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

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
EMBO Rep ; 24(9): e56624, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37440685

RESUMEN

The ability of stem cells to switch between quiescent and proliferative states is crucial for maintaining tissue homeostasis and regeneration. Drosophila quiescent neural stem cells (qNSCs) extend a primary protrusion that is enriched in acentrosomal microtubules and can be regenerated upon injury. Arf1 promotes microtubule growth, reactivation (exit from quiescence), and regeneration of qNSC protrusions upon injury. However, how Arf1 is regulated in qNSCs remains elusive. Here, we show that the microtubule minus-end binding protein Patronin/CAMSAP promotes acentrosomal microtubule growth and quiescent NSC reactivation. Patronin is important for the localization of Arf1 at Golgi and physically associates with Arf1, preferentially with its GDP-bound form. Patronin is also required for the regeneration of qNSC protrusion, likely via the regulation of microtubule growth. Finally, Patronin functions upstream of Arf1 and its effector Msps/XMAP215 to target the cell adhesion molecule E-cadherin to NSC-neuropil contact sites during NSC reactivation. Our findings reveal a novel link between Patronin/CAMSAP and Arf1 in the regulation of microtubule growth and NSC reactivation. A similar mechanism might apply to various microtubule-dependent systems in mammals.


Asunto(s)
Proteínas de Drosophila , Células-Madre Neurales , Animales , Proteínas Asociadas a Microtúbulos/metabolismo , Drosophila/metabolismo , Microtúbulos/metabolismo , Proteínas de Drosophila/metabolismo , Células-Madre Neurales/metabolismo , Mamíferos/metabolismo
2.
PLoS Genet ; 16(4): e1008653, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32324743

RESUMEN

Neural stem cells (NSCs) are crucial for development, regeneration, and repair of the nervous system. Most NSCs in mammalian adult brains are quiescent, but in response to extrinsic stimuli, they can exit from quiescence and become reactivated to give rise to new neurons. The delicate balance between NSC quiescence and activation is important for adult neurogenesis and NSC maintenance. However, how NSCs transit between quiescence and activation remains largely elusive. Here, we discuss our current understanding of the molecular mechanisms underlying the reactivation of quiescent NSCs. We review recent advances on signaling pathways originated from the NSC niche and their crosstalk in regulating NSC reactivation. We also highlight new intrinsic paradigms that control NSC reactivation in Drosophila and mammalian systems. We also discuss emerging evidence on modeling human neurodevelopmental disorders using NSCs.


Asunto(s)
Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Trastornos del Neurodesarrollo/patología , Transducción de Señal , Animales , Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Humanos , Insulina/metabolismo , Trastornos del Neurodesarrollo/terapia , Nicho de Células Madre
3.
Sci Adv ; 10(30): eadl4694, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39047090

RESUMEN

The transitioning of neural stem cells (NSCs) between quiescent and proliferative states is fundamental for brain development and homeostasis. Defects in NSC reactivation are associated with neurodevelopmental disorders. Drosophila quiescent NSCs extend an actin-rich primary protrusion toward the neuropil. However, the function of the actin cytoskeleton during NSC reactivation is unknown. Here, we reveal the fine filamentous actin (F-actin) structures in the protrusions of quiescent NSCs by expansion and super-resolution microscopy. We show that F-actin polymerization promotes the nuclear translocation of myocardin-related transcription factor, a microcephaly-associated transcription factor, for NSC reactivation and brain development. F-actin polymerization is regulated by a signaling cascade composed of G protein-coupled receptor Smog, G protein αq subunit, Rho1 guanosine triphosphatase, and Diaphanous (Dia)/Formin during NSC reactivation. Further, astrocytes secrete a Smog ligand folded gastrulation to regulate Gαq-Rho1-Dia-mediated NSC reactivation. Together, we establish that the Smog-Gαq-Rho1 signaling axis derived from astrocytes, an NSC niche, regulates Dia-mediated F-actin dynamics in NSC reactivation.


Asunto(s)
Actinas , Astrocitos , Proteínas de Drosophila , Células-Madre Neurales , Receptores Acoplados a Proteínas G , Transducción de Señal , Animales , Actinas/metabolismo , Astrocitos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Citoesqueleto de Actina/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Unión al GTP rho/metabolismo
4.
bioRxiv ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38903085

RESUMEN

The transitioning of neural stem cells (NSCs) between quiescent and proliferative states is fundamental for brain development and homeostasis. Defects in NSC reactivation are associated with neurodevelopmental disorders. Drosophila quiescent NSCs extend an actin-rich primary protrusion toward the neuropil. However, the function of the actin cytoskeleton during NSC reactivation is unknown. Here, we reveal the fine F-actin structures in the protrusions of quiescent NSCs by expansion and super-resolution microscopy. We show that F-actin polymerization promotes the nuclear translocation of Mrtf, a microcephaly-associated transcription factor, for NSC reactivation and brain development. F-actin polymerization is regulated by a signaling cascade composed of G-protein-coupled receptor (GPCR) Smog, G-protein αq subunit, Rho1 GTPase, and Diaphanous (Dia)/Formin during NSC reactivation. Further, astrocytes secrete a Smog ligand Fog to regulate Gαq-Rho1-Dia-mediated NSC reactivation. Together, we establish that the Smog-Gαq-Rho1 signaling axis derived from astrocytes, a NSC niche, regulates Dia-mediated F-actin dynamics in NSC reactivation.

5.
J Cell Biol ; 216(5): 1371-1386, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28400443

RESUMEN

The cell cortex is essential to maintain animal cell shape, and contractile forces generated within it by nonmuscle myosin II (NMY-2) drive cellular morphogenetic processes such as cytokinesis. The role of actin cross-linking proteins in cortical dynamics is still incompletely understood. Here, we show that the evolutionarily conserved actin bundling/cross-linking protein plastin is instrumental for the generation of potent cortical actomyosin contractility in the Caenorhabditis elegans zygote. PLST-1 was enriched in contractile structures and was required for effective coalescence of NMY-2 filaments into large contractile foci and for long-range coordinated contractility in the cortex. In the absence of PLST-1, polarization was compromised, cytokinesis was delayed or failed, and 50% of embryos died during development. Moreover, mathematical modeling showed that an optimal amount of bundling agents enhanced the ability of a network to contract. We propose that by increasing the connectivity of the F-actin meshwork, plastin enables the cortex to generate stronger and more coordinated forces to accomplish cellular morphogenesis.


Asunto(s)
Actinas/metabolismo , Caenorhabditis elegans/citología , Polaridad Celular , Citocinesis , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Actomiosina/metabolismo , Animales , Caenorhabditis elegans/embriología , Forma de la Célula , Morfogénesis , Factores de Tiempo , Cigoto/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA