Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37254876

RESUMEN

RAS/MAPK gene dysfunction underlies various cancers and neurocognitive disorders. Although the roles of RAS/MAPK genes have been well studied in cancer, less is known about their function during neurodevelopment. There are many genes that work in concert to regulate RAS/MAPK signaling, suggesting that if common brain phenotypes could be discovered they could have a broad impact on the many other disorders caused by distinct RAS/MAPK genes. We assessed the cellular and molecular consequences of hyperactivating the RAS/MAPK pathway using two distinct genes in a cell type previously implicated in RAS/MAPK-mediated cognitive changes, cortical GABAergic interneurons. We uncovered some GABAergic core programs that are commonly altered in each of the mutants. Notably, hyperactive RAS/MAPK mutants bias developing cortical interneurons towards those that are somatostatin positive. The increase in somatostatin-positive interneurons could also be prevented by pharmacological inhibition of the core RAS/MAPK signaling pathway. Overall, these findings present new insights into how different RAS/MAPK mutations can converge on GABAergic interneurons, which may be important for other RAS/MAPK genes and related disorders.


Asunto(s)
Transducción de Señal , Somatostatina , Alelos , Somatostatina/genética , Somatostatina/metabolismo , Transducción de Señal/genética , Sistema de Señalización de MAP Quinasas/genética , Interneuronas/metabolismo , Neuronas GABAérgicas/metabolismo
2.
Development ; 147(14)2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32586977

RESUMEN

Cortical interneurons (CINs) originate in the ganglionic eminences (GEs) and migrate tangentially to the cortex guided by different attractive and repulsive cues. Once inside the cortex, the cellular and molecular mechanisms determining the migration of CINs along the rostrocaudal axis are less well understood. Here, we investigated the cortical distribution of CINs originating in the medial and caudal GEs at different time points. Using molecular and genetic labeling, we showed that, in the mouse, early- and late-born CINs (E12 versus E15) are differentially distributed along the rostrocaudal axis. Specifically, late-born CINs are preferentially enriched in cortical areas closer to their respective sites of origin in the medial or caudal GE. Surprisingly, our in vitro experiments failed to show a preferential migration pattern along the rostrocaudal axis for medial- or caudal-born CINs. Moreover, in utero transplantation experiments suggested that the rostrocaudal dispersion of CINs depends on the developmental stage of the host brain and is limited by the migration time and the increasing size of the developing brain. These data suggest that the embryonic expansion of the cortex contributes to the rostrocaudal distribution of CINs.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Corteza Cerebral/citología , Neuronas GABAérgicas/citología , Animales , Encéfalo/embriología , Encéfalo/fisiología , Linaje de la Célula , Movimiento Celular , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Neuronas GABAérgicas/metabolismo , Ratones , Ratones Noqueados , Tamaño de los Órganos , Corteza Somatosensorial/citología , Corteza Somatosensorial/embriología , Corteza Somatosensorial/metabolismo , Factor Nuclear Tiroideo 1/deficiencia , Factor Nuclear Tiroideo 1/genética , Factor Nuclear Tiroideo 1/metabolismo , Ácido gamma-Aminobutírico/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(45): 28384-28392, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33122441

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL). The main proliferating component of KS tumors is a cell of endothelial origin termed the spindle cell. Spindle cells are predominantly latently infected with only a small percentage of cells undergoing viral replication. As there is no direct treatment for latent KSHV, identification of host vulnerabilities in latently infected endothelial cells could be exploited to inhibit KSHV-associated tumor cells. Using a pooled CRISPR-Cas9 lentivirus library, we identified host factors that are essential for the survival or proliferation of latently infected endothelial cells in culture, but not their uninfected counterparts. Among the many host genes identified, there was an enrichment in genes localizing to the mitochondria, including genes involved in mitochondrial translation. Antibiotics that inhibit bacterial and mitochondrial translation specifically inhibited the expansion of latently infected endothelial cells and led to increased cell death in patient-derived PEL cell lines. Direct inhibition of mitochondrial respiration or ablation of mitochondrial genomes leads to increased death in latently infected cells. KSHV latent infection decreases mitochondrial numbers, but there are increases in mitochondrial size, genome copy number, and transcript levels. We found that multiple gene products of the latent locus localize to the mitochondria. During latent infection, KSHV significantly alters mitochondrial biology, leading to enhanced sensitivity to inhibition of mitochondrial respiration, which provides a potential therapeutic avenue for KSHV-associated cancers.


Asunto(s)
Sistemas CRISPR-Cas , Infecciones por Herpesviridae/genética , Herpesvirus Humano 8/genética , Mitocondrias/metabolismo , Latencia del Virus/genética , Línea Celular , Proliferación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Células Endoteliales/metabolismo , Herpesvirus Humano 8/fisiología , Humanos , Linfoma de Efusión Primaria/genética , Sarcoma de Kaposi , Replicación Viral
4.
Proc Natl Acad Sci U S A ; 117(11): 6189-6195, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32123116

RESUMEN

Neurofibromatosis 1 (NF1) is caused by mutations in the NF1 gene, which encodes the protein, neurofibromin, an inhibitor of Ras activity. Cortical GABAergic interneurons (CINs) are implicated in NF1 pathology, but the cellular and molecular changes to CINs are unknown. We deleted mouse Nf1 from the medial ganglionic eminence, which gives rise to both oligodendrocytes and CINs that express somatostatin and parvalbumin. Nf1 loss led to a persistence of immature oligodendrocytes that prevented later-generated oligodendrocytes from occupying the cortex. Moreover, molecular and cellular properties of parvalbumin (PV)-positive CINs were altered by the loss of Nf1, without changes in somatostatin (SST)-positive CINs. We discovered that loss of Nf1 results in a dose-dependent decrease in Lhx6 expression, the transcription factor necessary to establish SST+ and PV+ CINs, which was rescued by the MEK inhibitor SL327, revealing a mechanism whereby a neurofibromin/Ras/MEK pathway regulates a critical CIN developmental milestone.


Asunto(s)
Corteza Cerebral/patología , Neuronas GABAérgicas/patología , Interneuronas/patología , Proteínas con Homeodominio LIM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Factores de Transcripción/metabolismo , Aminoacetonitrilo/administración & dosificación , Aminoacetonitrilo/análogos & derivados , Animales , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Neuronas GABAérgicas/metabolismo , Humanos , Interneuronas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Eminencia Media/citología , Ratones , Ratones Noqueados , Neurofibromatosis 1/genética , Neurofibromina 1/metabolismo , Neuroglía/citología , Parvalbúminas/metabolismo , Cultivo Primario de Células , Somatostatina/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo
5.
PLoS Pathog ; 16(6): e1008634, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32555637

RESUMEN

Kaposi's Sarcoma Herpesvirus (KSHV) is present in the main tumor cells of Kaposi's Sarcoma (KS), the spindle cells, which are of endothelial origin. KSHV is also associated with two B-cell lymphomas, Primary Effusion Lymphoma (PEL) and Multicentric Castleman's Disease. In KS and PEL, KSHV is primarily latent in the infected cells, expressing only a few genes. Although KSHV infection is required for KS and PEL, it is unclear how latent gene expression contributes to their formation. Proliferation of cancer cells occurs despite multiple checkpoints intended to prevent dysregulated cell growth. The first of these checkpoints, caused by shortening of telomeres, results in replicative senescence, where cells are metabolically active, but no longer divide. We found that human dermal lymphatic endothelial cells (LECs) are more susceptible to KSHV infection than their blood-specific endothelial cell counterparts and maintain KSHV latency to higher levels during passage. Importantly, KSHV infection of human LECs but not human BECs promotes their continued proliferation beyond this first checkpoint of replicative senescence. The latently expressed viral cyclin homolog is essential for KSHV-induced bypass of senescence in LECs. These data suggest that LECs may be an important reservoir for KSHV infection and may play a role during KS tumor development and that the viral cyclin is a critical oncogene for this process.


Asunto(s)
Senescencia Celular , Ciclinas/metabolismo , Células Endoteliales/metabolismo , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas Virales/metabolismo , Ciclinas/genética , Células Endoteliales/patología , Células Endoteliales/virología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/patología , Herpesvirus Humano 8/genética , Humanos , Proteínas Virales/genética
6.
Am J Med Genet A ; 188(2): 556-568, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34726335

RESUMEN

Predicting genotype-to-phenotype correlations from genomic variants has been challenging, particularly for genes that have a complex balance of dominant and recessive inheritance for phenotypes. Variants in NMDA receptor components GRIN1, GRIN2A, and GRIN2B cause a myriad of dominant disease phenotypes, with the most common being epilepsy and autism spectrum disorder. Starting from the analysis of a variant of uncertain significance (VUS, GRIN2A G760S), we realized the need for tools to map dominant variants for the components of the NMDA receptor. Some variants within GRIN1, GRIN2A, and GRIN2B exert dominant epilepsy and developmental delay, yet other amino acid variants are conserved and predicted to alter protein function but do not have dominant phenotypes. Common variant annotation tools are not powered to determine pathogenic dominant outcomes. To address this gap, we integrated sequence and structural analyses for GRIN1, GRIN2A, and GRIN2B. Using this approach, we determined that paralog homology mapping and topology can segregate dominant variants, with an elevation of intermolecular contacts between the subunits. Furthermore, demonstrating the general utility of our methodology, we show that 25 VUS within ClinVar also reach a dominant variant annotation, including the GRIN2A G760S variant. Our work suggests paralog homology and protein topology as a powerful strategy within the receptor complex to resolve dominant genetic variants relative to variants that would fit a recessive inheritance, requiring two damaging variants. These strategies should be tested in additional dominant genetic disorders to determine the broader utility.


Asunto(s)
Trastorno del Espectro Autista , Epilepsia , Epilepsia/genética , Humanos , N-Metilaspartato/genética , Fenotipo , Receptores de N-Metil-D-Aspartato/genética
7.
J Neurosci ; 40(11): 2215-2227, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-31988060

RESUMEN

Manipulations that enhance GABAergic inhibition have been associated with improved behavioral phenotypes in autism models, suggesting that autism may be treated by correcting underlying deficits of inhibition. Interneuron transplantation is a method for increasing recipient synaptic inhibition, and it has been considered a prospective therapy for conditions marked by deficient inhibition, including neuropsychiatric disorders. It is unknown, however, whether interneuron transplantation may be therapeutically effective only for conditions marked by reduced inhibition, and it is also unclear whether transplantation improves behavioral phenotypes solely by normalizing underlying circuit defects. To address these questions, we studied the effects of interneuron transplantation in male and female mice lacking the autism-associated gene, Pten, in GABAergic interneurons. Pten mutant mice exhibit social behavior deficits, elevated synaptic inhibition in prefrontal cortex, abnormal baseline and social interaction-evoked electroencephalogram (EEG) signals, and an altered composition of cortical interneuron subtypes. Transplantation of wild-type embryonic interneurons from the medial ganglionic eminence into the prefrontal cortex of neonatal Pten mutants rescued social behavior despite exacerbating excessive levels of synaptic inhibition. Furthermore, transplantation did not normalize recipient EEG signals measured during baseline states. Interneuron transplantation can thus correct behavioral deficits even when those deficits are associated with elevated synaptic inhibition. Moreover, transplantation does not exert therapeutic effects solely by restoring wild-type circuit states. Our findings indicate that interneuron transplantation could offer a novel cell-based approach to autism treatment while challenging assumptions that effective therapies must reverse underlying circuit defects.SIGNIFICANCE STATEMENT Imbalances between neural excitation and inhibition are hypothesized to contribute to the pathophysiology of autism. Interneuron transplantation is a method for altering recipient inhibition, and it has been considered a prospective therapy for neuropsychiatric disorders, including autism. Here we examined the behavioral and physiological effects of interneuron transplantation in a mouse genetic model of autism. They demonstrate that transplantation rescues recipient social interaction deficits without correcting a common measure of recipient inhibition, or circuit-level physiological measures. These findings demonstrate that interneuron transplantation can exert therapeutic behavioral effects without necessarily restoring wild-type circuit states, while highlighting the potential of interneuron transplantation as an autism therapy.


Asunto(s)
Trastorno Autístico/cirugía , Trasplante de Tejido Encefálico , Trasplante de Tejido Fetal , Neuronas GABAérgicas/fisiología , Interneuronas/trasplante , Inhibición Neural/fisiología , Fosfohidrolasa PTEN/deficiencia , Conducta Social , Animales , Trastorno Autístico/fisiopatología , Trastorno Autístico/psicología , Modelos Animales de Enfermedad , Electroencefalografía , Conducta Exploratoria , Femenino , Masculino , Aprendizaje por Laberinto , Eminencia Media/citología , Eminencia Media/embriología , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/fisiología , Técnicas de Placa-Clamp , Fenotipo , Corteza Prefrontal/fisiopatología , Distribución Aleatoria , Sinapsis/fisiología
8.
J Neurosci ; 39(1): 177-192, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30377227

RESUMEN

The CCCTC-binding factor (CTCF) is a central regulator of chromatin topology recently linked to neurodevelopmental disorders such as intellectual disability, autism, and schizophrenia. The aim of this study was to identify novel roles of CTCF in the developing mouse brain. We provide evidence that CTCF is required for the expression of the LIM homeodomain factor LHX6 involved in fate determination of cortical interneurons (CINs) that originate in the medial ganglionic eminence (MGE). Conditional Ctcf ablation in the MGE of mice of either sex leads to delayed tangential migration, abnormal distribution of CIN in the neocortex, a marked reduction of CINs expressing parvalbumin and somatostatin (Sst), and an increased number of MGE-derived cells expressing Lhx8 and other markers of basal forebrain projection neurons. Likewise, Ctcf-null MGE cells transplanted into the cortex of wild-type hosts generate fewer Sst-expressing CINs and exhibit lamination defects that are efficiently rescued upon reexpression of LHX6. Collectively, these data indicate that CTCF regulates the dichotomy between Lhx6 and Lhx8 to achieve correct specification and migration of MGE-derived CINs.SIGNIFICANCE STATEMENT This work provides evidence that CCCTC-binding factor (CTCF) controls an early fate decision point in the generation of cortical interneurons mediated at least in part by Lhx6. Importantly, the abnormalities described could reflect early molecular and cellular events that contribute to human neurological disorders previously linked to CTCF, including schizophrenia, autism, and intellectual disability.


Asunto(s)
Factor de Unión a CCCTC/fisiología , Corteza Cerebral/fisiología , Interneuronas/fisiología , Eminencia Media/fisiología , Animales , Factor de Unión a CCCTC/genética , Recuento de Células , Movimiento Celular/genética , Movimiento Celular/fisiología , Corteza Cerebral/citología , Femenino , Proteínas con Homeodominio LIM/biosíntesis , Proteínas con Homeodominio LIM/genética , Masculino , Eminencia Media/citología , Ratones , Ratones Endogámicos C57BL , Neocórtex/citología , Neocórtex/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Parvalbúminas/metabolismo , Somatostatina/metabolismo , Telencéfalo/citología , Telencéfalo/crecimiento & desarrollo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Ácido gamma-Aminobutírico/fisiología
9.
Development ; 144(21): 3867-3878, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29089360

RESUMEN

Cortical interneurons are a diverse group of neurons that project locally and are crucial for regulating information processing and flow throughout the cortex. Recent studies in mice have advanced our understanding of how these neurons are specified, migrate and mature. Here, we evaluate new findings that provide insights into the development of cortical interneurons and that shed light on when their fate is determined, on the influence that regional domains have on their development, and on the role that key transcription factors and other crucial regulatory genes play in these events. We focus on cortical interneurons that are derived from the medial ganglionic eminence, as most studies have examined this interneuron population. We also assess how these data inform our understanding of neuropsychiatric disease and discuss the potential role of cortical interneurons in cell-based therapies.


Asunto(s)
Corteza Cerebral/citología , Interneuronas/citología , Animales , Linaje de la Célula , Modelos Biológicos , Factores de Tiempo , Transcripción Genética
10.
Development ; 144(15): 2837-2851, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28694260

RESUMEN

Distinct cortical interneuron (CIN) subtypes have unique circuit functions; dysfunction in specific subtypes is implicated in neuropsychiatric disorders. Somatostatin- and parvalbumin-expressing (SST+ and PV+) interneurons are the two major subtypes generated by medial ganglionic eminence (MGE) progenitors. Spatial and temporal mechanisms governing their cell-fate specification and differential integration into cortical layers are largely unknown. We provide evidence that Coup-TF1 and Coup-TF2 (Nr2f1 and Nr2f2) transcription factor expression in an arc-shaped progenitor domain within the MGE promotes time-dependent survival of this neuroepithelium and the time-dependent specification of layer V SST+ CINs. Coup-TF1 and Coup-TF2 autonomously repress PV+ fate in MGE progenitors, in part through directly driving Sox6 expression. These results have identified, in mouse, a transcriptional pathway that controls SST-PV fate.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Factor de Transcripción COUP I/metabolismo , Interneuronas/metabolismo , Neocórtex/citología , Animales , Factor de Transcripción COUP I/genética , Factor de Transcripción COUP II/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Parvalbúminas/genética , Parvalbúminas/metabolismo , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
11.
Dev Neurosci ; 42(5-6): 195-207, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33657557

RESUMEN

Autism spectrum disorder (ASD) manifests early in childhood. While genetic variants increase risk for ASD, a growing body of literature has established that in utero chemical exposures also contribute to ASD risk. These chemicals include air-based pollutants like diesel particulate matter (DPM). A combination of single-cell and direct transcriptomics of DPM-exposed human-induced pluripotent stem cell-derived cerebral organoids revealed toxicogenomic effects of DPM exposure during fetal brain development. Direct transcriptomics, sequencing RNA bases via Nanopore, revealed that cerebral organoids contain extensive RNA modifications, with DPM-altering cytosine methylation in oxidative mitochondrial transcripts expressed in outer radial glia cells. Single-cell transcriptomics further confirmed an oxidative phosphorylation change in cell groups such as outer radial glia upon DPM exposure. This approach highlights how DPM exposure perturbs normal mitochondrial function and cellular respiration during early brain development, which may contribute to developmental disorders like ASD by altering neurodevelopment.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Material Particulado/toxicidad , Células Madre Pluripotentes/efectos de los fármacos , Emisiones de Vehículos/toxicidad , Trastorno del Espectro Autista/etiología , Encéfalo/efectos de los fármacos , Femenino , Humanos , Exposición Materna/efectos adversos , Organoides , Análisis de Secuencia de ARN
12.
Proc Natl Acad Sci U S A ; 114(50): 13132-13137, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29180416

RESUMEN

Artificial muscles hold promise for safe and powerful actuation for myriad common machines and robots. However, the design, fabrication, and implementation of artificial muscles are often limited by their material costs, operating principle, scalability, and single-degree-of-freedom contractile actuation motions. Here we propose an architecture for fluid-driven origami-inspired artificial muscles. This concept requires only a compressible skeleton, a flexible skin, and a fluid medium. A mechanical model is developed to explain the interaction of the three components. A fabrication method is introduced to rapidly manufacture low-cost artificial muscles using various materials and at multiple scales. The artificial muscles can be programed to achieve multiaxial motions including contraction, bending, and torsion. These motions can be aggregated into systems with multiple degrees of freedom, which are able to produce controllable motions at different rates. Our artificial muscles can be driven by fluids at negative pressures (relative to ambient). This feature makes actuation safer than most other fluidic artificial muscles that operate with positive pressures. Experiments reveal that these muscles can contract over 90% of their initial lengths, generate stresses of ∼600 kPa, and produce peak power densities over 2 kW/kg-all equal to, or in excess of, natural muscle. This architecture for artificial muscles opens the door to rapid design and low-cost fabrication of actuation systems for numerous applications at multiple scales, ranging from miniature medical devices to wearable robotic exoskeletons to large deployable structures for space exploration.


Asunto(s)
Músculo Esquelético/fisiología , Papel , Robótica/métodos , Animales , Miembros Artificiales , Fenómenos Biomecánicos , Biomimética/economía , Biomimética/métodos , Humanos , Hidrodinámica , Robótica/economía
13.
Adv Funct Mater ; 29(7)2019 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-31372108

RESUMEN

Fluidic soft sensors have been widely used in wearable devices for human motion capturing. However, thus far, the biocompatibility of the conductive liquid, the linearity of the sensing signal, and the hysteresis between the loading and release processes have limited the sensing quality as well as the applications of these sensors. In this paper, silicone based strain and force sensors composed of a novel biocompatible conductive liquid (potassium iodide and glycerol solution) are introduced. The strain sensors exhibit negligible hysteresis up to 5 Hz, with a gauge factor of 2.2 at 1 Hz. The force sensors feature a novel multi-functional layered structure, with micro-cylinder-filled channels to achieve high linearity, low hysteresis (5.3% hysteresis at 1 Hz), and good sensitivity (100% resistance increase at a 5 N load). The sensors' gauge factors are stable at various temperatures and humidity levels. These bio-compatible, low hysteresis, and high linearity sensors are promising for safe and reliable diagnostic devices, wearable motion capture, and compliant human-computer interfaces.

14.
Cereb Cortex ; 28(11): 3868-3879, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29028946

RESUMEN

Human mutations in CNTNAP2 are associated with an array of neuropsychiatric and neurological syndromes, including speech and language disorders, epilepsy, and autism spectrum disorder (ASD). We examined Cntnap2's expression and function in GABAergic cortical interneurons (CINs), where its RNA is present at highest levels in chandelier neurons, PV+ neurons and VIP+ neurons. In vivo functions were studied using both constitutive Cntnap2 null mice and a transplantation assay, the latter to assess cell autonomous phenotypes of medial ganglionic eminence (MGE)-derived CINs. We found that Cntnap2 constitutive null mutants had normal numbers of MGE-derived CINs, but had reduced PV+ CINs. Transplantation assays showed that Cntnap2 cell autonomously regulated the physiology of parvalbumin (PV)+, fast-spiking CINs; no phenotypes were observed in somatostatin+, regular spiking, CINs. We also tested the effects of 4 human CNTNAP2 ASD missense mutations in vivo, and found that they impaired PV+ CIN development. Together, these data reveal that reduced CNTNAP2 function impairs PV+ CINs, a cell type with important roles in regulating cortical circuits.


Asunto(s)
Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Alelos , Animales , Trastorno del Espectro Autista , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Missense , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Parvalbúminas/metabolismo , Proteína Reelina , Serina Endopeptidasas/metabolismo , Corteza Somatosensorial/fisiología , Telencéfalo/crecimiento & desarrollo
15.
Cereb Cortex ; 28(11): 3797-3815, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29028947

RESUMEN

The postnatal functions of the Dlx1&2 transcription factors in cortical interneurons (CINs) are unknown. Here, using conditional Dlx1, Dlx2, and Dlx1&2 knockouts (CKOs), we defined their roles in specific CINs. The CKOs had dendritic, synaptic, and survival defects, affecting even PV+ CINs. We provide evidence that DLX2 directly drives Gad1, Gad2, and Vgat expression, and show that mutants had reduced mIPSC amplitude. In addition, the mutants formed fewer GABAergic synapses on excitatory neurons and had reduced mIPSC frequency. Furthermore, Dlx1/2 CKO had hypoplastic dendrites, fewer excitatory synapses, and reduced excitatory input. We provide evidence that some of these phenotypes were due to reduced expression of GRIN2B (a subunit of the NMDA receptor), a high confidence Autism gene. Thus, Dlx1&2 coordinate key components of CIN postnatal development by promoting their excitability, inhibitory output, and survival.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Neuronas GABAérgicas/fisiología , Proteínas de Homeodominio/fisiología , Interneuronas/fisiología , Sinapsis/fisiología , Factores de Transcripción/fisiología , Ácido gamma-Aminobutírico/biosíntesis , Animales , Corteza Cerebral/citología , Femenino , Neuronas GABAérgicas/citología , Regulación del Desarrollo de la Expresión Génica , Glutamato Descarboxilasa/metabolismo , Proteínas de Homeodominio/genética , Interneuronas/citología , Masculino , Ratones Noqueados , Potenciales Postsinápticos Miniatura , Factores de Transcripción/genética , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo
16.
Cereb Cortex ; 27(9): 4303-4313, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27497284

RESUMEN

Prenatally, the cytokine CXCL12 regulates cortical interneuron migration, whereas its postnatal functions are poorly understood. Here, we report that CXCL12 is expressed postnatally in layer V pyramidal neurons and localizes on their cell bodies in the medial prefrontal cortex (mPFC), while its receptors CXCR4/CXCR7 localize to the axon terminals of parvalbumin (PV) interneurons. Conditionally eliminating CXCL12 in neonatal layer V pyramidal neurons led to decreased axon targeting and reduced inhibitory perisomatic synapses from PV+ basket interneurons onto layer V pyramidal neurons. Consequently, the mPFC of Cxcl12 conditional mutants displayed attenuated inhibitory postsynaptic currents onto layer V pyramidal neurons. Thus, postnatal CXCL12 signaling promotes a specific interneuron circuit that inhibits mPFC activity.


Asunto(s)
Quimiocina CXCL12/metabolismo , Interneuronas/metabolismo , Corteza Prefrontal/metabolismo , Sinapsis/fisiología , Animales , Axones/metabolismo , Quimiocina CXCL12/genética , Potenciales Postsinápticos Inhibidores/fisiología , Ratones Transgénicos , Parvalbúminas/metabolismo , Células Piramidales/fisiología , Receptores CXCR4/metabolismo , Ácido gamma-Aminobutírico/metabolismo
17.
Hum Mol Genet ; 24(17): 5024-39, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26071365

RESUMEN

Cleft palate is a common birth defect in humans. Therefore, understanding the molecular genetics of palate development is important from both scientific and medical perspectives. Lhx6 and Lhx8 encode LIM homeodomain transcription factors, and inactivation of both genes in mice resulted in profound craniofacial defects including cleft secondary palate. The initial outgrowth of the palate was severely impaired in the mutant embryos, due to decreased cell proliferation. Through genome-wide transcriptional profiling, we discovered that p57(Kip2) (Cdkn1c), encoding a cell cycle inhibitor, was up-regulated in the prospective palate of Lhx6(-/-);Lhx8(-/-) mutants. p57(Kip2) has been linked to Beckwith-Wiedemann syndrome and IMAGe syndrome in humans, which are developmental disorders with increased incidents of palate defects among the patients. To determine the molecular mechanism underlying the regulation of p57(Kip2) by the Lhx genes, we combined chromatin immunoprecipitation, in silico search for transcription factor-binding motifs, and in vitro reporter assays with putative cis-regulatory elements. The results of these experiments indicated that LHX6 and LHX8 regulated p57(Kip2) via both direct and indirect mechanisms, with the latter mediated by Forkhead box (FOX) family transcription factors. Together, our findings uncovered a novel connection between the initiation of palate development and a cell cycle inhibitor via LHX. We propose a model in which Lhx6 and Lhx8 negatively regulate p57(Kip2) expression in the prospective palate area to allow adequate levels of cell proliferation and thereby promote normal palate development. This is the first report elucidating a molecular genetic pathway downstream of Lhx in palate development.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas con Homeodominio LIM/genética , Proteínas del Tejido Nervioso/genética , Hueso Paladar/embriología , Hueso Paladar/metabolismo , Factores de Transcripción/genética , Animales , Proliferación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Proteínas con Homeodominio LIM/metabolismo , Maxilar/embriología , Maxilar/metabolismo , Ratones , Mutación , Proteínas del Tejido Nervioso/metabolismo , Organogénesis/genética , Hueso Paladar/patología , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional
18.
J Neurosci ; 34(35): 11519-25, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25164650

RESUMEN

GABAergic projections from the neocortex to subcortical structures have been poorly characterized. Using Dlxi12b-Cre mice, we found anatomical evidence for GABAergic neurons that project from the mouse medial prefrontal cortex (mPFC) to multiple subcortical targets. We used a combination of patch-clamp electrophysiology, optogenetics, and pharmacology to confirm that Dlxi12b-labeled projections from the mPFC to the nucleus accumbens (NAcc) release GABA and do not corelease glutamate. Furthermore, optogenetic stimulation of these GABAergic projections from mPFC to NAcc induces avoidance behavior in a real-time place preference task, suggesting that these long-range projecting GABAergic neurons can transmit aversive signals. Finally, we found evidence for heterogeneous histochemical and/or electrophysiological properties of long-range projecting GABAergic neurons in the mPFC. Some of these neurons were labeled in parvalbumin-Cre and vasoactive intestinal peptide-Cre mice. We also used a novel intersectional targeting strategy to label GABAergic neurons in the mPFC that project to NAcc and found that these neurons have fast-spiking properties and express parvalbumin. These results define possible functions and properties for a class of long-range projecting GABAergic neurons in the neocortex.


Asunto(s)
Reacción de Prevención/fisiología , Neuronas GABAérgicas/citología , Vías Nerviosas/citología , Núcleo Accumbens/citología , Corteza Prefrontal/citología , Animales , Conducta Animal/fisiología , Femenino , Masculino , Ratones , Vías Nerviosas/fisiología , Núcleo Accumbens/fisiología , Técnicas de Placa-Clamp , Corteza Prefrontal/fisiología
19.
Dev Biol ; 385(1): 94-106, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24157949

RESUMEN

The progenitor zones of the embryonic mouse ventral telencephalon give rise to GABAergic and cholinergic neurons. We have shown previously that two LIM-homeodomain (LIM-HD) transcription factors, Lhx6 and Lhx8, that are downstream of Nkx2.1, are critical for the development of telencephalic GABAergic and cholinergic neurons. Here we investigate the role of Ldb1, a nuclear protein that binds directly to all LIM-HD factors, in the development of these ventral telencephalon derived neurons. We show that Ldb1 is expressed in the Nkx2.1 cell lineage during embryonic development and in mature neurons. Conditional deletion of Ldb1 causes defects in the expression of a series of genes in the ventral telencephalon and severe impairment in the tangential migration of cortical interneurons from the ventral telencephalon. Similar to the phenotypes observed in Lhx6 or Lhx8 mutant mice, the Ldb1 conditional mutants show a reduction in the number of both GABAergic and cholinergic neurons in the telencephalon. Furthermore, our analysis reveals defects in the development of the parvalbumin-positive neurons in the globus pallidus and striatum of the Ldb1 mutants. These results provide evidence that Ldb1 plays an essential role as a transcription co-regulator of Lhx6 and Lhx8 in the control of mammalian telencephalon development.


Asunto(s)
Neuronas Colinérgicas/metabolismo , Proteínas de Unión al ADN/metabolismo , Neuronas GABAérgicas/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas Nucleares/metabolismo , Telencéfalo/embriología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Globo Pálido/embriología , Proteínas Hedgehog/biosíntesis , Proteínas Hedgehog/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Telencéfalo/crecimiento & desarrollo , Factor Nuclear Tiroideo 1
20.
Elife ; 122024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38536959

RESUMEN

The cell-type-specific expression of ligand/receptor and cell-adhesion molecules is a fundamental mechanism through which neurons regulate connectivity. Here, we determine a functional relevance of the long-established mutually exclusive expression of the receptor tyrosine kinase Kit and the trans-membrane protein Kit Ligand by discrete populations of neurons in the mammalian brain. Kit is enriched in molecular layer interneurons (MLIs) of the cerebellar cortex (i.e., stellate and basket cells), while cerebellar Kit Ligand is selectively expressed by a target of their inhibition, Purkinje cells (PCs). By in vivo genetic manipulation spanning embryonic development through adulthood, we demonstrate that PC Kit Ligand and MLI Kit are required for, and capable of driving changes in, the inhibition of PCs. Collectively, these works in mice demonstrate that the Kit Ligand/Kit receptor dyad sustains mammalian central synapse function and suggest a rationale for the affiliation of Kit mutation with neurodevelopmental disorders.


Asunto(s)
Células de Purkinje , Factor de Células Madre , Ratones , Animales , Células de Purkinje/fisiología , Factor de Células Madre/metabolismo , Cerebelo/fisiología , Corteza Cerebelosa/metabolismo , Interneuronas/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Mamíferos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA