Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 172
Filtrar
1.
J Neurosci ; 41(43): 8876-8886, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34503995

RESUMEN

Cortical parvalbumin-expressing (Pvalb+) neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. This class of inhibitory neurons undergoes extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. While several transcription factors, such as Nkx2-1, Lhx6, and Sox6, are known to be necessary for the differentiation of progenitors into Pvalb+ neurons, which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons' innervation and synaptic function remains largely unknown. Because Sox6 is continuously expressed in Pvalb+ neurons until adulthood, we used conditional knock-out strategies to investigate its putative role in the postnatal maturation and synaptic function of cortical Pvalb+ neurons in mice of both sexes. We found that early postnatal loss of Sox6 in Pvalb+ neurons leads to failure of synaptic bouton growth, whereas later removal in mature Pvalb+ neurons in the adult causes shrinkage of already established synaptic boutons. Paired recordings between Pvalb+ neurons and pyramidal neurons revealed reduced release probability and increased failure rate of Pvalb+ neurons' synaptic output. Furthermore, Pvalb+ neurons lacking Sox6 display reduced expression of full-length tropomyosin-receptor kinase B (TrkB), a key modulator of GABAergic transmission. Once re-expressed in neurons lacking Sox6, TrkB was sufficient to rescue the morphologic synaptic phenotype. Finally, we showed that Sox6 mRNA levels were increased by motor training. Our data thus suggest a constitutive role for Sox6 in the maintenance of synaptic output from Pvalb+ neurons into adulthood.SIGNIFICANCE STATEMENT Cortical parvalbumin-expressing (Pvalb+) inhibitory neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. These inhibitory neurons undergo extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. However, it remains largely unknown which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons. Here, we show that the transcription factor Sox6 cell-autonomously regulates the synaptic maintenance and output of Pvalb+ neurons until adulthood, leaving unaffected other maturational features of this neuronal population.


Asunto(s)
Corteza Cerebral/metabolismo , Neuronas/metabolismo , Parvalbúminas/biosíntesis , Factores de Transcripción SOXD/biosíntesis , Sinapsis/metabolismo , Animales , Animales Recién Nacidos , Corteza Cerebral/citología , Femenino , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Parvalbúminas/genética , Factores de Transcripción SOXD/genética , Sinapsis/genética
2.
Neuroimmunomodulation ; 29(4): 391-401, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35272296

RESUMEN

INTRODUCTION: The prenatal/perinatal exposure to infections may trigger neurodevelopmental alterations that lead to neuropsychiatric disorders such as autism spectrum disorder (ASD). Previous evidence points to long-term behavioral consequences, such as autistic-like behaviors in rodents induced by lipopolysaccharide (LPS) pre- and postnatal (PN) exposure during critical neurodevelopmental periods. Additionally, sex influences the prevalence and symptoms of ASD. Despite this, the mechanisms underlying this influence are poorly understood. We aim to study sex influences in behavioral and neurotrophic/inflammatory alterations triggered by LPS neonatal exposure in juvenile mice at an approximate age of ASD diagnosis in humans. METHODS: Swiss male and female mice on PN days 5 and 7 received a single daily injection of 500 µg/kg LPS from Escherichia coli or sterile saline (control group). We conducted behavioral determinations of locomotor activity, repetitive behavior, anxiety-like behavior, social interaction, and working memory in animals on PN25 (equivalent to 3-5 years old of the human). To determine BDNF levels in the prefrontal cortex and hippocampus, we used animals on PN8 (equivalent to a human term infant) and PN25. In addition, we evaluated iba-1 (microglia marker), TNFα, and parvalbumin expression on PN25. RESULTS: Male juvenile mice presented repetitive behavior, anxiety, and working memory deficits. Females showed social impairment and working memory deficits. In the neurochemical analysis, we detected lower BDNF levels in brain areas of female mice that were more evident in juvenile mice. Only LPS-challenged females presented a marked hippocampal expression of the microglial activation marker, iba-1, and increased TNFα levels, accompanied by a lower parvalbumin expression. DISCUSSION/CONCLUSION: Male and female mice presented distinct behavioral alterations. However, LPS-challenged juvenile females showed the most prominent neurobiological alterations related to autism, such as increased microglial activation and parvalbumin impairment. Since these sex-sensitive alterations seem to be age-dependent, a better understanding of changes induced by the exposure to specific risk factors throughout life represents essential targets for developing strategies for autism prevention and precision therapy.


Asunto(s)
Trastorno del Espectro Autista , Conducta Animal , Animales , Femenino , Masculino , Ratones , Embarazo , Trastorno del Espectro Autista/inmunología , Trastorno del Espectro Autista/fisiopatología , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Lipopolisacáridos/toxicidad , Trastornos de la Memoria/inmunología , Trastornos de la Memoria/fisiopatología , Parvalbúminas/biosíntesis , Factor de Necrosis Tumoral alfa , Enfermedades del Sistema Nervioso/inmunología , Enfermedades del Sistema Nervioso/fisiopatología , Microglía/inmunología , Factores Sexuales , Factores de Edad
3.
Metab Brain Dis ; 36(6): 1267-1275, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33783673

RESUMEN

Infection with the deadly rabies virus (RABV) leads to alteration of cellular gene expression. The RABV, similar to other neurodegenerative diseases may be implicated in neuronal death due to an imbalance in Ca2+ homeostasis. Parvalbumin (PV) and Secretagogin (Scgn), two members of the Calcium-Binding Proteins (CBPs) are useful neuronal markers responsible for calcium regulation and buffering with possible protective roles against infections. This study investigated whether infection with rabies virus causes variance in expression levels of PV and Scgn using the Challenge virus standard (CVS) and Nigerian Street Rabies virus (SRV) strains. Forty-eight, 4-week-old BALB/c mice strains were divided into two test groups and challenged with Rabies virus (RABV) infection and one control group. The presence of RABV antigen was verified by direct fluorescent antibody test (DFAT) and real-time quantitative PCR (qRT-PCR) was used to assess PV and Scgn gene expression. Infection with both virus strains resulted in significant (p < 0.05) increases in expression during early infection. Mid-infection phase caused reduced expression for both genes. However, as infection progressed to the terminal phase, a lower increase in expression was measured. Gene expression and viral load correlation indicated no positive relationship. Neurons with these CBPs may have a greater capacity to buffer calcium and be more resistant to degenerative changes caused by RABV. This implies that, when PV and Scgn expression levels are kept adequately high, the integrity of neurons may be maintained and degeneration caused by RABV infection may be prevented or stopped, hence, these are possible constituents of effective rabies therapy.


Asunto(s)
Encéfalo/metabolismo , Parvalbúminas/biosíntesis , Virus de la Rabia , Rabia/metabolismo , Secretagoginas/biosíntesis , Animales , Femenino , Regulación de la Expresión Génica/genética , Ratones , Ratones Endogámicos BALB C , Parvalbúminas/genética , Rabia/virología , Secretagoginas/genética , Carga Viral
4.
Cereb Cortex ; 28(2): 411-420, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28122807

RESUMEN

An interneuron alteration has been proposed as a source for the modified balance of excitation / inhibition in the cerebral cortex in autism. We previously demonstrated a decreased number of parvalbumin (PV)-expressing interneurons in prefrontal cortex in autism. PV-expressing interneurons include chandelier (Ch) and basket (Bsk) cells. We asked whether the decreased PV+ interneurons affected both Ch cells and Bsk cells in autism. The lack of single markers to specifically label Ch cells or Bsk cells presented an obstacle for addressing this question. We devised a method to discern between PV-Ch and PV-Bsk cells based on the differential expression of Vicia villosa lectin (VVA). VVA binds to N-acetylgalactosamine, that is present in the perineuronal net surrounding some cell types where it plays a role in intercellular communication. N-acetylgalactosamine is present in the perineuronal net surrounding Bsk but not Ch cells. We found that the number of Ch cells is consistently decreased in the prefrontal cortex of autistic (n = 10) when compared with control (n = 10) cases, while the number of Bsk cells is not as severely affected. This finding expand our understanding of GABAergic system functioning in the human cerebral cortex in autism, which will impact translational research directed towards providing better treatment paradigms for individuals with autism.


Asunto(s)
Trastorno Autístico/patología , Neuronas/patología , Corteza Prefrontal/patología , Adolescente , Adulto , Trastorno Autístico/metabolismo , Recuento de Células/métodos , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neuronas/metabolismo , Parvalbúminas/biosíntesis , Corteza Prefrontal/citología , Corteza Prefrontal/metabolismo , Adulto Joven
5.
Cereb Cortex ; 28(10): 3399-3413, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28968898

RESUMEN

The GABAergic system is regulated by the brain-derived neurotrophic factor (BDNF)/Tropomyosin-related kinase B (TrkB) pathway, but the cell-intrinsic role of TrkB signaling in parvalbumin cortical interneuron development and function is unclear. We performed conditional ablation of the TrkB receptor in parvalbumin-expressing (PV) interneurons to study whether postnatal loss of TrkB in parvalbumin cells affects their survival, connectivity, spontaneous and evoked neuronal activity and behavior. Using in vivo recordings of local field potentials, we found reduced gamma oscillations in the sensory cortex of PVcre+; TrkBF/F conditional knockout mice (TrkB cKO), along with increased firing of putative excitatory neurons. There was a significant downregulation in parvalbumin neuron number in cerebral and cerebellar cortices of TrkB cKO mice. In addition, inhibitory synaptic connections between basket cells and pyramidal neurons were profoundly reduced in the neocortex of TrkB cKO mice and there was a loss of cortical volume. TrkB cKO mice also showed profound hyperactivity, stereotypies, motor deficits and learning/memory defects. Our findings demonstrate that the targeting and/or synapse formation of PV-expressing basket cells with principal excitatory neurons require TrkB signaling in parvalbumin cells. Disruption of this signaling has major consequences for parvalbumin interneuron connectivity, network dynamics, cognitive and motor behavior.


Asunto(s)
Conducta Animal , Corteza Cerebral/citología , Corteza Cerebral/fisiopatología , Interneuronas , Glicoproteínas de Membrana/genética , Neuronas , Proteínas Tirosina Quinasas/genética , Animales , Fenómenos Electrofisiológicos/genética , Potenciales Evocados/fisiología , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/psicología , Glicoproteínas de Membrana/deficiencia , Trastornos de la Memoria/genética , Trastornos de la Memoria/psicología , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos del Movimiento/genética , Trastornos del Movimiento/psicología , Neocórtex/citología , Parvalbúminas/biosíntesis , Parvalbúminas/genética , Proteínas Tirosina Quinasas/deficiencia , Células Piramidales , Análisis de Supervivencia
6.
PLoS Genet ; 12(5): e1006035, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27171438

RESUMEN

During postnatal life the cerebral cortex passes through critical periods of plasticity allowing its physiological adaptation to the environment. In the visual cortex, critical period onset and closure are influenced by the non-cell autonomous activity of the Otx2 homeoprotein transcription factor, which regulates the maturation of parvalbumin-expressing inhibitory interneurons (PV cells). In adult mice, the maintenance of a non-plastic adult state requires continuous Otx2 import by PV cells. An important source of extra-cortical Otx2 is the choroid plexus, which secretes Otx2 into the cerebrospinal fluid. Otx2 secretion and internalization requires two small peptidic domains that are part of the DNA-binding domain. Thus, mutating these "transfer" sequences also modifies cell autonomous transcription, precluding this approach to obtain a cell autonomous-only mouse. Here, we develop a mouse model with inducible secretion of an anti-Otx2 single-chain antibody to trap Otx2 in the extracellular milieu. Postnatal secretion of this single-chain antibody by PV cells delays PV maturation and reduces plasticity gene expression. Induced adult expression of this single-chain antibody in cerebrospinal fluid decreases Otx2 internalization by PV cells, strongly induces plasticity gene expression and reopens physiological plasticity. We provide the first mammalian genetic evidence for a signaling mechanism involving intercellular transfer of a homeoprotein transcription factor. Our single-chain antibody mouse model is a valid strategy for extracellular neutralization that could be applied to other homeoproteins and signaling molecules within and beyond the nervous system.


Asunto(s)
Especificidad de Anticuerpos/inmunología , Interneuronas/inmunología , Factores de Transcripción Otx/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Especificidad de Anticuerpos/genética , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Ratones , Plasticidad Neuronal/inmunología , Factores de Transcripción Otx/genética , Parvalbúminas/biosíntesis , Transducción de Señal , Anticuerpos de Cadena Única/genética , Corteza Visual/inmunología , Corteza Visual/metabolismo
7.
Neurobiol Dis ; 112: 24-34, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29337050

RESUMEN

Loss of function mutations in the SCN1A gene, which encodes the voltage-gated sodium channel Nav1.1, have been described in the majority of Dravet syndrome patients presenting with epileptic seizures, hyperactivity, autistic traits, and cognitive decline. We previously reported predominant Nav1.1 expression in parvalbumin-expressing (PV+) inhibitory neurons in juvenile mouse brain and observed epileptic seizures in mice with selective deletion of Scn1a in PV+ cells mediated by PV-Cre transgene expression (Scn1afl/+/PV-Cre-TG). Here we investigate the behavior of Scn1afl/+/PV-Cre-TG mice using a comprehensive battery of behavioral tests. We observed that Scn1afl/+/PV-Cre-TG mice display hyperactive behavior, impaired social novelty recognition, and altered spatial memory. We also generated Scn1afl/+/SST-Cre-KI mice with a selective Scn1a deletion in somatostatin-expressing (SST+) inhibitory neurons using an SST-IRES-Cre knock-in driver line. We observed that Scn1afl/+/SST-Cre-KI mice display no spontaneous convulsive seizures and that Scn1afl/+/SST-Cre-KI mice have a lowered threshold temperature for hyperthermia-induced seizures, although their threshold values are much higher than those of Scn1afl/+/PV-Cre-TG mice. We finally show that Scn1afl/+/SST-Cre-KI mice exhibited no noticeable behavioral abnormalities. These observations suggest that impaired Nav1.1 function in PV+ interneurons is critically involved in the pathogenesis of hyperactivity, autistic traits, and cognitive decline, as well as epileptic seizures, in Dravet syndrome.


Asunto(s)
Conducta Exploratoria/fisiología , Relaciones Interpersonales , Canal de Sodio Activado por Voltaje NAV1.1/genética , Parvalbúminas/biosíntesis , Parvalbúminas/genética , Memoria Espacial/fisiología , Animales , Eliminación de Gen , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.1/deficiencia
8.
J Neurosci Res ; 96(7): 1186-1207, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29314192

RESUMEN

In rodents, the dorsolateral striatum regulates voluntary movement by integrating excitatory inputs from the motor-related cerebral cortex and thalamus to produce contingent inhibitory output to other basal ganglia nuclei. Striatal parvalbumin (PV)-producing interneurons receiving this excitatory input then inhibit medium spiny neurons (MSNs) and modify their outputs. To understand basal ganglia function in motor control, it is important to reveal the precise synaptic organization of motor-related cortical and thalamic inputs to striatal PV interneurons. To examine which domains of the PV neurons receive these excitatory inputs, we used male bacterial artificial chromosome transgenic mice expressing somatodendritic membrane-targeted green fluorescent protein in PV neurons. An anterograde tracing study with the adeno-associated virus vector combined with immunodetection of pre- and postsynaptic markers visualized the distribution of the excitatory appositions on PV dendrites. Statistical analysis revealed that the density of thalamostriatal appositions along the dendrites was significantly higher on the proximal than distal dendrites. In contrast, there was no positional preference in the density of appositions from axons of the dorsofrontal cortex. Population observations of thalamostriatal and corticostriatal appositions by immunohistochemistry for pathway-specific vesicular glutamate transporters confirmed that thalamic inputs preferentially, and cortical ones less preferentially, made apposition on proximal dendrites of PV neurons. This axodendritic organization suggests that PV neurons produce fast and reliable inhibition of MSNs in response to thalamic inputs and process excitatory inputs from motor cortices locally and plastically, possibly together with other GABAergic and dopaminergic dendritic inputs, to modulate MSN inhibition.


Asunto(s)
Cuerpo Estriado/fisiología , Dendritas/fisiología , Interneuronas/metabolismo , Interneuronas/fisiología , Parvalbúminas/biosíntesis , Tálamo/fisiología , Animales , Axones/metabolismo , Corteza Cerebral/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Dendritas/metabolismo , Ácido Glutámico , Masculino , Ratones , Ratones Transgénicos , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiología , Terminales Presinápticos/metabolismo , Terminales Presinápticos/fisiología , Sinapsis/metabolismo , Sinapsis/fisiología , Tálamo/metabolismo
9.
Proc Natl Acad Sci U S A ; 111(30): E3139-48, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024183

RESUMEN

Haploinsufficiency of the voltage-gated sodium channel NaV1.1 causes Dravet syndrome, an intractable developmental epilepsy syndrome with seizure onset in the first year of life. Specific heterozygous deletion of NaV1.1 in forebrain GABAergic-inhibitory neurons is sufficient to cause all the manifestations of Dravet syndrome in mice, but the physiological roles of specific subtypes of GABAergic interneurons in the cerebral cortex in this disease are unknown. Voltage-clamp studies of dissociated interneurons from cerebral cortex did not detect a significant effect of the Dravet syndrome mutation on sodium currents in cell bodies. However, current-clamp recordings of intact interneurons in layer V of neocortical slices from mice with haploinsufficiency in the gene encoding the NaV1.1 sodium channel, Scn1a, revealed substantial reduction of excitability in fast-spiking, parvalbumin-expressing interneurons and somatostatin-expressing interneurons. The threshold and rheobase for action potential generation were increased, the frequency of action potentials within trains was decreased, and action-potential firing within trains failed more frequently. Furthermore, the deficit in excitability of somatostatin-expressing interneurons caused significant reduction in frequency-dependent disynaptic inhibition between neighboring layer V pyramidal neurons mediated by somatostatin-expressing Martinotti cells, which would lead to substantial disinhibition of the output of cortical circuits. In contrast to these deficits in interneurons, pyramidal cells showed no differences in excitability. These results reveal that the two major subtypes of interneurons in layer V of the neocortex, parvalbumin-expressing and somatostatin-expressing, both have impaired excitability, resulting in disinhibition of the cortical network. These major functional deficits are likely to contribute synergistically to the pathophysiology of Dravet syndrome.


Asunto(s)
Potenciales de Acción , Epilepsias Mioclónicas/metabolismo , Neuronas GABAérgicas/metabolismo , Neocórtex/metabolismo , Parvalbúminas/biosíntesis , Células Piramidales/metabolismo , Somatostatina/biosíntesis , Animales , Modelos Animales de Enfermedad , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/patología , Neuronas GABAérgicas/patología , Regulación de la Expresión Génica , Ratones , Ratones Mutantes , Canal de Sodio Activado por Voltaje NAV1.1/biosíntesis , Canal de Sodio Activado por Voltaje NAV1.1/genética , Neocórtex/patología , Parvalbúminas/genética , Células Piramidales/patología , Somatostatina/genética
10.
J Neurosci ; 35(34): 11830-47, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26311767

RESUMEN

Compelling evidence suggests that pathological activity of the external globus pallidus (GPe), a nucleus in the basal ganglia, contributes to the motor symptoms of a variety of movement disorders such as Parkinson's disease. Recent studies have challenged the idea that the GPe comprises a single, homogenous population of neurons that serves as a simple relay in the indirect pathway. However, we still lack a full understanding of the diversity of the neurons that make up the GPe. Specifically, a more precise classification scheme is needed to better describe the fundamental biology and function of different GPe neuron classes. To this end, we generated a novel multicistronic BAC (bacterial artificial chromosome) transgenic mouse line under the regulatory elements of the Npas1 gene. Using a combinatorial transgenic and immunohistochemical approach, we discovered that parvalbumin-expressing neurons and Npas1-expressing neurons in the GPe represent two nonoverlapping cell classes, amounting to 55% and 27% of the total GPe neuron population, respectively. These two genetically identified cell classes projected primarily to the subthalamic nucleus and to the striatum, respectively. Additionally, parvalbumin-expressing neurons and Npas1-expressing neurons were distinct in their autonomous and driven firing characteristics, their expression of intrinsic ion conductances, and their responsiveness to chronic 6-hydroxydopamine lesion. In summary, our data argue that parvalbumin-expressing neurons and Npas1-expressing neurons are two distinct functional classes of GPe neurons. This work revises our understanding of the GPe, and provides the foundation for future studies of its function and dysfunction. SIGNIFICANCE STATEMENT: Until recently, the heterogeneity of the constituent neurons within the external globus pallidus (GPe) was not fully appreciated. We addressed this knowledge gap by discovering two principal GPe neuron classes, which were identified by their nonoverlapping expression of the markers parvalbumin and Npas1. Our study provides evidence that parvalbumin and Npas1 neurons have different topologies within the basal ganglia.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Globo Pálido/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Neuronas/clasificación , Neuronas/metabolismo , Parvalbúminas/biosíntesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Femenino , Globo Pálido/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/análisis , Neuronas/química , Parvalbúminas/análisis
11.
J Neurosci ; 34(43): 14375-87, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25339750

RESUMEN

Accumulating evidence strongly implicates the transcriptional coactivator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in the pathophysiology of multiple neurological disorders, but the downstream gene targets of PGC-1α in the brain have remained enigmatic. Previous data demonstrate that PGC-1α is primarily concentrated in inhibitory neurons and that PGC-1α is required for the expression of the interneuron-specific Ca(2+)-binding protein parvalbumin (PV) throughout the cortex. To identify other possible transcriptional targets of PGC-1α in neural tissue, we conducted a microarray on neuroblastoma cells overexpressing PGC-1α, mined results for genes with physiological relevance to interneurons, and measured cortical gene and protein expression of these genes in mice with underexpression and overexpression of PGC-1α. We observed bidirectional regulation of novel PGC-1α-dependent transcripts spanning synaptic [synaptotagmin 2 (Syt2) and complexin 1 (Cplx1)], structural [neurofilament heavy chain (Nefh)], and metabolic [neutral cholesterol ester hydrolase 1 (Nceh1), adenylate kinase 1 (Ak1), inositol polyphosphate 5-phosphatase J (Inpp5j), ATP synthase mitochondrial F1 complex O subunit (Atp5o), phytanol-CoA-2hydroxylase (Phyh), and ATP synthase mitrochondrial F1 complex α subunit 1 (Atp5a1)] functions. The neuron-specific genes Syt2, Cplx1, and Nefh were developmentally upregulated in an expression pattern consistent with that of PGC-1α and were expressed in cortical interneurons. Conditional deletion of PGC-1α in PV-positive neurons significantly decreased cortical transcript expression of these genes, promoted asynchronous GABA release, and impaired long-term memory. Collectively, these data demonstrate that PGC-1α is required for normal PV-positive interneuron function and that loss of PGC-1α in this interneuron subpopulation could contribute to cortical dysfunction in disease states.


Asunto(s)
Interneuronas/metabolismo , Parvalbúminas/biosíntesis , Factores de Transcripción/biosíntesis , Transcripción Genética/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma
12.
Biochem J ; 462(1): 67-75, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24844465

RESUMEN

Secisbp2 [SECIS (selenocysteine insertion sequence)-binding protein 2] binds to SECIS elements located in the 3'-UTR region of eukaryotic selenoprotein mRNAs. It facilitates the incorporation of the rare amino acid selenocysteine in response to UGA codons. Inactivation of Secisbp2 in hepatocytes greatly reduced selenoprotein levels. Neuron-specific inactivation of Secisbp2 (CamK-Cre; Secisbp2fl/fl) reduced cerebral expression of selenoproteins to a lesser extent than inactivation of tRNA[Ser]Sec. This allowed us to study the development of cortical PV (parvalbumin)+ interneurons, which are completely lost in tRNA[Ser]Sec mutants. PV+ interneuron density was reduced in the somatosensory cortex, hippocampus and striatum. In situ hybridization for Gad67 (glutamic acid decarboxylase 67) confirmed the reduction of GABAergic (where GABA is γ-aminobutyric acid) interneurons. Because of the obvious movement phenotype involving a broad dystonic gait, we suspected basal ganglia dysfunction. Tyrosine hydroxylase expression was normal in substantia nigra neurons and their striatal terminals. However the densities of striatal PV+ and Gad67+ neurons were decreased by 65% and 49% respectively. Likewise, the density of striatal cholinergic neurons was reduced by 68%. Our observations demonstrate that several classes of striatal interneurons depend on selenoprotein expression. These findings may offer an explanation for the movement phenotype of selenoprotein P-deficient mice and the movement disorder and mental retardation described in a patient carrying SECISBP2 mutations.


Asunto(s)
Cuerpo Estriado/metabolismo , Interneuronas/fisiología , Proteínas de Unión al ARN/genética , Selenoproteínas/biosíntesis , Animales , Glutamato Descarboxilasa/biosíntesis , Ratones , Trastornos del Movimiento/genética , Parvalbúminas/biosíntesis , Selenocisteína/metabolismo , Corteza Somatosensorial/metabolismo
13.
J Neurosci ; 33(27): 11145-54, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23825418

RESUMEN

A characteristic feature in the primary visual cortex is that visual responses are suppressed as a stimulus extends beyond the classical receptive field. Here, we examined the role of inhibitory neurons expressing somatostatin (SOM⁺) or parvalbumin (PV⁺) on surround suppression and preferred receptive field size. We recorded multichannel extracellular activity in V1 of transgenic mice expressing channelrhodopsin in SOM⁺ neurons or PV⁺ neurons. Preferred size and surround suppression were measured using drifting square-wave gratings of varying radii and at two contrasts. Consistent with findings in primates, we found that the preferred size was larger for lower contrasts across all cortical depths, whereas the suppression index (SI) showed a trend to decrease with contrast. We then examined the effect of these metrics on units that were suppressed by photoactivation of either SOM⁺ or PV⁺ neurons. When activating SOM⁺ neurons, we found a significant increase in SI at cortical depths >400 µm, whereas activating PV⁺ neurons caused a trend toward lower SIs regardless of cortical depth. Conversely, activating PV⁺ neurons significantly increased preferred size across all cortical depths, similar to lowering contrast, whereas activating SOM⁺ neurons had no systematic effect on preferred size across all depths. These data suggest that SOM⁺ and PV⁺ neurons contribute differently to spatial integration. Our findings are compatible with the notion that SOM⁺ neurons mediate surround suppression, particularly in deeper cortex, whereas PV⁺ activation decreases the drive of the input to cortex and therefore resembles the effects on spatial integration of lowering contrast.


Asunto(s)
Sensibilidad de Contraste/fisiología , Neuronas/metabolismo , Parvalbúminas/biosíntesis , Somatostatina/biosíntesis , Corteza Visual/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Transgénicos , Estimulación Luminosa/métodos , Percepción Espacial/fisiología
14.
J Neurosci ; 33(28): 11724-33, 2013 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23843539

RESUMEN

Inhibitory neurons have been shown to perform a variety of functions within brain circuits, including shaping response functions in target cells. Still, how the properties of specific inhibitory neuron classes relate to their local circuits remains unclear. To better understand the distribution and origins of orientation selectivity in inhibitory neurons expressing the calcium binding protein parvalbumin (PV) in the mouse primary visual cortex, we labeled PV(+) neurons with red fluorescent protein (RFP) and targeted them for cell-attached electrophysiological recordings. PV(+) neurons could be broadly tuned or sharply tuned for orientation but tended to be more broadly tuned than unlabeled neurons on average. The dendritic morphology of PV(+) cells, revealed by intracellular labeling, was strongly correlated with tuning: highly tuned PV(+) neurons had shorter dendrites that branched nearer to the soma and had smaller dendritic fields overall, whereas broadly tuned PV(+) neurons had longer dendrites that branched farther from the soma, producing larger dendritic fields. High-speed two-photon calcium imaging of visual responses showed that the orientation preferences of highly tuned PV(+) neurons resembled the preferred orientations of neighboring cells. These results suggest that the diversity of the local neighborhood and the nature of dendritic sampling may both contribute to the response selectivity of PV(+) neurons.


Asunto(s)
Dendritas/metabolismo , Regulación de la Expresión Génica , Inhibición Neural/fisiología , Parvalbúminas/biosíntesis , Estimulación Luminosa/métodos , Corteza Visual/citología , Corteza Visual/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo
15.
Anesthesiology ; 121(1): 79-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24589481

RESUMEN

BACKGROUND: A prolonged isoflurane exposure may lead to cognitive decline in rodents. Neuregulin 1 (NRG1)-ErbB4 signaling plays a key role in the modulation of hippocampal synaptic plasticity through regulating the neurotransmission. The authors hypothesized that hippocampal NRG1-ErbB4 signaling is involved in isoflurane-induced cognitive impairments in aged mice. METHODS: Fourteen-month-old C57BL/6 mice were randomized to receive 100% O2 exposure, vehicle injection after 100% O2 exposure, vehicle injection after exposure to isoflurane carried by 100% O2, NRG1-ß1 injection after exposure to isoflurane carried by 100% O2, and NRG1-ß1 and an ErbB4 inhibitor AG1478 injection after exposure to isoflurane carried by 100% O2. Fear conditioning test was used to assess the cognitive function of mice 48-h postexposure. The brain tissues were harvested 48-h postexposure to determine the levels of NRG1, ErbB4, p-ErbB4, parvalbumin, and glutamic acid decarboxylase 67 in the hippocampus using Western blotting, enzyme-linked immunosorbent assay, and immunofluorescence. RESULTS: The percentage of freezing time to context was decreased from 50.28 ± 11.53% to 30.82 ± 10.00%, and the hippocampal levels of NRG1, p-ErbB4/ErbB4, parvalbumin, and glutamic acid decarboxylase 67 were decreased from 172.79 ± 20.85 ng/g, 69.15 ± 12.20%, 101.68 ± 11.21%, and 104.71 ± 6.85% to 112.92 ± 16.65 ng/g, 42.26 ± 9.71%, 75.89 ± 10.26%, and 73.87 ± 16.89%, respectively, after isoflurane exposure. NRG1-ß1 attenuated the isoflurane-induced hippocampus-dependent cognitive impairment and the declines in the hippocampal NRG1, p-ErbB4/ErbB4, parvalbumin, and glutamic acid decarboxylase 67. AG1478 inhibited the rescuing effects of NRG1-ß1. CONCLUSION: Disruption of NRG1-ErbB4 signaling in the parvalbumin-positive interneurons might, at least partially, contribute to the isoflurane-induced hippocampus-dependent cognitive impairment after exposure to isoflurane carried by 100% O2 in aged mice.


Asunto(s)
Anestésicos por Inhalación/farmacología , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/fisiopatología , Receptores ErbB/efectos de los fármacos , Hipocampo/fisiopatología , Isoflurano/farmacología , Neurregulina-1/efectos de los fármacos , Envejecimiento/fisiología , Animales , Western Blotting , Regulación hacia Abajo/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Conducta Exploratoria/efectos de los fármacos , Miedo/psicología , Técnica del Anticuerpo Fluorescente , Glutamato Descarboxilasa/biosíntesis , Inyecciones Intraventriculares , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Parvalbúminas/biosíntesis , Parvalbúminas/metabolismo , Receptor ErbB-4 , Transducción de Señal/efectos de los fármacos
16.
J Pineal Res ; 54(1): 58-68, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22823500

RESUMEN

Melatonin has shown particular promise as a neuroprotective agent to prevent motoneuron death in animal models of both amyotrophic lateral sclerosis (ALS) and spinal cord injuries (SCI). However, an understanding of the roles of endogenous melatonin receptors including MT1, MT2, and orphan G-protein receptor 50 (GPR50) in neuroprotection is lacking. To address this deficiency, we utilized plasmids for transfection and overexpression of individual melatonin receptors in the ventral spinal cord 4.1 (VSC4.1) motoneuron cell line. Receptor-mediated cytoprotection following exposure to glutamate at a toxic level (25 µm) was determined by assessing cell viability, apoptosis, and intracellular free Ca(2+) levels. Our findings indicate a novel role for MT1 and MT2 for increasing expression of the calcium-binding proteins calbindin D28K and parvalbumin. Increased levels of calbindin D28K and parvalbumin in VSC4.1 cells overexpressing MT1 and MT2 were associated with cytoprotective effects including inhibition of proapoptotic signaling, downregulation of inflammatory factors, and expression of prosurvival markers. Interestingly, the neuroprotective effects conferred by overexpression of MT1 and/or MT2 were also associated with increases in the estrogen receptor ß (ERß): estrogen receptor α (ERα) ratio and upregulation of angiogenic factors. GPR50 did not exhibit cytoprotective effects. To further confirm the involvement of the melatonin receptors, we silenced both MT1 and MT2 in VSC4.1 cells using RNA interference technology. Knockdown of MT1 and MT2 led to an increase in glutamate toxicity, which was only partially reversed by melatonin treatment. Taken together, our findings suggest that the neuroprotection against glutamate toxicity exhibited by melatonin may depend on MT1 and MT2 but not GPR50.


Asunto(s)
Ácido Glutámico/toxicidad , Neuronas Motoras/efectos de los fármacos , Receptores de Melatonina/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Calbindinas/biosíntesis , Línea Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ratones , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Parvalbúminas/biosíntesis , ARN Interferente Pequeño/farmacología , Receptor de Melatonina MT1/genética , Receptor de Melatonina MT2/genética , Receptores Acoplados a Proteínas G/efectos de los fármacos
17.
Can J Physiol Pharmacol ; 91(11): 966-72, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24117265

RESUMEN

Pharmacological preconditioning (PPC) with mitochondrial ATP-sensitive K(+) channel openers such as diazoxide, provides protection against ischemia in cardiac muscle, skeletal muscle, and other tissues. Effects on Ca(2+) homeostasis during the late phase of PPC have been described in cardiomyocytes, but no information is available regarding intracellular Ca(2+) changes in skeletal muscle fibers during late PPC. Intracellular Ca(2+) signals were measured in single fibers of adult mouse skeletal muscle, with fluorescent probes, 48 h after the administration of diazoxide. Parvalbumin levels in the myofibers were quantitated by Western blot. Diazoxide induction of late PPC was confirmed by partial protection of muscles from peroxide-induced damage. Late PPC was associated with a significant decrease in the duration of Ca(2+) signals during single twitches and tetanus with no changes in peak values. This effect was prevented by the reactive oxygen species (ROS) scavenger tiron. Late PPC was accompanied by a 30% increase in parvalbumin levels, and this effect was also blocked by tiron. Our data show, for the first time, a role of parvalbumin in late PPC in skeletal muscle.


Asunto(s)
Canales KATP/agonistas , Músculo Esquelético/efectos de los fármacos , Parvalbúminas/biosíntesis , Algoritmos , Compuestos de Anilina , Animales , Western Blotting , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diazóxido/farmacología , Estimulación Eléctrica , Colorantes Fluorescentes , Homeostasis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Fibras Musculares Esqueléticas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Especies Reactivas de Oxígeno , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Xantenos
18.
J Neurosci ; 31(4): 1193-203, 2011 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-21273404

RESUMEN

Human cortical excitability can be modified by repetitive transcranial magnetic stimulation (rTMS), but the cellular mechanisms are largely unknown. Here, we show that the pattern of delivery of theta-burst stimulation (TBS) (continuous versus intermittent) differently modifies electric activity and protein expression in the rat neocortex. Intermittent TBS (iTBS), but not continuous TBS (cTBS), enhanced spontaneous neuronal firing and EEG gamma band power. Sensory evoked cortical inhibition increased only after iTBS, although both TBS protocols increased the first sensory response arising from the resting cortical state. Changes in the cortical expression of the calcium-binding proteins parvalbumin (PV) and calbindin D-28k (CB) indicate that changes in spontaneous and evoked cortical activity following rTMS are in part related to altered activity of inhibitory systems. By reducing PV expression in the fast-spiking interneurons, iTBS primarily affected the inhibitory control of pyramidal cell output activity, while cTBS, by reducing CB expression, more likely affected the dendritic integration of synaptic inputs controlled by other classes of inhibitory interneurons. Calretinin, the third major calcium-binding protein expressed by another class of interneurons was not affected at all. We conclude that different patterns of TBS modulate the activity of inhibitory cell classes differently, probably depending on the synaptic connectivity and the preferred discharge pattern of these inhibitory neurons.


Asunto(s)
Corteza Cerebral/fisiología , Potenciales de Acción , Animales , Calbindina 2 , Calbindinas , Electroencefalografía , Potenciales Evocados Somatosensoriales , Interneuronas/fisiología , Masculino , Inhibición Neural , Parvalbúminas/biosíntesis , Células Piramidales/fisiología , Ratas , Proteína G de Unión al Calcio S100/biosíntesis , Estimulación Magnética Transcraneal
19.
Eur J Neurosci ; 35(6): 838-54, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22429243

RESUMEN

To examine inputs to parvalbumin (PV)-producing interneurons, we generated transgenic mice expressing somatodendritic membrane-targeted green fluorescent protein specifically in the interneurons, and completely visualized their dendrites and somata. Using immunolabeling for vesicular glutamate transporter (VGluT)1, VGluT2, and vesicular GABA transporter, we found that VGluT1-positive terminals made contacts 4- and 3.1-fold more frequently with PV-producing interneurons than VGluT2-positive and GABAergic terminals, respectively, in the primary somatosensory cortex. Even in layer 4, where VGluT2-positive terminals were most densely distributed, VGluT1-positive inputs to PV-producing interneurons were 2.4-fold more frequent than VGluT2-positive inputs. Furthermore, although GABAergic inputs to PV-producing interneurons were as numerous as VGluT2-positive inputs in most cortical layers, GABAergic inputs clearly preferred the proximal dendrites and somata of the interneurons, indicating that the sites of GABAergic inputs were more optimized than those of VGluT2-positive inputs. Simulation analysis with a PV-producing interneuron model compatible with the present morphological data revealed a plausible reason for this observation, by showing that GABAergic and glutamatergic postsynaptic potentials evoked by inputs to distal dendrites were attenuated to 60 and 87%, respectively, of those evoked by somatic inputs. As VGluT1-positive and VGluT2-positive axon terminals were presumed to be cortical and thalamic glutamatergic inputs, respectively, cortical excitatory inputs to PV-producing interneurons outnumbered the thalamic excitatory and intrinsic inhibitory inputs more than two-fold in any cortical layer. Although thalamic inputs are known to evoke about two-fold larger unitary excitatory postsynaptic potentials than cortical ones, the present results suggest that cortical inputs control PV-producing interneurons at least as strongly as thalamic inputs.


Asunto(s)
Dendritas/ultraestructura , Interneuronas/ultraestructura , Modelos Neurológicos , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Interneuronas/metabolismo , Ratones , Ratones Transgénicos , Microscopía Inmunoelectrónica , Técnicas de Cultivo de Órganos , Parvalbúminas/biosíntesis , Técnicas de Placa-Clamp
20.
J Neurosci ; 30(1): 110-5, 2010 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20053893

RESUMEN

GABAergic synaptic inputs targeting cortical principal cells undergo marked changes in their functional properties from depolarizing at early postnatal life to hyperpolarizing at mature stages. In contrast, the nature of GABA(A) receptor-mediated signaling in interneurons during maturation of neuronal networks is controversial. By using gramicidin perforated-patch and whole-cell recordings from LIM homeobox 6 (Lhx6)-positive dentate gyrus perisomatic-targeting parvalbumin-expressing interneurons (PV-INs), we show that signaling at first formed GABAergic synapses at postnatal day 3 (P3) is excitatory and switches to shunting during the course of the first to second postnatal week. GABAergic synaptic inputs at P3-P6 reliably evoke action potentials in 65% of Lhx6-EGFP-expressing perisomatic-targeting cells and boost spike induction upon conjoint activation of glutamatergic fibers. Thus, GABAergic inputs change their functional role during maturation. They facilitate the recruitment of perisomatic-targeting INs in early postnatal circuits when network connectivity and synaptic glutamate receptor-mediated excitation are low and control spike timing at later stages when connectivity and glutamate-mediated drive are high.


Asunto(s)
Hipocampo/fisiología , Interneuronas/citología , Interneuronas/fisiología , Parvalbúminas/biosíntesis , Receptores de GABA-A/fisiología , Potenciales de Acción/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Hipocampo/citología , Hipocampo/metabolismo , Interneuronas/metabolismo , Ratones , Ratones Transgénicos , Receptores de GABA-A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA