Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(40): e2210649119, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36161950

RESUMO

Molecular pathways that intrinsically regulate neuronal maintenance are poorly understood, but rare pathogenic mutations that underlie neurodegenerative disease can offer important insights into the mechanisms that facilitate lifelong neuronal function. Here, we leverage a rat model to demonstrate directly that the TFG p.R106C variant implicated previously in complicated forms of hereditary spastic paraplegia (HSP) underlies progressive spastic paraparesis with accompanying ventriculomegaly and thinning of the corpus callosum, consistent with disease phenotypes identified in adolescent patients. Analyses of primary cortical neurons obtained from CRISPR-Cas9-edited animals reveal a kinetic delay in biosynthetic secretory protein transport from the endoplasmic reticulum (ER), in agreement with prior induced pluripotent stem cell-based studies. Moreover, we identify an unexpected role for TFG in the trafficking of Rab4A-positive recycling endosomes specifically within axons and dendrites. Impaired TFG function compromises the transport of at least a subset of endosomal cargoes, which we show results in down-regulated inhibitory receptor signaling that may contribute to excitation-inhibition imbalances. In contrast, the morphology and trafficking of other organelles, including mitochondria and lysosomes, are unaffected by the TFG p.R106C mutation. Our findings demonstrate a multifaceted role for TFG in secretory and endosomal protein sorting that is unique to cells of the central nervous system and highlight the importance of these pathways to maintenance of corticospinal tract motor neurons.


Assuntos
Endossomos , Neurônios Motores , Transporte Proteico , Animais , Ratos , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Neurônios Motores/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteínas/metabolismo , Paraplegia Espástica Hereditária/metabolismo
2.
Bioessays ; 40(7): e1800004, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29741780

RESUMO

Most metazoan organisms have evolved a mildly acidified and calcium diminished sorting hub in the early secretory pathway commonly referred to as the Endoplasmic Reticulum-Golgi intermediate compartment (ERGIC). These membranous vesicular-tubular clusters are found tightly juxtaposed to ER subdomains that are competent for the production of COPII-coated transport carriers. In contrast to many unicellular systems, metazoan COPII carriers largely transit just a few hundred nanometers to the ERGIC, prior to COPI-dependent transport on to the cis-Golgi. The mechanisms underlying formation and maintenance of ERGIC membranes are poorly defined. However, recent evidence suggests an important role for Trk-fused gene (TFG) in regulating the integrity of the ER/ERGIC interface. Moreover, in the absence of cytoskeletal elements to scaffold tracks on which COPII carriers might move, TFG appears to promote anterograde cargo transport by locally tethering COPII carriers adjacent to ERGIC membranes. This action, regulated in part by the intrinsically disordered domain of TFG, provides sufficient time for COPII coat disassembly prior to heterotypic membrane fusion and cargo delivery to the ERGIC.


Assuntos
Retículo Endoplasmático/genética , Complexo de Golgi/genética , Organelas/genética , Via Secretória/genética , Animais , Fusão de Membrana/genética , Moduladores de Transporte de Membrana/metabolismo , Organelas/metabolismo , Transporte Proteico/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
3.
Exp Eye Res ; 158: 124-136, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27185161

RESUMO

Integrins are a family of heterodimeric transmembrane receptors that mediate adhesion to the extracellular matrix (ECM). In addition to their role as adhesion receptors, integrins can act as ''bidirectional signal transducers'' that coordinate a large number of cellular activities in response to the extracellular environment and intracellular signaling events. This bidirectional signaling helps maintain tissue homeostasis. Dysregulated bidirectional signaling, however, could trigger the propagation of feedback loops that can lead to the establishment of a disease state such as glaucoma. Here we discuss the role of integrins and bidirectional signaling as they relate to the glaucomatous phenotype with special emphasis on the αvß3 integrin. We present evidence that this particular integrin may have a significant impact on the pathogenesis of glaucoma.


Assuntos
Matriz Extracelular/metabolismo , Glaucoma/metabolismo , Integrinas/fisiologia , Transdução de Sinais/fisiologia , Malha Trabecular/metabolismo , Animais , Glaucoma/fisiopatologia , Humanos , Integrina alfaVbeta3/fisiologia , Limbo da Córnea/metabolismo , Disco Óptico/metabolismo
4.
Exp Cell Res ; 347(2): 301-11, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27539661

RESUMO

We previously demonstrated that an αvß5 integrin/FAK- mediated pathway regulated the phagocytic properties of human trabecular meshwork (HTM) cells. Here we demonstrate that this process is mediated by Rac-1 and a previously unreported signaling pathway that utilizes the Tiam1 as well as a novel ILK/RhoG/ELMO2 signaling pathway. Phagocytosis in both a TM-1 cell line and normal HTM cells was mediated by Rac1 and could be significantly decreased by >75% using the Rac1 inhibitor EHop-016. Knockdown of Rac1 in TM-1 cells also inhibited phagocytosis by 40% whereas overexpression of a constitutively active Rac1 or stimulation with PDGF increased phagocytosis by 83% and 32% respectively. Tiam1 was involved in regulating phagocytosis. Knockdown of Tiam1 inhibited phagocytosis by 72% while overexpression of Tiam1 C1199 increased phagocytosis by 75%. Other upstream effectors of Rac1 found to be involved included ELMO2, RhoG, and ILK. Knockdowns of ELMO2, ILK, and RhoG caused a reduction in phagocytosis by 51%, 55% and 46% respectively. In contrast, knockdown of Vav2 and Dock1 or overexpression of Vav2 Y159/172F did not cause a significant change in phagocytosis. These data suggest a novel link between Tiam1 and RhoG/ILK /ELMO2 pathway as upstream effectors of the Rac1-mediated phagocytic process in TM cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Fagocitose , Proteínas Serina-Treonina Quinases/metabolismo , Malha Trabecular/citologia , Malha Trabecular/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Linhagem Celular , Técnicas de Silenciamento de Genes , Humanos , Modelos Biológicos , Fagocitose/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-vav/metabolismo , RNA Interferente Pequeno/metabolismo , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T
5.
Sci Rep ; 11(1): 2013, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33479349

RESUMO

Nε-lysine acetylation in the ER is an essential component of the quality control machinery. ER acetylation is ensured by a membrane transporter, AT-1/SLC33A1, which translocates cytosolic acetyl-CoA into the ER lumen, and two acetyltransferases, ATase1 and ATase2, which acetylate nascent polypeptides within the ER lumen. Dysfunctional AT-1, as caused by gene mutation or duplication events, results in severe disease phenotypes. Here, we used two models of AT-1 dysregulation to investigate dynamics of the secretory pathway: AT-1 sTg, a model of systemic AT-1 overexpression, and AT-1S113R/+, a model of AT-1 haploinsufficiency. The animals displayed reorganization of the ER, ERGIC, and Golgi apparatus. In particular, AT-1 sTg animals displayed a marked delay in Golgi-to-plasma membrane protein trafficking, significant alterations in Golgi-based N-glycan modification, and a marked expansion of the lysosomal network. Collectively our results indicate that AT-1 is essential to maintain proper organization and engagement of the secretory pathway.


Assuntos
Acetilcoenzima A/genética , Acetiltransferases/genética , Retículo Endoplasmático/genética , Proteínas de Membrana Transportadoras/genética , Acetilcoenzima A/metabolismo , Acetilação , Autofagia/genética , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica/genética , Complexo de Golgi/genética , Complexo de Golgi/patologia , Haploinsuficiência/genética , Humanos , Lisossomos/genética , Mutação/genética , Processamento de Proteína Pós-Traducional/genética , Transporte Proteico/genética , Via Secretória/genética
6.
Invest Ophthalmol Vis Sci ; 60(12): 3897-3913, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31529121

RESUMO

Purpose: Fibronectin fibrillogenesis is an integrin-mediated process that may contribute to the pathogenesis of primary open-angle glaucoma (POAG). Here, we examined the effects of αvß3 integrins on fibrillogenesis in immortalized TM-1 cells and human trabecular meshwork (HTM) cells. Methods: TM-1 cells overexpressing wild-type ß3 (WTß3) or constitutively active ß3 (CAß3) integrin subunits were generated. Control cells were transduced with an empty vector (EV). Deoxycholic acid (DOC) extraction of monolayers, immunofluorescence microscopy, and On-cell western analyses were used to determine levels of fibronectin fibrillogenesis and fibronectin fibril composition (EDA+ and EDB+ fibronectins) and conformation. αvß3 and α5ß1 Integrin levels were determined using fluorescence-activated cell sorting (FACS). Cilengitide and an adenovirus vector expressing WTß3 or CAß3 integrin subunits were used to examine the role of αvß3 integrin in HTM cells. The role of the canonical α5ß1 integrin-mediated pathway in fibrillogenesis was determined using the fibronectin-binding peptide FUD, the ß1 integrin function-blocking antibody 13, and the Rho kinase (ROCK) inhibitor Y27632. Results: Activation of αvß3 integrin enhanced the assembly of fibronectin into DOC-insoluble fibrils in both TM-1 and HTM cells. The formation of fibronectin fibrils was dependent on α5ß1 integrin and could be inhibited by FUD. However, fibrillogenesis was unaffected by Y27632. Fibrils assembled by CAß3 cells also contained high levels of EDA+ and EDB+ fibronectin and fibronectin that was stretched. Conclusions: αvß3 Integrin signaling altered the deposition and structure of fibronectin fibrils using a ß1 integrin/ROCK-independent mechanism. Thus, αvß3 integrins could play a significant role in altering the function of fibronectin matrices in POAG.


Assuntos
Fibrilinas/biossíntese , Fibronectinas/metabolismo , Integrina alfaVbeta3/metabolismo , Malha Trabecular/metabolismo , Quinases Associadas a rho/metabolismo , Amidas/farmacologia , Western Blotting , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Vetores Genéticos , Humanos , Microscopia de Fluorescência , Piridinas/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Transfecção , Quinases Associadas a rho/antagonistas & inibidores
7.
Cell Rep ; 24(9): 2248-2260, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30157421

RESUMO

Length-dependent axonopathy of the corticospinal tract causes lower limb spasticity and is characteristic of several neurological disorders, including hereditary spastic paraplegia (HSP) and amyotrophic lateral sclerosis. Mutations in Trk-fused gene (TFG) have been implicated in both diseases, but the pathomechanisms by which these alterations cause neuropathy remain unclear. Here, we biochemically and genetically define the impact of a mutation within the TFG coiled-coil domain, which underlies early-onset forms of HSP. We find that the TFG (p.R106C) mutation alters compaction of TFG ring complexes, which play a critical role in the export of cargoes from the endoplasmic reticulum (ER). Using CRISPR-mediated genome editing, we engineered human stem cells that express the mutant form of TFG at endogenous levels and identified specific defects in secretion from the ER and axon fasciculation following neuronal differentiation. Together, our data highlight a key role for TFG-mediated protein transport in the pathogenesis of HSP.


Assuntos
Fasciculação Axônica/genética , Proteínas/genética , Proteínas/metabolismo , Paraplegia Espástica Hereditária/genética , Paraplegia Espástica Hereditária/metabolismo , Axônios/metabolismo , Axônios/patologia , Sequência de Bases , Humanos , Mutação , Neurônios/metabolismo , Neurônios/patologia , Transporte Proteico , Paraplegia Espástica Hereditária/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA