Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
CNS Neurosci Ther ; 30(6): e14782, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38828651

RESUMEN

BACKGROUND: The thalamus system plays critical roles in the regulation of reversible unconsciousness induced by general anesthetics, especially the arousal stage of general anesthesia (GA). But the function of thalamus in GA-induced loss of consciousness (LOC) is little known. The thalamic reticular nucleus (TRN) is the only GABAergic neurons-composed nucleus in the thalamus, which is composed of parvalbumin (PV) and somatostatin (SST)-expressing GABAergic neurons. The anterior sector of TRN (aTRN) is indicated to participate in the induction of anesthesia, but the roles remain unclear. This study aimed to reveal the role of the aTRN in propofol and isoflurane anesthesia. METHODS: We first set up c-Fos straining to monitor the activity variation of aTRNPV and aTRNSST neurons during propofol and isoflurane anesthesia. Subsequently, optogenetic tools were utilized to activate aTRNPV and aTRNSST neurons to elucidate the roles of aTRNPV and aTRNSST neurons in propofol and isoflurane anesthesia. Electroencephalogram (EEG) recordings and behavioral tests were recorded and analyzed. Lastly, chemogenetic activation of the aTRNPV neurons was applied to confirm the function of the aTRN neurons in propofol and isoflurane anesthesia. RESULTS: c-Fos straining showed that both aTRNPV and aTRNSST neurons are activated during the LOC period of propofol and isoflurane anesthesia. Optogenetic activation of aTRNPV and aTRNSST neurons promoted isoflurane induction and delayed the recovery of consciousness (ROC) after propofol and isoflurane anesthesia, meanwhile chemogenetic activation of the aTRNPV neurons displayed the similar effects. Moreover, optogenetic and chemogenetic activation of the aTRN neurons resulted in the accumulated burst suppression ratio (BSR) during propofol and isoflurane GA, although they represented different effects on the power distribution of EEG frequency. CONCLUSION: Our findings reveal that the aTRN GABAergic neurons play a critical role in promoting the induction of propofol- and isoflurane-mediated GA.


Asunto(s)
Anestesia General , Estado de Conciencia , Neuronas GABAérgicas , Isoflurano , Propofol , Propofol/farmacología , Isoflurano/farmacología , Animales , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/fisiología , Ratones , Estado de Conciencia/efectos de los fármacos , Estado de Conciencia/fisiología , Masculino , Electroencefalografía , Anestésicos por Inhalación/farmacología , Núcleos Talámicos Anteriores/efectos de los fármacos , Núcleos Talámicos Anteriores/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Anestésicos Intravenosos/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Optogenética
2.
Acta Pharmacol Sin ; 45(6): 1160-1174, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38438581

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) regulate pain pathways with various outcomes depending on receptor subtypes, neuron types, and locations. But it remains unknown whether α4ß2 nAChRs abundantly expressed in the substantia nigra pars reticulata (SNr) have potential to mitigate hyperalgesia in pain states. We observed that injection of nAChR antagonists into the SNr reduced pain thresholds in naïve mice, whereas injection of nAChR agonists into the SNr relieved hyperalgesia in mice, subjected to capsaicin injection into the lower hind leg, spinal nerve injury, chronic constriction injury, or chronic nicotine exposure. The analgesic effects of nAChR agonists were mimicked by optogenetic stimulation of cholinergic inputs from the pedunculopontine nucleus (PPN) to the SNr, but attenuated upon downregulation of α4 nAChRs on SNr GABAergic neurons and injection of dihydro-ß-erythroidine into the SNr. Chronic nicotine-induced hyperalgesia depended on α4 nAChRs in SNr GABAergic neurons and was associated with the reduction of ACh release in the SNr. Either activation of α4 nAChRs in the SNr or optogenetic stimulation of the PPN-SNr cholinergic projection mitigated chronic nicotine-induced hyperalgesia. Interestingly, mechanical stimulation-induced ACh release was significantly attenuated in mice subjected to either capsaicin injection into the lower hind leg or SNI. These results suggest that α4 nAChRs on GABAergic neurons mediate a cholinergic analgesic circuit in the SNr, and these receptors may be effective therapeutic targets to relieve hyperalgesia in acute and chronic pain, and chronic nicotine exposure.


Asunto(s)
Neuronas GABAérgicas , Hiperalgesia , Ratones Endogámicos C57BL , Receptores Nicotínicos , Animales , Receptores Nicotínicos/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/fisiología , Masculino , Hiperalgesia/metabolismo , Hiperalgesia/tratamiento farmacológico , Ratones , Porción Reticular de la Sustancia Negra/metabolismo , Porción Reticular de la Sustancia Negra/efectos de los fármacos , Nicotina/farmacología , Analgésicos/farmacología , Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Capsaicina/farmacología , Acetilcolina/metabolismo , Optogenética , Umbral del Dolor/efectos de los fármacos
3.
Elife ; 122023 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-37665123

RESUMEN

Cortical GABAergic interneurons (INs) represent a diverse population of mainly locally projecting cells that provide specialized forms of inhibition to pyramidal neurons and other INs. Most recent work on INs has focused on subtypes distinguished by expression of Parvalbumin (PV), Somatostatin (SST), or Vasoactive Intestinal Peptide (VIP). However, a fourth group that includes neurogliaform cells (NGFCs) has been less well characterized due to a lack of genetic tools. Here, we show that these INs can be accessed experimentally using intersectional genetics with the gene Id2. We find that outside of layer 1 (L1), the majority of Id2 INs are NGFCs that express high levels of neuropeptide Y (NPY) and exhibit a late-spiking firing pattern, with extensive local connectivity. While much sparser, non-NGFC Id2 INs had more variable properties, with most cells corresponding to a diverse group of INs that strongly expresses the neuropeptide CCK. In vivo, using silicon probe recordings, we observed several distinguishing aspects of NGFC activity, including a strong rebound in activity immediately following the cortical down state during NREM sleep. Our study provides insights into IN diversity and NGFC distribution and properties, and outlines an intersectional genetics approach for further study of this underappreciated group of INs.


Asunto(s)
Neuronas GABAérgicas , Interneuronas , Neuropéptidos , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Neuropéptido Y/metabolismo , Neuropéptidos/metabolismo , Parvalbúminas/metabolismo , Células Piramidales/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
4.
Nat Neurosci ; 26(9): 1529-1540, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37524978

RESUMEN

Fluctuations in reproductive hormone levels are associated with mood disruptions in women, such as in postpartum and perimenopausal depression. However, the neural circuit mechanisms remain unclear. Here we report that medial preoptic area (MPOA) GABAergic neurons mediate multifaceted depressive-like behaviors in female mice after ovarian hormone withdrawal (HW), which can be attributed to downregulation of activity in Esr1 (estrogen receptor-1)-expressing GABAergic neurons. Enhancing activity of these neurons ameliorates depressive-like behaviors in HW-treated mice, whereas reducing their activity results in expression of these behaviors. Two separate subpopulations mediate different symptoms: a subpopulation projecting to the ventral tegmental area (VTA) mediates anhedonia and another projecting to the periaqueductal gray mediates immobility. These projections enhance activity of dopaminergic neurons in the VTA and serotonergic neurons in the dorsal raphe, respectively, with increased release of dopamine and serotonin, possibly through disinhibition mechanisms. Thus, the MPOA is a hub that mediates depressive-like behaviors resulting from transitions in reproductive hormone levels.


Asunto(s)
Área Preóptica , Área Tegmental Ventral , Ratones , Femenino , Animales , Área Preóptica/fisiología , Área Tegmental Ventral/fisiología , Neuronas Dopaminérgicas/fisiología , Neuronas GABAérgicas/fisiología
5.
J Neuroendocrinol ; 35(6): e13302, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37280378

RESUMEN

Polycystic ovary syndrome (PCOS) is a female endocrine disorder that is associated with prenatal exposure to excess androgens. In prenatally androgenized (PNA) mice that model PCOS, GABAergic neural transmission to and innervation of GnRH neurons is increased. Evidence suggests that elevated GABAergic innervation originates in the arcuate nucleus (ARC). We hypothesized that GABA-GnRH circuit abnormalities are a direct consequence of PNA, resulting from DHT binding to androgen receptor (AR) in the prenatal brain. However, whether prenatal ARC neurons express AR at the time of PNA treatment is presently unknown. We used RNAScope in situ hybridization to localize AR mRNA (Ar)-expressing cells in healthy gestational day (GD) 17.5 female mouse brains and to assess coexpression levels in specific neuronal phenotypes. Our study revealed that less than 10% of ARC GABA cells expressed Ar. In contrast, we found that ARC kisspeptin neurons, critical regulators of GnRH neurons, were highly colocalized with Ar. Approximately 75% of ARC Kiss1-expressing cells also expressed Ar at GD17.5, suggesting that ARC kisspeptin neurons are potential targets of PNA. Investigating other neuronal populations in the ARC we found that ~50% of pro-opiomelanocortin (Pomc) cells, 22% of tyrosine hydroxylase (Th) cells, 8% of agouti-related protein (Agrp) cells and 8% of somatostatin (Sst) cells express Ar. Lastly, RNAscope in coronal sections showed Ar expression in the medial preoptic area (mPOA), and the ventral part of the lateral septum (vLS). These Ar-expressing regions were highly GABAergic, and 22% of GABA cells in the mPOA and 25% of GABA cells in the vLS also expressed Ar. Our findings identify specific neuronal phenotypes in the ARC, mPOA, and vLS that are androgen sensitive in late gestation. PNA-induced functional changes in these neurons may be related to the development of impaired central mechanisms associated with PCOS-like features.


Asunto(s)
Andrógenos , Síndrome del Ovario Poliquístico , Humanos , Ratones , Femenino , Embarazo , Animales , Andrógenos/metabolismo , Receptores Androgénicos/metabolismo , Kisspeptinas/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas GABAérgicas/fisiología , Encéfalo/metabolismo , Virilismo/metabolismo
6.
Front Neural Circuits ; 17: 1122314, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37035504

RESUMEN

Fear learning and memory rely on dynamic interactions between the excitatory and inhibitory neuronal populations that make up the prefrontal cortical, amygdala, and hippocampal circuits. Whereas inhibition of excitatory principal cells (PCs) by GABAergic neurons restrains their excitation, inhibition of GABAergic neurons promotes the excitation of PCs through a process called disinhibition. Specifically, GABAergic interneurons that express parvalbumin (PV+) and somatostatin (SOM+) provide inhibition to different subcellular domains of PCs, whereas those that express the vasoactive intestinal polypeptide (VIP+) facilitate disinhibition of PCs by inhibiting PV+ and SOM+ interneurons. Importantly, although the main connectivity motifs and the underlying network functions of PV+, SOM+, and VIP+ interneurons are replicated across cortical and limbic areas, these inhibitory populations play region-specific roles in fear learning and memory. Here, we provide an overview of the fear processing in the amygdala, hippocampus, and prefrontal cortex based on the evidence obtained in human and animal studies. Moreover, focusing on recent findings obtained using genetically defined imaging and intervention strategies, we discuss the population-specific functions of PV+, SOM+, and VIP+ interneurons in fear circuits. Last, we review current insights that integrate the region-specific inhibitory and disinhibitory network patterns into fear memory acquisition and fear-related disorders.


Asunto(s)
Interneuronas , Aprendizaje , Animales , Humanos , Aprendizaje/fisiología , Interneuronas/fisiología , Miedo/fisiología , Memoria , Neuronas GABAérgicas/fisiología , Parvalbúminas , Péptido Intestinal Vasoactivo
7.
Neurosci Res ; 178: 33-40, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35189175

RESUMEN

GABAergic neurons are classified into multiple subtypes based on morphology, physiological properties, and gene expression profiles. Although traditionally defined axo-axonic cells (AACs) are a unique type of interneuron that expresses parvalbumin and innervates the axon initial segment (AIS) of pyramidal neurons, a genetic marker for AACs in the basolateral amygdala (BLA) has not been identified. Here, we show that vasoactive intestinal peptide receptor 2 (Vipr2)-expressing interneurons exhibit anatomical and electrophysiological properties of AACs in the BLA. Using a reporter mouse expressing fluorescent proteins specifically in Vipr2+ cells, we analyzed the distribution, postsynaptic targeting and electrophysical properties of Vipr2+ cells in the BLA. More than half of the Vipr2+ cells showed parvalbumin immunoreactivity and innervated the AIS of pyramidal neurons in the BLA of Vipr2-tdTomato mice. Notably, most of the Vipr2+ cells showed fast-spiking properties. Furthermore, the use of a Cre-dependent adeno-associated virus led to more selective labeling of AACs in the BLA. These results suggest that AACs are genetically identifiable in the BLA without anatomical or physiological analysis.


Asunto(s)
Complejo Nuclear Basolateral , Animales , Axones/metabolismo , Complejo Nuclear Basolateral/metabolismo , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Ratones , Parvalbúminas/genética , Parvalbúminas/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo
8.
Science ; 375(6579): eabk2346, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35084970

RESUMEN

The human cortex contains inhibitory interneurons derived from the medial ganglionic eminence (MGE), a germinal zone in the embryonic ventral forebrain. How this germinal zone generates sufficient interneurons for the human brain remains unclear. We found that the human MGE (hMGE) contains nests of proliferative neuroblasts with ultrastructural and transcriptomic features that distinguish them from other progenitors in the hMGE. When dissociated hMGE cells are transplanted into the neonatal mouse brain, they reform into nests containing proliferating neuroblasts that generate young neurons that migrate extensively into the mouse forebrain and mature into different subtypes of functional interneurons. Together, these results indicate that the nest organization and sustained proliferation of neuroblasts in the hMGE provide a mechanism for the extended production of interneurons for the human forebrain.


Asunto(s)
Interneuronas/fisiología , Eminencia Media/embriología , Células-Madre Neurales/fisiología , Neurogénesis , Prosencéfalo/citología , Animales , Animales Recién Nacidos , Movimiento Celular , Proliferación Celular , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/fisiología , Perfilación de la Expresión Génica , Edad Gestacional , Humanos , Interneuronas/citología , Eminencia Media/citología , Eminencia Media/crecimiento & desarrollo , Ratones , Células-Madre Neurales/trasplante , Prosencéfalo/embriología , Prosencéfalo/crecimiento & desarrollo , Trasplante Heterólogo
9.
Biochim Biophys Acta Mol Cell Res ; 1869(1): 119146, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34599984

RESUMEN

Gaba-ergic neurons are a diverse cell class with extensive influence over cortical processing, but their role in experience-dependent plasticity is not completely understood. Here we addressed the role of cortical somatostatin- (SOM-INs) and vasoactive intestinal polypeptide- (VIP-INs) containing interneurons in a Pavlovian conditioning where stimulation of the vibrissae is used as a conditioned stimulus and tail shock as unconditioned one. This procedure induces a plastic change observed as an enlargement of the cortical functional representation of vibrissae activated during conditioning. Using layer-targeted, cell-selective DREADD transductions, we examined the involvement of SOM-INs and VIP-INs activity in learning-related plastic changes. Under optical recordings, we injected DREADD-expressing vectors into layer IV (L4) barrels or layer II/III (L2/3) areas corresponding to the activated vibrissae. The activity of the interneurons was modulated during all conditioning sessions, and functional 2-deoxyglucose (2DG) maps were obtained 24 h after the last session. In mice with L4 but not L2/3 SOM-INs suppressed during conditioning, the plastic change of whisker representation was absent. The behavioral effect of conditioning was disturbed. Both L4 SOM-INs excitation and L2/3 VIP-INs inhibition during conditioning did not affect the plasticity or the conditioned response. We found the activity of L4 SOM-INs is indispensable in the formation of learning-induced plastic change. We propose that L4 SOM-INs may provide disinhibition by blocking L4 parvalbumin interneurons, allowing a flow of information into upper cortical layers during learning.


Asunto(s)
Interneuronas/fisiología , Aprendizaje , Inhibición Neural , Plasticidad Neuronal , Corteza Somatosensorial/fisiología , Animales , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Moduladores del Transporte de Membrana/farmacología , Ratones , Corteza Somatosensorial/citología , Somatostatina/genética , Somatostatina/metabolismo , Vibrisas/inervación , Vibrisas/fisiología
10.
J Neurophysiol ; 126(6): 2119-2129, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34817244

RESUMEN

Neuroimmune signaling is increasingly identified as a critical component of various illnesses, including chronic pain, substance use disorder, and depression. However, the underlying neural mechanisms remain unclear. Proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α), may play a role by modulating synaptic function and long-term plasticity. The midbrain structure periaqueductal gray (PAG) plays a well-established role in pain processing, and although TNF-α inhibitors have emerged as a therapeutic strategy for pain-related disorders, the impact of TNF-α on PAG neuronal activity has not been thoroughly characterized. Recent studies have identified subpopulations of ventrolateral PAG (vlPAG) with opposing effects on nociception, with dopamine (DA) neurons driving pain relief in contrast to GABA neurons. Therefore, we used slice physiology to examine the impact of TNF-α on neuronal activity of both these subpopulations. We focused on female mice since the PAG is a sexually dimorphic region and most studies use male subjects, limiting our understanding of mechanistic variations in females. We selectively targeted GABA and DA neurons using transgenic reporter lines. Following exposure to TNF-α, there was an increase in excitability of GABA neurons along with a reduction in glutamatergic synaptic transmission. In DA neurons, TNF-α exposure resulted in a robust decrease in excitability along with a modest reduction in glutamatergic synaptic transmission. Interestingly, TNF-α had no effect on inhibitory transmission onto DA neurons. Collectively, these data suggest that TNF-α differentially affects the function of GABA and DA neurons in female mice and enhances our understanding of how TNF-α-mediated signaling modulates vlPAG function.NEW & NOTEWORTHY This study describes the effects of TNF-α on two distinct subpopulations of neurons in the vlPAG. We show that TNF-α alters both neuronal excitability and glutamatergic synaptic transmission on GABA and dopamine neurons within the vlPAG of female mice. This provides critical new information on the role of TNF-α in the potential modulation of pain, since activation of vlPAG GABA neurons drives nociception, whereas activation of dopamine neurons drives analgesia.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Neuronas GABAérgicas/fisiología , Sustancia Gris Periacueductal/fisiología , Transmisión Sináptica/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Neuronas Dopaminérgicas/efectos de los fármacos , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Sustancia Gris Periacueductal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
11.
Pharmacol Rep ; 73(4): 1096-1108, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34426901

RESUMEN

The role of adenosine A2A receptor (A2AR) and striatal-enriched protein tyrosine phosphatase (STEP) interactions in the striatal-pallidal GABA neurons was recently discussed in relation to A2AR overexpression and cocaine-induced increases of brain adenosine levels. As to phosphorylation, combined activation of A2AR and metabotropic glutamate receptor 5 (mGluR5) in the striatal-pallidal GABA neurons appears necessary for phosphorylation of the GluA1 unit of the AMPA receptor to take place. Robert Yasuda (J Neurochem 152: 270-272, 2020) focused on finding a general mechanism by which STEP activation is enhanced by increased A2AR transmission in striatal-pallidal GABA neurons expressing A2AR and dopamine D2 receptor. In his Editorial, he summarized in a clear way the significant effects of A2AR activation on STEP in the dorsal striatal-pallidal GABA neurons which involves a rise of intracellular levels of calcium causing STEP activation through its dephosphorylation. However, the presence of the A2AR in an A2AR-fibroblast growth factor receptor 1 (FGFR1) heteroreceptor complex can be required in the dorsal striatal-pallidal GABA neurons for the STEP activation. Furthermore, Won et al. (Proc Natl Acad Sci USA 116: 8028-8037, 2019) found in mass spectrometry experiments that the STEP splice variant STEP61 can bind to mGluR5 and inactivate it. In addition, A2AR overexpression can lead to increased formation of A2AR-mGluR5 heterocomplexes in ventral striatal-pallidal GABA neurons. It involves enhanced facilitatory allosteric interactions leading to increased Gq-mediated mGluR5 signaling activating STEP. The involvement of both A2AR and STEP in the actions of cocaine on synaptic downregulation was also demonstrated. The enhancement of mGluR5 protomer activity by the A2AR protomer in A2AR-mGluR5 heterocomplexes in the nucleus accumbens shell appears to have a novel significant role in STEP mechanisms by both enhancing the activation of STEP and being a target for STEP61.


Asunto(s)
Neuronas GABAérgicas/fisiología , Fosforilación/genética , Fosforilación/fisiología , Células del Asta Posterior/fisiología , Receptor de Adenosina A2A/metabolismo , Animales , Cocaína/farmacología , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/patología , Neuronas GABAérgicas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Células del Asta Posterior/efectos de los fármacos , Subunidades de Proteína/efectos de los fármacos , Proteínas Tirosina Fosfatasas/genética , Receptor de Adenosina A2A/genética , Receptor del Glutamato Metabotropico 5/genética
12.
J Neuroendocrinol ; 33(8): e12999, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34216402

RESUMEN

Although polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility worldwide, the aetiology of the disorder remains poorly defined. Animal-based evidence highlights the brain as a prime suspect in both the development and maintenance of PCOS. Prenatally androgenised (PNA) models of PCOS exhibit excessive GABAergic wiring associated with PCOS-like reproductive deficits in adulthood, with aberrant brain wiring detected as early as postnatal day (P) 25, prior to disease onset, in the PNA mouse. The mechanisms underlying this aberrant brain wiring remain unknown. Microglia, the immune cells of the brain, are regulators of neuronal wiring across development, mediating both the formation and removal of neuronal inputs. Here, we tested the hypothesis that microglia play a role in the excessive GABAergic wiring that leads to PCOS-like features in the PNA brain. Using specific immunolabelling, microglia number and morphology associated with activation states were analysed in PNA and vehicle-treated controls across developmental timepoints, including embryonic day 17.5, P0, P25 and P60 (n = 7-14 per group), and in two regions of the hypothalamus implicated in fertility regulation. At P0, fewer amoeboid microglia were observed in the rostral preoptic area (rPOA) of PNA mice. However, the greatest changes were observed at P25, with PNA mice exhibiting fewer total microglia, and specifically fewer "sculpting" microglia, in the rPOA. Based on these findings, we assessed microglia-mediated refinement of GABAergic synaptic terminals at two developmental stages of peak synaptic refinement: P7 and P15 (n = 7 per group). PNA mice showed a reduction in the uptake of GABAergic synaptic material at P15. These findings reveal time-specific changes in the microglia population and refinement of GABAergic inputs in a mouse model of PCOS driven by prenatal androgen excess and suggest a role for microglia in shaping the atypical brain wiring associated with the development of PCOS features.


Asunto(s)
Encéfalo/patología , Microglía/fisiología , Síndrome del Ovario Poliquístico/psicología , Animales , Modelos Animales de Enfermedad , Femenino , Neuronas GABAérgicas/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Síndrome del Ovario Poliquístico/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/patología , Efectos Tardíos de la Exposición Prenatal/psicología
13.
Nat Commun ; 12(1): 3321, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-34059669

RESUMEN

Autism spectrum disorder (ASD) is a common neurodevelopmental disorder. The mechanisms underlying ASD are unclear. Astrocyte alterations are noted in ASD patients and animal models. However, whether astrocyte dysfunction is causal or consequential to ASD-like phenotypes in mice is unresolved. Type 2 inositol 1,4,5-trisphosphate 6 receptors (IP3R2)-mediated Ca2+ release from intracellular Ca2+ stores results in the activation of astrocytes. Mutations of the IP3R2 gene are associated with ASD. Here, we show that both IP3R2-null mutant mice and astrocyte-specific IP3R2 conditional knockout mice display ASD-like behaviors, such as atypical social interaction and repetitive behavior. Furthermore, we show that astrocyte-derived ATP modulates ASD-like behavior through the P2X2 receptors in the prefrontal cortex and possibly through GABAergic synaptic transmission. These findings identify astrocyte-derived ATP as a potential molecular player in the pathophysiology of ASD.


Asunto(s)
Adenosina Trifosfato/metabolismo , Astrocitos/patología , Trastorno del Espectro Autista/patología , Señalización del Calcio/fisiología , Receptores de Inositol 1,4,5-Trifosfato/deficiencia , Animales , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Conducta Animal , Calcio/metabolismo , Modelos Animales de Enfermedad , Neuronas GABAérgicas/fisiología , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Masculino , Ratones , Ratones Noqueados , Corteza Prefrontal/citología , Corteza Prefrontal/patología , Corteza Prefrontal/fisiopatología , Transmisión Sináptica/fisiología
14.
Epilepsia ; 62(3): 683-697, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33617692

RESUMEN

OBJECTIVE: This study was undertaken to investigate how gain of function (GOF) of slack channel due to a KCNT1 pathogenic variant induces abnormal neuronal cortical network activity and generates specific electroencephalographic (EEG) patterns of epilepsy in infancy with migrating focal seizures. METHODS: We used detailed microscopic computational models of neurons to explore the impact of GOF of slack channel (explicitly coded) on each subtype of neurons and on a cortical micronetwork. Then, we adapted a thalamocortical macroscopic model considering results obtained in detailed models and immature properties related to epileptic brain in infancy. Finally, we compared simulated EEGs resulting from the macroscopic model with interictal and ictal patterns of affected individuals using our previously reported EEG markers. RESULTS: The pathogenic variants of KCNT1 strongly decreased the firing rate properties of γ-aminobutyric acidergic (GABAergic) interneurons and, to a lesser extent, those of pyramidal cells. This change led to hyperexcitability with increased synchronization in a cortical micronetwork. At the macroscopic scale, introducing slack GOF effect resulted in epilepsy of infancy with migrating focal seizures (EIMFS) EEG interictal patterns. Increased excitation-to-inhibition ratio triggered seizure, but we had to add dynamic depolarizing GABA between somatostatin-positive interneurons and pyramidal cells to obtain migrating seizure. The simulated migrating seizures were close to EIMFS seizures, with similar values regarding the delay between the different ictal activities (one of the specific EEG markers of migrating focal seizures due to KCNT1 pathogenic variants). SIGNIFICANCE: This study illustrates the interest of biomathematical models to explore pathophysiological mechanisms bridging the gap between the functional effect of gene pathogenic variants and specific EEG phenotype. Such models can be complementary to in vitro cellular and animal models. This multiscale approach provides an in silico framework that can be further used to identify candidate innovative therapies.


Asunto(s)
Epilepsia/genética , Neuronas GABAérgicas/fisiología , Proteínas del Tejido Nervioso/genética , Canales de potasio activados por Sodio/genética , Convulsiones/genética , Simulación por Computador , Electroencefalografía , Epilepsia/etiología , Epilepsia/fisiopatología , Mutación con Ganancia de Función/genética , Humanos , Lactante , Convulsiones/etiología , Convulsiones/fisiopatología
15.
Neuron ; 109(5): 778-787.e3, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33472037

RESUMEN

Fast synaptic transmission relies upon the activation of ionotropic receptors by neurotransmitter release to evoke postsynaptic potentials. Glutamate and GABA play dominant roles in driving highly dynamic activity in synaptically connected neuronal circuits, but ionotropic receptors for other neurotransmitters are also expressed in the neocortex, including nicotinic receptors, which are non-selective cation channels gated by acetylcholine. To study the function of non-glutamatergic excitation in neocortex, we used two-photon microscopy to target whole-cell membrane potential recordings to different types of genetically defined neurons in layer 2/3 of primary somatosensory barrel cortex in awake head-restrained mice combined with pharmacological and optogenetic manipulations. Here, we report a prominent nicotinic input, which selectively depolarizes a subtype of GABAergic neuron expressing vasoactive intestinal peptide leading to disinhibition during active sensorimotor processing. Nicotinic disinhibition of somatosensory cortex during active sensing might contribute importantly to integration of top-down and motor-related signals necessary for tactile perception and learning.


Asunto(s)
Neuronas GABAérgicas/fisiología , Receptores Nicotínicos/fisiología , Corteza Somatosensorial/fisiología , Percepción del Tacto/fisiología , Animales , Conducta Animal , Femenino , Masculino , Potenciales de la Membrana , Neuronas/fisiología , Optogenética , Tacto/fisiología , Péptido Intestinal Vasoactivo/análisis , Vibrisas/fisiología
16.
Mol Psychiatry ; 26(7): 3093-3107, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33087855

RESUMEN

Alcohol use disorder (AUD) and anxiety disorders are frequently comorbid and share mechanisms that could be therapeutic targets. To facilitate mechanistic studies, we adapted an inhibitory avoidance-based "2-hit" rat model of posttraumatic stress disorder (PTSD) and identified predictors and biomarkers of comorbid alcohol (ethanol)/PTSD-like symptoms in these animals. Stressed Wistar rats received a single footshock on two occasions. The first footshock occurred when rats crossed into the dark chamber of a shuttle box. Forty-eight hours later, rats received the second footshock in a familiar (FAM) or novel (NOV) context. Rats then received 4 weeks of two-bottle choice (2BC) ethanol access. During subsequent abstinence, PTSD-like behavior responses, GABAergic synaptic transmission in the central amygdala (CeA), and circulating cytokine levels were measured. FAM and NOV stress more effectively increased 2BC drinking in males and females, respectively. Stressed male rats, especially drinking-vulnerable individuals (≥0.8 g/kg average 2-h ethanol intake with >50% ethanol preference), showed higher fear overgeneralization in novel contexts, increased GABAergic transmission in the CeA, and a profile of increased G-CSF, GM-CSF, IL-13, IL-6, IL-17a, leptin, and IL-4 that discriminated between stress context (NOV > FAM > Control). However, drinking-resilient males showed the highest G-CSF, IL-13, and leptin levels. Stressed females showed increased acoustic startle and decreased sleep maintenance, indicative of hyperarousal, with increased CeA GABAergic transmission in NOV females. This paradigm promotes key features of PTSD, including hyperarousal, fear generalization, avoidance, and sleep disturbance, with comorbid ethanol intake, in a sex-specific fashion that approximates clinical comorbidities better than existing models, and identifies increased CeA GABAergic signaling and a distinct pro-hematopoietic, proinflammatory, and pro-atopic cytokine profile that may aid in treatment.


Asunto(s)
Alcoholismo , Citocinas/sangre , Neuronas GABAérgicas/fisiología , Factores Sexuales , Trastornos por Estrés Postraumático , Transmisión Sináptica , Consumo de Bebidas Alcohólicas , Amígdala del Cerebelo , Animales , Femenino , Masculino , Ratas , Ratas Wistar
17.
Sci Rep ; 10(1): 17730, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33082413

RESUMEN

KRAS mutations are associated with rare cases of neurodevelopmental disorders that can cause intellectual disabilities. Previous studies showed that mice expressing a mutant KRAS have impaired the development and function of GABAergic inhibitory neurons, which may contribute to behavioural deficits in the mutant mice. However, the underlying cellular mechanisms and the role of excitatory neurons in these behavioural deficits in adults are not fully understood. Herein, we report that neuron type-specific expression of a constitutively active mutant KRASG12V in either excitatory or inhibitory neurons resulted in spatial memory deficits in adult mice. In inhibitory neurons, KRASG12V induced ERK activation and enhanced GABAergic synaptic transmission. Expressing KRASG12V in inhibitory neurons also impaired long-term potentiation in the hippocampal Shaffer-collateral pathway, which could be rescued by picrotoxin treatment. In contrast, KRASG12V induced ERK activation and neuronal cell death in excitatory neurons, which might have contributed to the severe behavioural deficits. Our results showed that both excitatory and inhibitory neurons are involved in mutant KRAS-associated learning deficits in adults via distinct cellular mechanisms.


Asunto(s)
Neuronas GABAérgicas/fisiología , Hipocampo/fisiología , Discapacidad Intelectual/genética , Mutación/genética , Trastornos del Neurodesarrollo/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Adulto , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Aprendizaje , Memoria , Ratones , Especificidad de Órganos , Picrotoxina/farmacología , Transducción de Señal
18.
FASEB J ; 34(12): 16567-16580, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33094857

RESUMEN

The growth and differentiation of neurons are critical events in the establishment of proper neuron connectivity and function. Planarians have a remarkable ability to completely regenerate a functional nervous system from a pluripotent stem cell population. Thus, planarians provide a powerful model to identify genes required for neuronal differentiation in vivo. The Wnt/Ca2+ signaling pathway is crucial for cancer development, arousing inflammatory responses, and neurodegeneration. We analyzed the expression patterns and RNAi phenotypes for members of the Wnt/Ca2+ signaling pathway in the planarian, Dugesia japonica. The expression of DjWnt5a, DjPLC-ß, DjCamKII, and DjCaln during regeneration was surprisingly similar and revealing in the regenerated brain. RNAi knockdown of DjWnt5a, DjPLC-ß, DjCamKII, and DjCaln led to defects in regenerated brains including brain partial deletions, incompact phenotypes at the posterior of the new brain, and lateral branches, which could not regenerate. Furthermore, the expressions of GAD and the number of GABAergic neurons decreased. Together, these results suggest that the Wnt/Ca2+ signaling pathway is required for GABAergic neuron regeneration.


Asunto(s)
Calcio/metabolismo , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Planarias/metabolismo , Planarias/fisiología , Transducción de Señal/fisiología , Vía de Señalización Wnt/fisiología , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/fisiología
19.
Elife ; 92020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32452766

RESUMEN

Optogenetic techniques for neural inactivation are valuable for linking neural activity to behavior but they have serious limitations in macaques. To achieve powerful and temporally precise neural inactivation, we used an adeno-associated viral (AAV) vector carrying the channelrhodopsin-2 gene under the control of a Dlx5/6 enhancer, which restricts expression to GABAergic neurons. We tested this approach in the primary visual cortex, an area where neural inactivation leads to interpretable behavioral deficits. Optical stimulation modulated spiking activity and reduced visual sensitivity profoundly in the region of space represented by the stimulated neurons. Rebound firing, which can have unwanted effects on neural circuits following inactivation, was not observed, and the efficacy of the optogenetic manipulation on behavior was maintained across >1000 trials. We conclude that this inhibitory cell-type-specific optogenetic approach is a powerful and spatiotemporally precise neural inactivation tool with broad utility for probing the functional contributions of cortical activity in macaques.


Asunto(s)
Neuronas GABAérgicas/fisiología , Optogenética , Corteza Visual/fisiología , Animales , Channelrhodopsins/genética , Dependovirus , Macaca mulatta
20.
Neurobiol Dis ; 141: 104942, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32423877

RESUMEN

Recent studies have demonstrated an active role for neurons in glioma progression. Specifically, peritumoral neurons establish functional excitatory synapses with glioma cells, and optogenetic stimulation of cortical pyramidal neurons drives tumor progression. However, the specific role of different subsets of cortical neurons, such as GABAergic interneurons, remains unexplored. Here, we directly compared the effects of optogenetic stimulation of pyramidal cells vs. fast-spiking, GABAergic neurons. In mice inoculated with GL261 cells into the motor cortex, we show that optogenetic stimulation of pyramidal neurons enhances glioma cell proliferation. In contrast, optogenetic stimulation of fast-spiking, parvalbumin-positive interneurons reduces proliferation as measured by BrdU incorporation and Ki67 immunolabelling. Since both principal cells and fast-spiking interneurons are directly activated by sensory afferent input, we next placed tumors in the occipital cortex to test the impact of visual stimulation/deprivation. We report that total lack of visual input via dark rearing enhances the density of proliferating glioma cells, while daily visual stimulation by gratings of different spatial frequencies and contrast reduces tumor growth. The effects of sensory input are region-specific, as visual deprivation has no significant effect on tumor proliferation in mice with gliomas in the motor cortex. We also report that sensory stimulation combined with temozolomide administration delays the loss of visual responses in peritumoral neurons. Altogether, these data demonstrate complex effects of different neuronal subtypes in the control of glioma proliferation.


Asunto(s)
Neoplasias Encefálicas/fisiopatología , Proliferación Celular , Neuronas GABAérgicas/fisiología , Glioma/fisiopatología , Células Piramidales/fisiología , Animales , Línea Celular Tumoral , Ratones Endogámicos C57BL , Corteza Motora/fisiopatología , Optogenética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA