Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Brain ; 14(1): 158, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34645511

RESUMEN

Alterations in the canonical processing of Amyloid Precursor Protein generate proteoforms that contribute to the onset of Alzheimer's Disease. Modified composition of γ-secretase or mutations in its subunits has been directly linked to altered generation of Amyloid beta. Despite biochemical evidence about the role of γ-secretase in the generation of APP, the molecular origin of how spatial heterogeneity in the generation of proteoforms arises is not well understood. Here, we evaluated the localization of Nicastrin, a γ-secretase subunit, at nanometer sized functional zones of the synapse. With the help of super resolution microscopy, we confirm that Nicastrin is organized into nanodomains of high molecular density within an excitatory synapse. A similar nanoorganization was also observed for APP and the catalytic subunit of γ-secretase, Presenilin 1, that were discretely associated with Nicastrin nanodomains. Though Nicastrin is a functional subunit of γ-secretase, the Nicastrin and Presenilin1 nanodomains were either colocalized or localized independent of each other. The Nicastrin and Presenilin domains highlight a potential independent regulation of these molecules different from their canonical secretase function. The collisions between secretases and substrate molecules decide the probability and rate of product formation for transmembrane proteolysis. Our observations of secretase nanodomains indicate a spatial difference in the confinement of substrate and secretases, affecting the local probability of product formation by increasing their molecular availability, resulting in differential generation of proteoforms even within single synapses.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/química , Glicoproteínas de Membrana/química , Sinapsis/química , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Endocitosis , Neuronas GABAérgicas/química , Neuronas GABAérgicas/ultraestructura , Microscopía/métodos , Proteínas del Tejido Nervioso/análisis , Densidad Postsináptica/química , Densidad Postsináptica/ultraestructura , Presenilina-1/química , Dominios Proteicos , Células Piramidales/química , Células Piramidales/ultraestructura , Sinapsis/ultraestructura
2.
Mol Brain ; 14(1): 151, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34583741

RESUMEN

The atypical chemokine receptor 3, ACKR3, is a G protein-coupled receptor, which does not couple to G proteins but recruits ßarrestins. At present, ACKR3 is considered a target for cancer and cardiovascular disorders, but less is known about the potential of ACKR3 as a target for brain disease. Further, mouse lines have been created to identify cells expressing the receptor, but there is no tool to visualize and study the receptor itself under physiological conditions. Here, we engineered a knock-in (KI) mouse expressing a functional ACKR3-Venus fusion protein to directly detect the receptor, particularly in the adult brain. In HEK-293 cells, native and fused receptors showed similar membrane expression, ligand induced trafficking and signaling profiles, indicating that the Venus fusion does not alter receptor signaling. We also found that ACKR3-Venus enables direct real-time monitoring of receptor trafficking using resonance energy transfer. In ACKR3-Venus knock-in mice, we found normal ACKR3 mRNA levels in the brain, suggesting intact gene transcription. We fully mapped receptor expression across 14 peripheral organs and 112 brain areas and found that ACKR3 is primarily localized to the vasculature in these tissues. In the periphery, receptor distribution aligns with previous reports. In the brain there is notable ACKR3 expression in endothelial vascular cells, hippocampal GABAergic interneurons and neuroblast neighboring cells. In conclusion, we have generated Ackr3-Venus knock-in mice with a traceable ACKR3 receptor, which will be a useful tool to the research community for interrogations about ACKR3 biology and related diseases.


Asunto(s)
Proteínas Bacterianas/genética , Encéfalo/irrigación sanguínea , Técnicas de Sustitución del Gen , Genes Reporteros , Proteínas Luminiscentes/genética , Receptores CXCR/genética , Animales , Proteínas Bacterianas/análisis , Proteínas Bacterianas/farmacocinética , Biomarcadores , Sistemas de Computación , Células Endoteliales/química , Células Endoteliales/citología , Neuronas GABAérgicas/química , Neuronas GABAérgicas/citología , Células HEK293 , Humanos , Interneuronas/química , Interneuronas/citología , Ligandos , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/farmacocinética , Ratones , Especificidad de Órganos , Receptores CXCR/análisis , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacocinética , Distribución Tisular , beta-Arrestina 1/metabolismo
3.
Front Neural Circuits ; 15: 714780, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34366798

RESUMEN

Anatomical and physiological studies have described the cortex as a six-layer structure that receives, elaborates, and sends out information exclusively as excitatory output to cortical and subcortical regions. This concept has increasingly been challenged by several anatomical and functional studies that showed that direct inhibitory cortical outputs are also a common feature of the sensory and motor cortices. Similar to their excitatory counterparts, subsets of Somatostatin- and Parvalbumin-expressing neurons have been shown to innervate distal targets like the sensory and motor striatum and the contralateral cortex. However, no evidence of long-range VIP-expressing neurons, the third major class of GABAergic cortical inhibitory neurons, has been shown in such cortical regions. Here, using anatomical anterograde and retrograde viral tracing, we tested the hypothesis that VIP-expressing neurons of the mouse auditory and motor cortices can also send long-range projections to cortical and subcortical areas. We were able to demonstrate, for the first time, that VIP-expressing neurons of the auditory cortex can reach not only the contralateral auditory cortex and the ipsilateral striatum and amygdala, as shown for Somatostatin- and Parvalbumin-expressing long-range neurons, but also the medial geniculate body and both superior and inferior colliculus. We also demonstrate that VIP-expressing neurons of the motor cortex send long-range GABAergic projections to the dorsal striatum and contralateral cortex. Because of its presence in two such disparate cortical areas, this would suggest that the long-range VIP projection is likely a general feature of the cortex's network.


Asunto(s)
Corteza Auditiva/metabolismo , Vías Auditivas/metabolismo , Neuronas GABAérgicas/metabolismo , Corteza Motora/fisiología , Péptido Intestinal Vasoactivo/biosíntesis , Animales , Corteza Auditiva/química , Vías Auditivas/química , Femenino , Neuronas GABAérgicas/química , Masculino , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos
4.
Biochem Pharmacol ; 192: 114711, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34324871

RESUMEN

Ample evidence indicates that maternal immune activation (MIA) during gestation is linked to an increased risk for neurodevelopmental and psychiatric disorders, such as autism spectrum disorder (ASD), anxiety and depression, in offspring. However, the underlying mechanism for such a link remains largely elusive. Here, we performed RNA sequencing (RNA-seq) to examine the transcriptional profiles changes in mice in response to MIA and identified that the expression of Scn1a gene, encoding the pore-forming α-subunit of the brain voltage-gated sodium channel type-1 (NaV1.1) primarily in fast-spiking inhibitory interneurons, was significantly decreased in the medial prefrontal cortex (mPFC) of juvenile offspring after MIA. Moreover, diminished excitatory drive onto interneurons causes reduction of spontaneous gamma-aminobutyric acid (GABA)ergic neurotransmission in the mPFC of MIA offspring, leading to hyperactivity in this brain region. Remarkably, treatment with low-dose benzodiazepines clonazepam, an agonist of GABAA receptors, completely prevented the behavioral abnormalities, including stereotypies, social deficits, anxiety- and depression-like behavior, via increasing inhibitory neurotransmission as well as decreasing neural activity in the mPFC of MIA offspring. Our results demonstrate that decreased expression of NaV1.1 in the mPFC leads to abnormalities in maternal inflammation-related behaviors and provides a potential therapeutic strategy for the abnormal behavioral phenotypes observed in the offspring exposed to MIA.


Asunto(s)
Clonazepam/uso terapéutico , Moduladores del GABA/uso terapéutico , Neuronas GABAérgicas/inmunología , Trastornos Mentales/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Transmisión Sináptica/inmunología , Animales , Clonazepam/farmacología , Femenino , Moduladores del GABA/farmacología , Agonistas de Receptores de GABA-A/farmacología , Agonistas de Receptores de GABA-A/uso terapéutico , Neuronas GABAérgicas/química , Neuronas GABAérgicas/efectos de los fármacos , Masculino , Trastornos Mentales/inducido químicamente , Trastornos Mentales/prevención & control , Ratones , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.1/biosíntesis , Canal de Sodio Activado por Voltaje NAV1.1/inmunología , Poli I-C/toxicidad , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/inmunología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/prevención & control , Receptores de GABA-A/inmunología , Transmisión Sináptica/efectos de los fármacos
5.
Anesthesiology ; 135(3): 463-481, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34259824

RESUMEN

BACKGROUND: The γ-aminobutyric acid-mediated (GABAergic) inhibitory system in the brain is critical for regulation of sleep-wake and general anesthesia. The lateral septum contains mainly GABAergic neurons, being cytoarchitectonically divided into the dorsal, intermediate, and ventral parts. This study hypothesized that GABAergic neurons of the lateral septum participate in the control of wakefulness and promote recovery from anesthesia. METHODS: By employing fiber photometry, chemogenetic and optogenetic neuronal manipulations, anterograde tracing, in vivo electrophysiology, and electroencephalogram/electromyography recordings in adult male mice, the authors measured the role of lateral septum GABAergic neurons to the control of sleep-wake transition and anesthesia emergence and the corresponding neuron circuits in arousal and emergence control. RESULTS: The GABAergic neurons of the lateral septum exhibited high activities during the awake state by in vivo fiber photometry recordings (awake vs. non-rapid eye movement sleep: 3.3 ± 1.4% vs. -1.3 ± 1.2%, P < 0.001, n = 7 mice/group; awake vs. anesthesia: 2.6 ± 1.2% vs. -1.3 ± 0.8%, P < 0.001, n = 7 mice/group). Using chemogenetic stimulation of lateral septum GABAergic neurons resulted in a 100.5% increase in wakefulness and a 51.2% reduction in non-rapid eye movement sleep. Optogenetic activation of these GABAergic neurons promoted wakefulness from sleep (median [25th, 75th percentiles]: 153.0 [115.9, 179.7] s to 4.0 [3.4, 4.6] s, P = 0.009, n = 5 mice/group) and accelerated emergence from isoflurane anesthesia (514.4 ± 122.2 s vs. 226.5 ± 53.3 s, P < 0.001, n = 8 mice/group). Furthermore, the authors demonstrated that the lateral septum GABAergic neurons send 70.7% (228 of 323 cells) of monosynaptic projections to the ventral tegmental area GABAergic neurons, preferentially inhibiting their activities and thus regulating wakefulness and isoflurane anesthesia depth. CONCLUSIONS: The results uncover a fundamental role of the lateral septum GABAergic neurons and their circuit in maintaining awake state and promoting general anesthesia emergence time.


Asunto(s)
Anestesia/métodos , Neuronas GABAérgicas/fisiología , Núcleos Septales/fisiología , Sueño/fisiología , Vigilia/fisiología , Animales , Neuronas GABAérgicas/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Optogenética/métodos , Núcleos Septales/química
6.
J Neurosci ; 41(35): 7492-7508, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34244365

RESUMEN

Many patients with chronic pain conditions suffer from depression. The mechanisms underlying pain-induced depression are still unclear. There are critical links of medial prefrontal cortex (mPFC) synaptic function to depression, with signaling through the endocannabinoid (eCB) system as an important contributor. We hypothesized that afferent noxious inputs after injury compromise activity-dependent eCB signaling in the mPFC, resulting in depression. Depression-like behaviors were tested in male and female rats with traumatic neuropathy [spared nerve injury (SNI)], and neuronal activity in the mPFC was monitored using the immediate early gene c-fos and in vivo electrophysiological recordings. mPFC eCB Concentrations were determined using mass spectrometry, and behavioral and electrophysiological experiments were used to evaluate the role of alterations in eCB signaling in depression after pain. SNI-induced pain induced the development of depression phenotypes in both male and female rats. Pyramidal neurons in mPFC showed increased excitability followed by reduced excitability in the onset and prolonged phases of pain, respectively. Concentrations of the eCBs, 2-arachidonoylglycerol (2-AG) in the mPFC, were elevated initially after SNI, and our results indicate that this resulted in a loss of CB1R function on GABAergic interneurons in the mPFC. These data suggest that excessive release of 2-AG as a result of noxious stimuli triggers use-dependent loss of function of eCB signaling leading to excessive GABA release in the mPFC, with the final result being behavioral depression.SIGNIFICANCE STATEMENT Pain has both somatosensory and affective components, so the complexity of mechanisms underlying chronic pain is best represented by a biopsychosocial model that includes widespread CNS dysfunction. Many patients with chronic pain conditions develop depression. The mechanism by which pain causes depression is unclear. Although manipulation of the eCB signaling system as an avenue for providing analgesia per se has not shown much promise in previous studies. An important limitation of past research has been inadequate consideration of the dynamic nature of the connection between pain and depression as they develop. Here, we show that activity-dependent synthesis of eCBs during the initial onset of persistent pain is the critical link leading to depression when pain is persistent.


Asunto(s)
Dolor Crónico/fisiopatología , Depresión/etiología , Endocannabinoides/fisiología , Neuralgia/fisiopatología , Corteza Prefrontal/fisiopatología , Animales , Mapeo Encefálico , Dolor Crónico/complicaciones , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/psicología , Depresión/fisiopatología , Conducta Alimentaria , Femenino , Neuronas GABAérgicas/química , Gabapentina/uso terapéutico , Genes fos , Hiperalgesia/fisiopatología , Hiperalgesia/psicología , Interneuronas/química , Imagen por Resonancia Magnética , Masculino , Neuralgia/complicaciones , Neuralgia/tratamiento farmacológico , Neuralgia/psicología , Nocicepción/fisiología , Prueba de Campo Abierto , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/análisis , Neuropatía Ciática/fisiopatología , Neuropatía Ciática/psicología , Organismos Libres de Patógenos Específicos , Natación
7.
J Chem Neuroanat ; 116: 101997, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34182088

RESUMEN

In the last few years we assist to an unexpected deluge of genomic data on hypothalamic development and structure. Perhaps most surprisingly, the Lateral Zone has received much attention too. The new information focuses first of all on transcriptional heterogeneity. Many already known and a number of hitherto unknown lateral hypothalamic neurons have been described to an enormous degree of detail. Maybe the most surprising novel discoveries are two: First, some restricted regions of the embryonic forebrain neuroepithelium generate specific LHA neurons, either GABAergic or glutamatergic. Second, evidence is mounting that supports the existence of numerous kinds of "bilingual" lateral hypothalamic neurons, expressing (and releasing) glutamate and GABA both as well as assorted neuropeptides. This is not accepted by all, and it could be that genomic researchers need a common set of rules to interpret their data (sensitivity, significance, age of analysis). In any case, some of the new results appear to confirm hypotheses about the ability of the hypothalamus and in particular its Lateral Zone to achieve physiological flexibility on a fixed connectivity ("biochemical switching"). Furthermore, the results succinctly reviewed here are the basis for future advances, since the transcriptional databases generated can now be mined e.g. for adhesion genes, to figure out the causes of the peculiar histology of the Lateral Zone; or for ion channel genes, to clarify present and future electrophysiological data. And with the specific expression data about small subpopulations of neurons, their connections can now be specifically labeled, revealing novel relations with functional significance.


Asunto(s)
Neuronas GABAérgicas/química , Neuronas GABAérgicas/metabolismo , Ácido Glutámico/metabolismo , Área Hipotalámica Lateral/crecimiento & desarrollo , Área Hipotalámica Lateral/metabolismo , Neurogénesis/fisiología , Animales , Ácido Glutámico/análisis , Humanos , Área Hipotalámica Lateral/química , Factores de Transcripción/análisis , Factores de Transcripción/biosíntesis
8.
Brain Res ; 1762: 147425, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33737065

RESUMEN

The amygdala plays a crucial role in anxiety-related behavior and various neuropsychiatric disorders. The offspring of dams, administered methylazoxymethanol acetate (MAM) intraperitoneally at gestational day 15, exhibit micrencephaly and anxiety-related behavior, such as hyperactivity in rearing and crossing behavior, alongside a distinct Fos expression profile in the basolateral (BLA) and central amygdala. However, the histochemical underpinnings of these changes remain to be elucidated. To determine the histochemical alterations in MAM-induced model rats, we performed Nissl staining, immunohistochemistry for parvalbumin (PV) or calbindin (Calb), and immunohistochemistry for PV in conjunction with in situ hybridization for glutamate decarboxylase (GAD). We compared immunoreactivity in the BLA between normal and MAM-induced model rats and observed a significant decrease in the number of PV-positive neurons in MAM-induced model rats; however, no significant differences in the number of Nissl- and Calb-positive neurons were observed. We did not detect any significant between-group differences with regards to the effects of environmental enrichment on the number of PV-positive neurons in the BLA. Double-labeling for GAD and PV revealed that many PV-positive neurons colocalized with digoxigenin-GAD65/67 signals. In addition, GAD/PV double-positive neurons and the total number of GAD-positive neurons in the BLA were lower in the MAM-induced model rats. These results indicate that histochemical alterations observed in the BLA of the MAM-induced model rats may attribute to an aberrant GABAergic inhibitory system.


Asunto(s)
Complejo Nuclear Basolateral/metabolismo , Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Acetato de Metilazoximetanol/análogos & derivados , Microcefalia/metabolismo , Parvalbúminas/metabolismo , Animales , Complejo Nuclear Basolateral/química , Complejo Nuclear Basolateral/efectos de los fármacos , Carcinógenos/toxicidad , Femenino , Neuronas GABAérgicas/química , Neuronas GABAérgicas/efectos de los fármacos , Interneuronas/química , Interneuronas/efectos de los fármacos , Masculino , Acetato de Metilazoximetanol/toxicidad , Microcefalia/inducido químicamente , Microcefalia/psicología , Parvalbúminas/análisis , Embarazo , Ratas , Ratas Sprague-Dawley
9.
Cereb Cortex ; 31(6): 3064-3081, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33570093

RESUMEN

Many developmental syndromes have been linked to genetic mutations that cause abnormal ERK/MAPK activity; however, the neuropathological effects of hyperactive signaling are not fully understood. Here, we examined whether hyperactivation of MEK1 modifies the development of GABAergic cortical interneurons (CINs), a heterogeneous population of inhibitory neurons necessary for cortical function. We show that GABAergic-neuron specific MEK1 hyperactivation in vivo leads to increased cleaved caspase-3 labeling in a subpopulation of immature neurons in the embryonic subpallial mantle zone. Adult mutants displayed a significant loss of parvalbumin (PV), but not somatostatin, expressing CINs and a reduction in perisomatic inhibitory synapses on excitatory neurons. Surviving mutant PV-CINs maintained a typical fast-spiking phenotype but showed signs of decreased intrinsic excitability that coincided with an increased risk of seizure-like phenotypes. In contrast to other mouse models of PV-CIN loss, we discovered a robust increase in the accumulation of perineuronal nets, an extracellular structure thought to restrict plasticity. Indeed, we found that mutants exhibited a significant impairment in the acquisition of behavioral response inhibition capacity. Overall, our data suggest PV-CIN development is particularly sensitive to hyperactive MEK1 signaling, which may underlie certain neurological deficits frequently observed in ERK/MAPK-linked syndromes.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Neuronas GABAérgicas/metabolismo , Inhibición Psicológica , MAP Quinasa Quinasa 1/metabolismo , Parvalbúminas/metabolismo , Animales , Corteza Cerebral/química , Electroencefalografía/métodos , Desarrollo Embrionario/fisiología , Neuronas GABAérgicas/química , Locomoción/fisiología , MAP Quinasa Quinasa 1/análisis , Ratones , Técnicas de Cultivo de Órganos , Parvalbúminas/análisis , Transducción de Señal/fisiología
10.
Neurosci Lett ; 745: 135621, 2021 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-33421491

RESUMEN

The Basolateral amygdala (BLA) and central nucleus of the amygdala (CEA) have been proved to play a key role in the control of anxiety, stress and fear-related behaviors. BLA is a cortex-like complex consisting of both γ-aminobutyric acidergic (GABAergic) interneurons and glutamatergic neurons. The CEA is a striatum-like output of the amygdala, consisting almost exclusively of GABAergic medium spiny neurons. In this study, we explored the morphology and axonal projections of the GABAergic neurons in BLA and CEA, using conditional anterograde axonal tracing, immunohistochemistry, and VGAT-Cre transgenic mice to further understand their functional roles. We found that the axonal projections of GABAergic neurons from the BLA mainly distributed to the forebrain, whilst GABAergic neurons from the CEA distributed to the forebrain, midbrain and brainstem. In the forebrain, the axonal projections of GABAergic neurons from the BLA projected to the anterior olfactory nucleus, the cerebral cortex, the septum, the striatum, the thalamus, the amygdala and the hippocampus. The axonal projections of GABAergic neurons from the CEA distributed to the nuclei of the prefrontal cortex, the bed nucleus of the stria terminalis, the hypothalamus and the thalamus. In the midbrain and brainstem, the axonal projections of GABAergic neurons from the CEA were found in the periaqueductal gray, the substantia nigra, and the locus coeruleus. These data reveal the neuroanatomical basis for exploring the function of GABAergic neurons in the BLA and CEA, particularly during the processing of fear-related behavior.


Asunto(s)
Complejo Nuclear Basolateral/fisiología , Núcleo Amigdalino Central/fisiología , Vías Eferentes/fisiología , Neuronas GABAérgicas/fisiología , Animales , Complejo Nuclear Basolateral/química , Núcleo Amigdalino Central/química , Vías Eferentes/química , Neuronas GABAérgicas/química , Células HEK293 , Humanos , Ratones , Ratones Transgénicos
11.
Neural Plast ; 2021: 8833087, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33510780

RESUMEN

Accumulating evidence implicates a role for brain structures outside the ascending auditory pathway in tinnitus, the phantom perception of sound. In addition to other factors such as age-dependent hearing loss, high-level sound exposure is a prominent cause of tinnitus. Here, we examined how noise exposure altered the distribution of excitatory and inhibitory synaptic inputs in the guinea pig hippocampus and determined whether these changes were associated with tinnitus. In experiment one, guinea pigs were overexposed to unilateral narrow-band noise (98 dB SPL, 2 h). Two weeks later, the density of excitatory (VGLUT-1/2) and inhibitory (VGAT) synaptic terminals in CA1, CA3, and dentate gyrus hippocampal subregions was assessed by immunohistochemistry. Overall, VGLUT-1 density primarily increased, while VGAT density decreased significantly in many regions. Then, to assess whether the noise-induced alterations were persistent and related to tinnitus, experiment two utilized a noise-exposure paradigm shown to induce tinnitus and assessed tinnitus development which was assessed using gap-prepulse inhibition of the acoustic startle (GPIAS). Twelve weeks after sound overexposure, changes in excitatory synaptic terminal density had largely recovered regardless of tinnitus status, but the recovery of GABAergic terminal density was dramatically different in animals expressing tinnitus relative to animals resistant to tinnitus. In resistant animals, inhibitory synapse density recovered to preexposure levels, but in animals expressing tinnitus, inhibitory synapse density remained chronically diminished. Taken together, our results suggest that noise exposure induces striking changes in the balance of excitatory and inhibitory synaptic inputs throughout the hippocampus and reveal a potential role for rebounding inhibition in the hippocampus as a protective factor leading to tinnitus resilience.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Ruido/efectos adversos , Acúfeno/metabolismo , Proteínas de Transporte Vesicular de Glutamato/metabolismo , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/metabolismo , Estimulación Acústica/efectos adversos , Animales , Vías Auditivas/metabolismo , Vías Auditivas/patología , Femenino , Neuronas GABAérgicas/química , Ácido Glutámico/análisis , Ácido Glutámico/metabolismo , Cobayas , Hipocampo/patología , Masculino , Sinapsis/química , Sinapsis/metabolismo , Acúfeno/patología , Proteínas de Transporte Vesicular de Glutamato/análisis , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/análisis
12.
Neuropharmacology ; 180: 108299, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32916145

RESUMEN

Predatory hunting is an important approach for animals to obtain valuable nutrition and energy, which critically depends on heightened arousal. Yet the neural substrates underlying predatory hunting remain largely undefined. Here, we report that basal forebrain (BF) GABAergic neurons play an important role in regulating predatory hunting. Our results showed that BF GABAergic neurons were activated during the prey (cricket)-hunting and food feeding in mice. Optogenetic activation of BF GABAergic neurons evoked immediate predatory-like actions to both artificial and natural preys, significantly reducing the attack latency while increasing the attack probability and the number of killed natural prey (crickets). Similar to the effect of activating the soma of BF GABAergic neurons, photoactivation of their terminals in the ventral tegmental area (VTA) also strongly promotes predatory hunting. Moreover, photoactivation of GABAergic BF - VTA pathway significantly increases the intake of various food in mice. By synchronous recording of electroencephalogram and electromyogram, we showed that photoactivation of GABAergic BF - VTA pathway induces instant arousal and maintains long-term wakefulness. In summary, our results clearly demonstrated that the GABAergic BF is a key neural substrate for predatory hunting, and promotes this behavior through GABAergic BF - VTA pathway.


Asunto(s)
Nivel de Alerta/fisiología , Prosencéfalo Basal/metabolismo , Neuronas GABAérgicas/metabolismo , Conducta Predatoria/fisiología , Animales , Prosencéfalo Basal/química , Electroencefalografía/métodos , Neuronas GABAérgicas/química , Gryllidae , Masculino , Ratones , Ratones Endogámicos C57BL , Optogenética/métodos
13.
Front Neural Circuits ; 14: 29, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32547372

RESUMEN

To understand functional neuronal circuits for emotion in the basal forebrain, patterns of neuronal activation were examined in mice by immunohistochemistry of immediate-early gene products (Zif268/Egr1 and c-Fos). In all mice examined, clusters of 30-50 neurons expressing Zif268 were found on both sides in the area between the extended amygdala (EA) and globus pallidus (GP), generally designated as sublenticular extended amygdala (SLEA). The clusters consisted of 79.9 ± 3.0% of GABAergic neurons in GAD65-mCherry mice. The expression of the cholinergic marker choline acetyltransferase and the GP markers parvalbumin, proenkephalin, and FoxP2 indicated that these neurons were different from known types of neurons in the EA and GP; therefore, we named them the sublenticular extended amygdalar Zif268/Egr1-expressing neuronal cluster (SLEA-zNC). Sublenticular extended amygdalar Zif268/Egr1-expressing neuronal clusters participated in stress processing because increasing numbers of cells were observed in SLEA-zNCs after exposure to restraint stress (RS), the induction of which was suppressed by diazepam treatment. Mapping SLEA-zNCs showed that their positions and arrangement varied individually; SLEA-zNCs were distributed asymmetrically and tended to be situated mainly in the middle region between the anterior commissure (AC) and posterior end of the GP. However, the total cell number in SLEA-zNCs was compatible between the right and left hemispheres after activation by RS. Therefore, SLEA-zNCs were distributed asymmetrically but were not lateralized. Because time courses of activation differed between the Zif268 and c-Fos, the sequential dual treatment of RSs enabled us to differentiate SLEA-zNCs activated by the first and second RS. The results supported that the same SLEA-zNCs responded to both the first and second RS, and this also applied for all SLEA-zNCs. Thus, we concluded that the cluster positions were invariable under RS in each mouse but were distributed differently between individual mice. We name these newly identified neuronal clusters as stress-related neuronal clusters, SLEA-zNCs, which are considered to be novel functional units of "islands of activation." Moreover, SLEA-zNCs were situated at different positions in all mice examined, showing individual differences in their positions.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Prosencéfalo Basal/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas/metabolismo , Estrés Psicológico/metabolismo , Amígdala del Cerebelo/química , Amígdala del Cerebelo/citología , Animales , Prosencéfalo Basal/química , Prosencéfalo Basal/citología , Femenino , Neuronas GABAérgicas/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/química , Restricción Física/efectos adversos , Restricción Física/psicología , Estrés Psicológico/psicología
14.
Front Neural Circuits ; 14: 26, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32587504

RESUMEN

Successful memory involves not only remembering over time but also keeping memories distinct. Computational models suggest that pattern separation appears as a highly efficient process to discriminate between overlapping memories. Furthermore, lesion studies have shown that the dentate gyrus (DG) participates in pattern separation. However, these manipulations did not allow identifying the neuronal mechanism underlying pattern separation. The development of different neurophotonics techniques, together with other genetic tools, has been useful for the study of the microcircuit involved in this process. It has been shown that less-overlapped information would generate distinct neuronal representations within the granule cells (GCs). However, because glutamatergic or GABAergic cells in the DG are not functionally or structurally homogeneous, identifying the specific role of the different subpopulations remains elusive. Then, understanding pattern separation requires the ability to manipulate a temporal and spatially specific subset of cells in the DG and ideally to analyze DG cells activity in individuals performing a pattern separation dependent behavioral task. Thus, neurophotonics and calcium imaging techniques in conjunction with activity-dependent promoters and high-resolution microscopy appear as important tools for this endeavor. In this work, we review how different neurophotonics techniques have been implemented in the elucidation of a neuronal network that supports pattern separation alone or in combination with traditional techniques. We discuss the limitation of these techniques and how other neurophotonic techniques could be used to complement the advances presented up to this date.


Asunto(s)
Simulación por Computador , Giro Dentado/fisiología , Memoria/fisiología , Modelos Neurológicos , Red Nerviosa/fisiología , Fenómenos Ópticos , Animales , Giro Dentado/química , Neuronas GABAérgicas/química , Neuronas GABAérgicas/fisiología , Humanos , Imagen Molecular/métodos , Red Nerviosa/química
15.
Front Neural Circuits ; 14: 17, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32390805

RESUMEN

A unique feature of the olfactory system is the continuous generation and integration of new neurons throughout adulthood. Adult-born neuron survival and integration is dependent on activity and sensory experience, which is largely mediated by early synaptic inputs that adult-born neurons receive upon entering the olfactory bulb (OB). As in early postnatal development, the first synaptic inputs onto adult-born neurons are GABAergic. However, the specific sources of early synaptic GABA and the influence of specific inputs on adult-born neuron development are poorly understood. Here, we use retrograde and anterograde viral tracing to reveal robust GABAergic projections from the basal forebrain horizontal limb of the diagonal band of Broca (HDB) to the granule cell layer (GCL) and glomerular layer (GL) of the mouse OB. Whole-cell electrophysiological recordings indicate that these projections target interneurons in the GCL and GL, including adult-born granule cells (abGCs). Recordings from birth-dated abGCs reveal a developmental time course in which HDB GABAergic input onto abGCs emerges as the neurons first enter the OB, and strengthens throughout the critical period of abGC development. Finally, we show that removing GABAergic signaling from HDB neurons results in decreased abGC survival. Together these data show that GABAergic projections from the HDB synapse onto immature abGCs in the OB to promote their survival through the critical period, thus representing a source of long-range input modulating plasticity in the adult OB.


Asunto(s)
Prosencéfalo Basal/fisiología , Neuronas GABAérgicas/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Bulbo Olfatorio/citología , Bulbo Olfatorio/fisiología , Factores de Edad , Animales , Prosencéfalo Basal/química , Supervivencia Celular/fisiología , Femenino , Neuronas GABAérgicas/química , Masculino , Ratones , Ratones Transgénicos , Bulbo Olfatorio/química , Vías Olfatorias/química , Vías Olfatorias/citología , Vías Olfatorias/fisiología
16.
Artículo en Inglés | MEDLINE | ID: mdl-32116571

RESUMEN

The piriform cortex (PC) is a key brain area involved in both processing and coding of olfactory information. It is implicated in various brain disorders, such as epilepsy, Alzheimer's disease, and autism. The PC consists of the anterior (APC) and posterior (PPC) parts, which are different anatomically and functionally. However, the direct input networks to specific neuronal populations within the APC and PPC remain poorly understood. Here, we mapped the whole-brain direct inputs to the two major neuronal populations, the excitatory glutamatergic principal neurons and inhibitory γ-aminobutyric acid (GABA)-ergic interneurons within the APC and PPC using the rabies virus (RV)-mediated retrograde trans-synaptic tracing system. We found that for both types of neurons, APC and PPC share some similarities in input networks, with dominant inputs originating from the olfactory region (OLF), followed by the cortical subplate (CTXsp), isocortex, cerebral nuclei (CNU), hippocampal formation (HPF) and interbrain (IB), whereas the midbrain (MB) and hindbrain (HB) were rarely labeled. However, APC and PPC also show distinct features in their input distribution patterns. For both types of neurons, the input proportion from the OLF to the APC was higher than that to the PPC; while the PPC received higher proportions of inputs from the HPF and CNU than the APC did. Overall, our results revealed the direct input networks of both excitatory and inhibitory neuronal populations of different PC subareas, providing a structural basis to analyze the diverse PC functions.


Asunto(s)
Neuronas GABAérgicas/fisiología , Ácido Glutámico/fisiología , Corteza Piriforme/citología , Corteza Piriforme/fisiología , Animales , Recuento de Células/métodos , Femenino , Neuronas GABAérgicas/química , Glutamato Descarboxilasa/análisis , Glutamato Descarboxilasa/fisiología , Ácido Glutámico/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Corteza Piriforme/química , Proteína 2 de Transporte Vesicular de Glutamato/análisis , Proteína 2 de Transporte Vesicular de Glutamato/fisiología
17.
J Comp Neurol ; 528(8): 1436-1456, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31808567

RESUMEN

In the primate thalamus, the parvocellular ventral anterior nucleus (VApc) and the centromedian nucleus (CM) receive GABAergic projections from the internal globus pallidus (GPi) and glutamatergic inputs from motor cortices. In this study, we used electron microscopy to assess potential structural changes in GABAergic and glutamatergic microcircuits in the VApc and CM of MPTP-treated parkinsonian monkeys. The intensity of immunostaining for GABAergic markers in VApc and CM did not differ between control and parkinsonian monkeys. In the electron microscope, three major types of terminals were identified in both nuclei: (a) vesicular glutamate transporter 1 (vGluT1)-positive terminals forming asymmetric synapses (type As), which originate from the cerebral cortex, (b) GABAergic terminals forming single symmetric synapses (type S1), which likely arise from the reticular nucleus and GABAergic interneurons, and (c) GABAergic terminals forming multiple symmetric synapses (type S2), which originate from GPi. The density of As terminals outnumbered that of S1 and S2 terminals in VApc and CM of control and parkinsonian animals. No significant change was found in the abundance and synaptic connectivity of S1 and S2 terminals in VApc or CM of MPTP-treated monkeys, while the prevalence of "As" terminals in VApc of parkinsonian monkeys was 51.4% lower than in controls. The cross-sectional area of vGluT1-positive boutons in both VApc and CM of parkinsonian monkeys was significantly larger than in controls, but their pattern of innervation of thalamic cells was not altered. Our findings suggest that the corticothalamic system undergoes significant synaptic remodeling in the parkinsonian state.


Asunto(s)
Neuronas GABAérgicas/fisiología , Red Nerviosa/fisiología , Trastornos Parkinsonianos/metabolismo , Núcleos Talámicos Ventrales/fisiología , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Animales , Femenino , Neuronas GABAérgicas/química , Neuronas GABAérgicas/ultraestructura , Ácido Glutámico/análisis , Ácido Glutámico/metabolismo , Macaca mulatta , Masculino , Red Nerviosa/química , Red Nerviosa/ultraestructura , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/patología , Núcleos Talámicos Ventrales/química , Núcleos Talámicos Ventrales/ultraestructura , Proteína 1 de Transporte Vesicular de Glutamato/análisis
18.
Eur J Neurosci ; 50(11): 3732-3749, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31374129

RESUMEN

γ-Aminobutyric acid (GABA) neurons in the ventral tegmental area (VTA) provide local inhibitory control of dopamine neuron activity and send long-range projections to several target regions including the nucleus accumbens. They play diverse roles in reward and aversion, suggesting that they be comprised of several functionally distinct sub-groups, but our understanding of this diversity has been limited by a lack of molecular markers that might provide genetic entry points for cell type-specific investigations. To address this, we conducted transcriptional profiling of GABA neurons and dopamine neurons using immunoprecipitation of tagged polyribosomes (RiboTag) and RNAseq. First, we directly compared these two transcriptomes in order to obtain a list of genes enriched in GABA neurons compared with dopamine neurons. Next, we created a novel bioinformatic approach, that used the PANTHER (Protein ANalysis THrough Evolutionary Relationships) gene ontology database and VTA gene expression data from the Allen Mouse Brain Atlas, from which we obtained 6 candidate genes: Cbln4, Rxfp3, Rora, Gpr101, Trh and Nrp2. As a final step, we verified the selective expression of these candidate genes in sub-groups of GABA neurons in the VTA (and neighbouring substantia nigra pars compacta) using immunolabelling. Taken together, our study provides a valuable toolbox for the future investigation of GABA neuron sub-groups in the VTA.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes/fisiología , Área Tegmental Ventral/metabolismo , Ácido gamma-Aminobutírico/biosíntesis , Animales , Neuronas GABAérgicas/química , Expresión Génica , Ratones , Ratones Transgénicos , Área Tegmental Ventral/química , Ácido gamma-Aminobutírico/genética
19.
J Neurosci ; 39(23): 4527-4549, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-30926750

RESUMEN

The medial septum implements cortical theta oscillations, a 5-12 Hz rhythm associated with locomotion and paradoxical sleep reflecting synchronization of neuronal assemblies such as place cell sequence coding. Highly rhythmic burst-firing parvalbumin-positive GABAergic medial septal neurons are strongly coupled to theta oscillations and target cortical GABAergic interneurons, contributing to coordination within one or several cortical regions. However, a large population of medial septal neurons of unidentified neurotransmitter phenotype and with unknown axonal target areas fire with a low degree of rhythmicity. We investigated whether low-rhythmic-firing neurons (LRNs) innervated similar or different cortical regions to high-rhythmic-firing neurons (HRNs) and assessed their temporal dynamics in awake male mice. The majority of LRNs were GABAergic and parvalbumin-immunonegative, some expressing calbindin; they innervated interneurons mostly in the dentate gyrus (DG) and CA3. Individual LRNs showed several distinct firing patterns during immobility and locomotion, forming a parallel inhibitory stream for the modulation of cortical interneurons. Despite their fluctuating firing rates, the preferred firing phase of LRNs during theta oscillations matched the highest firing probability phase of principal cells in the DG and CA3. In addition, as a population, LRNs were markedly suppressed during hippocampal sharp-wave ripples, had a low burst incidence, and several of them did not fire on all theta cycles. Therefore, CA3 receives GABAergic input from both HRNs and LRNs, but the DG receives mainly LRN input. We propose that distinct GABAergic LRNs contribute to changing the excitability of the DG and CA3 during memory discrimination via transient disinhibition of principal cells.SIGNIFICANCE STATEMENT For the encoding and recall of episodic memories, nerve cells in the cerebral cortex are activated in precisely timed sequences. Rhythmicity facilitates the coordination of neuronal activity and these rhythms are detected as oscillations of different frequencies such as 5-12 Hz theta oscillations. Degradation of these rhythms, such as through neurodegeneration, causes memory deficits. The medial septum, a part of the basal forebrain that innervates the hippocampal formation, contains high- and low-rhythmic-firing neurons (HRNs and LRNs, respectively), which may contribute differentially to cortical neuronal coordination. We discovered that GABAergic LRNs preferentially innervate the dentate gyrus and the CA3 area of the hippocampus, regions important for episodic memory. These neurons act in parallel with the HRNs mostly via transient inhibition of inhibitory neurons.


Asunto(s)
Región CA3 Hipocampal/fisiología , Giro Dentado/fisiología , Neuronas GABAérgicas/fisiología , Vías Nerviosas/fisiología , Tabique del Cerebro/citología , Potenciales de Acción , Animales , Región CA3 Hipocampal/citología , Calbindinas/análisis , Giro Dentado/citología , Neuronas GABAérgicas/química , Masculino , Memoria Episódica , Recuerdo Mental/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/análisis , Parvalbúminas/análisis , Carrera , Tabique del Cerebro/fisiología , Ritmo Teta/fisiología , Vigilia
20.
J Comp Neurol ; 527(14): 2215-2232, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30847931

RESUMEN

Transgenic animals have become a widely used model to identify and study specific cell types in whole organs. Promotor-driven reporter gene labeling of the cells under investigation has promoted experimental efficacy to a large degree. However, rigorous assessment of transgene expression specificity in these animal models is highly recommended to validate cellular identity and to isolate potentially mislabeled cell populations. Here, we report on one such mislabeled neuron population in a widely used transgenic mouse line in which GABAergic somatostatin-expressing interneurons (SOMpos INs) are labeled by eGFP (so-called GIN mouse, FVB-Tg(GadGFP)45704Swn/J). These neurons represent a subpopulation of all SOMpos INs. However, we report here on GFP labeling of non-GABAergic neurons in the nucleus endopiriformis of this mouse line.


Asunto(s)
Claustro/metabolismo , Neuronas GABAérgicas/metabolismo , Glutamato Descarboxilasa/biosíntesis , Proteínas Fluorescentes Verdes/biosíntesis , Corteza Piriforme/metabolismo , Animales , Claustro/química , Neuronas GABAérgicas/química , Expresión Génica , Glutamato Descarboxilasa/análisis , Glutamato Descarboxilasa/genética , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Corteza Piriforme/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...