Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Hum Gene Ther ; 29(7): 771-784, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29325457

RESUMO

Gene therapy is a promising approach in the treatment of inherited and common complex disorders of the retina. Preclinical and clinical studies have validated the use of adeno-associated viral vectors (AAV) as a safe and efficient delivery vehicle for gene transfer. Retinal pigment epithelium and rods-and to a lesser extent, cone photoreceptors-can be efficiently targeted with AAV. Other retinal cell types however are more challenging targets. The aim of this study was to characterize the transduction profile and efficiency of in silico designed, synthetic Anc80 AAVs for retinal gene transfer. Three Anc80 variants were evaluated for retinal targeting in mice and primates following subretinal delivery. In the murine retina Anc80L65 demonstrated high level of retinal pigment epithelium and photoreceptor targeting with comparable cone photoreceptor affinity compared to other AAVs. Remarkably, Anc80L65 enhanced transduction kinetics with visible expression as early as day 1 and steady state mRNA levels at day 3. Inner retinal tropism of Anc80 variants demonstrated distinct transduction patterns of Müller glia, retinal ganglion cells and inner nuclear layer neurons. Finally, murine findings with Anc80L65 qualitatively translated to the Rhesus macaque in terms of cell targets, levels and onset of expression. Our findings support the use of Anc80L65 for therapeutic subretinal gene delivery.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/administração & dosagem , Retina/metabolismo , Doenças Retinianas/terapia , Animais , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Humanos , Macaca mulatta , Camundongos , Retina/efeitos dos fármacos , Retina/patologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Doenças Retinianas/genética , Doenças Retinianas/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Transdução Genética
2.
Nat Med ; 18(10): 1503-10, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22983396

RESUMO

The discovery of potent inhibitors of the BRAF proto-oncogene has revolutionized therapy for melanoma harboring mutations in BRAF, yet NRAS-mutant melanoma remains without an effective therapy. Because direct pharmacological inhibition of the RAS proto-oncogene has thus far been unsuccessful, we explored systems biology approaches to identify synergistic drug combination(s) that can mimic RAS inhibition. Here, leveraging an inducible mouse model of NRAS-mutant melanoma, we show that pharmacological inhibition of mitogen-activated protein kinase kinase (MEK) activates apoptosis but not cell-cycle arrest, which is in contrast to complete genetic neuroblastoma RAS homolog (NRAS) extinction, which triggers both of these effects. Network modeling pinpointed cyclin-dependent kinase 4 (CDK4) as a key driver of this differential phenotype. Accordingly, combined pharmacological inhibition of MEK and CDK4 in vivo led to substantial synergy in therapeutic efficacy. We suggest a gradient model of oncogenic NRAS signaling in which the output is gated, resulting in the decoupling of discrete downstream biological phenotypes as a result of incomplete inhibition. Such a gated signaling model offers a new framework to identify nonobvious coextinction target(s) for combined pharmacological inhibition in NRAS-mutant melanomas.


Assuntos
Apoptose/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Melanoma/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Benzimidazóis/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Quinase 4 Dependente de Ciclina/metabolismo , Doxiciclina/farmacologia , Feminino , Genes ras , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Knockout , Camundongos Nus , Pessoa de Meia-Idade , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais/efeitos dos fármacos
3.
Glycobiology ; 21(2): 152-61, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20855470

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) is a heparan sulfate (HS)-binding factor. GDNF is produced by somatic Sertoli cells, where it signals to maintain spermatogonial stem cells (SSCs) and reproduction. Here, we investigate the roles of extracellular HS 6-O-endosulfatases (Sulfs), Sulf1 and Sulf2, in the matrix transmission of GDNF from Sertoli cells to SSCs. Although Sulfs are not required for testis formation, Sulf deficiency leads to the accelerated depletion of SSCs, a testis phenotype similar to that of GDNF+/- mice. Mechanistically, we show that Sulfs are expressed in GDNF-producing Sertoli cells. In addition, reduced Sulf activity profoundly worsens haplo-deficient GDNF phenotypes in our genetic studies. These findings establish a critical role of Sulfs in promoting GDNF signaling and support a model in which Sulfs regulate the bioavailability of GDNF by enzymatically remodeling HS 6-O-desulfation to release GDNF from matrix sequestration. Further, Sertoli cell-specific transcriptional factor Wilm's tumor 1 (WT1) directly activates the transcription of both Sulf1 and Sulf2 genes. Together, our studies not only identify Sulfs as essential regulators of GDNF signaling in the SSC niche, but also as direct downstream targets of WT1, thus establishing a physiological role of WT1 in Sertoli cells.


Assuntos
Células de Sertoli/metabolismo , Espermatogônias , Sulfatases , Sulfotransferases , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Masculino , Camundongos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Fatores de Processamento de RNA , Ratos , Transdução de Sinais/fisiologia , Espermatogônias/metabolismo , Nicho de Células-Tronco/metabolismo , Células-Tronco/metabolismo , Sulfatases/genética , Sulfatases/metabolismo , Sulfotransferases/genética , Sulfotransferases/metabolismo
4.
Am J Respir Cell Mol Biol ; 44(3): 293-301, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20139349

RESUMO

Our study investigates the innervation of the respiratory tract during mouse embryonic development, with a focus on the identification of cell origin and essential developmental signals for the resident, or intrinsic, neurons. Using lineage tracing, we show that these intrinsic neurons are exclusively derived from neural crest cells, and cluster to form ganglia that reside in the dorsal trachea and medial bronchi with diminishing frequency. Comparisons of intrinsic neurogenesis between wild-type, glial cell-derived neurotrophic factor (GDNF)(-/-), neurturin(-/-), and tyrosine kinase receptor Ret(-/-) embryos, in combination with lung organ cultures, identified that Ret signaling, redundantly activated by GDNF family members, is required for intrinsic neurogenesis in the trachea and primary bronchi. In contrast, Ret deficiency exerts no effect on the innervation of the rest of the respiratory tract, suggesting that innervation by neurons whose cell bodies are located outside of the lung (so-called extrinsic neurons) is independent of Ret signaling. Furthermore, although the trachea, the esophagus, and their intrinsic neurons share foregut endoderm and a neural crest cell origin, respectively, the signals required for their intrinsic neurogenesis are divergent. Together, our results not only establish the neural crest lineage of intrinsic neurons in the respiratory tract, but also identify regional differences in the abundance and developmental signals of intrinsic neurons along the respiratory tract and in the esophagus.


Assuntos
Crista Neural/citologia , Neurogênese , Sistema Respiratório/embriologia , Alelos , Animais , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Imuno-Histoquímica/métodos , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurturina/biossíntese , Gravidez , Proteínas Proto-Oncogênicas c-ret/biossíntese , Sistema Respiratório/metabolismo
5.
Dev Biol ; 311(2): 464-77, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17920055

RESUMO

Heparan sulfate proteoglycans (HSPGs) are required during muscle regeneration for regulating extracellular signaling pathways. HSPGs interact with growth factors and receptors through heparan sulfate (HS) chains. However, the regulatory mechanisms that control HS sulfation to affect the growth factor-dependent proliferation and differentiation of satellite cells are yet unknown. Here we report the essential functions of extracellular HS 6-O-endosulfatases (Sulfs) during muscle regeneration. We show that quiescent and activated satellite cells differentially express mouse Sulf1 (MSulf1) and MSulf2. MSulfs are not required for the formation of skeletal muscles and satellite cells, but they have redundant, essential roles to promote muscle regeneration, as MSulf double mutant mice exhibit delayed myogenic differentiation and prolonged Pax7 expression after cardiotoxin-induced skeletal muscle injury, while single MSulf knockouts regenerate normally. HS structural analysis demonstrates that Sulfs are regulatory HS-modifying enzymes that control HS 6-O-desulfation of activated satellite cells. Mechanistically, we show that MSulfs repress FGF2 signaling in activated satellite cells, leading us to propose that MSulfs are growth factor signaling sensors to control the proliferation to differentiation switch of satellite cells to initiate differentiation during regeneration. Our results establish Sulfs as essential regulators of HS-dependent growth factor signaling in the adult muscle stem cell niche.


Assuntos
Diferenciação Celular/fisiologia , Músculo Esquelético , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/fisiologia , Transdução de Sinais/fisiologia , Sulfatases/metabolismo , Sulfotransferases/metabolismo , Animais , Ciclo Celular/fisiologia , Células Cultivadas , Dissacarídeos/química , Proteoglicanas de Heparan Sulfato/química , Proteoglicanas de Heparan Sulfato/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Células Satélites de Músculo Esquelético/citologia , Sulfatases/genética , Sulfotransferases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA