Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.459
Filtrar
1.
Dis Model Mech ; 17(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38785269

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in MECP2, which encodes methyl-CpG-binding protein 2, a transcriptional regulator of many genes, including brain-derived neurotrophic factor (BDNF). BDNF levels are lower in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of LM22A-4, a brain-penetrant, small-molecule ligand of the BDNF receptor TrkB (encoded by Ntrk2), on dendritic spine density and form in hippocampal pyramidal neurons and on behavioral phenotypes in female Mecp2 heterozygous (HET) mice. A 4-week systemic treatment of Mecp2 HET mice with LM22A-4 restored spine volume in MeCP2-expressing neurons to wild-type (WT) levels, whereas spine volume in MeCP2-lacking neurons remained comparable to that in neurons from female WT mice. Female Mecp2 HET mice engaged in aggressive behaviors more than WT mice, the levels of which were reduced to WT levels by the 4-week LM22A-4 treatment. These data provide additional support to the potential usefulness of novel therapies not only for RTT but also to other BDNF-related disorders.


Asunto(s)
Conducta Animal , Espinas Dendríticas , Proteína 2 de Unión a Metil-CpG , Fenotipo , Receptor trkB , Síndrome de Rett , Animales , Síndrome de Rett/patología , Síndrome de Rett/tratamiento farmacológico , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Femenino , Receptor trkB/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Conducta Animal/efectos de los fármacos , Ligandos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/patología , Ratones , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/patología , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Heterocigoto , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Benzamidas
2.
Anesthesiology ; 140(6): 1192-1200, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38624275

RESUMEN

Tonic inhibition in mouse hippocampal CA1 pyramidal neurons is mediated by α5 subunit-containing γ-aminobutyric acid type A receptors. By Caraiscos VB, Elliott EM, You-Ten KE, Cheng VY, Belelli D, Newell JG, Jackson MF, Lambert JJ, Rosahl TW, Wafford KA, MacDonald JF, Orser BA. Proc Natl Acad Sci U S A 2004; 101:3662-7. Reprinted with permission. In this Classic Paper Revisited, the author recounts the scientific journey leading to a report published in the Proceedings of the National Academy of Sciences (PNAS) and shares several personal stories from her formative years and "research truths" that she has learned along the way. Briefly, the principal inhibitory neurotransmitter in the brain, γ-aminobutyric acid (GABA), was conventionally thought to regulate cognitive processes by activating synaptic GABA type A (GABAA) receptors and generating transient inhibitory synaptic currents. However, the author's laboratory team discovered a novel nonsynaptic form of tonic inhibition in hippocampal pyramidal neurons, mediated by extrasynaptic GABAA receptors that are pharmacologically distinct from synaptic GABAA receptors. This tonic current is highly sensitive to most general anesthetics, including sevoflurane and propofol, and likely contributes to the memory-blocking properties of these drugs. Before the publication in PNAS, the subunit composition of GABAA receptors that generate the tonic current was unknown. The team's research showed that GABAA receptors containing the α5 subunit (α5GABAARs) generated the tonic inhibitory current in hippocampal neurons. α5GABAARs are highly sensitive to GABA, desensitize slowly, and are thus well suited for detecting low, persistent, ambient concentrations of GABA in the extracellular space. Interest in α5GABAARs has surged since the PNAS report, driven by their pivotal roles in cognitive processes and their potential as therapeutic targets for treating various neurologic disorders.


Asunto(s)
Receptores de GABA-A , Animales , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo , Ratones , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Células Piramidales/metabolismo , Humanos , Sinapsis/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ácido gamma-Aminobutírico/metabolismo
3.
Mar Drugs ; 22(4)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38667787

RESUMEN

Porphyran, a sulfated polysaccharide found in various species of marine red algae, has been demonstrated to exhibit diverse bioactivities, including anti-inflammatory effects. However, the protective effects of porphyran against cerebral ischemia and reperfusion (IR) injury have not been investigated. The aim of this study was to examine the neuroprotective effects of porphyran against brain IR injury and its underlying mechanisms using a gerbil model of transient forebrain ischemia (IR in the forebrain), which results in pyramidal cell (principal neuron) loss in the cornu ammonis 1 (CA1) subregion of the hippocampus on day 4 after IR. Porphyran (25 and 50 mg/kg) was orally administered daily for one week prior to IR. Pretreatment with 50 mg/kg of porphyran, but not 25 mg/kg, significantly attenuated locomotor hyperactivity and protected pyramidal cells located in the CA1 area from IR injury. The pretreatment with 50 mg/kg of porphyran significantly suppressed the IR-induced activation and proliferation of microglia in the CA1 subregion. Additionally, the pretreatment significantly inhibited the overexpressions of nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing protein-3 (NLRP3) inflammasome complex, and pro-inflammatory cytokines (interleukin 1 beta and interleukin 18) induced by IR in the CA1 subregion. Overall, our findings suggest that porphyran exerts neuroprotective effects against brain IR injury, potentially by reducing the reaction (activation) and proliferation of microglia and reducing NLRP3 inflammasome-mediated neuroinflammation.


Asunto(s)
Región CA1 Hipocampal , Gerbillinae , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Enfermedades Neuroinflamatorias , Fármacos Neuroprotectores , Daño por Reperfusión , Sefarosa/análogos & derivados , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Masculino , Daño por Reperfusión/tratamiento farmacológico , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Modelos Animales de Enfermedad , Microglía/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Polisacáridos/farmacología , Neuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo
4.
Neuropharmacology ; 254: 109970, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38685343

RESUMEN

Pharmacological approaches to induce N-methyl-d-aspartate receptor (NMDAR) hypofunction have been intensively used to understand the aetiology and pathophysiology of schizophrenia. Yet, the precise cellular and molecular mechanisms that relate to brain network dysfunction remain largely unknown. Here, we used a set of complementary approaches to assess the functional network abnormalities present in male mice that underwent a 7-day subchronic phencyclidine (PCP 10 mg/kg, subcutaneously, once daily) treatment. Our data revealed that pharmacological intervention with PCP affected cognitive performance and auditory evoked gamma oscillations in the prefrontal cortex (PFC) mimicking endophenotypes of some schizophrenia patients. We further assessed PFC cellular function and identified altered neuronal intrinsic membrane properties, reduced parvalbumin (PV) immunostaining and diminished inhibition onto L5 PFC pyramidal cells. A decrease in the strength of optogenetically-evoked glutamatergic current at the ventral hippocampus to PFC synapse was also demonstrated, along with a weaker shunt of excitatory transmission by local PFC interneurons. On a macrocircuit level, functional ultrasound measurements indicated compromised functional connectivity within several brain regions particularly involving PFC and frontostriatal circuits. Herein, we reproduced a panel of schizophrenia endophenotypes induced by subchronic PCP application in mice. We further recapitulated electrophysiological signatures associated with schizophrenia and provided an anatomical reference to critical elements in the brain circuitry. Together, our findings contribute to a better understanding of the physiological underpinnings of deficits induced by subchronic NMDAR antagonist regimes and provide a test system for characterization of pharmacological compounds.


Asunto(s)
Modelos Animales de Enfermedad , Fenciclidina , Corteza Prefrontal , Receptores de N-Metil-D-Aspartato , Animales , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Masculino , Fenciclidina/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Ratones , Esquizofrenia/inducido químicamente , Esquizofrenia/fisiopatología , Esquizofrenia/metabolismo , Ratones Endogámicos C57BL , Parvalbúminas/metabolismo , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ritmo Gamma/efectos de los fármacos , Ritmo Gamma/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología
5.
J Physiol ; 602(9): 2047-2060, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38500302

RESUMEN

Adverse experiences in early life can induce maladaptive responses to acute stress in later life. Chronic social isolation during adolescence is an early life adversity that can precipitate stress-related psychiatric disorders. We found that male mice after 8 weeks of adolescent social isolation (SI) have markedly increased aggression after being exposed to 2 h of restraint stress (RS), which was accompanied by a significant increase of AMPA receptor- and NMDA receptor-mediated synaptic transmission in prefrontal cortex (PFC) pyramidal neurons of SIRS males. Compared to group-housed counterparts, SIRS males exhibited a significantly decreased level of histone H3 acetylation in PFC. Systemic administration of class I histone deacetylase inhibitors, romidepsin or MS-275, ameliorated the aggressive behaviour, as well as general social interaction deficits, of SIRS males. Electrophysiological recordings also found normalization of PFC glutamatergic currents by romidepsin treatment of SIRS male mice. These results revealed an epigenetic mechanism and intervention avenue for aggression induced by chronic social isolation. KEY POINTS: Adolescent chronic social isolation can precipitate stress-related psychiatric disorders. A significant increase of glutamatergic transmission is found in the prefrontal cortex (PFC) of socially isolated male mice exposed to an acute stress (SIRS). Treatment with class I histone deacetylase (HDAC) inhibitors ameliorates the aggressive behaviour and social interaction deficits of SIRS males, and normalizes glutamatergic currents in PFC neurons. It provides an epigenetic mechanism and intervention avenue for aberrant stress responses induced by chronic social isolation.


Asunto(s)
Agresión , Inhibidores de Histona Desacetilasas , Ratones Endogámicos C57BL , Corteza Prefrontal , Aislamiento Social , Estrés Psicológico , Animales , Masculino , Inhibidores de Histona Desacetilasas/farmacología , Aislamiento Social/psicología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Agresión/efectos de los fármacos , Ratones , Depsipéptidos/farmacología , Piridinas/farmacología , Benzamidas/farmacología , Restricción Física , Transmisión Sináptica/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Células Piramidales/metabolismo
6.
Cereb Cortex ; 33(13): 8179-8193, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-36967112

RESUMEN

Motor disturbances are observed in schizophrenia patients, but the neuroanatomical background is unknown. Our aim was to investigate the pyramidal cells of the primary motor cortex (BA 4) in both hemispheres of postmortem control and schizophrenia subjects-8 subjects in each group-with 2.5-5.5 h postmortem interval. The density and size of the Sternberger monoclonal incorporated antibody 32 (SMI32)-immunostained pyramidal cells in layer 3 and 5 showed no change; however, the proportion of larger pyramidal cells is decreased in layer 5. Giant pyramidal neurons (Betz cells) were investigated distinctively with SMI32- and parvalbumin (PV) immunostainings. In the right hemisphere of schizophrenia subjects, the density of Betz cells was decreased and their PV-immunopositive perisomatic input showed impairment. Part of the Betz cells contained PV in both groups, but the proportion of PV-positive cells has declined with age. The rat model of antipsychotic treatment with haloperidol and olanzapine showed no differences in size and density of SMI32-immunopositive pyramidal cells. Our results suggest that motor impairment of schizophrenia patients may have a morphological basis involving the Betz cells in the right hemisphere. These alterations can have neurodevelopmental and neurodegenerative explanations, but antipsychotic treatment does not explain them.


Asunto(s)
Lateralidad Funcional , Corteza Motora , Células Piramidales , Esquizofrenia , Anciano , Anciano de 80 o más Años , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ratas , Envejecimiento , Antipsicóticos/uso terapéutico , Autopsia , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Lateralidad Funcional/efectos de los fármacos , Haloperidol/farmacología , Haloperidol/uso terapéutico , Inmunohistoquímica , Filamentos Intermedios/metabolismo , Corteza Motora/efectos de los fármacos , Corteza Motora/patología , Olanzapina/farmacología , Olanzapina/uso terapéutico , Parvalbúminas/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/patología , Ratas Sprague-Dawley , Análisis de Regresión , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/patología
7.
Neurosci Lett ; 790: 136898, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36195298

RESUMEN

Noopept (NP) is a proline-containing dipeptide with nootropic and neuroprotective properties. We have previously shown that NP significantly increased the frequency of spontaneous IPSCs in hippocampal CA1 pyramidal cells mediated by the activation of inhibitory interneurons in stratum radiatum. The cholinergic system plays an important role in the performance of cognitive functions, furthermore multiple behavioral and clinical facts link NP with the cholinergic system. The present study was undertaken to reveal the possible interaction of NP with neuronal nicotinic acetylcholine receptors (nAChRs). Currents were recorded from rat hippocampal neurons using the whole-cell, patch-clamp technique. NP (5 µM) increased the action potential firing frequency recorded from GABAergic interneurons in the stratum radiatum (SR) of CA1 region. This effect was almost completely abolished by the application of the α7 nAChR-selective antagonists α-bungarotoxin (α-BGT; 6 nM) and methyllycaconitine (MLA; 20 nM). The increase in the frequency of spontaneous IPSCs in CA1 pyramidal cells induced by NP was also eliminated by α7 nAChRs antagonists. These results imply the involvement of α7 nAChRs in the modulation of hippocampal neuronal activity caused by NP and indicate that a7 nAChRs are an important site of action of NP.


Asunto(s)
Nootrópicos , Receptores Nicotínicos , Animales , Ratas , Bungarotoxinas , Dipéptidos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Interneuronas/metabolismo , Antagonistas Nicotínicos/farmacología , Nootrópicos/farmacología , Prolina/farmacología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas Sprague-Dawley , Receptores Nicotínicos/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
8.
Brain Res ; 1783: 147847, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35227652

RESUMEN

Menthol is a natural compound that evokes cold sensations by activating TRPM8 channels in peripheral sensory receptors. Little is known about the effects of this compound on brain neurons. It has been shown previously that menthol exerts antiepileptic effects in hippocampal neurons by enhancing GABA receptors. The aim of this patch-clamp study was to assess the effects of menthol on sodium currents, action potentials and epileptiform events in cortical neurons. Menthol inhibited fast voltage-gated sodium channels and neuronal excitability defined as the number of action potentials per depolarization step. The influence of menthol on epileptic events was also assessed in this study. Interictal epileptiform events lasting <2 s were recorded in zero magnesium high potassium proepileptic extracellular solution. The frequency of these epileptiform events was inhibited by menthol (200 µM). Ictal epileptic events lasting >100 s were recorded in zero magnesium proepileptic extracellular solution containing 4-AP. The frequency of these ictal events was potently decreased by menthol. TRPM8 channels were not involved in the inhibitory effect of menthol on ictal events because epileptic discharges persisted in the presence of the TRPM8 inhibitor AMTB. Moreover, ictal events were inhibited by therapeutic concentrations of the antiepileptic drug carbamazepine. Menthol and carbamazepine inhibited ictal events to a similar extent. This study showed that menthol exerts antiepileptic effects in cortical neurons.


Asunto(s)
Anticonvulsivantes , Epilepsia , Mentol , Células Piramidales , Anticonvulsivantes/farmacología , Carbamazepina , Epilepsia/tratamiento farmacológico , Humanos , Magnesio/farmacología , Mentol/farmacología , Corteza Prefrontal/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Canales Catiónicos TRPM/metabolismo
9.
Sci Rep ; 12(1): 3186, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35210456

RESUMEN

Sexual differentiation of the brain is influenced by testosterone and its metabolites during the perinatal period, when many aspects of brain development, including the maturation of GABAergic transmission, occur. Whether and how testosterone signaling during the perinatal period affects GABAergic transmission is unclear. Here, we analyzed GABAergic circuit functional markers in male, female, testosterone-treated female, and testosterone-insensitive male rats after the first postnatal week and in young adults. In the hippocampus, mRNA levels of proteins associated with GABA signaling were not significantly affected at postnatal day (P) 7 or P40. Conversely, membrane protein levels of KCC2, which are critical for determining inhibition strength, were significantly higher in females compared to males and testosterone-treated females at P7. Further, female and testosterone-insensitive male rats at P7 showed higher levels of the neurotrophin BDNF, which is a powerful regulator of neuronal function, including GABAergic transmission. Finally, spontaneous GABAergic currents in hippocampal CA1 pyramidal cells were more frequent in females and testosterone-insensitive males at P40. Overall, these results show that perinatal testosterone levels modulate GABAergic circuit function, suggesting a critical role of perinatal sex hormones in regulating network excitability in the adult hippocampus.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Células Piramidales/metabolismo , Simportadores/metabolismo , Transmisión Sináptica/efectos de los fármacos , Testosterona/farmacología , Síndrome de Resistencia Androgénica/genética , Animales , Animales Recién Nacidos/metabolismo , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Hipocampo/efectos de los fármacos , Masculino , Mutación , Neuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/metabolismo , Caracteres Sexuales
10.
Eur J Pharmacol ; 919: 174795, 2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-35122868

RESUMEN

N-methyl-D-aspartate (NMDA) receptors are affected by many pharmaceuticals. In this work, we studied the action of the serine protease inhibitors nafamostat, gabexate and camostat, and an antiprotozoal compound, furamidine, on native NMDA receptors in rat hippocampal pyramidal neurons. Nafamostat, furamidine and gabexate inhibited these receptors with IC50 values of 0.20 ± 0.04, 0.64 ± 0.13 and 16 ± 3 µM, respectively, whereas camostat was ineffective. Nafamostat and furamidine showed voltage-dependent inhibition, while gabexate showed practically voltage-independent inhibition. Nafamostat and furamidine demonstrated tail currents, implying a 'foot-in-the-door' mechanism of action; gabexate did not demonstrate any signs of 'foot-in-the-door' or trapping channel block. Gabexate action was also not competitive, suggesting allosteric inhibition of NMDA receptors. Furamidine and nafamostat are structurally similar to the previously studied diminazene and all three demonstrated a 'foot-in-the-door' mechanism. They have a rather rigid, elongated structures and cannot fold into more compact forms. By contrast, the gabexate molecule can fold, but its folded structure differs drastically from that of typical NMDA receptor blockers, in agreement with its voltage-independent inhibition. These findings provide a better understanding of the structural determinants of NMDA receptor antagonism, while also supporting the potential clinical repurposing of these drugs as neuroprotectors for glaucoma and other neurodegenerative diseases.


Asunto(s)
Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Inhibidores de Serina Proteinasa/farmacología , Animales , Benzamidinas/farmacología , Benzamidinas/uso terapéutico , Reposicionamiento de Medicamentos , Ésteres/farmacología , Ésteres/uso terapéutico , Gabexato/farmacología , Gabexato/uso terapéutico , Guanidinas/farmacología , Guanidinas/uso terapéutico , Hipocampo/efectos de los fármacos , Concentración 50 Inhibidora , Masculino , Modelos Animales , Enfermedades Neurodegenerativas/tratamiento farmacológico , Células Piramidales/efectos de los fármacos , Ratas , Ratas Wistar , Inhibidores de Serina Proteinasa/uso terapéutico
11.
Int J Mol Sci ; 23(3)2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-35163623

RESUMEN

In neurons, changes in Akt activity have been detected in response to the stimulation of transmembrane receptors. However, the mechanisms that lead to changes in neuronal function upon Akt inhibition are still poorly understood. In the present study, we interrogate how Akt inhibition could affect the activity of the neuronal Nav channels with while impacting intrinsic excitability. To that end, we employed voltage-clamp electrophysiological recordings in heterologous cells expressing the Nav1.6 channel isoform and in hippocampal CA1 pyramidal neurons in the presence of triciribine, an inhibitor of Akt. We showed that in both systems, Akt inhibition resulted in a potentiation of peak transient Na+ current (INa) density. Akt inhibition correspondingly led to an increase in the action potential firing of the CA1 pyramidal neurons that was accompanied by a decrease in the action potential current threshold. Complementary confocal analysis in the CA1 pyramidal neurons showed that the inhibition of Akt is associated with the lengthening of Nav1.6 fluorescent intensity along the axonal initial segment (AIS), providing a mechanism for augmented neuronal excitability. Taken together, these findings provide evidence that Akt-mediated signal transduction might affect neuronal excitability in a Nav1.6-dependent manner.


Asunto(s)
Potenciales de Acción , Hipocampo/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Animales , Células HEK293 , Hipocampo/metabolismo , Hipocampo/fisiología , Humanos , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/fisiología
12.
Neurol Res ; 44(2): 128-138, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34396932

RESUMEN

OBJECTIVES: Lithium exerts a broad neuroprotective effect on the brain. This study examined whether lithium exerts therapeutic effects on stroke by restoring neural connections at the ischemic core of cortices post brain insult. METHODS: We treated rats with lithium or vehicle (saline) every 24 h for the first 72 h, starting at the beginning of reperfusion after inducing middle cerebral artery occlusion (MCAO) in rats. Somatosensory evoked potential (SSEP) recording and behavioral testing were employed to evaluate the beneficial effects of lithium treatment. To examine the effects of lithium-induced neuroplasticity, we evaluated the dendritic morphology in cortex pyramidal cells and the primary neuronal cell culture that underwent brain insults and oxygen and glucose deprivation (OGD), respectively. RESULTS: The results demonstrated that rats subjected to MCAO had prolonged N1 latency and a decreased N1/P1 amplitude at the ipsilateral cortex. Four doses of lithium reduced the brain infarction volume and enhanced the SSEP amplitude. The results of neurobehavioral tests demonstrated that lithium treatment improved sensory function, as demonstrated by improved 28-point clinical scale scores. In vitro study results showed that lithium treatment increased the dendritic lengths and branches of cultured neurons and reversed the suppressive effects of OGD. The in vivo study results indicated that lithium treatment increased cortical spine density in various layers and resulted in the development of the dendritic structure in the contralateral hemisphere. CONCLUSION: Our study confirmed that neuroplasticity in cortical neurons is crucial for lithium-induced brain function 50 recovery after brain ischemia.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Potenciales Evocados Somatosensoriales/efectos de los fármacos , Infarto de la Arteria Cerebral Media/complicaciones , Accidente Cerebrovascular Isquémico/complicaciones , Compuestos de Litio/farmacología , Plasticidad Neuronal/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Células Piramidales/efectos de los fármacos , Daño por Reperfusión/etiología , Daño por Reperfusión/prevención & control , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Compuestos de Litio/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Ratas
13.
Neuropharmacology ; 202: 108846, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34687710

RESUMEN

Drugs that block N-methyl-d-aspartate receptors (NMDARs) suppress hippocampus-dependent memory formation; they also block long-term potentiation (LTP), a cellular model of learning and memory. However, the fractional block that is required to achieve these effects is unknown. Here, we measured the dose-dependent suppression of contextual memory in vivo by systemic administration of the competitive antagonist (R,S)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid (CPP); in parallel, we measured the concentration-dependent block by CPP of NMDAR-mediated synapses and LTP of excitatory synapses in hippocampal brain slices in vitro. We found that the dose of CPP that suppresses contextual memory in vivo (EC50 = 2.3 mg/kg) corresponds to a free concentration of 53 nM. Surprisingly, applying this concentration of CPP to hippocampal brain slices had no effect on the NMDAR component of evoked field excitatory postsynaptic potentials (fEPSPNMDA), or on LTP. Rather, the IC50 for blocking the fEPSPNMDA was 434 nM, and for blocking LTP was 361 nM - both nearly an order of magnitude higher. We conclude that memory impairment produced by systemically administered CPP is not due primarily to its blockade of NMDARs on hippocampal pyramidal neurons. Rather, systemic CPP suppresses memory formation by actions elsewhere in the memory-encoding circuitry.


Asunto(s)
Región CA1 Hipocampal/fisiología , Aprendizaje/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Memoria/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Relación Dosis-Respuesta a Droga , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Técnicas In Vitro , Masculino , Ratones Endogámicos C57BL
14.
Neurobiol Aging ; 109: 113-124, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34715442

RESUMEN

Layer 3 (L3) pyramidal neurons in aged rhesus monkey lateral prefrontal cortex (LPFC) exhibit significantly elevated excitability in vitro and reduced spine density compared to neurons in young subjects. The time-course of these alterations, and whether they can be ameliorated in middle age by the powerful anti-oxidant curcumin is unknown. We compared the properties of L3 pyramidal neurons from the LPFC of behaviorally characterized rhesus monkeys over the adult lifespan using whole-cell patch clamp recordings and neuronal reconstructions. Working memory (WM) impairment, neuronal hyperexcitability, and spine loss began in middle age. There was no significant relationship between neuronal properties and WM performance. Middle-aged subjects given curcumin exhibited better WM performance and less neuronal excitability compared to control subjects. These findings suggest that the appropriate time frame for intervention for age-related cognitive changes is early middle age, and points to the efficacy of curcumin in delaying WM decline. Because there was no relationship between excitability and behavior, the effects of curcumin on these measures appear to be independent.


Asunto(s)
Envejecimiento/efectos de los fármacos , Envejecimiento/patología , Curcumina/administración & dosificación , Curcumina/farmacología , Suplementos Dietéticos , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , Células Piramidales/efectos de los fármacos , Células Piramidales/patología , Factores de Edad , Envejecimiento/psicología , Animales , Femenino , Macaca mulatta , Masculino , Técnicas de Placa-Clamp , Células Piramidales/fisiología , Factores de Tiempo
15.
Biomed Pharmacother ; 145: 112446, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34808556

RESUMEN

Cordycepin (known as 3-deoxyadenosine, CRD), a natural product from the valuable traditional Chinese medicine Cordyceps militaris, has been reported to improve cognitive function and modulate neuroprotective effects on the central nervous system (CNS). However, the modulating mechanisms of cordycepin on information processing in hippocampal CA1 pyramidal neurons are not fully understood. To clarify how cordycepin modulates synaptic responses of pyramidal neurons in rat hippocampal CA1 region, we conducted an electrophysiological experiment using whole-cell patch-clamp technique. The spontaneous and miniature excitatory postsynaptic currents (sEPSCs and mEPSCs, respectively) and the spontaneous and miniature inhibitory postsynaptic currents (sIPSCs and mIPSCs, respectively) recorded by this technique evaluated pure single or multi-synapse responses and enabled us to accurately quantify how cordycepin influenced the pre and postsynaptic aspects of synaptic transmission. The present results showed that cordycepin significantly decreased the frequency of both glutamatergic and GABAergic postsynaptic currents without affecting the amplitude, while these inhibitory effects were antagonized by the A1 adenosine receptor antagonist (DPCPX), but not the A2A (ZM 241385), A2B (MRS1754) and A3 (MRS1191) adenosine receptor antagonists. Taken together, our results suggested that cordycepin had a clear presynaptic effect on glutamatergic and GABAergic transmission, and provided novel evidence that cordycepin suppresses the synaptic transmission through the activation of A1AR.


Asunto(s)
Desoxiadenosinas/farmacología , Fármacos Neuroprotectores/farmacología , Células Piramidales/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Femenino , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Células Piramidales/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A1/efectos de los fármacos , Receptor de Adenosina A1/metabolismo , Ácido gamma-Aminobutírico/metabolismo
16.
Neuropharmacology ; 206: 108926, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-34921828

RESUMEN

Perseveration is a characteristic of patients with obsessive-compulsive disorder (OCD). Clinically, neuronal activity in the lateral orbitofrontal cortex (OFC) is increased in OCD patients. Successful treatment with selective serotonin reuptake inhibitors (SSRIs) reduces activity in the lateral OFC of OCD patients, but the precise mechanisms underlying this effect are unclear. Previously, we reported that repeated injection of the dopamine D2 receptor agonist quinpirole (QNP) resulted in OCD-like deficits, including perseveration in a reversal learning task. QNP-treated mice showed hyperactivity in lateral OFC pyramidal neurons. The present study demonstrated that 4-week administration of an SSRI increased the rate of correct choice in a reversal learning task. Using the electrophysiological approach, we revealed that an SSRI decreased the activity of lateral OFC pyramidal neurons in QNP-treated mice by potentiating inhibitory inputs. The 4-week administration of an SSRI inhibited the potentiation of neuronal activity induced by a 5-HT2C receptor agonist. Additionally, both 4-week administration of SSRI and acute application of 5-HT2C receptor antagonist prevented the QNP-induced potentiation of inhibitory inputs to fast-spiking interneurons in the lateral OFC. Administration of a 5-HT2C receptor antagonist to mice for 4 days increased the rate of correct choice in a reversal learning task. Collectively, these results indicate that chronic SSRI ameliorated perseverative behavior in QNP-treated mice by modulating inhibitory inputs in the lateral OFC. Short-term 5-HT2C receptor blockade also ameliorated QNP-induced behavioral and neurological abnormalities by, at least in part, a common mechanism with chronic SSRI.


Asunto(s)
Conducta Animal/efectos de los fármacos , Trastorno Obsesivo Compulsivo/tratamiento farmacológico , Corteza Prefrontal/efectos de los fármacos , Receptor de Serotonina 5-HT2C/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Animales , Modelos Animales de Enfermedad , Interneuronas/efectos de los fármacos , Ratones , Células Piramidales/efectos de los fármacos , Aprendizaje Inverso/efectos de los fármacos , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Transducción de Señal/efectos de los fármacos
17.
Cell Rep ; 37(9): 109933, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34852233

RESUMEN

Pyramidal neurons in the anterior cingulate cortex (ACC), a prefrontal region involved in processing the affective components of pain, display hyperexcitability in chronic neuropathic pain conditions, and their silencing abolishes hyperalgesia. We show that dopamine, through D1 receptor (D1R) signaling, inhibits pyramidal neurons of mouse ACC by modulation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Activation of Gs-coupled D1R by dopamine induces the opening of HCN channels at physiological membrane potentials, driving a significant decrease in input resistance and excitability. Systemic L-DOPA in chronic neuropathic mice rescues HCN channel activity, normalizes pyramidal excitability in ACC, and blocks mechanical and thermal allodynia. Moreover, microinjection of a selective D1R agonist in the ACC relieves the aversiveness of ongoing neuropathic pain, while an ACC D1R antagonist blocks gabapentin- and lidocaine-evoked antinociception. We conclude that dopaminergic inhibition via D1R in ACC plays an analgesic role in physiological conditions and is decreased in chronic pain.


Asunto(s)
Dopamina/metabolismo , Giro del Cíngulo/efectos de los fármacos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Levodopa/farmacología , Neuralgia/prevención & control , Canales de Potasio/metabolismo , Células Piramidales/efectos de los fármacos , Receptores de Dopamina D1/agonistas , Animales , Dopaminérgicos/farmacología , Giro del Cíngulo/metabolismo , Giro del Cíngulo/patología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Masculino , Potenciales de la Membrana , Neuralgia/etiología , Neuralgia/metabolismo , Neuralgia/patología , Canales de Potasio/genética , Células Piramidales/metabolismo , Células Piramidales/patología , Ratas , Ratas Sprague-Dawley
18.
Biochem Biophys Res Commun ; 585: 29-35, 2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-34781058

RESUMEN

Epidemiologic evidence has suggested a relationship between di (2-ethylhexyl) phthalate (DEHP) prenatal exposure and autism spectrum disorders (ASD), but the underlying mechanisms are still at large unknown. In this study, pregnant mice were intragastrically administered with DEHP once a day from GD 3 to GD 17 and the neurobehavioral changes of offspring were evaluated. In addition to the repetitive stereotyped behaviors, DEHP at the concentration of 50 mg/kg/day and above significantly impaired the sociability of the offspring (P < 0.05) and decreased the density of dendritic spines of pyramidal neurons in the prefrontal cortex (P < 0.05). At the same time, the expression of Nischarin protein in prefrontal lobe increased (P < 0.05). Similarly, after 12-h incubation of DEHP at the concentration of 100 nM, the total spine density, especially the mushroom and stubby spine populations, significantly decreased in the primary cultured prefrontal cortical neurons (P < 0.05). However, the inhibitory effect of DEHP were reversed by knockdown of Nischarin expression. Collectively, these results suggest that prenatal DEHP exposure induces Nischarin expression, causes dendritic spine loss, and finally leads to autism-like behavior in mouse offspring.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Dietilhexil Ftalato/toxicidad , Receptores de Imidazolina/metabolismo , Corteza Prefrontal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Trastorno del Espectro Autista/inducido químicamente , Línea Celular Tumoral , Células Cultivadas , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/fisiología , Femenino , Receptores de Imidazolina/genética , Ratones Endogámicos ICR , Plastificantes/toxicidad , Corteza Prefrontal/citología , Corteza Prefrontal/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Conducta Social
19.
PLoS Comput Biol ; 17(10): e1009435, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34597293

RESUMEN

In the hippocampus, episodic memories are thought to be encoded by the formation of ensembles of synaptically coupled CA3 pyramidal cells driven by sparse but powerful mossy fiber inputs from dentate gyrus granule cells. The neuromodulators acetylcholine and noradrenaline are separately proposed as saliency signals that dictate memory encoding but it is not known if they represent distinct signals with separate mechanisms. Here, we show experimentally that acetylcholine, and to a lesser extent noradrenaline, suppress feed-forward inhibition and enhance Excitatory-Inhibitory ratio in the mossy fiber pathway but CA3 recurrent network properties are only altered by acetylcholine. We explore the implications of these findings on CA3 ensemble formation using a hierarchy of models. In reconstructions of CA3 pyramidal cells, mossy fiber pathway disinhibition facilitates postsynaptic dendritic depolarization known to be required for synaptic plasticity at CA3-CA3 recurrent synapses. We further show in a spiking neural network model of CA3 how acetylcholine-specific network alterations can drive rapid overlapping ensemble formation. Thus, through these distinct sets of mechanisms, acetylcholine and noradrenaline facilitate the formation of neuronal ensembles in CA3 that encode salient episodic memories in the hippocampus but acetylcholine selectively enhances the density of memory storage.


Asunto(s)
Acetilcolina/farmacología , Región CA3 Hipocampal , Memoria , Norepinefrina/farmacología , Animales , Región CA3 Hipocampal/citología , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/fisiología , Biología Computacional , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Neurológicos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos
20.
J Integr Neurosci ; 20(3): 613-622, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34645094

RESUMEN

As a gamma-aminobutyric acid type A receptor agonist sevoflurane is a common general anesthetic used in anesthesia and affects the neural development in offspring. We hypothesized that sevoflurane could regulate interneurons via the neuregulin-1-epidermal growth factor receptor-4 (NRG1-ErbB4) pathway in the entorhinal cortex (ECT) of the middle pregnancy. Six female rats in middle pregnancy (14.5 days of pregnancy) were randomly and equally divided into sevoflurane (SeV) and control groups. The rats in the SeV group were exposed to 4% sevoflurane for 3 hours. The expression levels of NRG1 and ErbB4, parvalbumin (PV) and glutamic acid decarboxylase (GAD67), and N-methyl-D-aspartate receptor subunit 2A (NR2A) and subunit 2B (NR2B) in offspring were examined through immunohistochemistry. The pyramidal neurons in the ECT were examined via Golgi staining. The levels of NRG1 and ErbB4 were significantly decreased (P < 0.01) and the levels of PV and GAD67 (interneurons) were found to be decreased in the SeV group (P < 0.01). The level of NR2B was found to be increased while the level of NR2A being decreased in the SeV group (P < 0.01). The development of pyramidal neurons was abnormal in the SeV group (P < 0.05). Conclusively, prenatal sevoflurane exposure could lead to the disturbance of the interneurons by activating the NRG1-ErbB4 pathway and subsequently result in abnormal development of pyramidal neurons in middle pregnancy. Prenatal sevoflurane exposure in middle pregnancy could be potentially harmful to the neural development of rat offspring. This study may reveal a novel pathway in the influence mechanism of sevoflurane on rat offspring.


Asunto(s)
Corteza Entorrinal/efectos de los fármacos , Agonistas de Receptores de GABA-A/farmacología , Interneuronas/efectos de los fármacos , Neurregulina-1/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Células Piramidales/efectos de los fármacos , Receptor ErbB-4/efectos de los fármacos , Sevoflurano/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Agonistas de Receptores de GABA-A/administración & dosificación , Embarazo , Ratas , Sevoflurano/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...