Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 42(19): e113246, 2023 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-37575021

RESUMEN

Neuronal hyperactivity is a key feature of early stages of Alzheimer's disease (AD). Genetic studies in AD support that microglia act as potential cellular drivers of disease risk, but the molecular determinants of microglia-synapse engulfment associated with neuronal hyperactivity in AD are unclear. Here, using super-resolution microscopy, 3D-live imaging of co-cultures, and in vivo imaging of lipids in genetic models, we found that spines become hyperactive upon Aß oligomer stimulation and externalize phosphatidylserine (ePtdSer), a canonical "eat-me" signal. These apoptotic-like spines are targeted by microglia for engulfment via TREM2 leading to amelioration of Aß oligomer-induced synaptic hyperactivity. We also show the in vivo relevance of ePtdSer-TREM2 signaling in microglia-synapse engulfment in the hAPP NL-F knock-in mouse model of AD. Higher levels of apoptotic-like synapses in mice as well as humans that carry TREM2 loss-of-function variants were also observed. Our work supports that microglia remove hyperactive ePtdSer+ synapses in Aß-relevant context and suggest a potential beneficial role for microglia in the earliest stages of AD.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Ratones , Animales , Enfermedad de Alzheimer/genética , Microglía , Sinapsis , Modelos Animales de Enfermedad , Péptidos beta-Amiloides/genética , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/genética
2.
Nat Methods ; 20(10): 1581-1592, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37723246

RESUMEN

Here we report SUPPORT (statistically unbiased prediction utilizing spatiotemporal information in imaging data), a self-supervised learning method for removing Poisson-Gaussian noise in voltage imaging data. SUPPORT is based on the insight that a pixel value in voltage imaging data is highly dependent on its spatiotemporal neighboring pixels, even when its temporally adjacent frames alone do not provide useful information for statistical prediction. Such dependency is captured and used by a convolutional neural network with a spatiotemporal blind spot to accurately denoise voltage imaging data in which the existence of the action potential in a time frame cannot be inferred by the information in other frames. Through simulations and experiments, we show that SUPPORT enables precise denoising of voltage imaging data and other types of microscopy image while preserving the underlying dynamics within the scene.


Asunto(s)
Microscopía , Redes Neurales de la Computación , Relación Señal-Ruido , Distribución Normal , Procesamiento de Imagen Asistido por Computador/métodos
3.
Proc Natl Acad Sci U S A ; 120(12): e2220649120, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36920925

RESUMEN

Subthreshold depolarization enhances neurotransmitter release evoked by action potentials and plays a key role in modulating synaptic transmission by combining analog and digital signals. This process is known to be Ca2+ dependent. However, the underlying mechanism of how small changes in basal Ca2+ caused by subthreshold depolarization can regulate transmitter release triggered by a large increase in local Ca2+ is not well understood. This study aimed to investigate the source and signaling mechanisms of Ca2+ that couple subthreshold depolarization with the enhancement of glutamate release in hippocampal cultures and CA3 pyramidal neurons. Subthreshold depolarization increased presynaptic Ca2+ levels, the frequency of spontaneous release, and the amplitude of evoked release, all of which were abolished by blocking L-type Ca2+ channels. A high concentration of intracellular Ca2+ buffer or blockade of calmodulin abolished depolarization-induced increases in transmitter release. Estimation of the readily releasable pool size using hypertonic sucrose showed depolarization-induced increases in readily releasable pool size, and this increase was abolished by the blockade of calmodulin. Our results provide mechanistic insights into the modulation of transmitter release by subthreshold potential change and highlight the role of L-type Ca2+ channels in coupling subthreshold depolarization to the activation of Ca2+-dependent signaling molecules that regulate transmitter release.


Asunto(s)
Canales de Calcio Tipo L , Calcio , Potenciales Evocados , Ácido Glutámico , Potenciales de la Membrana , Canales de Calcio Tipo L/metabolismo , Ácido Glutámico/metabolismo , Calmodulina/metabolismo , Calcio/metabolismo , Terminales Presinápticos/metabolismo , Neurotransmisores/metabolismo , Animales , Ratas , Células Cultivadas , Hipocampo/citología , Neuronas/metabolismo , Ratas Sprague-Dawley , Transmisión Sináptica
4.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33372133

RESUMEN

Glutamate uptake into synaptic vesicles (SVs) depends on cation/H+ exchange activity, which converts the chemical gradient (ΔpH) into membrane potential (Δψ) across the SV membrane at the presynaptic terminals. Thus, the proper recruitment of cation/H+ exchanger to SVs is important in determining glutamate quantal size, yet little is known about its localization mechanism. Here, we found that secretory carrier membrane protein 5 (SCAMP5) interacted with the cation/H+ exchanger NHE6, and this interaction regulated NHE6 recruitment to glutamatergic presynaptic terminals. Protein-protein interaction analysis with truncated constructs revealed that the 2/3 loop domain of SCAMP5 is directly associated with the C-terminal region of NHE6. The use of optical imaging and electrophysiological recording showed that small hairpin RNA-mediated knockdown (KD) of SCAMP5 or perturbation of SCAMP5/NHE6 interaction markedly inhibited axonal trafficking and the presynaptic localization of NHE6, leading to hyperacidification of SVs and a reduction in the quantal size of glutamate release. Knockout of NHE6 occluded the effect of SCAMP5 KD without causing additional defects. Together, our results reveal that as a key regulator of axonal trafficking and synaptic localization of NHE6, SCAMP5 could adjust presynaptic strength by regulating quantal size at glutamatergic synapses. Since both proteins are autism candidate genes, the reduced quantal size by interrupting their interaction may underscore synaptic dysfunction observed in autism.


Asunto(s)
Ácido Glutámico/metabolismo , Proteínas de la Membrana/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Axones/metabolismo , Transporte Biológico , Línea Celular , Potenciales Postsinápticos Excitadores/fisiología , Células HEK293 , Humanos , Proteínas de la Membrana/fisiología , Técnicas de Placa-Clamp , Terminales Presinápticos/fisiología , Transporte de Proteínas , Intercambiadores de Sodio-Hidrógeno/fisiología , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo
5.
BMC Biol ; 21(1): 248, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37940973

RESUMEN

BACKGROUND: Open-top light-sheet microscopy (OT-LSM) is a specialized microscopic technique for the high-throughput cellular imaging of optically cleared, large-sized specimens, such as the brain. Despite the development of various OT-LSM techniques, achieving submicron resolution in all dimensions remains. RESULTS: We developed a high-resolution open-top axially swept LSM (HR-OTAS-LSM) for high-throughput and high-resolution imaging in all dimensions. High axial and lateral resolutions were achieved by using an aberration-corrected axially swept excitation light sheet in the illumination arm and a high numerical aperture (NA) immersion objective lens in the imaging arm, respectively. The high-resolution, high-throughput visualization of neuronal networks in mouse brain and retina specimens validated the performance of HR-OTAS-LSM. CONCLUSIONS: The proposed HR-OTAS-LSM method represents a significant advancement in the high-resolution mapping of cellular networks in biological systems such as the brain and retina.


Asunto(s)
Encéfalo , Neuronas , Ratones , Animales , Microscopía Fluorescente/métodos
6.
Opt Express ; 30(11): 19152-19164, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-36221700

RESUMEN

In microscopic imaging of biological tissues, particularly real-time visualization of neuronal activities, rapid acquisition of volumetric images poses a prominent challenge. Typically, two-dimensional (2D) microscopy can be devised into an imaging system with 3D capability using any varifocal lens. Despite the conceptual simplicity, such an upgrade yet requires additional, complicated device components and usually suffers from a reduced acquisition rate, which is critical to properly document rapid neurophysiological dynamics. In this study, we implemented an electrically tunable lens (ETL) in the line-scan confocal microscopy (LSCM), enabling the volumetric acquisition at the rate of 20 frames per second with a maximum volume of interest of 315 × 315 × 80 µm3. The axial extent of point-spread-function (PSF) was 17.6 ± 1.6 µm and 90.4 ± 2.1 µm with the ETL operating in either stationary or resonant mode, respectively, revealing significant depth axial penetration by the resonant mode ETL microscopy. We further demonstrated the utilities of the ETL system by volume imaging of both cleared mouse brain ex vivo samples and in vivo brains. The current study showed a successful application of resonant ETL for constructing a high-performance 3D axially scanning LSCM (asLSCM) system. Such advances in rapid volumetric imaging would significantly enhance our understanding of various dynamic biological processes.


Asunto(s)
Cristalino , Lentes , Animales , Electricidad , Ratones , Microscopía Confocal/métodos , Cintigrafía
7.
J Neurosci ; 40(44): 8426-8437, 2020 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-32989096

RESUMEN

Synaptic strength and reliability are determined by the number of vesicles released per action potential and the availability of release-competent vesicles in the readily releasable pool (RRP). Compared with release of a single vesicle (univesicular release), multivesicular release (MVR) would speed up RRP depletion, yet whether the RRP is refilled differently during the two different release modes has not been investigated. Here, we address this question by quantitative optical imaging with an axon-targeting glutamate sensor, iGluSnFRpre. We found that hippocampal synapses preferentially release multiple vesicles per action potential at high extracellular calcium or by paired-pulse stimulation. When MVR prevails, the RRP is recovered very rapidly with a time constant of 430 ms. This rapid recovery is mediated by dynamin-dependent endocytosis followed by direct reuse of retrieved vesicles. Furthermore, our simulation proved that the portion of retrieved vesicles that directly refill the RRP increases dramatically (>70%) in MVR compared with that in univesicular release (<10%). These results suggest that the contribution of rapid and direct recruitment of retrieved vesicle to the RRP changes dynamically with release mode at the level of individual synapses, which suggests a form of presynaptic homeostatic plasticity for reliable synaptic transmission during various synaptic activity.SIGNIFICANCE STATEMENT The number of vesicles released in response to an action potential and the number of release competent vesicles in the readily releasable pool (RRP) are the fundamental determinants of synaptic efficacy. Despite its functional advantages, releasing multiple vesicles, especially at small synapses, can deplete the RRP after a couple of action potentials. To prevent failure of synaptic transmission, the RRP should be refilled rapidly, yet whether the RRP replenishment process is regulated by the release mode has not been investigated. Here, using quantitative optical glutamate imaging and simulation, we demonstrate that the contribution of the fast refilling mechanism changes with release mode at the level of individual synapses, suggesting a rapid form of presynaptic homeostatic plasticity during various synaptic activity.


Asunto(s)
Hipocampo/fisiología , Sinapsis/fisiología , Vesículas Sinápticas/fisiología , Potenciales de Acción/fisiología , Animales , Axones/fisiología , Señalización del Calcio/fisiología , Simulación por Computador , Dinaminas/fisiología , Fenómenos Electrofisiológicos , Endocitosis , Ácido Glutámico/metabolismo , Ácido Glutámico/fisiología , Inmunohistoquímica , Cinética , Ratas , Transmisión Sináptica
8.
Proc Natl Acad Sci U S A ; 113(24): 6749-54, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27226294

RESUMEN

Neural Abelson-related gene-binding protein 2 (nArgBP2) was originally identified as a protein that directly interacts with synapse-associated protein 90/postsynaptic density protein 95-associated protein 3 (SAPAP3), a postsynaptic scaffolding protein critical for the assembly of glutamatergic synapses. Although genetic deletion of nArgBP2 in mice leads to manic/bipolar-like behaviors resembling many aspects of symptoms in patients with bipolar disorder, the actual function of nArgBP2 at the synapse is completely unknown. Here, we found that the knockdown (KD) of nArgBP2 by specific small hairpin RNAs (shRNAs) resulted in a dramatic change in dendritic spine morphology. Reintroducing shRNA-resistant nArgBP2 reversed these defects. In particular, nArgBP2 KD impaired spine-synapse formation such that excitatory synapses terminated mostly at dendritic shafts instead of spine heads in spiny neurons, although inhibitory synapse formation was not affected. nArgBP2 KD further caused a marked increase of actin cytoskeleton dynamics in spines, which was associated with increased Wiskott-Aldrich syndrome protein-family verprolin homologous protein 1 (WAVE1)/p21-activated kinase (PAK) phosphorylation and reduced activity of cofilin. These effects of nArgBP2 KD in spines were rescued by inhibiting PAK or activating cofilin combined with sequestration of WAVE. Together, our results suggest that nArgBP2 functions to regulate spine morphogenesis and subsequent spine-synapse formation at glutamatergic synapses. They also raise the possibility that the aberrant regulation of synaptic actin filaments caused by reduced nArgBP2 expression may contribute to the manifestation of the synaptic dysfunction observed in manic/bipolar disorder.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Espinas Dendríticas/metabolismo , Sinapsis/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Trastorno Bipolar/genética , Trastorno Bipolar/metabolismo , Técnicas de Silenciamiento del Gen , Ratones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Sinapsis/genética
9.
J Cell Sci ; 128(9): 1848-61, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25795301

RESUMEN

The 5-hydroxytryptamine (5-HT, also known as serotonin) subtype 6 receptor (5-HT6R, also known as HTR6) plays roles in cognition, anxiety and learning and memory disorders, yet new details concerning its regulation remain poorly understood. In this study, we found that 5-HT6R directly interacted with SNX14 and that this interaction dramatically increased internalization and degradation of 5-HT6R. Knockdown of endogenous SNX14 had the opposite effect. SNX14 is highly expressed in the brain and contains a putative regulator of G-protein signaling (RGS) domain. Although its RGS domain was found to be non-functional as a GTPase activator for Gαs, we found that it specifically bound to and sequestered Gαs, thus inhibiting downstream cAMP production. We further found that protein kinase A (PKA)-mediated phosphorylation of SNX14 inhibited its binding to Gαs and diverted SNX14 from Gαs binding to 5-HT6R binding, thus facilitating the endocytic degradation of the receptor. Therefore, our results suggest that SNX14 is a dual endogenous negative regulator in 5-HT6R-mediated signaling pathway, modulating both signaling and trafficking of 5-HT6R.


Asunto(s)
Neuronas/metabolismo , Receptores de Serotonina/metabolismo , Transducción de Señal , Nexinas de Clasificación/metabolismo , Animales , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citosol/metabolismo , Endocitosis , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteolisis , Ratas
10.
J Biol Chem ; 290(12): 7323-35, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25605715

RESUMEN

Recent studies have reported conflicting results regarding the role of ARF6 in dendritic spine development, but no clear answer for the controversy has been suggested. We found that ADP-ribosylation factor 6 (ARF6) either positively or negatively regulates dendritic spine formation depending on neuronal maturation and activity. ARF6 activation increased the spine formation in developing neurons, whereas it decreased spine density in mature neurons. Genome-wide microarray analysis revealed that ARF6 activation in each stage leads to opposite patterns of expression of a subset of genes that are involved in neuronal morphology. ARF6-mediated Rac1 activation via the phospholipase D pathway is the coincident factor in both stages, but the antagonistic RhoA pathway becomes involved in the mature stage. Furthermore, blocking neuronal activity in developing neurons using tetrodotoxin or enhancing the activity in mature neurons using picrotoxin or chemical long term potentiation reversed the effect of ARF6 on each stage. Thus, activity-dependent dynamic changes in ARF6-mediated spine structures may play a role in structural plasticity of mature neurons.


Asunto(s)
Factores de Ribosilacion-ADP/fisiología , Espinas Dendríticas , Neuronas/citología , Factor 6 de Ribosilación del ADP , Animales , Secuencia de Bases , Células Cultivadas , Hipocampo/citología , Hipocampo/embriología , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley
11.
Biochim Biophys Acta ; 1853(10 Pt A): 2432-43, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26149501

RESUMEN

The type I phosphatidylinositol 4-phosphate 5-kinase (PIP5K) family members and their lipid product, phosphatidylinositol 4,5-bisphosphate (PIP2) are important regulators of actin cytoskeleton. PIP5Kγ 90kDa (PIP5Kγ90), an isoform of PIP5K, localizes to focal adhesions (FAs) and is activated via its interaction with the cytoskeletal protein, talin. Currently, regulatory signaling pathways of talin-PIP5Kγ90 interaction related to FA dynamics and cell motility are not well understood. Considering the presence of Akt consensus motifs in PIP5Kγ90, we examined a potential link of Akt activation to talin-PIP5Kγ90 interaction. We found that Akt phosphorylated PIP5Kγ90 specifically at serine 555 (S555) in vitro and in epidermal growth factor (EGF)-treated cells phosphoinositide 3-kinase-dependently. EGF treatment suppressed talin-PIP5Kγ90 interaction and PIP2 levels. Similarly, a phosphomimetic mutant (S555D), but not non-phosphorylatable mutant (S555A), of PIP5Kγ90 had reduced talin binding affinity, lowered PIP2 levels, and was dislocated from FAs. The S555D mutant also caused decreases in actin stress fibers and vinculin-positive FAs. Moreover, assembly and disassembly of FAs were enhanced by S555D expression and EGF-induced cell migration was relatively low in S555A-expressing cells compared to wild-type-expressing cells. PIP5Kγ87, a PIP5Kγ splice variant lacking the talin binding motif, was phosphorylated by Akt, which, however, hardly affected PIP2 levels. Taken together, our results suggested that Akt-mediated PIP5Kγ90 S555 phosphorylation is a novel regulatory point for talin binding to control PIP2 level at the FAs, thereby modulating FA dynamics and cell motility.


Asunto(s)
Empalme Alternativo/fisiología , Movimiento Celular/fisiología , Adhesiones Focales/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Talina/metabolismo , Sustitución de Aminoácidos , Adhesiones Focales/genética , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Mutación Missense , Fosforilación/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas c-akt/genética , Talina/genética
12.
J Neurosci ; 34(30): 10085-95, 2014 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25057210

RESUMEN

Secretory carrier membrane protein 5 (SCAMP5), a recently identified candidate gene for autism, is brain specific and highly abundant in synaptic vesicles (SVs), but its function is currently unknown. Here, we found that knockdown (KD) of endogenous SCAMP5 by SCAMP5-specific shRNAs in cultured rat hippocampal neurons resulted in a reduction in total vesicle pool size as well as in recycling pool size, but the recycling/resting pool ratio was significantly increased. SCAMP5 KD slowed endocytosis after stimulation, but impaired it severely during strong stimulation. We also found that KD dramatically lowered the threshold of activity at which SV endocytosis became unable to compensate for the ongoing exocytosis occurring during a stimulus. Reintroducing shRNA-resistant SCAMP5 reversed these endocytic defects. Therefore, our results suggest that SCAMP5 functions during high neuronal activity when a heavy load is imposed on endocytosis. Our data also raise the possibility that the reduction in expression of SCAMP5 in autistic patients may be related to the synaptic dysfunction observed in autism.


Asunto(s)
Proteínas Portadoras/fisiología , Endocitosis/fisiología , Proteínas de la Membrana/fisiología , Neuronas/fisiología , Vesículas Sinápticas/fisiología , Animales , Proteínas Portadoras/genética , Endocitosis/genética , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Hipocampo/citología , Hipocampo/fisiología , Humanos , Masculino , Proteínas de la Membrana/genética , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/genética
13.
J Biol Chem ; 288(41): 29453-66, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-24003235

RESUMEN

SNX26, a brain-enriched RhoGAP, plays a key role in dendritic arborization during early neuronal development in the neocortex. In mature neurons, it is localized to dendritic spines, but little is known about its role in later stages of development. Our results show that SNX26 interacts with PSD-95 in dendritic spines of cultured hippocampal neurons, and as a GTPase-activating protein for Cdc42, it decreased the F-actin content in COS-7 cells and in dendritic spines of neurons. Overexpression of SNX26 resulted in a GTPase-activating protein activity-dependent decrease in total protrusions and spine density together with dramatic inhibition of filopodia-to-spine transformations. Such effects of SNX26 were largely rescued by a constitutively active mutant of Cdc42. Consistently, an shRNA-mediated knockdown of SNX26 significantly increased total protrusions and spine density, resulting in an increase in thin or stubby type spines at the expense of the mushroom spine type. Moreover, endogenous expression of SNX26 was shown to be bi-directionally modulated by neuronal activity. Therefore, we propose that in addition to its key role in neuronal development, SNX26 also has a role in the activity-dependent structural change of dendritic spines in mature neurons.


Asunto(s)
Espinas Dendríticas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Guanilato-Quinasas/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Nexinas de Clasificación/metabolismo , Actinas/metabolismo , Animales , Sitios de Unión , Células COS , Células Cultivadas , Chlorocebus aethiops , Homólogo 4 de la Proteína Discs Large , Proteínas Activadoras de GTPasa/genética , Guanilato-Quinasas/genética , Células HEK293 , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Humanos , Immunoblotting , Proteínas de la Membrana/genética , Ratones , Microscopía Confocal , Neuronas/citología , Unión Proteica , Seudópodos/metabolismo , Interferencia de ARN , Ratas , Nexinas de Clasificación/genética , Factores de Tiempo
14.
Pflugers Arch ; 466(10): 1987-98, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24413888

RESUMEN

Intracellular Ca(2+) signal is a key regulator of axonal growth during brain development. As transient receptor potential (TRP) channels are permeable to Ca(2+) and mediate numerous brain functions, it is conceivable that many TRP channels would regulate neuronal differentiation. We therefore screened TRP channels that are involved in the regulation of neurite growth. Among the TRP channels, the Trpm2 level was inversely associated with neurite growth. TRPM2 was highly expressed in embryonic brain. Pharmacological perturbation or knockdown of TRPM2 markedly increased the axonal growth, whereas its overexpression inhibited the axonal growth. Addition of ADP ribose, an endogenous activator of TRPM2, to PC12 cells significantly repressed the axonal growth. TRPM2 was actively involved in the neuronal retraction induced by cerebrospinal fluid-rich lysophosphatidic acid (LPA). More importantly, neurons isolated from the brain of Trpm2-deficient mice have significantly longer neurites with a greater number of spines than those obtained from the brain of wild-type mice. Therefore, we conclude that TRPM2 mediates the LPA-induced suppression of axonal growth, which provides a long-sought mechanism underlying the effect of LPA on neuronal development.


Asunto(s)
Encéfalo/metabolismo , Neuritas/metabolismo , Neurogénesis , Canales Catiónicos TRPM/metabolismo , Adenosina Difosfato Ribosa/farmacología , Animales , Encéfalo/citología , Encéfalo/embriología , Células Cultivadas , Células HEK293 , Humanos , Lisofosfolípidos/farmacología , Ratones , Neuritas/efectos de los fármacos , Células PC12 , Ratas , Canales Catiónicos TRPM/genética
15.
J Cell Sci ; 125(Pt 1): 67-80, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22250195

RESUMEN

Neural Wiskott-Aldrich syndrome protein (N-WASP) is involved in tight regulation of actin polymerization and dynamics. N-WASP activity is regulated by intramolecular interaction, binding to small GTPases and tyrosine phosphorylation. Here, we report on a novel regulatory mechanism; we demonstrate that N-WASP interacts with dual-specificity tyrosine-phosphorylation-regulated kinase 1A (Dyrk1A). In vitro kinase assays indicate that Dyrk1A directly phosphorylates the GTPase-binding domain (GBD) of N-WASP at three sites (Thr196, Thr202 and Thr259). Phosphorylation of the GBD by Dyrk1A promotes the intramolecular interaction of the GBD and verprolin, cofilin and acidic (VCA) domains of N-WASP, and subsequently inhibits Arp2/3-complex-mediated actin polymerization. Overexpression of either Dyrk1A or a phospho-mimetic N-WASP mutant inhibits filopodia formation in COS-7 cells. By contrast, the knockdown of Dyrk1A expression or overexpression of a phospho-deficient N-WASP mutant promotes filopodia formation. Furthermore, the overexpression of a phospho-mimetic N-WASP mutant significantly inhibits dendritic spine formation in primary hippocampal neurons. These findings suggest that Dyrk1A negatively regulates actin filament assembly by phosphorylating N-WASP, which ultimately promotes the intramolecular interaction of its GBD and VCA domains. These results provide insight on the mechanisms contributing to diverse actin-based cellular processes such as cell migration, endocytosis and neuronal differentiation.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Fosfotreonina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/química , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/química , Animales , Células COS , Chlorocebus aethiops , Dendritas/metabolismo , GTP Fosfohidrolasas/metabolismo , Expresión Génica , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/genética , Seudópodos/metabolismo , Ratas , Proteína del Síndrome de Wiskott-Aldrich/antagonistas & inhibidores , Proteína de Unión al GTP cdc42/metabolismo , Quinasas DyrK
16.
Biochem Biophys Res Commun ; 444(4): 644-50, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24491538

RESUMEN

CPAP is an essential component for centriole formation. Here, we report that CPAP is also critical for symmetric spindle pole formation during mitosis. We observed that pericentriolar material between the mitotic spindle poles were asymmetrically distributed in CPAP-depleted cells even with intact numbers of centrioles. The length of procentrioles was slightly reduced by CPAP depletion, but the length of mother centrioles was not affected. Surprisingly, the young mother centrioles of the CPAP-depleted cells are not fully matured, as evidenced by the absence of distal and subdistal appendage proteins. We propose that the selective absence of centriolar appendages at the young mother centrioles may be responsible for asymmetric spindle pole formation in CPAP-depleted cells. Our results suggest that the neural stem cells with CPAP mutations might form asymmetric spindle poles, which results in premature initiation of differentiation.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Mitosis , Polos del Huso/genética , Centriolos/genética , Centriolos/ultraestructura , Células HeLa , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética , Polos del Huso/ultraestructura
17.
Front Cell Neurosci ; 18: 1354900, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38440150

RESUMEN

nArgBP2, a protein whose disruption is implicated in intellectual disability, concentrates in excitatory spine-synapses. By forming a triad with GKAP and SHANK, it regulates spine structural rearrangement. We here find that GKAP and SHANK3 concentrate close to the synaptic contact, whereas nArgBP2 concentrates more centrally in the spine. The three proteins collaboratively form biomolecular condensates in living fibroblasts, exhibiting distinctive layered localizations. nArgBP2 concentrates in the inner phase, SHANK3 in the outer phase, and GKAP partially in both. Upon co-expression of GKAP and nArgBP2, they evenly distribute within condensates, with a notable peripheral localization of SHANK3 persisting when co-expressed with either GKAP or nArgBP2. Co-expression of SHANK3 and GKAP with CaMKIIα results in phase-in-phase condensates, with CaMKIIα at the central locus and SHANK3 and GKAP exhibiting peripheral localization. Additional co-expression of nArgBP2 maintains the layered organizational structure within condensates. Subsequent CaMKIIα activation disperses a majority of the condensates, with an even distribution of all proteins within the extant deformed condensates. Our findings suggest that protein segregation via phase separation may contribute to establishing layered organization in dendritic spines.

18.
Exp Mol Med ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951140

RESUMEN

The amyloid cascade hypothesis suggests that amyloid beta (Aß) contributes to initiating subsequent tau pathology in Alzheimer's disease (AD). However, the underlying mechanisms through which Aß contributes to tau uptake and propagation remain poorly understood. Here, we show that preexisting amyloid pathology accelerates the uptake of extracellular tau into neurons. Using quantitative proteomic analysis of endocytic vesicles, we reveal that Aß induces the internalization of fibroblast growth factor receptor 3 (FGFR3). Extracellular tau binds to the extracellular domain of FGFR3 and is internalized by the FGFR3 ligand, fibroblast growth factor 2 (FGF2). Aß accelerates FGF2 secretion from neurons, thereby inducing the internalization of tau-attached FGFR3. Knockdown of FGFR3 in the hippocampus reduces tau aggregation by decreasing tau uptake and improving memory function in AD model mice. These data suggest FGFR3 in neurons as a novel tau receptor and a key mediator of Aß-induced tau uptake in AD.

19.
Sci Adv ; 10(13): eadj9600, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38536932

RESUMEN

Recently identified human FOXP3lowCD45RA- inflammatory non-suppressive (INS) cells produce proinflammatory cytokines, exhibit reduced suppressiveness, and promote antitumor immunity unlike conventional regulatory T cells (Tregs). In spite of their implication in tumors, the mechanism for generation of FOXP3lowCD45RA- INS cells in vivo is unclear. We showed that the FOXP3lowCD45RA- cells in human tumors demonstrate attenuated expression of CRIF1, a vital mitochondrial regulator. Mice with CRIF1 deficiency in Tregs bore Foxp3lowINS-Tregs with mitochondrial dysfunction and metabolic reprograming. The enhanced glutaminolysis activated α-ketoglutarate-mTORC1 axis, which promoted proinflammatory cytokine expression by inducing EOMES and SATB1 expression. Moreover, chromatin openness of the regulatory regions of the Ifng and Il4 genes was increased, which facilitated EOMES/SATB1 binding. The increased α-ketoglutarate-derived 2-hydroxyglutarate down-regulated Foxp3 expression by methylating the Foxp3 gene regulatory regions. Furthermore, CRIF1 deficiency-induced Foxp3lowINS-Tregs suppressed tumor growth in an IFN-γ-dependent manner. Thus, CRIF1 deficiency-mediated mitochondrial dysfunction results in the induction of Foxp3lowINS-Tregs including FOXP3lowCD45RA- cells that promote antitumor immunity.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz , Enfermedades Mitocondriales , Neoplasias , Humanos , Ratones , Animales , Linfocitos T Reguladores , Ácidos Cetoglutáricos/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Citocinas/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo
20.
J Neurosci ; 32(26): 8845-54, 2012 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-22745485

RESUMEN

The blood-brain barrier (BBB), which is formed by adherens and tight junctions (TJs) of endothelial cells, maintains homeostasis of the brain. Disrupted intracellular Ca²âº homeostasis and breakdown of the BBB have been implicated in the pathogenesis of Alzheimer's disease (AD). The receptor for advanced glycation end products (RAGE) is known to interact with amyloid ß-peptide (Aß) and mediate Aß transport across the BBB, contributing to the deposition of Aß in the brain. However, molecular mechanisms underlying Aß-RAGE interaction-induced alterations in the BBB have not been identified. We found that Aß1₋42 induces enhanced permeability, disruption of zonula occludin-1 (ZO-1) expression in the plasma membrane, and increased intracellular calcium and matrix metalloproteinase (MMP) secretion in cultured endothelial cells. Neutralizing antibodies against RAGE and inhibitors of calcineurin and MMPs prevented Aß1₋42-induced changes in ZO-1, suggesting that Aß-RAGE interactions alter TJ proteins through the Ca²âº-calcineurin pathway. Consistent with these in vitro findings, we found disrupted microvessels near Aß plaque-deposited areas, elevated RAGE expression, and enhanced MMP secretion in microvessels of the brains of 5XFAD mice, an animal model for AD. We have identified a potential molecular pathway underlying Aß-RAGE interaction-induced breakage of BBB integrity. This pathway might play an important role in the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/patología , Calcineurina/metabolismo , Calcio/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal/fisiología , Uniones Estrechas/metabolismo , Factores de Edad , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Transporte Biológico , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular Transformada , Claudinas/metabolismo , Dextranos/metabolismo , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Células Endoteliales , Femenino , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Regulación de la Expresión Génica/genética , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Imagenología Tridimensional , Inmunosupresores/farmacología , Metaloproteinasas de la Matriz/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Microvasos/patología , Mutación/genética , Fragmentos de Péptidos/farmacología , Fosfoproteínas/metabolismo , Presenilina-1/genética , Inhibidores de Proteasas/farmacología , Receptor para Productos Finales de Glicación Avanzada , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tacrolimus/farmacología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Uniones Estrechas/ultraestructura , Transfección , Proteína de la Zonula Occludens-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA