Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Curr Med Chem ; 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37921176

RESUMO

BACKGROUND: Retinal pigment epithelium (RPE) 65 is a key enzyme in the visual cycle involved in the regeneration of 11-cis-retinal. Mutations in the human RPE65 gene cause Leber's congenital amaurosis (LCA), a severe form of an inherited retinal disorder. Animal models carrying Rpe65 mutations develop early-onset retinal degeneration. In particular, the cones degenerate faster than the rods. To date, gene therapy has been used successfully to treat RPE65-associated retinal disorders. However, gene therapy does not completely prevent progressive retinal degeneration in patients, possibly due to the vulnerability of cones in these patients. In the present study, we tested whether leukemia inhibitory factor (LIF), a trophic factor, protects cones in rd12 mice harboring a nonsense mutation in Rpe65. METHODS: LIF was administrated to rd12 mice by intravitreal microinjection. Apoptosis of retinal cells was analyzed by TUNEL assay. The degeneration of cone cells was evaluated by immunostaining of retinal sections and retinal flat-mounts. Signaling proteins regulated by LIF in the retinal and cultured cells were determined by immunoblotting. RESULTS: Intravitreal administration of LIF activated the STAT3 signaling pathway, thereby inhibiting photoreceptor apoptosis and preserving cones in rd12 mice. Niclosamide (NCL), an inhibitor of STAT3 signaling, effectively blocked STAT3 signaling and autophagy in cultured 661W cells treated with LIF. Co-administration of LIF with NCL to rd12 mice abolished the protective effect of LIF, suggesting that STAT3 signaling and autophagy mediate the protection. CONCLUSION: LIF is a potent factor that protects cones in rd12 mice. This finding implies that LIF can be used in combination with gene therapy to achieve better therapeutic outcomes for patients with RPE65-associated LCA.

2.
Biotechnol Genet Eng Rev ; : 1-17, 2023 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-36946780

RESUMO

Triptolide (TP) is involved in the progression of liver cancer. However, the detailed molecular network regulated through TP is still unclear. Long non-coding RNA (LncRNA) SLC9A3 exerts roles in various pathological progresses. Nevertheless, whether SLC9A3 affects the sensitivity of liver cancer cells to TP have not been uncovered. The content of SLC9A3-AS1 and miR-449b-5p was estimated by utilizing quantitative real-time polymerase-chain reaction (qRT-PCR). Cell counting kit 8 (CCK-8) assay was introduced to assess cell viability. Additionally, cell viability as well as invasion was tested via transwell assay. The direct binding between miR-449b-5p and SLC9A3-AS1 or LDHA was confirmed through luciferase reporter gene assay. Moreover, glycolysis rate was tested by calculating the uptake of glucose in addition to the production of lactate in Huh7 cells. LncRNA SLC9A3-AS1 was up-regulated in liver cancer tissue samples and cells. Knockdown of SLC9A3-AS1 notably further inhibited viability, migration as well as invasion in Huh7 cells. MiR-449b-5p was the direct downstream miRNA of SLC9A3-AS1 and was down-regulated by SLC9A3-AS1 in Huh7 cells. In addition, miR-449b-5p was reduced in liver cancer tissues and cells. Overexpressed miR-449b-5p increased the sensitivity of Huh7 cells to TP remarkably. Moreover, miR-449b-5p negatively regulated LDHA expression in Huh7 cells. This work proved that SLC9A3-AS1 increased the sensitivity of liver cancer cells to TP by regulating glycolysis rate mediated via miR-449b-5p/LDHA axis. These findings implied that TP is likely to be a potent agent for treating patients diagnosed with liver cancer.

3.
Biotechnol Genet Eng Rev ; : 1-18, 2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-36951619

RESUMO

Hepatocellular carcinoma (HCC) is identified as a common cancer type across the world and needs novel and efficient treatment. Tripterine, a well-known compound, exerts suppressive role in HCC development. However, the related molecular mechanism of tripterine in HCC remains unclear. The expression of MBNL1-AS1in HCC tissues and cells was measured via qRT-PCR assay. MTT assay was employed to estimate cell viability. Besides, cell migration as well as invasion was determined through transwell assay. Additionally, the binding ability of miR-708-5p and MBNL1-AS1or HK2 was proved by starBase database and luciferase reporter gene assay. Moreover, the HK2 level was detected by immunoblotting. MBNL1-AS1 was reduced in HCC tissues and cells. Overexpression of MBNL1-AS1 decreased the sensitivity of HCC cells to tripterine while MBNL1-AS1 silence played opposite effect. In addition, miR-708-5p was the target of MBNL1-AS1 and was down-regulated through MBNL1-AS1 in HCC cells. Moreover, miR-708-5p suppressed glycolysis rate and reduced the expression of vital glycolytic enzyme (HK2, LDHA and PKM2) in HCC cells. Furthermore, miR-708-5p reduced HK2 expression by binding to it directly. In this investigation, we proved that LncRNA MBNL1-AS1 increased the tripterine resistance of HCC cells at least partly by mediating miR-708-5p-related glycolysis. These findings revealed a potent therapeutic target for the treatment of HCC.

4.
Zool Res ; 43(3): 442-456, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35503560

RESUMO

Mutations in serologically defined colon cancer autoantigen protein 8 ( SDCCAG8) were first identified in retinal ciliopathy families a decade ago with unknown function. To investigate the pathogenesis of SDCCAG8-associated retinal ciliopathies in vivo, we employed CRISPR/Cas9-mediated homology-directed recombination (HDR) to generate two knock-in mouse models, Sdccag8Y236X/Y236X and Sdccag8E451GfsX467/E451GfsX467 , which carry truncating mutations of the mouse Sdccag8, corresponding to mutations that cause Bardet-Biedl syndrome (BBS) and Senior-Løken syndrome (SLS) (c.696T>G p.Y232X and c.1339-1340insG p.E447GfsX463) in humans, respectively. The two mutant Sdccag8 knock-in mice faithfully recapitulated human SDCCAG8-associated BBS phenotypes such as rod-cone dystrophy, cystic renal disorder, polydactyly, infertility, and growth retardation, with varied age of onset and severity depending on the hypomorphic strength of the Sdccag8 mutations. To the best of our knowledge, these knock-in mouse lines are the first BBS mouse models to present with the polydactyly phenotype. Major phototransduction protein mislocalization was also observed outside the outer segment after initiation of photoreceptor degeneration. Impaired cilia were observed in the mutant photoreceptors, renal epithelial cells, and mouse embryonic fibroblasts derived from the knock-in mouse embryos, suggesting that SDCCAG8 plays an essential role in ciliogenesis, and cilium defects are a primary driving force of SDCCAG8-associated retinal ciliopathies.


Assuntos
Síndrome de Bardet-Biedl , Ciliopatias , Polidactilia , Doenças dos Roedores , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Síndrome de Bardet-Biedl/genética , Síndrome de Bardet-Biedl/metabolismo , Síndrome de Bardet-Biedl/veterinária , Ciliopatias/genética , Ciliopatias/metabolismo , Ciliopatias/veterinária , Fibroblastos , Camundongos , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Polidactilia/veterinária
5.
Mol Genet Genomic Med ; 9(7): e1686, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33949806

RESUMO

BACKGROUND: Papilon-Lefevre syndrome (PLS; OMIM 245000) is a rare autosomal recessive disease characterized by aggressive periodontitis and palmoplantar keratoderma. The prevalence of PLS in the general population is one to four cases per million. Although the etiology and pathogenic mechanisms underlying PLS remain largely unclear, existing evidence shows loss-of-function mutations of the cathepsin C gene (CTSC; OMIM 602365) could cause PLS. Here we found a novel variant of the CTSC gene in a Chinese PLS family and predicted the effect of the variant on the physic-chemical characters and tertiary structure of the protein. METHODS: The 1-7 coding exons and exon-intron boundaries of CTSC gene of the proband and her family were amplified and sequenced directly, and Chromas was used to read sequencing files. Furthermore, the PolyPhen-2, PROVEAN, and Mutation Taster were utilized to predict the pathogenicity of the variant. Besides, the physic-chemical and structural characters of the protein were analyzed by ProtParam, ProtScale, and SWISS-MODEL. RESULTS: Our study identified a novel homozygous variant c.763T>C (p.Cys255Arg) in exon 6 of the CTSC gene, and it was a likely pathogenic variant as predicted by PolyPhen-2, PROVEAN, and Mutation Taster. Moreover, ProtParam and Protscale revealed the variant increased the isoelectric point and hydrophilicity of the protein, and the SWISS-MODEL analysis suggested the variant was located in a critical domain for protein activity. CONCLUSION: Our study analyzed a Chinese family with PLS and identified a novel missense variant in the CTSC gene. Besides, this study retrospectively summarized 113 variants of CTSC in the world and highlighted the features of 27 CTSC variants in Chinese PLS patients. In addition, this study paid much particular attention to the relationship between CTSC variants and different phenotypes.


Assuntos
Catepsina C/genética , Displasia Ectodérmica/genética , Criança , Displasia Ectodérmica/diagnóstico , Feminino , Homozigoto , Humanos , Mutação de Sentido Incorreto , Fenótipo
6.
Genet Med ; 21(10): 2345-2354, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31000793

RESUMO

PURPOSE: Primary open-angle glaucoma (POAG) is the leading cause of irreversible blindness worldwide and mutations in known genes can only explain 5-6% of POAG. This study was conducted to identify novel POAG-causing genes and explore the pathogenesis of this disease. METHODS: Exome sequencing was performed in a Han Chinese cohort comprising 398 sporadic cases with POAG and 2010 controls, followed by replication studies by Sanger sequencing. A heterozygous Ramp2 knockout mouse model was generated for in vivo functional study. RESULTS: Using exome sequencing analysis and replication studies, we identified pathogenic variants in receptor activity-modifying protein 2 (RAMP2) within three genetically diverse populations (Han Chinese, German, and Indian). Six heterozygous RAMP2 pathogenic variants (Glu39Asp, Glu54Lys, Phe103Ser, Asn113Lysfs*10, Glu143Lys, and Ser171Arg) were identified among 16 of 4763 POAG patients, whereas no variants were detected in any exon of RAMP2 in 10,953 control individuals. Mutant RAMP2s aggregated in transfected cells and resulted in damage to the AM-RAMP2/CRLR-cAMP signaling pathway. Ablation of one Ramp2 allele led to cAMP reduction and retinal ganglion cell death in mice. CONCLUSION: This study demonstrated that disruption of RAMP2/CRLR-cAMP axis could cause POAG and identified a potential therapeutic intervention for POAG.


Assuntos
Glaucoma de Ângulo Aberto/genética , Proteína 2 Modificadora da Atividade de Receptores/genética , Animais , Povo Asiático , Células COS , Proteína Semelhante a Receptor de Calcitonina/genética , Proteína Semelhante a Receptor de Calcitonina/metabolismo , China , Chlorocebus aethiops , Estudos de Coortes , AMP Cíclico/genética , Predisposição Genética para Doença/genética , Glaucoma de Ângulo Aberto/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Mutação/genética , Linhagem , Polimorfismo de Nucleotídeo Único , Proteína 2 Modificadora da Atividade de Receptores/metabolismo , Sequenciamento do Exoma/métodos
7.
Mol Med Rep ; 18(3): 2866-2872, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30015904

RESUMO

Lung cancer is the most common type of cancer worldwide, the most prevalent form of which is non­small cell lung cancer (NSCLC). MicroRNAs (miRs) are involved in the progression of NSCLC; however, the specific function of miR­140­5p in NSCLC remains unclear. The present study demonstrated that miR­140­5p was downregulated in the tumor tissues of patients with NSCLC, and it was associated with a poor prognosis. Furthermore, miR­140­5p significantly suppressed cell migration and invasion of the NSCLC cell line A549. In addition, the direct regulatory effect of miR­140­5p on vascular endothelial growth factor­A (VEGFA) was predicted by TargetScan and verified using a luciferase reporter gene assay. The present study also hypothesized that miR­140­5p may inhibit the expression of phosphorylated­protein kinase B by targeting VEGFA. In conclusion, miR­140­5p may be a potential target for the development of anti­neoplastic therapies in lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , MicroRNAs/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Regiões 3' não Traduzidas , Células A549 , Antagomirs/metabolismo , Sequência de Bases , Sítios de Ligação , Movimento Celular , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Alinhamento de Sequência , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética
8.
Nat Genet ; 48(6): 640-7, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27089177

RESUMO

Polypoidal choroidal vasculopathy (PCV), a subtype of 'wet' age-related macular degeneration (AMD), constitutes up to 55% of cases of wet AMD in Asian patients. In contrast to the choroidal neovascularization (CNV) subtype, the genetic risk factors for PCV are relatively unknown. Exome sequencing analysis of a Han Chinese cohort followed by replication in four independent cohorts identified a rare c.986A>G (p.Lys329Arg) variant in the FGD6 gene as significantly associated with PCV (P = 2.19 × 10(-16), odds ratio (OR) = 2.12) but not with CNV (P = 0.26, OR = 1.13). The intracellular localization of FGD6-Arg329 is distinct from that of FGD6-Lys329. In vitro, FGD6 could regulate proangiogenic activity, and oxidized phospholipids increased expression of FGD6. FGD6-Arg329 promoted more abnormal vessel development in the mouse retina than FGD6-Lys329. Collectively, our data suggest that oxidized phospholipids and FGD6-Arg329 might act synergistically to increase susceptibility to PCV.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Mutação de Sentido Incorreto , Degeneração Macular Exsudativa/genética , Células Cultivadas , China , Estudos de Coortes , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Etnicidade , Perfilação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Polimorfismo de Nucleotídeo Único , Frações Subcelulares/metabolismo
9.
Sci Rep ; 6: 20914, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26861912

RESUMO

Neovascular age-related macular degeneration (AMD) and polypoidal choroidal vasculopathy (PCV) are leading causes of blindness in aging populations. This study was conducted to investigate the associations of chromosome 6p21.3 region, including CFB-SKIV2L-TNXB-FKBPL-NOTCH4 genes, with both neovascular AMD and PCV. Six single nucleotide polymorphisms (SNPs) in this region and two known AMD-associated SNPs in CFH (rs800292) and HTRA1 (rs11200638) were genotyped in a Han Chinese cohort composed of 490 neovascular AMD patients, 419 PCV patients and 1316 controls. Among the SNPs, TNXB rs12153855 and FKBPL rs9391734 conferred an increased susceptibility to neovascular AMD (P = 2.8 × 10(-4) and 0.001, OR = 1.80 and 1.76, respectively), while SKIV2L exerted a protective effect on neovascular AMD (P = 2.2 × 10(-4), OR = 0.49). Rs12153855C and rs9391734A alleles could further increase the susceptibility to AMD in subjects with rs800292, rs11200638 and rs429608 risk alleles. However, only the association of SKIV2L rs429608 remained significant after adjusting for rs800292, rs11200638 and the other 5 SNPs. The protective haplotype AATGAG exhibited significant association with neovascular AMD (permutation P = 0.015, OR = 0.34). None of the SNPs in this region was associated with PCV. Association profiles of 6p21.3 region showed discrepancy between neovascular AMD and PCV, indicating possible molecular and pathological differences between these two retinal disorders.


Assuntos
Neovascularização de Coroide/genética , Cromossomos Humanos Par 6 , Estudos de Associação Genética , Variação Genética , Degeneração Macular/genética , Idoso , Idoso de 80 Anos ou mais , Alelos , Estudos de Casos e Controles , Neovascularização de Coroide/patologia , DNA Helicases/genética , Feminino , Frequência do Gene , Loci Gênicos , Genótipo , Humanos , Imunofilinas/genética , Desequilíbrio de Ligação , Degeneração Macular/patologia , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Proteínas Proto-Oncogênicas/genética , Receptor Notch4 , Receptores Notch/genética , Proteínas de Ligação a Tacrolimo , Tenascina/genética
10.
J Biol Chem ; 291(13): 7142-55, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-26814127

RESUMO

Arf-like protein 3 (ARL3) is a ubiquitous small GTPase expressed in ciliated cells of plants and animals. Germline deletion ofArl3in mice causes multiorgan ciliopathy reminiscent of Bardet-Biedl or Joubert syndromes. As photoreceptors are elegantly compartmentalized and have cilia, we probed the function of ARL3 (ADP-ribosylation factor (Arf)-like 3 protein) by generating rod photoreceptor-specific (prefix(rod)) and retina-specific (prefix(ret))Arl3deletions. In predegenerate(rod)Arl3(-/-)mice, lipidated phototransduction proteins showed trafficking deficiencies, consistent with the role of ARL3 as a cargo displacement factor for lipid-binding proteins. By contrast,(ret)Arl3(-/-)rods and cones expressing Cre recombinase during embryonic development formed neither connecting cilia nor outer segments and degenerated rapidly. Absence of cilia infers participation of ARL3 in ciliogenesis and axoneme formation. Ciliogenesis was rescued, and degeneration was reversed in part by subretinal injection of adeno-associated virus particles expressing ARL3-EGFP. The conditional knock-out phenotypes permitted identification of two ARL3 functions, both in the GTP-bound form as follows: one as a regulator of intraflagellar transport participating in photoreceptor ciliogenesis and the other as a cargo displacement factor transporting lipidated protein to the outer segment. Surprisingly, a farnesylated inositol polyphosphate phosphatase only trafficked from the endoplasmic reticulum to the Golgi, thereby excluding it from a role in photoreceptor cilia physiology.


Assuntos
Fatores de Ribosilação do ADP/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Fatores de Ribosilação do ADP/deficiência , Fatores Etários , Animais , Cílios/metabolismo , Cílios/patologia , Dependovirus/genética , Eletrorretinografia , Embrião de Mamíferos , Proteínas do Olho/genética , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Integrases/genética , Integrases/metabolismo , Transdução de Sinal Luminoso , Camundongos , Camundongos Knockout , Organogênese/genética , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Bastonetes/patologia
11.
Biomaterials ; 58: 63-71, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25941783

RESUMO

Dendritic cell (DC) based vaccines have shown promising results in the immunotherapy of cancers and other diseases. How to track the in vivo fate of DC vaccines will provide important insights to the final therapeutic results. In this study, we chose magnetic resonance imaging (MRI) to track murine DCs migration to the draining lymph node under a clinical 3 T scanner. Different from labeling immature DCs usually reported in literature, this study instead labeled matured DC with superparamagnetic iron oxide (SPIO) nanoparticle based imaging probes. The labeling process did not show negative impacts on cell viability, morphology, and surface biomarker expression. To overcome the imaging challenges brought by the limitations of the scanner, the size of lymph node, and the number of labeled cell, we optimized MRI pulse sequences. As a result, the signal reduction, caused either by gelatin phantoms containing as low as 12 SPIO-laden cells in each voxel or by the homing SPIO-laden DCs within the draining nodes after footpad injection of only 1 × 10(5) cells, can be clearly depicted under a 3 T MR scanner. Overall, the MRI labeling probes offer a low-toxic and high-efficient MR imaging platform for the assessment of DC-based immunotherapies.


Assuntos
Rastreamento de Células/métodos , Células Dendríticas/citologia , Imageamento por Ressonância Magnética , Animais , Biomarcadores , Vacinas Anticâncer/química , Cátions , Movimento Celular , Sobrevivência Celular , Meios de Contraste , Células Dendríticas/efeitos dos fármacos , Dextranos/química , Gelatina/química , Imunoterapia/métodos , Ferro/química , Linfonodos/patologia , Nanopartículas de Magnetita/química , Camundongos , Camundongos Endogâmicos BALB C , Nanocompostos , Nanopartículas/química , Imagens de Fantasmas , Fenótipo
12.
FASEB J ; 29(3): 932-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25422369

RESUMO

The retinitis pigmentosa 2 polypeptide (RP2) functions as a GTPase-activating protein (GAP) for ARL3 (Arf-like protein 3), a small GTPase. ARL3 is an effector of phosphodiesterase 6 Δ (PDE6D), a prenyl-binding protein and chaperone of prenylated protein in photoreceptors. Mutations in the human RP2 gene cause X-linked retinitis pigmentosa (XLRP) and cone-rod dystrophy (XL-CORD). To study mechanisms causing XLRP, we generated an RP2 knockout mouse. The Rp2h(-/-) mice exhibited a slowly progressing rod-cone dystrophy simulating the human disease. Rp2h(-/-) scotopic a-wave and photopic b-wave amplitudes declined at 1 mo of age and continued to decline over the next 6 mo. Prenylated PDE6 subunits and G-protein coupled receptor kinase 1 (GRK1) were unable to traffic effectively to the Rp2h(-/-) outer segments. Mechanistically, absence of RP2 GAP activity increases ARL3-GTP levels, forcing PDE6D to assume a predominantly "closed" conformation that impedes binding of lipids. Lack of interaction disrupts trafficking of PDE6 and GRK1 to their destination, the photoreceptor outer segments. We propose that hyperactivity of ARL3-GTP in RP2 knockout mice and human patients with RP2 null alleles leads to XLRP resembling recessive rod-cone dystrophy.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Proteínas do Olho/fisiologia , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas de Membrana/fisiologia , Prenilação de Proteína , Retinose Pigmentar/metabolismo , Animais , Formação de Anticorpos , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/metabolismo , Cílios/metabolismo , Eletrorretinografia , Feminino , Proteínas de Ligação ao GTP , Guanosina Trifosfato/metabolismo , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Fotorreceptoras de Vertebrados/metabolismo , Transporte Proteico , Coelhos , Retinose Pigmentar/patologia
13.
Vision Res ; 75: 19-25, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22960045

RESUMO

Expressed ubiquitously, PrBP/δ functions as chaperone/co-factor in the transport of a subset of prenylated proteins. PrBP/δ features an immunoglobulin-like ß-sandwich fold for lipid binding, and interacts with diverse partners. PrBP/δ binds both C-terminal C15 and C20 prenyl side chains of phototransduction polypeptides and small GTP-binding (G) proteins of the Ras superfamily. PrBP/δ also interacts with the small GTPases, ARL2 and ARL3, which act as release factors (GDFs) for prenylated cargo. Targeted deletion of the mouse Pde6d gene encoding PrBP/δ resulted in impeded trafficking to the outer segments of GRK1 and cone PDE6 which are predicted to be farnesylated and geranylgeranylated, respectively. Rod and cone transducin trafficking was largely unaffected. These trafficking defects produce progressive cone-rod dystrophy in the Pde6d(-/-) mouse.


Assuntos
Proteínas de Transporte/fisiologia , Chaperonas Moleculares/fisiologia , Diester Fosfórico Hidrolases/fisiologia , Células Fotorreceptoras de Vertebrados/fisiologia , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Humanos , Camundongos , Transporte Proteico/fisiologia
14.
Vision Res ; 75: 26-32, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23000199

RESUMO

The mechanism by which myristoylated proteins are targeted to specific subcellular membrane compartments is poorly understood. Two novel acyl-binding proteins, UNC119A and UNC119B, have been shown recently to function as chaperones/co-factors in the transport of myristoylated G protein α-subunits and src-type tyrosine kinases. UNC119 polypeptides feature an immunoglobulin-like ß-sandwich fold that forms a hydrophobic pocket capable of binding lauroyl (C12) and myristoyl (C14) side chains. UNC119A in rod photoreceptors facilitates the transfer of transducin α subunits (Tα) from inner segment to outer segment membranes by forming an intermediate diffusible UNC119-Tα complex. Similar complexes are formed in other sensory neurons, as the G proteins ODR-3 and GPA-13 in Caenorhabditis elegans unc-119 mutants traffic inappropriately. UNC119B knockdown in IMCD3 cells prevents trafficking ofmyristoylated nephrocystin-3 (NPHP3), a protein associated with nephronophthisis, to cilia. Further, UNC119A was shown to transport myristoylated src-type tyrosine kinases to cell membranes and to affect T-cell receptor (TCR) and interleukin-5 receptor (IL-5R) activities. These interactions establish UNC119 polypeptides as novel lipid-binding chaperones with specificity for a diverse subset of myristoylated proteins.


Assuntos
Proteína de Transporte de Acila/fisiologia , Proteína de Transporte de Acila/química , Animais , Caenorhabditis elegans/fisiologia , Cílios/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Chaperonas Moleculares/fisiologia , Estrutura Terciária de Proteína , Transporte Proteico/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Quinases da Família src/metabolismo
15.
J Neurosci ; 28(15): 4008-14, 2008 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-18400900

RESUMO

Lecithin retinol acyl transferase (LRAT) and retinal pigment epithelium protein 65 (RPE65) are key enzymes of the retinoid cycle. In Lrat(-/-) and Rpe65(-/-) mice, models of human Leber congenital amaurosis, the retinoid cycle is disrupted and 11-cis-retinal, the chromophore of visual pigments, is not produced. The Lrat(-/-) and Rpe65(-/-) retina phenotype presents with rapid sectorial cone degeneration, and the visual pigments, S-opsin and M/L-opsin, fail to traffic to cone outer segments appropriately. In contrast, rod opsin traffics normally in mutant rods. Concomitantly, guanylate cyclase 1, cone T alpha-subunit, cone phosphodiesterase 6alpha' (PDE6alpha'), and GRK1 (G-protein-coupled receptor kinase 1; opsin kinase) are not transported to Lrat(-/-) and Rpe65(-/-) cone outer segments. Aberrant localization of these membrane-associated proteins was evident at postnatal day 15, before the onset of ventral and central cone degeneration. Protein levels of cone T alpha and cone PDE6alpha' were reduced, whereas their transcript levels were unchanged, suggesting posttranslational degradation. In an Rpe65(-/-)Rho(-/-) double knock-out model, trafficking of cone pigments and membrane-associated cone phototransduction polypeptides to the outer segments proceeded normally after 11-cis-retinal administration. These results suggest that ventral and central cone opsins must be regenerated with 11-cis-retinal to permit transport to the outer segments. Furthermore, the presence of 11-cis-retinal is essential for proper transport of several membrane-associated cone phototransduction polypeptides in these cones.


Assuntos
Proteínas de Membrana/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Retinaldeído/metabolismo , Aciltransferases/deficiência , Aciltransferases/metabolismo , Animais , Cegueira/congênito , Cegueira/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Modelos Animais de Doenças , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Guanilato Ciclase/metabolismo , Camundongos , Camundongos Knockout , Degeneração Neural/etiologia , Degeneração Neural/fisiopatologia , Isoformas de Proteínas/metabolismo , Transporte Proteico/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Células Fotorreceptoras Retinianas Cones/fisiopatologia , Pigmentos da Retina/metabolismo , Retinaldeído/deficiência , Retinaldeído/farmacologia , Opsinas de Bastonetes/metabolismo , Fatores de Tempo , Visão Ocular , cis-trans-Isomerases
16.
Vision Res ; 48(3): 442-52, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17949773

RESUMO

We discuss putative mechanisms of membrane protein transport in photoreceptors based on Pde6d and Gucy2e/Gucy2f knockout mice. Knockout of the Pde6d gene encoding PrBP/delta, a prenyl binding protein present in the retina at relatively high levels, was shown to impair transport of G-protein coupled receptor kinase 1 (GRK1) and cone phosphodiesterase alpha' subunit (PDE6alpha') to the rod and cone outer segments. Other prenylated proteins are minimally affected, suggesting some specificity of interaction. Knockout of the Gucy2e gene encoding guanylate cyclase 1 (GC1) disrupted transport of G-protein coupled receptor kinase 1 (GRK1), cone PDE6alpha', cone transducin alpha and gamma subunits (cTalpha and cTgamma) to the cone outer segments, while a GC1/GC2 double knockout prevented transport of rod PDE6, but not transducin, GRK1, or rhodopsin, to the rod outer segments. These knockout phenotypes suggest that PrBP/delta functions in extracting prenylated proteins from the endoplasmic reticulum (ER) where they dock after prenylation, and that GC-bearing membranes may co-transport peripheral membrane proteins in vesicles. We conclude that distinct pathways have evolved in rods and cones for transport of integral and peripherally membrane-associated proteins.


Assuntos
Modelos Animais , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Visão Ocular/fisiologia , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/fisiologia , Regulação para Baixo , Retículo Endoplasmático/metabolismo , Deleção de Genes , Guanilato Ciclase/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Neopreno/metabolismo , Transporte Proteico/fisiologia , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo
18.
J Biol Chem ; 280(19): 18822-32, 2005 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-15755727

RESUMO

The retinoid cycle is a recycling system that replenishes the 11-cis-retinal chromophore of rhodopsin and cone pigments. Photoreceptor-specific retinol dehydrogenase (prRDH) catalyzes reduction of all-trans-retinal to all-trans-retinol and is thought to be a key enzyme in the retinoid cycle. We disrupted mouse prRDH (human gene symbol RDH8) gene expression by targeted recombination and generated a homozygous prRDH knock-out (prRDH-/-) mouse. Histological analysis and electron microscopy of retinas from 6- to 8-week-old prRDH-/- mice revealed no structural differences of the photoreceptors or inner retina. For brief light exposure, absence of prRDH did not affect the rate of 11-cis-retinal regeneration or the decay of Meta II, the activated form of rhodopsin. Absence of prRDH, however, caused significant accumulation of all-trans-retinal following exposure to bright lights and delayed recovery of rod function as measured by electroretinograms and single cell recordings. Retention of all-trans-retinal resulted in slight overproduction of A2E, a condensation product of all-trans-retinal and phosphatidylethanolamine. We conclude that prRDH is an enzyme that catalyzes reduction of all-trans-retinal in the rod outer segment, most noticeably at higher light intensities and prolonged illumination, but is not an essential enzyme of the retinoid cycle.


Assuntos
Oxirredutases do Álcool/fisiologia , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Retinoides/metabolismo , Oxirredutases do Álcool/metabolismo , Animais , Southern Blotting , Catálise , Linhagem Celular , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , DNA Complementar/metabolismo , Eletroforese em Gel de Poliacrilamida , Eletrorretinografia , Olho/metabolismo , Vetores Genéticos , Genótipo , Humanos , Immunoblotting , Imuno-Histoquímica , Insetos , Cinética , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Modelos Químicos , Modelos Genéticos , Mutação , Fosfatidiletanolaminas/metabolismo , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Recombinação Genética , Retinaldeído/química , Retinoides/química , Rodopsina/química , Rodopsina/metabolismo , Fatores de Tempo , Transgenes , Vitamina A/metabolismo
19.
Methods Enzymol ; 403: 42-56, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16473576

RESUMO

A 17-kDa prenyl-binding protein, PrBP(PDEdelta), is highly conserved among various species from human to Caenorhabditis elegans. First identified as a putative regulatory delta subunit of the cyclic nucleotide phosphodiesterase (PDE6) purified from mammalian photoreceptor cells, PrBP(PDEdelta) has been hypothesized to reduce activation of PDE6 by the heterotrimeric G-protein, transducin, thereby desensitizing the photoresponse. However, recent work shows that PrBP(PDEdelta) interacts with numerous prenylated proteins at their farnesylated or geranylgeranylated C-termini, as well as with non-prenylated proteins. These polypeptides include small GTPases such as Rab13, Ras, Rap, and Rho6, as well as components involved in phototransduction (e.g., rod and cone PDE6, rod and cone opsin kinases). Expression of PrBP(PDEdelta) in tissues and organisms not expressing PDE6, the demonstration of multiple interacting partners with PrBP(PDEdelta), and its low abundance in rod outer segments all argue against it being a regulatory PDE6 subunit. This raises intriguing questions as to its physiological functions. In this chapter, we review the current status of PrBP(PDEdelta) and describe some of the assays used to determine these interactions in detail. In mammalian photoreceptors, the results are consistent with a role of PrBP(PDEdelta) in the transport of prenylated proteins from their site of synthesis in the inner segment to the outer segment where phototransduction occurs.


Assuntos
Diester Fosfórico Hidrolases/metabolismo , Anfíbios , Animais , Western Blotting , Bovinos , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6 , DNA Complementar , Humanos , Camundongos , Ligação Proteica , Espécies Reativas de Oxigênio , Retina/enzimologia , Segmento Externo da Célula Bastonete/enzimologia , Técnicas do Sistema de Duplo-Híbrido
20.
J Biol Chem ; 279(1): 407-13, 2004 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-14561760

RESUMO

Bovine PDEdelta was originally copurified with rod cGMP phosphodiesterase (PDE) and shown to interact with prenylated, carboxymethylated C-terminal Cys residues. Other studies showed that PDEdelta can interact with several small GTPases including Rab13, Ras, Rap, and Rho6, all of which are prenylated, as well as the N-terminal portion of retinitis pigmentosa GTPase regulator and Arl2/Arl3, which are not prenylated. We show by immunocytochemistry with a PDEdelta-specific antibody that PDEdelta is present in rods and cones. We find by yeast two-hybrid screening with a PDEdelta bait that it can interact with farnesylated rhodopsin kinase (GRK1) and that prenylation is essential for this interaction. In vitro binding assays indicate that both recombinant farnesylated GRK1 and geranylgeranylated GRK7 co-precipitate with a glutathione S-transferase-PDEdelta fusion protein. Using fluorescence resonance energy transfer techniques exploiting the intrinsic tryptophan fluorescence of PDEdelta and dansylated prenyl cysteines as fluorescent ligands, we show that PDEdelta specifically binds geranylgeranyl and farnesyl moieties with a Kd of 19.06 and 0.70 microm, respectively. Our experiments establish that PDEdelta functions as a prenyl-binding protein interacting with multiple prenylated proteins.


Assuntos
3',5'-GMP Cíclico Fosfodiesterases/metabolismo , Cisteína , Células Fotorreceptoras de Vertebrados/enzimologia , Células Fotorreceptoras Retinianas Bastonetes/enzimologia , 3',5'-GMP Cíclico Fosfodiesterases/análise , 3',5'-GMP Cíclico Fosfodiesterases/química , Sequência de Aminoácidos , Animais , Bovinos , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6 , Transferência Ressonante de Energia de Fluorescência , GTP Fosfo-Hidrolases/metabolismo , Imuno-Histoquímica , Cinética , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Prenilação de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA