Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38617238

RESUMEN

To survive, organisms must adapt to a staggering diversity of environmental signals, ranging from sensory information to pathogenic infection, across the lifespan. At the same time, organisms intrinsically generate biological oscillations, such as circadian rhythms, without input from the environment. While the nervous system is well-suited to integrate extrinsic and intrinsic cues, how the brain balances these influences to shape biological function system-wide is not well understood at the molecular level. Here, we demonstrate that the cytokine receptor Fn14, previously identified as a mediator of sensory experience-dependent synaptic refinement during brain development, regulates neuronal activity and function in adult mice in a time-of-day-dependent manner. We show that a subset of excitatory pyramidal (PYR) neurons in the CA1 subregion of the hippocampus increase Fn14 expression when neuronal activity is heightened. Once expressed, Fn14 constrains the activity of these same PYR neurons, suggesting that Fn14 operates as a molecular brake on neuronal activity. Strikingly, differences in PYR neuron activity between mice lacking or expressing Fn14 were most robust at daily transitions between light and dark, and genetic ablation of Fn14 caused aberrations in circadian rhythms, sleep-wake states, and sensory-cued and spatial memory. At the cellular level, microglia contacted fewer, but larger, excitatory synapses in CA1 in the absence of Fn14, suggesting that these brain-resident immune cells may dampen neuronal activity by modifying synaptic inputs onto PYR neurons. Finally, mice lacking Fn14 exhibited heightened susceptibility to chemically induced seizures, implicating Fn14 in disorders characterized by hyperexcitation, such as epilepsy. Altogether, these findings reveal that cytokine receptors that mediates inflammation in the periphery, such as Fn14, can also play major roles in healthy neurological function in the adult brain downstream of both extrinsic and intrinsic cues.

2.
bioRxiv ; 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38644996

RESUMEN

Retinal waves represent an early form of patterned spontaneous neural activity in the visual system. These waves originate in the retina before eye-opening and propagate throughout the visual system, influencing the assembly and maturation of subcortical visual brain regions. However, because it is technically challenging to ablate retina-derived cortical waves without inducing compensatory activity, the role these waves play in the development of the visual cortex remains unclear. To address this question, we used targeted conditional genetics to disrupt cholinergic retinal waves and their propagation to select regions of primary visual cortex, which largely prevented compensatory patterned activity. We find that loss of cholinergic retinal waves without compensation impaired the molecular and synaptic maturation of excitatory neurons located in the input layers of visual cortex, as well as layer 1 interneurons. These perinatal molecular and synaptic deficits also relate to functional changes observed at later ages. We find that the loss of perinatal cholinergic retinal waves causes abnormal visual cortex retinotopy, mirroring changes in the retinotopic organization of gene expression, and additionally impairs the processing of visual information. We further show that retinal waves are necessary for higher order processing of sensory information by impacting the state-dependent activity of layer 1 interneurons, a neuronal type that shapes neocortical state-modulation, as well as for state-dependent gain modulation of visual responses of excitatory neurons. Together, these results demonstrate that a brief targeted perinatal disruption of patterned spontaneous activity alters early cortical gene expression as well as synaptic and physiological development, and compromises both fundamental and, notably, higher-order functions of visual cortex after eye-opening.

3.
bioRxiv ; 2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37662250

RESUMEN

Oligodendrocyte precursor cells (OPCs) sculpt neural circuits through the phagocytic engulfment of synapses during development and in adulthood. However, precise techniques for analyzing synapse engulfment by OPCs are limited. Here, we describe a two-pronged cell biological approach for quantifying synapse engulfment by OPCs which merges low- and high-throughput methodologies. In the first method, an adeno-associated virus encoding a pH-sensitive, fluorescently-tagged synaptic marker is expressed in neurons in vivo. This construct allows for the differential labeling of presynaptic inputs that are contained outside of and within acidic phagolysosomal compartments. When followed by immunostaining for markers of OPCs and synapses in lightly fixed tissue, this approach enables the quantification of synapses engulfed by around 30-50 OPCs within a given experiment. In the second method, OPCs isolated from dissociated brain tissue are fixed, incubated with fluorescent antibodies against presynaptic proteins, and then analyzed by flow cytometry. This approach enables the quantification of presynaptic material within tens of thousands of OPCs in less than one week. These methods extend beyond the current imaging-based engulfment assays designed to quantify synaptic phagocytosis by brain-resident immune cells, microglia. Through the integration of these methods, the engulfment of synapses by OPCs can be rigorously quantified at both the individual and populational levels. With minor modifications, these approaches can be adapted to study synaptic phagocytosis by numerous glial cell types in the brain.

4.
Trends Neurosci ; 46(8): 628-639, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37286422

RESUMEN

Oligodendrocyte precursor cells (OPCs) are non-neuronal brain cells that give rise to oligodendrocytes, glia that myelinate the axons of neurons in the brain. Classically known for their contributions to myelination via oligodendrogenesis, OPCs are increasingly appreciated to play diverse roles in the nervous system, ranging from blood vessel formation to antigen presentation. Here, we review emerging literature suggesting that OPCs may be essential for the establishment and remodeling of neural circuits in the developing and adult brain via mechanisms that are distinct from the production of oligodendrocytes. We discuss the specialized features of OPCs that position these cells to integrate activity-dependent and molecular cues to shape brain wiring. Finally, we place OPCs within the context of a growing field focused on understanding the importance of communication between neurons and glia in the contexts of both health and disease.


Asunto(s)
Células Precursoras de Oligodendrocitos , Células Precursoras de Oligodendrocitos/fisiología , Diferenciación Celular/fisiología , Neuronas/fisiología , Axones , Oligodendroglía/fisiología , Vaina de Mielina/fisiología
5.
Nat Rev Neurosci ; 24(7): 395, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37147381
7.
Nat Neurosci ; 25(10): 1273-1278, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36171430

RESUMEN

Oligodendrocyte precursor cells (OPCs) give rise to myelinating oligodendrocytes throughout life, but the functions of OPCs are not limited to oligodendrogenesis. Here we show that OPCs contribute to thalamocortical presynapse elimination in the developing and adult mouse visual cortex. OPC-mediated synapse engulfment increases in response to sensory experience during neural circuit refinement. Our data suggest that OPCs may regulate synaptic connectivity in the brain independently of oligodendrogenesis.


Asunto(s)
Células Precursoras de Oligodendrocitos , Animales , Encéfalo , Diferenciación Celular/fisiología , Ratones , Ratones Transgénicos , Células Precursoras de Oligodendrocitos/fisiología , Oligodendroglía/fisiología , Sinapsis
8.
Immunity ; 55(8): 1334-1336, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35947976

RESUMEN

In this issue of Immunity, Bi et al. identify a microglia-neuron signaling axis that is critical for maintaining central control of the sympathetic nervous system. They find that platelet growth factor B released by microglia acts on neurons via PDGFRα to regulate sympathetic outflow. Disrupting this pathway leads to neuronal excitability, highlighting a promising therapeutic target to modulate sympathetic outflow and reduce hypertension.


Asunto(s)
Hipertensión , Sistema Nervioso Simpático , Humanos , Hipertensión/metabolismo , Neuronas/fisiología , Sistema Nervioso Simpático/metabolismo
9.
Front Immunol ; 12: 703527, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276699

RESUMEN

Intercellular signaling molecules such as cytokines and their receptors enable immune cells to communicate with one another and their surrounding microenvironments. Emerging evidence suggests that the same signaling pathways that regulate inflammatory responses to injury and disease outside of the brain also play powerful roles in brain development, plasticity, and function. These observations raise the question of how the same signaling molecules can play such distinct roles in peripheral tissues compared to the central nervous system, a system previously thought to be largely protected from inflammatory signaling. Here, we review evidence that the specialized roles of immune signaling molecules such as cytokines in the brain are to a large extent shaped by neural activity, a key feature of the brain that reflects active communication between neurons at synapses. We discuss the known mechanisms through which microglia, the resident immune cells of the brain, respond to increases and decreases in activity by engaging classical inflammatory signaling cascades to assemble, remodel, and eliminate synapses across the lifespan. We integrate evidence from (1) in vivo imaging studies of microglia-neuron interactions, (2) developmental studies across multiple neural circuits, and (3) molecular studies of activity-dependent gene expression in microglia and neurons to highlight the specific roles of activity in defining immune pathway function in the brain. Given that the repurposing of signaling pathways across different tissues may be an important evolutionary strategy to overcome the limited size of the genome, understanding how cytokine function is established and maintained in the brain could lead to key insights into neurological health and disease.


Asunto(s)
Encéfalo/inmunología , Citocinas/inmunología , Microglía/inmunología , Neurogénesis/inmunología , Transducción de Señal/inmunología , Sinapsis/inmunología , Humanos , Plasticidad Neuronal/inmunología
10.
Neuron ; 108(3): 451-468.e9, 2020 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-32931754

RESUMEN

Sensory experience remodels neural circuits in the early postnatal brain through mechanisms that remain to be elucidated. Applying a new method of ultrastructural analysis to the retinogeniculate circuit, we find that visual experience alters the number and structure of synapses between the retina and the thalamus. These changes require vision-dependent transcription of the receptor Fn14 in thalamic relay neurons and the induction of its ligand TWEAK in microglia. Fn14 functions to increase the number of bulbous spine-associated synapses at retinogeniculate connections, likely contributing to the strengthening of the circuit that occurs in response to visual experience. However, at retinogeniculate connections near TWEAK-expressing microglia, TWEAK signals via Fn14 to restrict the number of bulbous spines on relay neurons, leading to the elimination of a subset of connections. Thus, TWEAK and Fn14 represent an intercellular signaling axis through which microglia shape retinogeniculate connectivity in response to sensory experience.


Asunto(s)
Microglía/fisiología , Microglía/ultraestructura , Plasticidad Neuronal/fisiología , Sinapsis/fisiología , Sinapsis/ultraestructura , Animales , Citocina TWEAK/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Electrónica de Transmisión/métodos , Neuronas/metabolismo , Neuronas/ultraestructura , Estimulación Luminosa , Receptor de TWEAK/metabolismo , Vías Visuales/fisiología , Vías Visuales/ultraestructura
11.
Nat Neurosci ; 22(7): 1075-1088, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209379

RESUMEN

Microglia rapidly respond to changes in neural activity and inflammation to regulate synaptic connectivity. The extracellular signals, particularly neuron-derived molecules, that drive these microglial functions at synapses remain a key open question. Here we show that whisker lesioning, known to dampen cortical activity, induces microglia-mediated synapse elimination. This synapse elimination is dependent on signaling by CX3CR1, the receptor for microglial fractalkine (also known as CXCL1), but not complement receptor 3. Furthermore, mice deficient in CX3CL1 have profound defects in synapse elimination. Single-cell RNA sequencing revealed that Cx3cl1 is derived from cortical neurons, and ADAM10, a metalloprotease that cleaves CX3CL1 into a secreted form, is upregulated specifically in layer IV neurons and in microglia following whisker lesioning. Finally, inhibition of ADAM10 phenocopies Cx3cr1-/- and Cx3cl1-/- synapse elimination defects. Together, these results identify neuron-to-microglia signaling necessary for cortical synaptic remodeling and reveal that context-dependent immune mechanisms are utilized to remodel synapses in the mammalian brain.


Asunto(s)
Proteína ADAM10/fisiología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Receptor 1 de Quimiocinas CX3C/fisiología , Quimiocina CX3CL1/fisiología , Proteínas de la Membrana/fisiología , Microglía/fisiología , Corteza Sensoriomotora/fisiopatología , Tacto/fisiología , Vibrisas/lesiones , Proteína ADAM10/antagonistas & inhibidores , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Receptor 1 de Quimiocinas CX3C/deficiencia , Receptor 1 de Quimiocinas CX3C/genética , Recuento de Células , Femenino , Regulación de la Expresión Génica , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas Analíticas Microfluídicas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Corteza Sensoriomotora/metabolismo , Corteza Sensoriomotora/patología , Transducción de Señal/fisiología , Análisis de la Célula Individual , Transcriptoma , Vibrisas/fisiología
12.
Neuron ; 99(3): 525-539.e10, 2018 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-30033152

RESUMEN

Sensory experience influences the establishment of neural connectivity through molecular mechanisms that remain unclear. Here, we employ single-nucleus RNA sequencing to investigate the contribution of sensory-driven gene expression to synaptic refinement in the dorsal lateral geniculate nucleus of the thalamus, a region of the brain that processes visual information. We find that visual experience induces the expression of the cytokine receptor Fn14 in excitatory thalamocortical neurons. By combining electrophysiological and structural techniques, we show that Fn14 is dispensable for early phases of refinement mediated by spontaneous activity but that Fn14 is essential for refinement during a later, experience-dependent period of development. Refinement deficits in mice lacking Fn14 are associated with functionally weaker and structurally smaller retinogeniculate inputs, indicating that Fn14 mediates both functional and anatomical rearrangements in response to sensory experience. These findings identify Fn14 as a molecular link between sensory-driven gene expression and vision-sensitive refinement in the brain.


Asunto(s)
Cuerpos Geniculados/metabolismo , Células Ganglionares de la Retina/metabolismo , Receptor de TWEAK/biosíntesis , Percepción Visual/fisiología , Animales , Femenino , Expresión Génica , Cuerpos Geniculados/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Tracto Óptico/crecimiento & desarrollo , Tracto Óptico/metabolismo , Retina/metabolismo , Receptor de TWEAK/genética
13.
Nat Neurosci ; 21(7): 1017, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29752482

RESUMEN

In the version of this article initially published, the x-axis labels in Fig. 3c read Vglut, Gad1/2, Aldh1l1 and Pecam1; they should have read Vglut+, Gad1/2+, Aldh1l1+ and Pecam1+. In Fig. 4, the range values were missing from the color scales; they are, from left to right, 4-15, 0-15, 4-15 and 0-15 in Fig. 4a and 4-15, 4-15 and 4-8 in Fig. 4h. In the third paragraph of the main text, the phrase reading "Previous approaches have analyzed a limited number of inhibitory cell types, thus masking the full diversity of excitatory populations" should have read "Previous approaches have analyzed a limited number of inhibitory cell types and masked the full diversity of excitatory populations." In the second paragraph of Results section "Diversity of experience-regulated ERGs," the phrase reading "thus suggesting considerable divergence within the gene expression program responding to early stimuli" should have read "thus suggesting considerable divergence within the early stimulus-responsive gene expression program." In the fourth paragraph of Results section "Excitatory neuronal LRGs," the sentence reading "The anatomical organization of these cell types into sublayers, coupled with divergent transcriptional responses to a sensory stimulus, suggested previously unappreciated functional subdivisions located within the laminae of the mouse visual cortex and resembling the cytoarchitecture in higher mammals" should have read "The anatomical organization of these cell types into sublayers, coupled with divergent transcriptional responses to a sensory stimulus, suggests previously unappreciated functional subdivisions located within the laminae of the mouse visual cortex, resembling the cytoarchitecture in higher mammals." In the last sentence of the Results, "sensory-responsive genes" should have read "sensory-stimulus-responsive genes." The errors have been corrected in the HTML and PDF versions of the article.

14.
Proc Natl Acad Sci U S A ; 115(5): E1051-E1060, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29343640

RESUMEN

Coordinated changes in gene expression underlie the early patterning and cell-type specification of the central nervous system. However, much less is known about how such changes contribute to later stages of circuit assembly and refinement. In this study, we employ single-cell RNA sequencing to develop a detailed, whole-transcriptome resource of gene expression across four time points in the developing dorsal lateral geniculate nucleus (LGN), a visual structure in the brain that undergoes a well-characterized program of postnatal circuit development. This approach identifies markers defining the major LGN cell types, including excitatory relay neurons, oligodendrocytes, astrocytes, microglia, and endothelial cells. Most cell types exhibit significant transcriptional changes across development, dynamically expressing genes involved in distinct processes including retinotopic mapping, synaptogenesis, myelination, and synaptic refinement. Our data suggest that genes associated with synapse and circuit development are expressed in a larger proportion of nonneuronal cell types than previously appreciated. Furthermore, we used this single-cell expression atlas to identify the Prkcd-Cre mouse line as a tool for selective manipulation of relay neurons during a late stage of sensory-driven synaptic refinement. This transcriptomic resource provides a cellular map of gene expression across several cell types of the LGN, and offers insight into the molecular mechanisms of circuit development in the postnatal brain.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Cuerpos Geniculados/embriología , Cuerpos Geniculados/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Transcriptoma , Animales , Axones/fisiología , Encéfalo/embriología , Perfilación de la Expresión Génica , Ratones , Microscopía Electrónica de Rastreo , Neurogénesis , Retina/fisiología , Análisis de Secuencia de ARN , Programas Informáticos , Vías Visuales/fisiología
15.
Nat Neurosci ; 21(1): 120-129, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29230054

RESUMEN

Activity-dependent transcriptional responses shape cortical function. However, a comprehensive understanding of the diversity of these responses across the full range of cortical cell types, and how these changes contribute to neuronal plasticity and disease, is lacking. To investigate the breadth of transcriptional changes that occur across cell types in the mouse visual cortex after exposure to light, we applied high-throughput single-cell RNA sequencing. We identified significant and divergent transcriptional responses to stimulation in each of the 30 cell types characterized, thus revealing 611 stimulus-responsive genes. Excitatory pyramidal neurons exhibited inter- and intralaminar heterogeneity in the induction of stimulus-responsive genes. Non-neuronal cells showed clear transcriptional responses that may regulate experience-dependent changes in neurovascular coupling and myelination. Together, these results reveal the dynamic landscape of the stimulus-dependent transcriptional changes occurring across cell types in the visual cortex; these changes are probably critical for cortical function and may be sites of deregulation in developmental brain disorders.


Asunto(s)
Neuroglía/fisiología , Neuronas/fisiología , Transcripción Genética/fisiología , Transcriptoma/fisiología , Corteza Visual/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación de la Expresión Génica/fisiología , Ontología de Genes , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Inhibición Neural/fisiología , Neuronas/citología , Acoplamiento Neurovascular/fisiología , Estimulación Luminosa , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/fisiología , Análisis de la Célula Individual/métodos , Estadísticas no Paramétricas , Vías Visuales
16.
J Neurosci ; 34(23): 7999-8009, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24899721

RESUMEN

Dendritic arbors are complex neuronal structures that receive and process synaptic inputs. One mechanism regulating dendrite differentiation is Semaphorin/Plexin signaling, specifically through binding of soluble Sema3A to Neuropilin/PlexinA coreceptors. Here we show that the protein Farp1 [FERM, RhoGEF (ARHGEF), and pleckstrin domain protein 1], a Rac1 activator previously identified as a synaptogenic signaling protein, contributes to establishing dendrite tip number and total dendritic branch length in maturing rat neurons and is sufficient to promote dendrite complexity. Aiming to define its upstream partners, our results support that Farp1 interacts with the Neuropilin-1/PlexinA1 complex and colocalizes with PlexinA1 along dendritic shafts. Functionally, Farp1 is required by Sema3A to promote dendritic arborization of hippocampal neurons, and Sema3A regulates dendritic F-actin distribution via Farp1. Unexpectedly, Sema3A also requires neuronal activity to promote dendritic complexity, presumably because silencing neurons leads to a proteasome-dependent reduction of PlexinA1 in dendrites. These results provide new insights into how activity and soluble cues cooperate to refine dendritic morphology through intracellular signaling pathways.


Asunto(s)
Dendritas/fisiología , Neuronas/citología , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Semaforina-3A/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Dendritas/efectos de los fármacos , Antagonistas del GABA/farmacología , Hipocampo/citología , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Neuronas/efectos de los fármacos , Picrotoxina/farmacología , Ratas , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Semaforina-3A/genética , Semaforina-3A/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología , Transfección
17.
J Cell Biol ; 199(6): 985-1001, 2012 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-23209303

RESUMEN

Synaptic adhesion organizes synapses, yet the signaling pathways that drive and integrate synapse development remain incompletely understood. We screened for regulators of these processes by proteomically analyzing synaptic membranes lacking the synaptogenic adhesion molecule SynCAM 1. This identified FERM, Rho/ArhGEF, and Pleckstrin domain protein 1 (Farp1) as strongly reduced in SynCAM 1 knockout mice. Farp1 regulates dendritic filopodial dynamics in immature neurons, indicating roles in synapse formation. Later in development, Farp1 is postsynaptic and its 4.1 protein/ezrin/radixin/moesin (FERM) domain binds SynCAM 1, assembling a synaptic complex. Farp1 increases synapse number and modulates spine morphology, and SynCAM 1 requires Farp1 for promoting spines. In turn, SynCAM 1 loss reduces the ability of Farp1 to elevate spine density. Mechanistically, Farp1 activates the GTPase Rac1 in spines downstream of SynCAM 1 clustering, and promotes F-actin assembly. Farp1 furthermore triggers a retrograde signal regulating active zone composition via SynCAM 1. These results reveal a postsynaptic signaling pathway that engages transsynaptic interactions to coordinate synapse development.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Citoesqueleto/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Sinapsis/fisiología , Actinas/metabolismo , Animales , Molécula 1 de Adhesión Celular , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Espinas Dendríticas/fisiología , Células HEK293 , Hipocampo/citología , Hipocampo/fisiología , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/fisiología , Ratones , Ratones Noqueados , Neurogénesis , Neuronas/citología , Neuronas/fisiología , Neuropéptidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteómica , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
18.
J Biol Chem ; 286(2): 942-51, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21036901

RESUMEN

Cross-talk between Gα(i)- and Gα(q)-linked G-protein-coupled receptors yields synergistic Ca(2+) responses in a variety of cell types. Prior studies have shown that synergistic Ca(2+) responses from macrophage G-protein-coupled receptors are primarily dependent on phospholipase Cß3 (PLCß3), with a possible contribution of PLCß2, whereas signaling through PLCß4 interferes with synergy. We here show that synergy can be induced by the combination of Gßγ and Gα(q) activation of a single PLCß isoform. Synergy was absent in macrophages lacking both PLCß2 and PLCß3, but it was fully reconstituted following transduction with PLCß3 alone. Mechanisms of PLCß-mediated synergy were further explored in NIH-3T3 cells, which express little if any PLCß2. RNAi-mediated knockdown of endogenous PLCßs demonstrated that synergy in these cells was dependent on PLCß3, but PLCß1 and PLCß4 did not contribute, and overexpression of either isoform inhibited Ca(2+) synergy. When synergy was blocked by RNAi of endogenous PLCß3, it could be reconstituted by expression of either human PLCß3 or mouse PLCß2. In contrast, it could not be reconstituted by human PLCß3 with a mutation of the Y box, which disrupted activation by Gßγ, and it was only partially restored by human PLCß3 with a mutation of the C terminus, which partly disrupted activation by Gα(q). Thus, both Gßγ and Gα(q) contribute to activation of PLCß3 in cells for Ca(2+) synergy. We conclude that Ca(2+) synergy between Gα(i)-coupled and Gα(q)-coupled receptors requires the direct action of both Gßγ and Gα(q) on PLCß and is mediated primarily by PLCß3, although PLCß2 is also competent.


Asunto(s)
Señalización del Calcio/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Fosfolipasa C beta/metabolismo , Animales , Complemento C5a/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Mutagénesis , Células 3T3 NIH , Fosfolipasa C beta/genética , ARN Interferente Pequeño , Receptores Purinérgicos P2/metabolismo , Uridina Difosfato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...