Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 705
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(40): e2202536119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36161898

RESUMEN

Through synaptic connections, long-range circuits transmit information among neurons and connect different brain regions to form functional motifs and execute specific functions. Tracing the synaptic distribution of specific neurons requires submicron-level resolution information. However, it is a great challenge to map the synaptic terminals completely because these fine structures span multiple regions, even in the whole brain. Here, we develop a pipeline including viral tracing, sample embedding, fluorescent micro-optical sectional tomography, and big data processing. We mapped the whole-brain distribution and architecture of long projections of the parvalbumin neurons in the basal forebrain at the synaptic level. These neurons send massive projections to multiple downstream regions with subregional preference. With three-dimensional reconstruction in the targeted areas, we found that synaptic degeneration was inconsistent with the accumulation of amyloid-ß plaques but was preferred in memory-related circuits, such as hippocampal formation and thalamus, but not in most hypothalamic nuclei in 8-month-old mice with five familial Alzheimer's disease mutations. Our pipeline provides a platform for generating a whole-brain atlas of cell-type-specific synaptic terminals in the physiological and pathological brain, which can provide an important resource for the study of the organizational logic of specific neural circuits and the circuitry changes in pathological conditions.


Asunto(s)
Enfermedad de Alzheimer , Prosencéfalo Basal , Neuronas , Sinapsis , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Prosencéfalo Basal/ultraestructura , Modelos Animales de Enfermedad , Ratones , Mutación , Neuroimagen , Neuronas/ultraestructura , Parvalbúminas/análisis , Sinapsis/ultraestructura
2.
J Neurosci ; 41(34): 7182-7196, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34253625

RESUMEN

Up states are the best studied example of an emergent neural dynamic regime. Computational models based on a single class of inhibitory neurons indicate that Up states reflect bistable dynamic systems in which positive feedback is stabilized by strong inhibition and predict a paradoxical effect in which increased drive to inhibitory neurons results in decreased inhibitory activity. To date, however, computational models have not incorporated empirically defined properties of parvalbumin (PV) and somatostatin (SST) neurons. Here we first experimentally characterized the frequency-current (F-I) curves of pyramidal (Pyr), PV, and SST neurons from mice of either sex, and confirmed a sharp difference between the threshold and slopes of PV and SST neurons. The empirically defined F-I curves were incorporated into a three-population computational model that simulated the empirically derived firing rates of pyramidal, PV, and SST neurons. Simulations revealed that the intrinsic properties were sufficient to predict that PV neurons are primarily responsible for generating the nontrivial fixed points representing Up states. Simulations and analytical methods demonstrated that while the paradoxical effect is not obligatory in a model with two classes of inhibitory neurons, it is present in most regimes. Finally, experimental tests validated predictions of the model that the Pyr ↔ PV inhibitory loop is stronger than the Pyr ↔ SST loop.SIGNIFICANCE STATEMENT Many cortical computations, such as working memory, rely on the local recurrent excitatory connections that define cortical circuit motifs. Up states are among the best studied examples of neural dynamic regimes that rely on recurrent excitatory excitation. However, this positive feedback must be held in check by inhibition. To address the relative contribution of PV and SST neurons, we characterized the intrinsic input-output differences between these classes of inhibitory neurons and, using experimental and theoretical methods, show that the higher threshold and gain of PV leads to a dominant role in network stabilization.


Asunto(s)
Neuronas/fisiología , Potenciales de Acción , Animales , Simulación por Computador , Retroalimentación Fisiológica , Ratones , Modelos Neurológicos , Neuronas/química , Neuronas/clasificación , Optogenética , Parvalbúminas/análisis , Células Piramidales/química , Células Piramidales/fisiología , Somatostatina/análisis , Transfección
3.
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
4.
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
5.
J Neurosci ; 41(13): 2944-2963, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33593859

RESUMEN

Synchronous activity of cortical inhibitory interneurons expressing parvalbumin (PV) underlies expression of cortical γ rhythms. Paradoxically, deficient PV inhibition is associated with increased broadband γ power in the local field potential. Increased baseline broadband γ is also a prominent characteristic in schizophrenia and a hallmark of network alterations induced by NMDAR antagonists, such as ketamine. Whether enhanced broadband γ is a true rhythm, and if so, whether rhythmic PV inhibition is involved or not, is debated. Asynchronous and increased firing activities are thought to contribute to broadband power increases spanning the γ band. Using male and female mice lacking NMDAR activity specifically in PV neurons to model deficient PV inhibition, we here show that neuronal activity with decreased synchronicity is associated with increased prefrontal broadband γ power. Specifically, reduced spike time precision and spectral leakage of spiking activity because of higher firing rates (spike "contamination") affect the broadband γ band. Desynchronization was evident at multiple time scales, with reduced spike entrainment to the local field potential, reduced cross-frequency coupling, and fragmentation of brain states. Local application of S(+)-ketamine in (control) mice with intact NMDAR activity in PV neurons triggered network desynchronization and enhanced broadband γ power. However, our investigations suggest that disparate mechanisms underlie increased broadband γ power caused by genetic alteration of PV interneurons and ketamine-induced power increases in broadband γ. Our study confirms that enhanced broadband γ power can arise from asynchronous activities and demonstrates that long-term deficiency of PV inhibition can be a contributor.SIGNIFICANCE STATEMENT Brain oscillations are fundamental to the coordination of neuronal activity across neurons and structures. γ oscillations (30-80 Hz) have received particular attention through their association with perceptual and cognitive processes. Synchronous activity of inhibitory parvalbumin (PV) interneurons generates cortical γ oscillation, but, paradoxically, PV neuron deficiency is associated with increases in γ oscillations. We here reconcile this conundrum and show how deficient PV inhibition can lead to increased and asynchronous excitatory firing, contaminating the local field potential and manifesting as increased γ power. Thus, increased γ power does not always reflect a genuine rhythm. Further, we show that ketamine-induced γ increases are caused by separate network mechanisms.


Asunto(s)
Potenciales de Acción/fisiología , Encéfalo/metabolismo , Ritmo Gamma/fisiología , Interneuronas/metabolismo , Red Nerviosa/metabolismo , Animales , Química Encefálica/fisiología , Femenino , Interneuronas/química , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Red Nerviosa/química , Parvalbúminas/análisis , Parvalbúminas/metabolismo , Receptores de N-Metil-D-Aspartato/análisis , Receptores de N-Metil-D-Aspartato/metabolismo
6.
J Comp Neurol ; 529(11): 2827-2841, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33576496

RESUMEN

During development, the visual system maintains a high capacity for modification by expressing characteristics permissive for plasticity, enabling neural circuits to be refined by visual experience to achieve their mature form. This period is followed by the emergence of characteristics that stabilize the brain to consolidate for lifetime connections that were informed by experience. Attenuation of plasticity potential is thought to derive from an accumulation of plasticity-inhibiting characteristics that appear at ages beyond the peak of plasticity. Perineuronal nets (PNNs) are molecular aggregations that primarily surround fast-spiking inhibitory neurons called parvalbumin (PV) cells, which exhibit properties congruent with a plasticity inhibitor. In this study, we examined the development of PNNs and PV cells in the primary visual cortex of a highly visual mammal, and assessed the impact that 10 days of darkness had on both characteristics. Here, we show that labeling for PV expression emerges earlier and reaches adult levels sooner than PNNs. We also demonstrate that darkness, a condition known to enhance plasticity, significantly reduces the density of PNNs and the size of PV cell somata but does not alter the number of PV cells in the visual cortex. The darkness-induced reduction of PV cell size occurred irrespective of whether neurons were surrounded by a PNN, suggesting that PNNs have a restricted capacity to inhibit plasticity. Finally, we show that PV cells surrounded by a PNN were significantly larger than those without one, supporting the view that PNNs may mediate trophic support to the cells they surround.


Asunto(s)
Oscuridad , Red Nerviosa/crecimiento & desarrollo , Neuronas/fisiología , Parvalbúminas/fisiología , Corteza Visual Primaria/crecimiento & desarrollo , Factores de Edad , Animales , Gatos , Red Nerviosa/química , Neuronas/química , Parvalbúminas/análisis , Corteza Visual Primaria/química , Corteza Visual Primaria/citología
7.
Food Chem ; 337: 127986, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-32920269

RESUMEN

We have developed a novel approach that involves inception-resnet network (IRN) modeling based on infrared spectroscopy (IR) for rapid and specific detection of the fish allergen parvalbumin. SDS-PAGE and ELISA were used to validate the new method. Through training and learning with parvalbumin IR spectra from 16 fish species, IRN, support vector machine (SVM), and random forest (RF) models were successfully established and compared. The IRN model extracted highly representative features from the IR spectra, leading to high accuracy in recognizing parvalbumin (up to 97.3%) in a variety of seafood matrices. The proposed infrared spectroscopic IRN (IR-IRN) method was rapid (~20 min, cf. ELISA ~4 h) and required minimal expert knowledge for application. Thus, it could be extended for large-scale field screening and identification of parvalbumin or other potential allergens in complex food matrices.


Asunto(s)
Productos Pesqueros/análisis , Proteínas de Peces/análisis , Redes Neurales de la Computación , Parvalbúminas/análisis , Espectrofotometría Infrarroja/estadística & datos numéricos , Alérgenos/química , Animales , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Peces/inmunología , Análisis de los Alimentos/métodos , Análisis de los Alimentos/estadística & datos numéricos , Hipersensibilidad a los Alimentos , Ratones Endogámicos BALB C , Parvalbúminas/inmunología , Reproducibilidad de los Resultados , Espectrofotometría Infrarroja/métodos , Máquina de Vectores de Soporte
8.
FASEB J ; 34(9): 12239-12254, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-33000527

RESUMEN

α-Synuclein (α-syn)-induced neurotoxicity has been generally accepted as a key step in the pathogenesis of Parkinson's disease (PD). Microtubule-associated protein tau, which is considered second only to α-syn, has been repeatedly linked with PD in association studies. However, the underlying interaction between these two PD-related proteins in vivo remains unclear. To investigate how the expression of tau affects α-syn-induced neurodegeneration in vivo, we generated triple transgenic mice that overexpressed α-syn A53T mutation in the midbrain dopaminergic neurons (mDANs) with different expression levels of tau. Here, we found that tau had no significant effect on the A53T α-syn-mediated mDANs degeneration. However, tau knockout could modestly promote the formation of α-syn aggregates, accelerate the severe and progressive degeneration of parvalbumin-positive (PV+) neurons in substantia nigra pars reticulata (SNR), accompanied with anxiety-like behavior in aged PD-related α-syn A53T mice. The mechanisms may be associated with A53T α-syn-mediated specifically successive impairment of N-methyl-d-aspartate receptor subunit 2B (NR2B), postsynaptic density-95 (PSD-95) and microtubule-associated protein 1A (MAP1A) in PV+ neurons. Our study indicates that MAP1A may play a beneficial role in preserving the survival of PV+ neurons, and that inhibition of the impairment of NR2B/PSD-95/MAP1A pathway, may be a novel and preferential option to ameliorate α-syn-induced neurodegeneration.


Asunto(s)
Mutación , Degeneración Nerviosa , Enfermedad de Parkinson/etiología , Parvalbúminas/análisis , Sustancia Negra/patología , alfa-Sinucleína/genética , Proteínas tau/fisiología , Animales , Homólogo 4 de la Proteína Discs Large/fisiología , Proteínas de Homeodominio/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/fisiología , Enfermedad de Parkinson/patología , Fragmentos de Péptidos/fisiología , Agregado de Proteínas , Receptores de N-Metil-D-Aspartato/fisiología , Factores de Transcripción/fisiología , alfa-Sinucleína/fisiología , Proteínas tau/química , Proteínas tau/genética
9.
Cereb Cortex ; 30(12): 6108-6120, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-32676666

RESUMEN

Chronic symptoms indicating excess cortical excitability follow mild traumatic brain injury, particularly repetitive mild traumatic brain injury (rmTBI). Yet mechanisms underlying post-traumatic excitation/inhibition (E/I) ratio abnormalities may differ between the early and late post-traumatic phases. We therefore measured seizure threshold and cortical gamma-aminobutyric acid (GABA) and glutamate (Glu) concentrations, 1 and 6 weeks after rmTBI in mice. We also analyzed the structure of parvalbumin-positive interneurons (PVIs), their perineuronal nets (PNNs), and their electroencephalography (EEG) signature (gamma frequency band power). For mechanistic insight, we measured cortical oxidative stress, reflected in the reduced/oxidized glutathione (GSH/GSSG) ratio. We found that seizure susceptibility increased both early and late after rmTBI. However, whereas increased Glu dominated the E/I 1 week after rmTBI, Glu concentration normalized and the E/I was instead characterized by depressed GABA, reduced per-PVI parvalbumin expression, and reduced gamma EEG power at the 6-week post-rmTBI time point. Oxidative stress was increased early after rmTBI, where transient PNN degradation was noted, and progressed throughout the monitoring period. We conclude that GSH depletion, perhaps triggered by early Glu-mediated excitotoxicity, leads to late post-rmTBI loss of PVI-dependent cortical inhibitory tone. We thus propose dampening of Glu signaling, maintenance of redox state, and preservation of PVI inhibitory capacity as therapeutic targets for post-rmTBI treatment.


Asunto(s)
Conmoción Encefálica/complicaciones , Encéfalo/fisiopatología , Ácido Glutámico/metabolismo , Interneuronas/fisiología , Estrés Oxidativo , Convulsiones/fisiopatología , Ácido gamma-Aminobutírico/metabolismo , Animales , Encéfalo/metabolismo , Ritmo Gamma , Masculino , Ratones Endogámicos C57BL , Parvalbúminas/análisis , Convulsiones/etiología , Convulsiones/metabolismo
10.
Fungal Biol ; 124(6): 592-600, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32448450

RESUMEN

Parvalbumins play crucial physiological roles in neuromuscular systems of vertebrates, such as cell-cycle, development of neurons, contraction of muscles, and regulation of intracellular calcium. To perform these neuromuscular functions, parvalbumin may be in associated with other proteins including calbindin, carbonic anhydrase, and cytochrome oxidase. Humans may show an IgE-specific hypersensitivity to parvalbumins after consumption of some distinct fish species. While this protein is abundant in fish muscles, literature review of publications related to fish parvalbumins, do not point to the presence of parvalbumins in eukaryotic microbes. In this study, we propose that distantly related parvalbumins may be found in some non-fish species. Bioinformatics studies such as multiple sequence alignment (MSA), phylogenetic analysis as well as molecular-based experiments indicate that, at least two parvalbumins sequences (UniProt IDs: A0A178F775 and A0A178F7E4) with EF-hand domains and Ca2+-binding sites could be identified in Trichophyton violaceum, a pathogenic fungal species. It was determined that both genes consisted of a single exon and encoded for parvalbumin proteins possessing conserved amino acid motifs. Antigenicity prediction revealed antigenic sites located in both sides of the Ca2+-binding site of the first EF-hand domain. Our phylogenetic analysis revealed that one of parvalbumins (UniProt ID: 0A178F775) can be evolved to other parvalbumins in T. violaceum (UniProt ID: A0A178F7E4) and fish species through evolutionary phenomenon. To confirm our in-silico findings, we designed three primer pairs to detect one of the T. violaceum parvalbumins (UniProt ID: A0A178F7E4) by polymerase chain reaction (PCR); one primer pair showed a strong and specific band in agarose gel electrophoresis. To evaluate the specificity of the method, the primers were tested on extracted DNA from Trichophyton rubrum and T. mentagrophytes. The results demonstrated that the evaluated parvalbumin gene (UniProt ID: A0A178F7E4) was T. violaceum-specific and this pathogenic fungus can be differentiated from T. rubrum and T. mentagrophytes through identification of parvalbumin genes. Further studies are necessary to unravel the biochemical and physiological functions of parvalbumins in T. violaceum.


Asunto(s)
Arthrodermataceae/química , Arthrodermataceae/genética , Proteínas Fúngicas/genética , Parvalbúminas/genética , Animales , Antígenos Fúngicos , Evolución Molecular , Proteínas de Peces/química , Proteínas de Peces/genética , Peces , Proteínas Fúngicas/análisis , Proteínas Fúngicas/química , Proteínas Fúngicas/inmunología , Genes Fúngicos , Parvalbúminas/análisis , Parvalbúminas/química , Parvalbúminas/inmunología , Filogenia
11.
Brain Struct Funct ; 225(3): 1135-1152, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32266458

RESUMEN

The Kv3.1b potassium channel subunit, which facilitates the fast-spiking phenotype characteristic of parvalbumin (PV)-expressing inhibitory interneurons, is also expressed by subpopulations of excitatory neurons in macaque cortex. We have previously shown that V1 neurons expressing Kv3.1b but not PV or GABA were largely concentrated within layers 4Cα and 4B of V1, suggesting laminar or pathway specificity. In the current study, the distribution and pattern of co-immunoreactivity of GABA, PV, and Kv3.1b across layers in extrastriate cortical areas V2 and MT of the macaque monkey were measured using the same triple immunofluorescence labeling, confocal microscopy, and partially automated cell-counting strategies used in V1. For comparison, densities of the overall cell and neuronal populations were also measured for each layer of V2 and MT using tissue sections immunofluorescence labeled for the pan-neuronal marker NeuN. GABAergic neurons accounted for 14% of the total neuronal population in V2 and 25% in MT. Neurons expressing Kv3.1b but neither GABA nor PV were present in both areas. This subpopulation was most prevalent in the lowest subcompartment of layer 3, comprising 5% of the total neuronal population in layer 3C of both areas, and 41% and 36% of all Kv3.1b+ neurons in this layer in V2 and MT, respectively. The prevalence and laminar distribution of this subpopulation were remarkably consistent between V2 and MT and showed a striking similarity to the patterns observed previously in V1, suggesting a common contribution to the cortical circuit across areas.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Neuronas/metabolismo , Canales de Potasio Shaw/análisis , Corteza Visual/metabolismo , Animales , Recuento de Células , Femenino , Macaca fascicularis , Macaca nemestrina , Masculino , Parvalbúminas/análisis , Vías Visuales/metabolismo , Ácido gamma-Aminobutírico/análisis
12.
Brain Struct Funct ; 225(3): 909-923, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32144495

RESUMEN

Amygdala plays crucial roles in emotional learning. The lateral amygdala (LA) is the input station of the amygdala, where learning related plasticity occurs. The LA is cortical like in nature in terms of its cellular make up, composed of a majority of principal cells and a minority of interneurons with distinct subtypes defined by morphology, intrinsic electrophysiological properties and neurochemical expression profile. The specific functions served by LA interneuron subtypes remain elusive. This study aimed to elucidate the interneuron subtype mediating feedback inhibition. Electrophysiological evidence involving antidromic activation of recurrent LA circuitry via basolateral amygdala stimulation and paired recordings implicate low-threshold spiking interneurons in feedback inhibition. Recordings in somatostatin-cre animals crossed with tdtomato mice have revealed remarkable similarities between a subset of SOM+ interneurons and LTS interneurons. This study concludes that LTS interneurons, most of which are putatively SOM+, mediate feedback inhibition in the LA. Parallels with cortical areas and potential implications for information processing and plasticity are discussed.


Asunto(s)
Potenciales de Acción , Complejo Nuclear Basolateral/fisiología , Interneuronas/fisiología , Animales , Complejo Nuclear Basolateral/citología , Complejo Nuclear Basolateral/metabolismo , Interneuronas/citología , Interneuronas/metabolismo , Masculino , Glicoproteínas de Membrana , Parvalbúminas/análisis , Receptores de Interleucina-1 , Somatostatina/análisis
13.
Brain Struct Funct ; 225(3): 1019-1032, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32189114

RESUMEN

Williams syndrome (WS) is a rare neurodevelopmental disorder caused by the hemideletion of approximately 25-28 genes at 7q11.23. Its unusual social and cognitive phenotype is most strikingly characterized by the disinhibition of social behavior, in addition to reduced global IQ, with a relative sparing of language ability. Hypersociality and increased social approach behavior in WS may represent a unique inability to inhibit responses to specific social stimuli, which is likely associated with abnormalities of frontostriatal circuitry. The striatum is characterized by a diversity of interneuron subtypes, including inhibitory parvalbumin-positive interneurons (PV+) and excitatory cholinergic interneurons (Ch+). Animal model research has identified an important role for these specialized cells in regulating social approach behavior. Previous research in humans identified a depletion of interneuron subtypes associated with neuropsychiatric disorders. Here, we examined the density of PV+ and Ch+ interneurons in the striatum of 13 WS and neurotypical (NT) subjects. We found a significant reduction in the density of Ch+ interneurons in the medial caudate nucleus and nucleus accumbens, important regions receiving cortical afferents from the orbitofrontal and ventromedial prefrontal cortex, and circuitry involved in language and reward systems. No significant difference in the distribution of PV+ interneurons was found. The pattern of decreased Ch+ interneuron densities in WS differs from patterns of interneuron depletion found in other disorders.


Asunto(s)
Neuronas Colinérgicas/patología , Cuerpo Estriado/patología , Interneuronas/patología , Síndrome de Williams/patología , Adolescente , Adulto , Anciano , Colina O-Acetiltransferasa/análisis , Femenino , Humanos , Masculino , Persona de Mediana Edad , Parvalbúminas/análisis , Adulto Joven
14.
Brain Struct Funct ; 225(3): 935-954, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32107637

RESUMEN

Accumulation of amyloid ß oligomers (AßO) in Alzheimer's disease (AD) impairs hippocampal theta and gamma oscillations. These oscillations are important in memory functions and depend on distinct subtypes of hippocampal interneurons such as somatostatin-positive (SST) and parvalbumin-positive (PV) interneurons. Here, we investigated whether AßO causes dysfunctions in SST and PV interneurons by optogenetically manipulating them during theta and gamma oscillations in vivo in AßO-injected SST-Cre or PV-Cre mice. Hippocampal in vivo multi-electrode recordings revealed that optogenetic activation of channelrhodopsin-2 (ChR2)-expressing SST and PV interneurons in AßO-injected mice selectively restored AßO-induced reduction of the peak power of theta and gamma oscillations, respectively, and resynchronized CA1 pyramidal cell (PC) spikes. Moreover, SST and PV interneuron spike phases were resynchronized relative to theta and gamma oscillations, respectively. Whole-cell voltage-clamp recordings in CA1 PC in ex vivo hippocampal slices from AßO-injected mice revealed that optogenetic activation of SST and PV interneurons enhanced spontaneous inhibitory postsynaptic currents (IPSCs) selectively at theta and gamma frequencies, respectively. Furthermore, analyses of the stimulus-response curve, paired-pulse ratio, and short-term plasticity of SST and PV interneuron-evoked IPSCs ex vivo showed that AßO increased the initial GABA release probability to depress SST/PV interneuron's inhibitory input to CA1 PC selectively at theta and gamma frequencies, respectively. Our results reveal frequency-specific and interneuron subtype-specific presynaptic dysfunctions of SST and PV interneurons' input to CA1 PC as the synaptic mechanisms underlying AßO-induced impairments of hippocampal network oscillations and identify them as potential therapeutic targets for restoring hippocampal network oscillations in early AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Ritmo Gamma , Hipocampo/fisiología , Interneuronas/fisiología , Ritmo Teta , Péptidos beta-Amiloides/administración & dosificación , Animales , Ritmo Gamma/efectos de los fármacos , Técnicas de Sustitución del Gen , Hipocampo/efectos de los fármacos , Interneuronas/efectos de los fármacos , Ratones , Optogenética , Parvalbúminas/análisis , Somatostatina/análisis , Ritmo Teta/efectos de los fármacos
15.
J Neurosci ; 40(11): 2314-2331, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32005764

RESUMEN

Distinct components of working memory are coordinated by different classes of inhibitory interneurons in the PFC, but the role of cholecystokinin (CCK)-positive interneurons remains enigmatic. In humans, this major population of interneurons shows histological abnormalities in schizophrenia, an illness in which deficient working memory is a core defining symptom and the best predictor of long-term functional outcome. Yet, CCK interneurons as a molecularly distinct class have proved intractable to examination by typical molecular methods due to widespread expression of CCK in the pyramidal neuron population. Using an intersectional approach in mice of both sexes, we have succeeded in labeling, interrogating, and manipulating CCK interneurons in the mPFC. Here, we describe the anatomical distribution, electrophysiological properties, and postsynaptic connectivity of CCK interneurons, and evaluate their role in cognition. We found that CCK interneurons comprise a larger proportion of the mPFC interneurons compared with parvalbumin interneurons, targeting a wide range of neuronal subtypes with a distinct connectivity pattern. Phase-specific optogenetic inhibition revealed that CCK, but not parvalbumin, interneurons play a critical role in the retrieval of working memory. These findings shine new light on the relationship between cortical CCK interneurons and cognition and offer a new set of tools to investigate interneuron dysfunction and cognitive impairments associated with schizophrenia.SIGNIFICANCE STATEMENT Cholecystokinin-expressing interneurons outnumber other interneuron populations in key brain areas involved in cognition and memory, including the mPFC. However, they have proved intractable to examination as experimental techniques have lacked the necessary selectivity. To the best of our knowledge, the present study is the first to report detailed properties of cortical cholecystokinin interneurons, revealing their anatomical organization, electrophysiological properties, postsynaptic connectivity, and behavioral function in working memory.


Asunto(s)
Colecistoquinina/fisiología , Interneuronas/fisiología , Memoria a Corto Plazo/fisiología , Recuerdo Mental/fisiología , Corteza Prefrontal/fisiología , Animales , Conducta Apetitiva/fisiología , Aprendizaje Discriminativo/fisiología , Discriminación en Psicología/fisiología , Femenino , Genes Reporteros , Interneuronas/clasificación , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/análisis , Odorantes , Optogenética , Parvalbúminas/análisis , Técnicas de Placa-Clamp , Recompensa , Esquizofrenia/fisiopatología , Olfato/fisiología , Potenciales Sinápticos/fisiología
16.
Hear Res ; 388: 107883, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31981822

RESUMEN

Animal-studies associate age-related hearing loss (presbycusis) with decreasing number of spiral ganglion neurons (SGNs) in Rosenthal's canal (RC) of cochlea. The excitatory neurotransmitter for SGNs is glutamate (through its receptor NMDAR 2B), which can be neurotoxic through Ca2+ overload. Neurotoxicity is balanced by calcium-binding proteins (CBPs) like Parvalbumin (PV), which is the predominant CBP of the SGNs. To estimate the volume of the RC and total number of SGNs that are immunoreactive to PV and NMDAR 2B, we used unbiased stereology in 35 human cochleae derived from cadavers of persons from 2nd to 8th decade of life (subsequently statistically divided into two groups) and compared them to the total number of cresyl violet (CV) stained SGNs. We also estimated the volume of individual neurons and their nuclei. Regression analysis was made on estimated parameters against age. Hierarchical-cluster analysis was done on the neuronal against neuronal nuclear volumes.The average volume of the RC did not change with increasing age (p = 0.4115). The total number of SGNs (CV-stained and those separately expressing PV and NMDAR 2B) significantly decreased with age (p < 0.001). We identified three distinct populations of neurons on the basis of their volumes among SGNs. Thus, there is significant age-related decline in the total number of SGNs, which starts early in life. It may be due to ambient noise and inadequate neutralisation of excitotoxicity.


Asunto(s)
Envejecimiento/metabolismo , Neuronas/química , Parvalbúminas/análisis , Presbiacusia/metabolismo , Receptores de N-Metil-D-Aspartato/análisis , Ganglio Espiral de la Cóclea/química , Adolescente , Adulto , Factores de Edad , Anciano , Envejecimiento/patología , Benzoxazinas , Cadáver , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Neuronas/patología , Presbiacusia/patología , Ganglio Espiral de la Cóclea/patología , Coloración y Etiquetado , Adulto Joven
17.
Brain Struct Funct ; 225(1): 387-401, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31873798

RESUMEN

Inhibitory interneurons in the cerebral cortex contain specific proteins or peptides characteristic for a certain interneuron subtype. In mice, three biochemical markers constitute non-overlapping interneuron populations, which account for 80-90% of all inhibitory cells. These interneurons express parvalbumin (PV), somatostatin (SST), or vasoactive intestinal peptide (VIP). SST is not only a marker of a specific interneuron subtype, but also an important neuropeptide that participates in numerous biochemical and signalling pathways in the brain via somatostatin receptors (SSTR1-5). In the nervous system, SST acts as a neuromodulator and neurotransmitter affecting, among others, memory, learning, and mood. In the sensory cortex, the co-localisation of GABA and SST is found in approximately 30% of interneurons. Considering the importance of interactions between inhibitory interneurons in cortical plasticity and the possible GABA and SST co-release, it seems important to investigate the localisation of different SSTRs on cortical interneurons. Here, we examined the distribution of SSTR1-5 on barrel cortex interneurons containing PV, SST, or VIP. Immunofluorescent staining using specific antibodies was performed on brain sections from transgenic mice that expressed red fluorescence in one specific interneuron subtype (PV-Ai14, SST-Ai14, and VIP-Ai14 mice). SSTRs expression on PV, SST, and VIP interneurons varied among the cortical layers and we found two patterns of SSTRs distribution in L4 of barrel cortex. We also demonstrated that, in contrast to other interneurons, PV cells did not express SSTR2, but expressed other SSTRs. SST interneurons, which were not found to make chemical synapses among themselves, expressed all five SSTR subtypes.


Asunto(s)
Interneuronas/química , Receptores de Somatostatina/análisis , Corteza Somatosensorial/química , Animales , Interneuronas/citología , Interneuronas/metabolismo , Masculino , Ratones Transgénicos , Parvalbúminas/análisis , Receptores de Somatostatina/metabolismo , Corteza Somatosensorial/citología , Corteza Somatosensorial/metabolismo , Somatostatina/análisis , Péptido Intestinal Vasoactivo/análisis
18.
Anat Histol Embryol ; 48(4): 334-339, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31016783

RESUMEN

The olfactory bulb (OB) shows special characteristics in its phylogenetic cortical structure and synaptic pattern. In the OB, gamma-aminobutyric acid (GABA), as an inhibitory neurotransmitter, is secreted from GABAergic neurons which contain parvalbumin (a calcium-binding protein). Many studies on the distribution of parvalbumin-immunoreactive neurons in the rodent OB have been published but poorly reported in the avian OB. Therefore, in this study, we compared the structure of the OB and distribution of parvalbumin-immunoreactive neurons in the OB between the rat and pigeon using cresyl violet staining and immunohistochemistry for parvalbumin, respectively. Fundamentally, the pigeon OB showed layers like those of the rat OB; however, some layers were not clear like in the rat OB. Parvalbumin-immunoreactive neurons in the pigeon OB were predominantly distributed in the external plexiform layer like that in the rat OB; however, the neurons did not have long processes like those in the rat. Furthermore, parvalbumin-immunoreactive fibres were abundant in some layers; this finding was not shown in the rat OB. In brief, parvalbumin-immunoreactive neurons were found like those in the rat OB; however, parvalbumin-immunoreactive fibres were significantly abundant in the pigeon OB compared to those in the rat OB.


Asunto(s)
Columbidae/anatomía & histología , Bulbo Olfatorio/citología , Parvalbúminas/análisis , Ratas Sprague-Dawley/anatomía & histología , Animales , Benzoxazinas , Colorantes , Columbidae/metabolismo , Inmunohistoquímica/veterinaria , Masculino , Bulbo Olfatorio/química , Parvalbúminas/inmunología , Ratas , Ratas Sprague-Dawley/metabolismo , Coloración y Etiquetado/veterinaria
19.
Epilepsia ; 60(5): e52-e57, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30963545

RESUMEN

Cryptogenic temporal lobe epilepsy develops in the absence of identified brain injuries, infections, or structural malformations, and in these cases, an unidentified pre-existing abnormality may initiate febrile seizures, hippocampal sclerosis, and epilepsy. Although a role for GABAergic dysfunction in epilepsy is intuitively obvious, no causal relationship has been established. In this study, hippocampal GABA neurons were targeted for selective elimination to determine whether a focal hippocampal GABAergic defect in an otherwise normal brain can initiate cryptogenic temporal lobe epilepsy with hippocampal sclerosis. We used Stable Substance P-saporin conjugate (SSP-saporin) to target rat hippocampal GABA neurons, which selectively and constitutively express the neurokinin-1 receptors that internalize this neurotoxin. Bilateral and longitudinally extensive intrahippocampal microinjections of SSP-saporin caused no obvious behavioral effects for several days. However, starting ~4 days postinjection, rats exhibited episodes of immobilization, abnormal flurries of "wet-dog" shakes, and brief focal motor seizures characterized by facial automatisms and forepaw clonus. These clinically subtle behaviors stopped after ~4 days. Convulsive status epilepticus did not develop, and no deaths occurred. Months later, chronically implanted rats exhibited spontaneous focal motor seizures and extreme hippocampal sclerosis. These data suggest that hippocampal GABAergic dysfunction is epileptogenic and can produce the defining features of cryptogenic temporal lobe epilepsy.


Asunto(s)
Epilepsia del Lóbulo Temporal/inducido químicamente , Neuronas GABAérgicas/efectos de los fármacos , Hipocampo/efectos de los fármacos , Saporinas/toxicidad , Sustancia P/análogos & derivados , Animales , Enfermedad Crónica , Giro Dentado/química , Giro Dentado/efectos de los fármacos , Giro Dentado/patología , Hipocampo/química , Hipocampo/patología , Masculino , Parvalbúminas/análisis , Ratas , Ratas Sprague-Dawley , Saporinas/farmacología , Esclerosis , Sustancia P/farmacología , Sustancia P/toxicidad , Ácido gamma-Aminobutírico/fisiología
20.
J Neurosci ; 39(23): 4489-4510, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-30936240

RESUMEN

By virtue of their extensive axonal arborization and perisomatic synaptic targeting, cortical inhibitory parvalbumin (PV) cells strongly regulate principal cell output and plasticity and modulate experience-dependent refinement of cortical circuits during development. An interesting aspect of PV cell connectivity is its prolonged maturation time course, which is completed only by end of adolescence. The p75 neurotrophin receptor (p75NTR) regulates numerous cellular functions; however, its role on cortical circuit development and plasticity remains elusive, mainly because localizing p75NTR expression with cellular and temporal resolution has been challenging. By using RNAscope and a modified version of the proximity ligation assay, we found that p75NTR expression in PV cells decreases between the second and fourth postnatal week, at a time when PV cell synapse numbers increase dramatically. Conditional knockout of p75NTR in single PV neurons in vitro and in PV cell networks in vivo causes precocious formation of PV cell perisomatic innervation and perineural nets around PV cell somata, therefore suggesting that p75NTR expression modulates the timing of maturation of PV cell connectivity in the adolescent cortex. Remarkably, we found that PV cells still express p75NTR in adult mouse cortex of both sexes and that its activation is sufficient to destabilize PV cell connectivity and to restore cortical plasticity following monocular deprivation in vivo Together, our results show that p75NTR activation dynamically regulates PV cell connectivity, and represent a novel tool to foster brain plasticity in adults.SIGNIFICANCE STATEMENT In the cortex, inhibitory, GABA-releasing neurons control the output and plasticity of excitatory neurons. Within this diverse group, parvalbumin-expressing (PV) cells form the larger inhibitory system. PV cell connectivity develops slowly, reaching maturity only at the end of adolescence; however, the mechanisms controlling the timing of its maturation are not well understood. We discovered that the expression of the neurotrophin receptor p75NTR in PV cells inhibits the maturation of their connectivity in a cell-autonomous fashion, both in vitro and in vivo, and that p75NTR activation in adult PV cells promotes their remodeling and restores cortical plasticity. These results reveal a new p75NTR function in the regulation of the time course of PV cell maturation and in limiting cortical plasticity.


Asunto(s)
Envejecimiento/fisiología , Interneuronas/fisiología , Plasticidad Neuronal/fisiología , Receptores de Factor de Crecimiento Nervioso/fisiología , Maduración Sexual/fisiología , Corteza Visual/crecimiento & desarrollo , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Conectoma , Potenciales Evocados Visuales , Femenino , Neuronas GABAérgicas/citología , Regulación del Desarrollo de la Expresión Génica , Interneuronas/química , Interneuronas/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Parvalbúminas/análisis , Precursores de Proteínas/farmacología , Distribución Aleatoria , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Receptores de Factor de Crecimiento Nervioso/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Sinapsis/fisiología , Visión Monocular/fisiología , Corteza Visual/citología , Corteza Visual/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...