Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Neurobiol Dis ; 194: 106482, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38522590

RESUMEN

A growing number of clinical and animal studies suggest that the nucleus accumbens (NAc), especially the shell, is involved in the pathogenesis of temporal lobe epilepsy (TLE). However, the role of parvalbumin (PV) GABAergic neurons in the NAc shell involved in TLE is still unclear. In this study, we induced a spontaneous TLE model by intrahippocampal administration of kainic acid (KA), which generally induce acute seizures in first 2 h (acute phase) and then lead to spontaneous recurrent seizures after two months (chronic phase). We found that chemogenetic activation of NAc shell PV neurons could alleviate TLE seizures by reducing the number and period of focal seizures (FSs) and secondary generalized seizures (sGSs), while selective inhibition of PV exacerbated seizure activity. Ruby-virus mapping results identified that the hippocampus (ventral and dorsal) is one of the projection targets of NAc shell PV neurons. Chemogenetic activation of the NAc-Hip PV projection fibers can mitigate seizures while inhibition has no effect on seizure ictogenesis. In summary, our findings reveal that PV neurons in the NAc shell could modulate the seizures in TLE via a long-range NAc-Hip circuit. All of these results enriched the investigation between NAc and epilepsy, offering new targets for future epileptogenesis research and precision therapy.


Asunto(s)
Epilepsia del Lóbulo Temporal , Animales , Epilepsia del Lóbulo Temporal/patología , Núcleo Accumbens/metabolismo , Parvalbúminas/metabolismo , Convulsiones/patología , Hipocampo/patología , Neuronas GABAérgicas/metabolismo , Ácido Kaínico/toxicidad , Modelos Animales de Enfermedad
2.
Dev Psychobiol ; 66(2): e22468, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38351459

RESUMEN

There is considerable evidence of reorganization in the prefrontal cortex during adolescence in humans, as well as in rodents, where the cellular basis can be explored. Studies from my laboratory in the rat medial prefrontal cortex are reviewed here. In general, growth predominates before puberty. Pruning mainly occurs at puberty and after with decreases in the number of synapses, dendrites, and neurons. Perineuronal nets, extracellular structures that control plasticity, are pruned peripubertally only in female rats, which may further open the adolescent prefrontal cortex to environmental influences. This is supported by our recent evidence that exposure to mild stress early, but not late, in adolescence decreases prepulse inhibition. Additionally, exposure to methamphetamine in females early in adolescence increases the number of a major class of inhibitory interneurons, parvalbumin neurons, while the opposite occurs late in adolescence. In females, even estrogen receptor beta mRNA decreases at puberty in the prefrontal cortex. Interestingly, rats of both sexes perform better after puberty on a test of cognitive flexibility in the water maze. Thus, evidence is accruing that adolescence is not a single entity but rather an ongoing set of processes, and environmental effects will differ depending on timing and sex.


Asunto(s)
Neuronas , Maduración Sexual , Humanos , Masculino , Ratas , Femenino , Animales , Adolescente , Interneuronas/fisiología , Corteza Prefrontal/fisiología , Parvalbúminas
3.
Int J Neuropsychopharmacol ; 26(5): 309-321, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-36975001

RESUMEN

Redox biology and immune signaling play major roles in the body, including in brain function. A rapidly growing literature also suggests that redox and immune abnormalities are implicated in neuropsychiatric conditions such as schizophrenia (SZ), bipolar disorder, autism, and epilepsy. In this article we review this literature, its implications for the pathophysiology of SZ, and the potential for development of novel treatment interventions targeting redox and immune signaling. Redox biology and immune signaling in the brain are complex and not fully understood; in addition, there are discrepancies in the literature, especially in patient-oriented studies. Nevertheless, it is clear that abnormalities arise in SZ from an interaction between genetic and environmental factors during sensitive periods of brain development, and these abnormalities disrupt local circuits and long-range connectivity. Interventions that correct these abnormalities may be effective in normalizing brain function in psychotic disorders, especially in early phases of illness.


Asunto(s)
Trastorno Bipolar , Trastornos Psicóticos , Esquizofrenia , Humanos , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/genética , Estrés Oxidativo/fisiología , Oxidación-Reducción
4.
Proc Natl Acad Sci U S A ; 116(26): 13077-13086, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31182594

RESUMEN

Conditioned inhibition is an important process to suppress learned responses for optimal adaptation, but its underlying biological mechanism is poorly understood. Here we used safety learning (SL)/fear discrimination after fear conditioning as a conditioned inhibition model because it demonstrates the essential properties of summation and retardation. Activity of the dorsomedial prefrontal cortex (dmPFC) parvalbumin (PV) neurons bidirectionally regulates spiking levels of dmPFC excitatory neurons and fear states. Responses to safety cues are increased in dopaminergic (DA) neurons in the ventral tegmental area (VTA) and in PV neurons in dmPFC after SL. Plasticity in the VTA is implicated, since SL requires activation of N-methyl-d-aspartate receptors. Furthermore, in a posttraumatic stress disorder model, impaired SL is associated with impaired potentiation of VTA DA neuron activity. Our results demonstrate a DA-dependent learning process that targets prefrontal inhibitory neurons for suppression of learned responses, and have implications for the pathogenesis and treatment of various psychiatric diseases.


Asunto(s)
Condicionamiento Clásico/fisiología , Miedo/psicología , Inhibición Psicológica , Corteza Prefrontal/fisiología , Trastornos por Estrés Postraumático/psicología , Área Tegmental Ventral/fisiología , Animales , Señales (Psicología) , Modelos Animales de Enfermedad , Dopamina/metabolismo , Electrodos Implantados , Humanos , Masculino , Ratones , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Optogenética , Parvalbúminas/metabolismo , Corteza Prefrontal/citología , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/fisiología , Técnicas Estereotáxicas , Área Tegmental Ventral/citología
5.
J Neurosci ; 38(47): 10019-10041, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30249799

RESUMEN

The amygdala projects to hippocampus in pathways through which affective or social stimuli may influence learning and memory. We investigated the still unknown amygdalar termination patterns and their postsynaptic targets in hippocampus from system to synapse in rhesus monkeys of both sexes. The amygdala robustly innervated the stratum lacunosum-moleculare layer of cornu ammonis fields and uncus anteriorly. Sparser terminations in posterior hippocampus innervated the radiatum and pyramidal layers at the prosubicular/CA1 juncture. The terminations, which were larger than other afferents in the surrounding neuropil, position the amygdala to influence hippocampal input anteriorly, and its output posteriorly. Most amygdalar boutons (76-80%) innervated spines of excitatory hippocampal neurons, and most of the remaining innervated presumed inhibitory neurons, identified by morphology and label with parvalbumin or calretinin, which distinguished nonoverlapping neurochemical classes of hippocampal inhibitory neurons. In CA1, amygdalar axons innervated some calretinin neurons, which disinhibit pyramidal neurons. By contrast, in CA3 the amygdala innervated both calretinin and parvalbumin neurons; the latter strongly inhibit nearby excitatory neurons. In CA3, amygdalar pathways also made closely spaced dual synapses on excitatory neurons. The strong excitatory synapses in CA3 may facilitate affective context representations and trigger sharp-wave ripples associated with memory consolidation. When the amygdala is excessively activated during traumatic events, the specialized innervation of excitatory neurons and the powerful parvalbumin inhibitory neurons in CA3 may allow the suppression of activity of nearby neurons that receive weaker nonamygdalar input, leading to biased passage of highly charged affective stimuli and generalized fear.SIGNIFICANCE STATEMENT Strong pathways from the amygdala targeted the anterior hippocampus, and more weakly its posterior sectors, positioned to influence a variety of emotional and cognitive functions. In hippocampal field CA1, the amygdala innervated some calretinin neurons, which disinhibit excitatory neurons. By contrast, in CA3 the amygdala innervated calretinin as well as some of the powerful parvalbumin inhibitory neurons and may help balance the activity of neural ensembles to allow social interactions, learning, and memory. These results suggest that when the amygdala is hyperactive during emotional upheaval, it strongly activates excitatory hippocampal neurons and parvalbumin inhibitory neurons in CA3, which can suppress nearby neurons that receive weaker input from other sources, biasing the passage of stimuli with high emotional import and leading to generalized fear.


Asunto(s)
Amígdala del Cerebelo/fisiología , Hipocampo/fisiología , Red Nerviosa/fisiología , Amígdala del Cerebelo/química , Amígdala del Cerebelo/ultraestructura , Animales , Femenino , Hipocampo/química , Hipocampo/ultraestructura , Macaca mulatta , Masculino , Red Nerviosa/química , Red Nerviosa/ultraestructura , Vías Nerviosas/química , Vías Nerviosas/patología , Vías Nerviosas/ultraestructura , Terminales Presinápticos/química , Terminales Presinápticos/fisiología , Terminales Presinápticos/ultraestructura , Primates
6.
Proc Natl Acad Sci U S A ; 113(46): E7287-E7296, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27803317

RESUMEN

Rett syndrome (RTT) arises from loss-of-function mutations in methyl-CpG binding protein 2 gene (Mecp2), but fundamental aspects of its physiological mechanisms are unresolved. Here, by whole-cell recording of synaptic responses in MeCP2 mutant mice in vivo, we show that visually driven excitatory and inhibitory conductances are both reduced in cortical pyramidal neurons. The excitation-to-inhibition (E/I) ratio is increased in amplitude and prolonged in time course. These changes predict circuit-wide reductions in response reliability and selectivity of pyramidal neurons to visual stimuli, as confirmed by two-photon imaging. Targeted recordings reveal that parvalbumin-expressing (PV+) interneurons in mutant mice have reduced responses. PV-specific MeCP2 deletion alone recapitulates effects of global MeCP2 deletion on cortical circuits, including reduced pyramidal neuron responses and reduced response reliability and selectivity. Furthermore, MeCP2 mutant mice show reduced expression of the cation-chloride cotransporter KCC2 (K+/Cl- exporter) and a reduced KCC2/NKCC1 (Na+/K+/Cl- importer) ratio. Perforated patch recordings demonstrate that the reversal potential for GABA is more depolarized in mutant mice, but is restored by application of the NKCC1 inhibitor bumetanide. Treatment with recombinant human insulin-like growth factor-1 restores responses of PV+ and pyramidal neurons and increases KCC2 expression to normalize the KCC2/NKCC1 ratio. Thus, loss of MeCP2 in the brain alters both excitation and inhibition in brain circuits via multiple mechanisms. Loss of MeCP2 from a specific interneuron subtype contributes crucially to the cell-specific and circuit-wide deficits of RTT. The joint restoration of inhibition and excitation in cortical circuits is pivotal for functionally correcting the disorder.


Asunto(s)
Corteza Cerebral/fisiología , Interneuronas/fisiología , Células Piramidales/fisiología , Síndrome de Rett/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Factor I del Crecimiento Similar a la Insulina/farmacología , Interneuronas/efectos de los fármacos , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Parvalbúminas , Células Piramidales/efectos de los fármacos , Proteínas Recombinantes
7.
Neurobiol Dis ; 117: 149-160, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29894753

RESUMEN

Temporal lobe epilepsy (TLE) is the most common type of epilepsy and is often medically refractory. Previous studies suggest that selective pharmaco-genetic inhibition of pyramidal neurons has therapeutic value for the treatment of epilepsy, however there is a risk of disrupting normal physical functions. Here, we test whether pharmaco-genetic activation of parvalbumin neurons, which are transgenetically transduced with the modified muscarinic receptor hM3Dq can attenuate TLE. We found that pharmaco-genetic activation of hippocampal parvalbumin neurons in epileptogenic zone not only significantly extends the latency to different seizure stages and attenuates seizure activities in acute seizure model, but also greatly alleviates the severity of seizure onsets in two chronic epilepsy models. This manipulation did not affect the normal physical function evaluated in various cognitive tasks. Further, the activation of parvalbumin neurons produced an inhibition on parts of surrounding pyramidal neurons, and the direct inactivation of pyramidal neurons via the viral expression of a modified muscarinic receptor hM4Di produced a similar anti-ictogenic effect. Interestingly, pharmaco-genetic inactivation of pyramidal neurons was more sensitive to impair cognitive function. Those data demonstrated that pharmaco-genetic seizure attenuation through targeting parvalbumin neurons rather than pyramidal neurons may be a novel and relatively safe approach for treating refractory TLE.


Asunto(s)
Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/metabolismo , Neuronas/metabolismo , Parvalbúminas/metabolismo , Farmacogenética/métodos , Animales , Anticonvulsivantes/administración & dosificación , Relación Dosis-Respuesta a Droga , Epilepsia del Lóbulo Temporal/genética , Antagonistas de Receptores de GABA-A/administración & dosificación , Masculino , Ratones , Ratones Transgénicos , Neuronas/química , Neuronas/efectos de los fármacos , Parvalbúminas/análisis , Parvalbúminas/genética
8.
Cereb Cortex ; 26(11): 4242-4252, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27600839

RESUMEN

In the auditory system, early neural stations such as brain stem are characterized by strict tonotopy, which is used to deconstruct sounds to their basic frequencies. But higher along the auditory hierarchy, as early as primary auditory cortex (A1), tonotopy starts breaking down at local circuits. Here, we studied the response properties of both excitatory and inhibitory neurons in the auditory cortex of anesthetized mice. We used in vivo two photon-targeted cell-attached recordings from identified parvalbumin-positive neurons (PVNs) and their excitatory pyramidal neighbors (PyrNs). We show that PyrNs are locally heterogeneous as characterized by diverse best frequencies, pairwise signal correlations, and response timing. In marked contrast, neighboring PVNs exhibited homogenous response properties in pairwise signal correlations and temporal responses. The distinct physiological microarchitecture of different cell types is maintained qualitatively in response to natural sounds. Excitatory heterogeneity and inhibitory homogeneity within the same circuit suggest different roles for each population in coding natural stimuli.


Asunto(s)
Corteza Auditiva/citología , Mapeo Encefálico , Red Nerviosa/fisiología , Inhibición Neural/fisiología , Células Piramidales/fisiología , Estimulación Acústica , Animales , Estimulación Eléctrica , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Parvalbúminas/genética , Parvalbúminas/metabolismo , Técnicas de Placa-Clamp , Vocalización Animal/fisiología
9.
Neuroscience ; 538: 68-79, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38157976

RESUMEN

Inhibitory parvalbumin (PV) interneurons regulate the activity of neural circuits within brain regions involved in emotional processing, including the prefrontal cortex (PFC). Recently, rodent studies have implicated a stress-induced increase in prefrontal PV neuron activity in the development of anxiety behaviors, particularly in females. However, the mechanisms through which stress increases activity of prefrontal PV neurons remain unknown. The fast-spiking properties of PV neurons in part come from their expression of voltage-gated potassium (K+) ion channels, particularly Kv3.1 channels. We therefore suggest that stress-induced changes in Kv3.1 channels contribute to the appearance of an anxious phenotype following chronic stress in female mice. Here, we first showed that unpredictable chronic mild stress (UCMS) increased expression of Kv3.1 channels on prefrontal PV neurons in female mice, a potential mechanism underlying the previously observed hyperactivity of these neurons after stress. We then showed that female mice deficient in Kv3.1 channels displayed resilience to UCMS-induced anxiety-like behaviors. Altogether, our findings implicate Kv3.1 channels in the development of anxiety-like behaviors following UCMS, particularly in females, providing a novel mechanism to understand sex-specific vulnerabilities to stress-induced psychopathologies.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Masculino , Ratones , Femenino , Animales , Canales de Potasio con Entrada de Voltaje/metabolismo , Neuronas/metabolismo , Interneuronas/metabolismo , Encéfalo/metabolismo , Ansiedad/metabolismo , Canales de Potasio Shaw/metabolismo
10.
Brain Res ; 1841: 149122, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009061

RESUMEN

Women have a two-fold increased risk of developing Alzheimer's disease (AD) than men, yet the underlying mechanisms of this sex-specific vulnerability remain unknown. Here, we aimed at determining in the 5XFAD mouse model whether deficits in prefrontal-dependent cognitive functions, which are impacted in the preclinical stages of AD, appear earlier in females, and whether these cognitive deficits are associated with alterations in the activity of prefrontal parvalbumin (PV)-neurons that regulate prefrontal circuits activity. We observed that 3.5-month-old 5XFAD females, but not males, display impairments in spatial short-term recognition memory, a function that relies on the integrity of the prefrontal cortex. Hippocampal-dependent cognitive functions were intact in both sexes. We then observed that 5XFAD females have more prefrontal PV neurons expressing the marker of chronic activity FosB; this was inversely correlated with prefrontal-dependent cognitive performances. Our findings show for the first time sex-specific, early deregulation of prefrontal PV neurons activity, which is associated with early appearance of prefrontal-dependent cognitive functions in 5XFAD females providing a potential novel mechanism to the increased risk to AD in females.


Asunto(s)
Enfermedad de Alzheimer , Modelos Animales de Enfermedad , Trastornos de la Memoria , Ratones Transgénicos , Neuronas , Parvalbúminas , Corteza Prefrontal , Animales , Corteza Prefrontal/metabolismo , Parvalbúminas/metabolismo , Femenino , Neuronas/metabolismo , Masculino , Trastornos de la Memoria/fisiopatología , Trastornos de la Memoria/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Memoria Espacial/fisiología , Ratones , Reconocimiento en Psicología/fisiología , Hipocampo/metabolismo , Caracteres Sexuales , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Humanos , Ratones Endogámicos C57BL
11.
Environ Sci Pollut Res Int ; 31(20): 29385-29399, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38573577

RESUMEN

Fluorene-9-bisphenol (BHPF) is widely used in the manufacture of plastic products and potentially disrupts several physiological processes, but its biological effects on social behavior remain unknown. In this study, we investigated the effects of BHPF exposure on anxiety-like and social behavior in female mice and the potential mechanisms, thereby proposing a potential therapy strategy. We exposed female Balb/c mice to BHPF by oral gavage at different doses (0.5, 50 mg/kg bw/2-day) for 28 days, which were found BHPF (50 mg/kg) exposure affected motor activity in the open field test (OFT) and elevated cross maze (EPM), resulting in anxiety-like behaviors, as well as abnormal social behavioral deficits in the Social Interaction Test (SIT). Analysis of histopathological staining results showed that BHPF exposure caused damage to hippocampal neurons in the CA1/CA3/DG region and decreased Nissl pyramidal neurons in the CA1/CA3 regions of the hippocampus, as well as a decrease in parvalbumin neuron expression. In addition, BHPF exposure upregulated the expression of excitatory and inhibitory (E/I) vesicle transporter genes (Vglut1, Vglut2, VGAT, GAD67, Gabra) and axon growth gene (Dcc) in the mouse hippocampus. Interestingly, behavioral disturbances and E/I balance could be alleviated by exogenous melatonin (15 mg/kg bw/2-day) therapy. Our findings suggest that exogenous melatonin may be a potential therapy with protective potential for ameliorating or preventing BHPF-induced hippocampal neuronal damage and behavioral disturbances. This study provided new insight into the neurotoxicological effects on organisms exposed to endocrine-disrupting chemicals and aroused our vigilance in current environmental safety about chemical use.


Asunto(s)
Ansiedad , Fluorenos , Melatonina , Ratones Endogámicos BALB C , Conducta Social , Animales , Ratones , Ansiedad/inducido químicamente , Femenino , Fluorenos/toxicidad , Melatonina/farmacología , Conducta Animal/efectos de los fármacos , Hipocampo/efectos de los fármacos , Fenoles/toxicidad
12.
eNeuro ; 10(3)2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36808099

RESUMEN

Women are twice as likely as men to experience emotional dysregulation after stress, resulting in substantially higher psychopathology for equivalent lifetime stress exposure, yet the mechanisms underlying this vulnerability remain unknown. Studies suggest changes in medial prefrontal cortex (mPFC) activity as a potential contributor. Whether maladaptive changes in inhibitory interneurons participate in this process, and whether adaptations in response to stress differ between men and women, producing sex-specific changes in emotional behaviors and mPFC activity, remained undetermined. This study examined whether unpredictable chronic mild stress (UCMS) in mice differentially alters behavior and mPFC parvalbumin (PV) interneuron activity by sex, and whether the activity of these neurons drives sex-specific behavioral changes. Four weeks of UCMS increased anxiety-like and depressive-like behaviors associated with FosB activation in mPFC PV neurons, particularly in females. After 8 weeks of UCMS, both sexes displayed these behavioral and neural changes. Chemogenetic activation of PV neurons in UCMS-exposed and nonstressed males induced significant changes in anxiety-like behaviors. Importantly, patch-clamp electrophysiology demonstrated altered excitability and basic neural properties on the same timeline as the emergence of behavioral effects: changes in females after 4 weeks and in males after 8 weeks of UCMS. These findings show, for the first time, that sex-specific changes in the excitability of prefrontal PV neurons parallel the emergence of anxiety-like behavior, revealing a potential novel mechanism underlying the enhanced vulnerability of females to stress-induced psychopathology and supporting further investigation of this neuronal population to identify new therapeutic targets for stress disorders.


Asunto(s)
Ansiedad , Parvalbúminas , Masculino , Ratones , Femenino , Animales , Parvalbúminas/metabolismo , Ansiedad/patología , Neuronas/metabolismo , Trastornos de Ansiedad , Emociones , Interneuronas/fisiología , Corteza Prefrontal/metabolismo , Estrés Psicológico/patología
13.
Exp Neurol ; 360: 114289, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36471512

RESUMEN

BACKGROUND: Based on the theory of interhemispheric inhibition and the bimodal balance-recovery model in stroke, we explored the effects of excitation/inhibition (E/I) of parvalbumin (PV) neurons in the contralateral primary motor cortex (cM1) connecting the ipsilateral M1 (iM1) via the corpus callosum (cM1-CC-iM1) of ischemic stroke rats by optogenetic stimulation. METHODS: We tested this by injecting anterograde and retrograde virus in rats with middle cerebral artery occlusion (MCAO), and evaluated the neurological scores, motor behavior, volume of cerebral infarction and the E/I balance of the bilateral M1 two weeks after employing optogenetic treatment. RESULTS: We found that concentrations of Glu and GABA decreased and increased, respectively, in the iM1 of MCAO rats, and that the former increased in the cM1, suggesting E/I imbalance in bilateral M1 after ischemic stroke. Interestingly, optogenetic stimulation improved M1 E/I imbalance, as illustrated by the increase of Glu in the iM1 and the decrease of GABA in both iM1 and cM1, which were accompanied by an improvement in neurological deficit and motor dysfunction. In addition, we observed a reduced infarct volume, an increase in the expression of the NMDAR and AMPAR, and a decrease in GAD67 in the iM1 after intervention. CONCLUSIONS: Optogenetic modulation of PV neurons of the iM1-CC-cM1 improve E/I balance, leading to reduced neurological deficit and improved motor dysfunction following ischemic stroke in rats.


Asunto(s)
Accidente Cerebrovascular Isquémico , Corteza Motora , Rehabilitación de Accidente Cerebrovascular , Accidente Cerebrovascular , Humanos , Ratas , Animales , Parvalbúminas , Optogenética , Infarto de la Arteria Cerebral Media , Neuronas , Ácido gamma-Aminobutírico
14.
Front Neural Circuits ; 17: 1185095, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37396397

RESUMEN

Background: How volatile anesthetics work remains poorly understood. Modulations of synaptic neurotransmission are the direct cellular mechanisms of volatile anesthetics in the central nervous system. Volatile anesthetics such as isoflurane may reduce neuronal interaction by differentially inhibiting neurotransmission between GABAergic and glutamatergic synapses. Presynaptic voltage-dependent sodium channels (Nav), which are strictly coupled with synaptic vesicle exocytosis, are inhibited by volatile anesthetics and may contribute to the selectivity of isoflurane between GABAergic and glutamatergic synapses. However, it is still unknown how isoflurane at clinical concentrations differentially modulates Nav currents between excitatory and inhibitory neurons at the tissue level. Methods: In this study, an electrophysiological recording was applied in cortex slices to investigate the effects of isoflurane on Nav between parvalbumin (PV+) and pyramidal neurons in PV-cre-tdTomato and/or vglut2-cre-tdTomato mice. Results: Isoflurane at clinically relevant concentrations produced a hyperpolarizing shift in the voltage-dependent inactivation and slowed the recovery time from the fast inactivation in both cellular subtypes. Since the voltage of half-maximal inactivation was significantly depolarized in PV+ neurons compared to that of pyramidal neurons, isoflurane inhibited the peak Nav currents in pyramidal neurons more potently than those of PV+ neurons (35.95 ± 13.32% vs. 19.24 ± 16.04%, P = 0.036 by the Mann-Whitney test). Conclusions: Isoflurane differentially inhibits Nav currents between pyramidal and PV+ neurons in the prefrontal cortex, which may contribute to the preferential suppression of glutamate release over GABA release, resulting in the net depression of excitatory-inhibitory circuits in the prefrontal cortex.


Asunto(s)
Anestésicos por Inhalación , Isoflurano , Canales de Sodio Activados por Voltaje , Ratones , Animales , Isoflurano/farmacología , Parvalbúminas , Anestésicos por Inhalación/farmacología , Neuronas , Células Piramidales/fisiología , Corteza Prefrontal
15.
Epilepsia Open ; 8(3): 1002-1012, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37277986

RESUMEN

OBJECTIVE: Anterior nucleus of thalamus (ANT) has been widely accepted as a potential therapeutic target for drug-resistant epilepsy. Although increased volume of the ANT was also reported in patients with absence epilepsy, the relationship between the ANT and absence epilepsy has been barely illustrated. METHODS: Using chemogenetics, we evaluated the effect of ANT parvalbumin (PV) neurons on pentylenetetrazole (PTZ)-induced absence seizures in mice. RESULTS: We found that intraperitoneal injection of PTZ (30 mg/kg) can stably induce absence-like seizures characterized by bilaterally synchronous spike-wave discharges (SWDs). Selective activation of PV neurons in the ANT by chemogenetics could aggravate the severity of absence seizures, whereas selective inhibition of that cannot reverse this condition and even promote absence seizures as well. Moreover, chemogenetic inhibition of ANT PV neurons without administration of PTZ was also sufficient to generate SWDs. Analysis of background EEG showed that chemogenetic activation or inhibition of ANT PV neurons could both significantly increase the EEG power of delta oscillation in the frontal cortex, which might mediate the pro-seizure effect of ANT PV neurons. SIGNIFICANCE: Our findings indicated that either activation or inhibition of ANT PV neurons might disturb the intrinsic delta rhythms in the cortex and worsen absence seizures, which highlighted the importance of maintaining the activity of ANT PV neurons in absence seizure.


Asunto(s)
Núcleos Talámicos Anteriores , Epilepsia Tipo Ausencia , Animales , Ratones , Núcleos Talámicos Anteriores/fisiología , Neuronas/fisiología , Parvalbúminas/farmacología , Pentilenotetrazol/farmacología , Convulsiones
16.
Acta Neuropathol Commun ; 11(1): 34, 2023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36882863

RESUMEN

Mutations in the solute carrier family 6-member 8 (Slc6a8) gene, encoding the protein responsible for cellular creatine (Cr) uptake, cause Creatine Transporter Deficiency (CTD), an X-linked neurometabolic disorder presenting with intellectual disability, autistic-like features, and epilepsy. The pathological determinants of CTD are still poorly understood, hindering the development of therapies. In this study, we generated an extensive transcriptomic profile of CTD showing that Cr deficiency causes perturbations of gene expression in excitatory neurons, inhibitory cells, and oligodendrocytes which result in remodeling of circuit excitability and synaptic wiring. We also identified specific alterations of parvalbumin-expressing (PV+) interneurons, exhibiting a reduction in cellular and synaptic density, and a hypofunctional electrophysiological phenotype. Mice lacking Slc6a8 only in PV+ interneurons recapitulated numerous CTD features, including cognitive deterioration, impaired cortical processing and hyperexcitability of brain circuits, demonstrating that Cr deficit in PV+ interneurons is sufficient to determine the neurological phenotype of CTD. Moreover, a pharmacological treatment targeted to restore the efficiency of PV+ synapses significantly improved cortical activity in Slc6a8 knock-out animals. Altogether, these data demonstrate that Slc6a8 is critical for the normal function of PV+ interneurons and that impairment of these cells is central in the disease pathogenesis, suggesting a novel therapeutic venue for CTD.


Asunto(s)
Encefalopatías Metabólicas Innatas , Proteínas de Transporte de Membrana , Parvalbúminas , Animales , Ratones , Creatina , Neuronas , Proteínas de Transporte de Membrana/genética
17.
Cells ; 11(7)2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35406741

RESUMEN

Oxidative stress induced by brain ischemia upregulates transient receptor potential melastatin-like-7 (TRPM7) expression and currents, which could contribute to neurotoxicity and cell death. Accordingly, suppression of TRPM7 reduces neuronal death, tissue damage and motor deficits. However, the neuroprotective effects of TRPM7 suppression in different cell types have not been investigated. Here, we found that induction of ischemia resulted in loss of parvalbumin (PV) gamma-aminobutyric acid (GABAergic) neurons more than Ca2+/calmodulin-kinase II (CaMKII) glutamatergic neurons in the mouse cortex. Furthermore, brain ischemia increased TRPM7 expression in PV neurons more than that in CaMKII neurons. We generated two lines of conditional knockout mice of TRPM7 in GABAergic PV neurons (PV-TRPM7-/-) and in glutamatergic neurons (CaMKII-TRPM7-/-). Following exposure to brain ischemia, we found that deleting TRPM7 reduced the infarct volume in both lines of transgenic mice. However, the volume in PV-TRPM7-/- mice was more significantly lower than that in the control group. Neuronal survival of both GABAergic and glutamatergic neurons was increased in PV-TRPM7-/- mice; meanwhile, only glutamatergic neurons were protected in CaMKII-TRPM7-/-. At the behavioral level, only PV-TRPM7-/- mice exhibited significant reductions in neurological and motor deficits. Inflammatory mediators such as GFAP, Iba1 and TNF-α were suppressed in PV-TRPM7-/- more than in CaMKII-TRPM7-/-. Mechanistically, p53 and cleaved caspase-3 were reduced in both groups, but the reduction in PV-TRPM7-/- mice was more than that in CaMKII-TRPM7-/- following ischemia. Upstream from these signaling molecules, the Akt anti-oxidative stress signaling was activated only in PV-TRPM7-/- mice. Therefore, deleting TRPM7 in GABAergic PV neurons might have stronger neuroprotective effects against ischemia pathologies than doing so in glutamatergic neurons.


Asunto(s)
Isquemia Encefálica , Neuroprotección , Canales Catiónicos TRPM , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Neuronas GABAérgicas/metabolismo , Eliminación de Gen , Isquemia/metabolismo , Ratones , Parvalbúminas/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
18.
Neuron ; 110(24): 4108-4124.e6, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36223767

RESUMEN

Central sensitization caused by spinal disinhibition is a key mechanism of mechanical allodynia in neuropathic pain. However, the molecular mechanisms underlying spinal disinhibition after nerve injury remain unclear. Here, we show in mice that spared nerve injury (SNI), which induces mechanical hypersensitivity and neuropathic pain, triggers homeostatic reduction of inhibitory outputs from dorsal horn parvalbumin-positive (PV+) interneurons onto both primary afferent terminals and excitatory interneurons. The reduction in inhibitory outputs drives hyperactivation of the spinal cord nociceptive pathway, causing mechanical hypersensitivity. We identified the retinoic acid receptor RARα, a central regulator of homeostatic plasticity, as the key molecular mediator for this synaptic disinhibition. Deletion of RARα in spinal PV+ neurons or application of an RARα antagonist in the spinal cord prevented the development of SNI-induced mechanical hypersensitivity. Our results identify RARα as a crucial molecular effector for neuropathic pain and a potential target for its treatment.


Asunto(s)
Neuralgia , Médula Espinal , Ratones , Animales , Médula Espinal/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Neuronas/metabolismo , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Receptores de Ácido Retinoico
19.
Elife ; 112022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35576146

RESUMEN

Recent discoveries of extreme cellular diversity in the brain warrant rapid development of technologies to access specific cell populations within heterogeneous tissue. Available approaches for engineering-targeted technologies for new neuron subtypes are low yield, involving intensive transgenic strain or virus screening. Here, we present Specific Nuclear-Anchored Independent Labeling (SNAIL), an improved virus-based strategy for cell labeling and nuclear isolation from heterogeneous tissue. SNAIL works by leveraging machine learning and other computational approaches to identify DNA sequence features that confer cell type-specific gene activation and then make a probe that drives an affinity purification-compatible reporter gene. As a proof of concept, we designed and validated two novel SNAIL probes that target parvalbumin-expressing (PV+) neurons. Nuclear isolation using SNAIL in wild-type mice is sufficient to capture characteristic open chromatin features of PV+ neurons in the cortex, striatum, and external globus pallidus. The SNAIL framework also has high utility for multispecies cell probe engineering; expression from a mouse PV+ SNAIL enhancer sequence was enriched in PV+ neurons of the macaque cortex. Expansion of this technology has broad applications in cell type-specific observation, manipulation, and therapeutics across species and disease models.


Asunto(s)
Elementos de Facilitación Genéticos , Aprendizaje Automático , Neuronas , Análisis de Secuencia de ADN , Animales , Corteza Cerebral/metabolismo , Biología Computacional/métodos , Elementos de Facilitación Genéticos/genética , Globo Pálido , Ratones , Neuronas/metabolismo , Parvalbúminas/metabolismo , Análisis de Secuencia de ADN/métodos
20.
Cell Rep ; 36(6): 109503, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34380026

RESUMEN

The association between cause and effect is usually probabilistic. Memories triggered by ambiguous cues may be altered or biased into a more negative perception in psychiatric diseases. Understanding the formation and modulation of this probabilistic association is important for revealing the nature of aversive memory and alterations in brain diseases. We found that 50% conditioned and unconditioned stimuli (CS-US) association during Pavlovian fear conditioning results in reduced fear responses and neural spiking in the dorsomedial prefrontal cortex (dmPFC) due to enhanced inhibition from dmPFC parvalbumin (PV) neurons. Formation of probabilistic memory is associated with increased synaptic inputs to PV-neurons and requires activation of ventral hippocampus, which detects CS-US mismatch during conditioning. Stress prior to conditioning impairs the formation of probabilistic memory by abolishing PV-neuronal plasticity, while stress prior to memory retrieval reverts enhanced PV-neuron activity. In conclusion, PV-neurons tailor learned responses to fit brain state at the moment of retrieval.


Asunto(s)
Condicionamiento Clásico , Miedo/fisiología , Modelos Estadísticos , Inhibición Neural/fisiología , Corteza Prefrontal/fisiología , Animales , Hipocampo/fisiología , Masculino , Memoria/fisiología , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Parvalbúminas/metabolismo , Estrés Psicológico/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA