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










Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 10: 1015125, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36393857

RESUMEN

The six subunits (Elp1 to Elp6) Elongator complex promotes specific uridine modifications in tRNA's wobble site. Moreover, this complex has been indirectly involved in the regulation of α-tubulin acetylation in microtubules (MTs) via the stabilization of ATP-Citrate Lyase (Acly), the main cytosolic source of acetyl-CoA production in cells, a key substrate used for global protein acetylation. Here, we report additional evidence that Elongator activity is important for proper cytoskeleton remodeling as cells lacking expression of Elp1 show morphology impairment; including distinct neurite process formation and disorganization and instability of MTs. Here, we show that loss of Elongator results in a reduction of expression of the microtubule associated protein Tau (MAPT). Tau, is a well-known key MT regulator in neurons whose lysines can be competitively acetylated or ubiquitylated. Therefore, we tested whether Tau is an indirect acetylation target of Elongator. We found that a reduction of Elongator activity leads to a decrease of lysine acetylation on Tau that favors its proteasomal degradation. This phenotype was prevented by using selective deacetylase or proteasomal inhibitors. Moreover, our data demonstrate that Acly's activity regulates the mechanism underlying Tau mediated neurite morphology defects found in Elp1 KD since both Tau levels and neurites morphology are restored due to Acly overexpression. This suggests a possible involvement of both Tau and Acly dysfunction in Familial Dysautonomia (FD), which is an autosomal recessive peripheral neuropathy caused by mutation in the ELP1 gene that severely affects Elp1 expression levels in the nervous system in FD patients in a similar way as found previously in Elp1 KD neuroblastoma cells.

3.
Nat Commun ; 12(1): 5878, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34620845

RESUMEN

Microtubule (MT)-based transport is an evolutionary conserved process finely tuned by posttranslational modifications. Among them, α-tubulin acetylation, primarily catalyzed by a vesicular pool of α-tubulin N-acetyltransferase 1 (Atat1), promotes the recruitment and processivity of molecular motors along MT tracks. However, the mechanism that controls Atat1 activity remains poorly understood. Here, we show that ATP-citrate lyase (Acly) is enriched in vesicles and provide Acetyl-Coenzyme-A (Acetyl-CoA) to Atat1. In addition, we showed that Acly expression is reduced upon loss of Elongator activity, further connecting Elongator to Atat1 in a pathway regulating α-tubulin acetylation and MT-dependent transport in projection neurons, across species. Remarkably, comparable defects occur in fibroblasts from Familial Dysautonomia (FD) patients bearing an autosomal recessive mutation in the gene coding for the Elongator subunit ELP1. Our data may thus shine light on the pathophysiological mechanisms underlying FD.


Asunto(s)
ATP Citrato (pro-S)-Liasa/metabolismo , Transporte Axonal/fisiología , ATP Citrato (pro-S)-Liasa/genética , Acetilcoenzima A/metabolismo , Acetilación , Acetiltransferasas/genética , Animales , Transporte Axonal/genética , Drosophila melanogaster , Disautonomía Familiar/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Larva , Masculino , Ratones , Microtúbulos/metabolismo , Procesamiento Proteico-Postraduccional , Tubulina (Proteína)/metabolismo
4.
Sci Adv ; 5(12): eaax2705, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31897425

RESUMEN

Microtubules are polymerized dimers of α- and ß-tubulin that underlie a broad range of cellular activities. Acetylation of α-tubulin by the acetyltransferase ATAT1 modulates microtubule dynamics and functions in neurons. However, it remains unclear how this enzyme acetylates microtubules over long distances in axons. Here, we show that loss of ATAT1 impairs axonal transport in neurons in vivo, and cell-free motility assays confirm a requirement of α-tubulin acetylation for proper bidirectional vesicular transport. Moreover, we demonstrate that the main cellular pool of ATAT1 is transported at the cytosolic side of neuronal vesicles that are moving along axons. Together, our data suggest that axonal transport of ATAT1-enriched vesicles is the predominant driver of α-tubulin acetylation in axons.


Asunto(s)
Acetiltransferasas/metabolismo , Transporte Axonal/fisiología , Proteínas de Microtúbulos/metabolismo , Microtúbulos/metabolismo , Acetilación , Acetiltransferasas/genética , Animales , Drosophila melanogaster/metabolismo , Femenino , Células HEK293 , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Larva/fisiología , Locomoción , Masculino , Ratones , Ratones Noqueados , Proteínas de Microtúbulos/genética , Neuronas/metabolismo , Tubulina (Proteína)/metabolismo
5.
Cell Rep ; 23(8): 2429-2442, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29791853

RESUMEN

The protein p27Kip1 plays roles that extend beyond cell-cycle regulation during cerebral cortex development, such as the regulation of neuronal migration and neurite branching via signaling pathways that converge on the actin and microtubule cytoskeletons. Microtubule-dependent transport is essential for the maturation of neurons and the establishment of neuronal connectivity though synapse formation and maintenance. Here, we show that p27Kip1 controls the transport of vesicles and organelles along the axon of mice cortical projection neurons in vitro. Moreover, suppression of the p27Kip1 ortholog, dacapo, in Drosophila melanogaster disrupts axonal transport in vivo, leading to the reduction of locomotor activity in third instar larvae and adult flies. At the molecular level, p27Kip1 stabilizes the α-tubulin acetyltransferase 1, thereby promoting the acetylation of microtubules, a post-translational modification required for proper axonal transport.


Asunto(s)
Acetiltransferasas/metabolismo , Transporte Axonal , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Acetilación , Animales , Drosophila melanogaster/metabolismo , Estabilidad de Enzimas , Femenino , Células HEK293 , Histona Desacetilasa 6/metabolismo , Humanos , Masculino , Ratones , Microtúbulos/metabolismo , Modelos Biológicos , Actividad Motora , Neuronas/metabolismo , Unión Proteica
6.
PLoS One ; 10(10): e0138807, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26437462

RESUMEN

A splicing mutation in the IKBKAP gene causes Familial Dysautonomia (FD), affecting the IKAP protein expression levels and proper development and function of the peripheral nervous system (PNS). Here we found new molecular insights for the IKAP role and the impact of the FD mutation in the human PNS lineage by using a novel and unique human embryonic stem cell (hESC) line homozygous to the FD mutation originated by pre implantation genetic diagnosis (PGD) analysis. We found that IKBKAP downregulation during PNS differentiation affects normal migration in FD-hESC derived neural crest cells (NCC) while at later stages the PNS neurons show reduced intracellular colocalization between vesicular proteins and IKAP. Comparative wide transcriptome analysis of FD and WT hESC-derived neurons together with the analysis of human brains from FD and WT 12 weeks old embryos and experimental validation of the results confirmed that synaptic vesicular and neuronal transport genes are directly or indirectly affected by IKBKAP downregulation in FD neurons. Moreover we show that kinetin (a drug that corrects IKBKAP alternative splicing) promotes the recovery of IKAP expression and these IKAP functional associated genes identified in the study. Altogether, these results support the view that IKAP might be a vesicular like protein that might be involved in neuronal transport in hESC derived PNS neurons. This function seems to be mostly affected in FD-hESC derived PNS neurons probably reflecting some PNS neuronal dysfunction observed in FD.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación hacia Abajo , Disautonomía Familiar/genética , Células Madre Embrionarias Humanas/patología , Neuronas/metabolismo , Sistema Nervioso Periférico/patología , Vesículas Sinápticas/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/genética , Proteínas Portadoras/genética , Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Disautonomía Familiar/metabolismo , Disautonomía Familiar/patología , Feto , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Cinetina/farmacología , Masculino , Mutación , Cresta Neural/efectos de los fármacos , Cresta Neural/patología , Neuronas/efectos de los fármacos , Sistema Nervioso Periférico/efectos de los fármacos , Fenotipo , Vesículas Sinápticas/efectos de los fármacos , Factores de Elongación Transcripcional
7.
PLoS One ; 9(4): e94612, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24760006

RESUMEN

The splice site mutation in the IKBKAP gene coding for IKAP protein leads to the tissue-specific skipping of exon 20, with concomitant reduction in IKAP protein production. This causes the neurodevelopmental, autosomal-recessive genetic disorder - Familial Dysautonomia (FD). The molecular hallmark of FD is the severe reduction of IKAP protein in the nervous system that is believed to be the main reason for the devastating symptoms of this disease. Our recent studies showed that in the brain of two FD patients, genes linked to oligodendrocyte differentiation and/or myelin formation are significantly downregulated, implicating IKAP in the process of myelination. However, due to the scarcity of FD patient tissues, these results awaited further validation in other models. Recently, two FD mouse models that faithfully recapitulate FD were generated, with two types of mutations resulting in severely low levels of IKAP expression. Here we demonstrate that IKAP deficiency in these FD mouse models affects a similar set of genes as in FD patients' brains. In addition, we identified two new IKAP target genes involved in oligodendrocyte cells differentiation and myelination, further underscoring the essential role of IKAP in this process. We also provide proof that IKAP expression is needed cell-autonomously for the regulation of expression of genes involved in myelin formation since knockdown of IKAP in the Oli-neu oligodendrocyte precursor cell line results in similar deficiencies. Further analyses of these two experimental models will compensate for the lack of human postmortem tissues and will advance our understanding of the role of IKAP in myelination and the disease pathology.


Asunto(s)
Proteínas Portadoras/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Animales , Proteínas Portadoras/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Disautonomía Familiar/genética , Disautonomía Familiar/metabolismo , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , Ratones , Mutación , Vaina de Mielina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA