed that genes belonging to the Bmp/Smad and Wnt/Ca(2+) signaling pathways are expressed in the mature wild type retina and that their expression is significantly altered in the Nrl(-/-) retina. Chromatin immunoprecipitation analysis of adult retina identified Bmp4 and Smad4, which are down-regulated in the Nrl(-/-) retina, as possible direct transcriptional targets of Nrl. Consistent with these studies, Bmp4 and Smad4 are expressed in the mature rod photoreceptors of mouse retina. Modulation of Bmp4 and/or Smad4 by Nrl may provide a mechanism for integrating diverse cell signaling networks in rods. We hypothesize that Bmp/Smad and Wnt/Ca(2+) pathways participate in cell-cell communication in the mature retina, and expression changes observed in the Nrl(-/-) retina reflect their biased utilization in rod versus cone homeostasis.


Assuntos
Sinalização do Cálcio , Regulação da Expressão Gênica , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica , Receptores de Proteínas Morfogenéticas Ósseas , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/metabolismo , Proteínas do Olho , Perfilação da Expressão Gênica/métodos , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Doenças Retinianas/genética , Proteínas Smad , Transativadores/metabolismo , Proteínas Wnt
16.
Hum Mol Genet ; 13(14): 1487-503, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15163632

RESUMO

The rod photoreceptor-specific neural retina leucine zipper protein Nrl is essential for rod differentiation and plays a critical role in regulating gene expression. In the mouse retina, rods account for 97% of the photoreceptors; however, in the absence of Nrl (Nrl-/-), no rods are present and a concomitant increase in cones is observed. A functional all-cone mouse retina represents a unique opportunity to investigate, at the molecular level, differences between the two photoreceptor subtypes. Using mouse GeneChips (Affymetrix), we have generated expression profiles of the wild-type and Nrl-/- retina at three time-points representing distinct stages of photoreceptor differentiation. Comparative data analysis revealed 161 differentially expressed genes; of which, 78 exhibited significantly lower and 83 higher expression in the Nrl-/- retina. Hierarchical clustering was utilized to predict the function of these genes in a temporal context. The differentially expressed genes primarily encode proteins associated with signal transduction, transcriptional regulation, intracellular transport and other processes, which likely correspond to differences between rods and cones and/or retinal remodeling in the absence of rods. A significant number of these genes may serve as candidates for diseases involving rod or cone dysfunction. Chromatin immunoprecipitation assay showed that in addition to the rod phototransduction genes, Nrl might modulate the promoters of many functionally diverse genes in vivo. Our studies provide molecular insights into differences between rod and cone function, yield interesting candidates for retinal diseases and assist in identifying transcriptional regulatory targets of Nrl.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/biossíntese , Proteínas do Olho/biossíntese , Perfilação da Expressão Gênica , Retina/metabolismo , Animais , Apoptose/genética , Fatores de Transcrição de Zíper de Leucina Básica/genética , Cálcio/metabolismo , Imunoprecipitação da Cromatina , Proteínas do Olho/genética , Homeostase , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Células Fotorreceptoras Retinianas Cones , Doenças Retinianas/genética , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Estresse Fisiológico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA