Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; 36(25): e2400347, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38573812

RESUMO

Suppressing trap-assisted nonradiative losses through passivators is a prerequisite for efficient perovskite light-emitting diodes (PeLEDs). However, the complex bonding between passivators and perovskites severely suppresses the passivation process, which still lacks comprehensive understanding. Herein, the number, category, and degree of bonds between different functional groups and the perovskite are quantitatively assessed to study the passivation dynamics. Functional groups with high electrostatic potential and large steric hindrance prioritize strong bonding with organic cations and halides on the perfect surface, leading to suppressed coordination with bulky defects. By modulating the binding priorities and coordination capacity, hindrance from the intense interaction with perfect perovskite is significantly reduced, leading to a more direct passivation process. Consequently, the near-infrared PeLED without external light out-coupling demonstrates a record external quantum efficiency of 24.3% at a current density of 42 mA cm-2. In addition, the device exhibits a record-level-cycle ON/OFF switching of 20 000 and ultralong half-lifetime of 1126.3 h under 5 mA cm-2. An in-depth understanding of the passivators can offer new insights into the development of high-performance PeLEDs.

2.
Hum Mol Genet ; 19(19): 3797-805, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20634195

RESUMO

The reduced expression of the Sp4 gene in Sp4 hypomorphic mice resulted in subtle vacuolization in the hippocampus as well as deficits in sensorimotor gating and contextual memory, putative endophenotypes for schizophrenia and other psychiatric disorders. In this study, we examined both spatial learning/memory and hippocampal long-term potentiation (LTP) of Sp4 hypomorphic mice. Impaired spatial learning/memory and markedly reduced LTP were found. To corroborate the functional studies, the expression of N-methyl-D-aspartate (NMDA) glutamate receptors was investigated with both western blot and immunohistochemical analyses. The reduced expression of the Sp4 gene decreased the level of the NR1 subunit of NMDA receptors in Sp4 hypomorphic mice. In human, SP4 gene was found to be deleted sporadically in schizophrenia patients, corroborating evidence that polymorphisms of human SP4 gene are associated with schizophrenia and other psychiatric disorders. Impaired NMDA neurotransmission has been implicated in several human psychiatric disorders. As yet, it remains unclear how mutations of candidate susceptibility genes for these disorders may contribute to the disruption of NMDA neurotransmission. Sp4 hypomorphic mice could therefore serve as a genetic model to investigate impaired NMDA functions resulting from loss-of-function mutations of human SP4 gene in schizophrenia and/or other psychiatric disorders. Furthermore, aberrant expression of additional genes, besides NMDAR1, likely also contributes to the behavioral abnormalities in Sp4 hypomorphic mice. Thus, further investigation of the Sp4 pathway may provide novel insights in our understanding of a variety of neuropsychiatric disorders.


Assuntos
Endofenótipos/metabolismo , Transtornos Mentais/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Fator de Transcrição Sp4/metabolismo , Animais , Região CA1 Hipocampal/patologia , Região CA1 Hipocampal/fisiopatologia , Fenômenos Eletrofisiológicos , Deleção de Genes , Homozigoto , Humanos , Potenciação de Longa Duração/fisiologia , Aprendizagem em Labirinto , Transtornos Mentais/fisiopatologia , Camundongos , Subunidades Proteicas/metabolismo , Esquizofrenia/genética , Esquizofrenia/fisiopatologia , Fator de Transcrição Sp4/genética
4.
ScientificWorldJournal ; 9: 68-85, 2009 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-19151899

RESUMO

Corticotropin-releasing factor (CRF) is a 41-amino-acid neuropeptide involved in stress responses initiated from several brain areas, including the amygdala formation. Research shows a strong relationship between stress, brain CRF, and excessive alcohol consumption. Behavioral studies suggest that the central amygdala (CeA) is significantly involved in alcohol reward and dependence. We recently reported that the ethanol augmentation of GABAergic synaptic transmission in rat CeA involves CRF1 receptors, because both CRF and ethanol significantly enhanced the amplitude of evoked GABAergic inhibitory postsynaptic currents (IPSCs) in CeA neurons from wild-type (WT) and CRF2 knockout (KO) mice, but not in neurons of CRF1 KO mice. The present study extends these findings using selective CRF receptor ligands, gene KO models, and miniature IPSC (mIPSC) analysis to assess further a presynaptic role for the CRF receptors in mediating ethanol effects in the CeA. In whole-cell patch recordings of pharmacologically isolated GABAAergic IPSCs from slices of mouse CeA, both CRF and ethanol augmented evoked IPSCs in a concentration-dependent manner, with low EC50s. A CRF1 (but not CRF2) KO construct and the CRF1-selective nonpeptide antagonist NIH-3 (LWH-63) blocked the augmenting effect of both CRF and ethanol on evoked IPSCs. Furthermore, the new selective CRF1 agonist stressin1, but not the CRF2 agonist urocortin 3, also increased evoked IPSC amplitudes. Both CRF and ethanol decreased paired-pulse facilitation (PPF) of evoked IPSCs and significantly enhanced the frequency, but not the amplitude, of spontaneous miniature GABAergic mIPSCs in CeA neurons of WT mice, suggesting a presynaptic site of action. The PPF effect of ethanol was abolished in CeA neurons of CRF1 KO mice. The CRF1 antagonist NIH-3 blocked the CRF- and ethanol-induced enhancement of mIPSC frequency in CeA neurons. These data indicate that presynaptic CRF1 receptors play a critical role in permitting or mediating ethanol enhancement of GABAergic synaptic transmission in CeA, via increased vesicular GABA release, and thus may be a rational target for the treatment of alcohol abuse and alcoholism.


Assuntos
Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Etanol/farmacologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/biossíntese , Animais , Eletrofisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , Receptores de Hormônio Liberador da Corticotropina/agonistas , Receptores de Hormônio Liberador da Corticotropina/deficiência , Receptores de Hormônio Liberador da Corticotropina/genética
5.
Proc Natl Acad Sci U S A ; 105(27): 9397-402, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18599437

RESUMO

Emerging evidence suggests that myocyte enhancer factor 2 (MEF2) transcription factors act as effectors of neurogenesis in the brain, with MEF2C the predominant isoform in developing cerebrocortex. Here, we show that conditional knockout of Mef2c in nestin-expressing neural stem/progenitor cells (NSCs) impaired neuronal differentiation in vivo, resulting in aberrant compaction and smaller somal size. NSC proliferation and survival were not affected. Conditional null mice surviving to adulthood manifested more immature electrophysiological network properties and severe behavioral deficits reminiscent of Rett syndrome, an autism-related disorder. Our data support a crucial role for MEF2C in programming early neuronal differentiation and proper distribution within the layers of the neocortex.


Assuntos
Diferenciação Celular , Fatores de Regulação Miogênica/metabolismo , Neurônios/citologia , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Comportamento , Cognição , Eletrofisiologia , Desenvolvimento Embrionário , Fatores de Transcrição MEF2 , Camundongos , Camundongos Knockout , Mitose , Neocórtex/embriologia , Neocórtex/patologia , Neurônios/patologia , Fenótipo
6.
J Neurosci ; 28(26): 6557-68, 2008 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-18579729

RESUMO

Cell-based therapies require a reliable source of cells that can be easily grown, undergo directed differentiation, and remain viable after transplantation. Here, we generated stably transformed murine ES (embryonic stem) cells that express a constitutively active form of myocyte enhancer factor 2C (MEF2CA). MEF2C has been implicated as a calcium-dependent transcription factor that enhances survival and affects synapse formation of neurons as well as differentiation of cardiomyocytes. We now report that expression of MEF2CA, both in vitro and in vivo, under regulation of the nestin enhancer effectively produces "neuronal" progenitor cells that differentiate into a virtually pure population of neurons. Histological, electrophysiological, and behavioral analyses demonstrate that MEF2C-directed neuronal progenitor cells transplanted into a mouse model of cerebral ischemia can successfully differentiate into functioning neurons and ameliorate stroke-induced behavioral deficits.


Assuntos
Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Fatores de Regulação Miogênica/genética , Transplante de Células-Tronco/métodos , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Animais , Apoptose/genética , Isquemia Encefálica/terapia , Transplante de Tecido Encefálico/métodos , Diferenciação Celular/fisiologia , Linhagem Celular Transformada , Proliferação de Células , Sobrevivência Celular/genética , Células Cultivadas , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica/genética , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Fatores de Transcrição MEF2 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/etiologia , Degeneração Neural/fisiopatologia , Degeneração Neural/terapia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurônios/citologia , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Células-Tronco/citologia
7.
J Neurophysiol ; 99(1): 122-32, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18003876

RESUMO

Expression of the NR3A subunit with NR1/NR2 in Xenopus oocytes or mammalian cell lines leads to a reduction in N-methyl-d-aspartate (NMDA)-induced currents and decreased Mg(2+) sensitivity and Ca(2+) permeability compared with NR1/NR2 receptors. Consistent with these findings, neurons from NR3A knockout (KO) mice exhibit enhanced NMDA-induced currents. Recombinant NR3A can also form excitatory glycine receptors with NR1 in the absence of NR2. However, the effects of NR3A on channel properties in neurons and synaptic transmission have not been fully elucidated. To study physiological roles of NR3A subunits, we generated NR3A transgenic (Tg) mice. Cultured NR3A Tg neurons exhibited two populations of NMDA receptor (NMDAR) channels, reduced Mg(2+) sensitivity, and decreased Ca(2+) permeability in response to NMDA/glycine, but glycine alone did not elicit excitatory currents. In addition, NMDAR-mediated excitatory postsynaptic currents (EPSCs) in NR3A Tg hippocampal slices showed reduced Mg(2+) sensitivity, consistent with the notion that NR3A subunits incorporated into synaptic NMDARs. To study the function of endogenous NR3A subunits, we compared NMDAR-mediated EPSCs in NR3A KO and WT control mice. In NR3A KO mice, the ratio of the amplitudes of the NMDAR-mediated component to alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor-mediated component of the EPSC was significantly larger than that seen in WT littermates. This result suggests that NR3A subunits contributed to the NMDAR-mediated component of the EPSC in WT mice. Taken together, these results show that NR3A subunits contribute to NMDAR responses from both synaptic and extrasynaptic receptors, likely composed of NR1, NR2, and NR3 subunits.


Assuntos
Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Transmissão Sináptica/genética , Animais , Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Células Cultivadas , Córtex Cerebral/citologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Ativação do Canal Iônico/genética , Magnésio/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Ratos , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Proteínas Recombinantes de Fusão/genética , Transmissão Sináptica/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
8.
Pharmacol Ther ; 107(1): 80-98, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15963352

RESUMO

Considerable evidence suggests that the synapse is the most sensitive CNS element for ethanol effects. Although most alcohol research has focussed on the postsynaptic sites of ethanol action, especially regarding interactions with the glutamatergic and GABAergic receptors, few such studies have directly addressed the possible presynaptic loci of ethanol action, and even fewer describe effects on synaptic terminals. Nonetheless, there is burgeoning evidence that presynaptic terminals play a major role in ethanol effects. The methods used to verify such ethanol actions range from electrophysiological analysis of paired-pulse facilitation (PPF) and spontaneous and miniature synaptic potentials to direct recording of ion channel activity and transmitter/messenger release from acutely isolated synaptic terminals, and microscopic observation of vesicular release, with a focus predominantly on GABAergic, glutamatergic, and peptidergic synapses. The combined data suggest that acute ethanol administration can both increase and decrease the release of these transmitters from synaptic terminals, and more recent results suggest that prolonged or chronic ethanol treatment (CET) can also alter the function of presynaptic terminals. These new findings suggest that future analyses of synaptic effects of ethanol should attempt to ascertain the role of presynaptic terminals and their involvement in alcohol's behavioral actions. Other future directions should include an assessment of ethanol's effects on presynaptic signal transduction linkages and on the molecular machinery of transmitter release and exocytosis in general. Such studies could lead to the formulation of new treatment strategies for alcohol intoxication, alcohol abuse, and alcoholism.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Glutamatos , Terminações Pré-Sinápticas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ácido gama-Aminobutírico , Animais , Glutamatos/efeitos dos fármacos , Glutamatos/metabolismo , Glutamatos/fisiologia , Humanos , Transdução de Sinais/fisiologia , Ácido gama-Aminobutírico/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/fisiologia
9.
Science ; 303(5663): 1512-4, 2004 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-15001778

RESUMO

The central amygdala (CeA) plays a role in the relationship among stress, corticotropin-releasing factor (CRF), and alcohol abuse. In whole-cell recordings, both CRF and ethanol enhanced gamma-aminobutyric acid-mediated (GABAergic) neurotransmission in CeA neurons from wild-type and CRF2 receptor knockout mice, but not CRF1 receptor knockout mice. CRF1 (but not CRF2) receptor antagonists blocked both CRF and ethanol effects in wild-type mice. These data indicate that CRF1 receptors mediate ethanol enhancement of GABAergic synaptic transmission in the CeA, and they suggest a cellular mechanism underlying involvement of CRF in ethanol's behavioral and motivational effects.


Assuntos
Tonsila do Cerebelo/fisiologia , Etanol/farmacologia , Neurônios/fisiologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Consumo de Bebidas Alcoólicas , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Hormônio Liberador da Corticotropina/farmacologia , Relação Dose-Resposta a Droga , Potenciais Evocados/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de GABA-A/metabolismo , Estresse Psicológico/fisiopatologia
10.
Ann N Y Acad Sci ; 1003: 196-211, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14684447

RESUMO

Both the nucleus accumbens (NAcc) and central amygdala (CeA) are thought to play roles in tolerance to, and dependence on, abused drugs. Although our past studies in rat brain slices suggested a role for NMDA receptors (NMDARs) in NAcc neurons in the effects of acute and chronic opiate treatment, the cellular and molecular mechanisms remained unclear. Therefore, we examined the effects of morphine dependence on electrophysiological properties of NMDARs in freshly isolated NAcc neurons and on expression of mRNA coding for NR2A-C subunits using single-cell RT-PCR. Chronic morphine did not alter the affinity for NMDAR agonists glutamate, homoquinolinate, or NMDA, but decreased the affinity of the coagonist glycine. Chronic morphine altered the NMDAR inhibition by two NMDAR antagonists, 7-Cl-kynurenate and ifenprodil, but not that by d-APV or Mg2+. Chronic morphine accelerated the NMDA current desensitization rate in NAcc neurons. In single-cell RT-PCR, chronic morphine predominantly reduced the number of neurons expressing multiple NR2 subunits. Ethanol also alters NMDARs. We found that low ethanol concentrations (IC50 = 13 mM) inhibited NMDA currents and NMDA-EPSPs in most NAcc neurons in a slice preparation. NAcc neurons from ethanol-dependent rats showed enhanced NMDA sensitivity. In CeA neurons, acute ethanol decreased (by 10-25%) non-NMDA- and NMDA-EPSPs in most neurons. In CeA neurons from ethanol-dependent rats, acute ethanol decreased the non-NMDA-EPSPs to the same extent as in naïve rats, but inhibited (by 30-40%) NMDA-EPSPs significantly more than in controls, suggesting sensitization to ethanol. Preliminary studies with microdialysis and real-time PCR analysis support this idea: local ethanol administration in vivo had no effect on glutamate release, but chronic ethanol nearly tripled the expression of NR2B subunits (the most ethanol sensitive) in CeA. These combined findings suggest that changes in glutamatergic transmission in NAcc and CeA may underlie the neuroadaptions that lead to opiate and ethanol dependence.


Assuntos
Alcoolismo/fisiopatologia , Glutamatos/fisiologia , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Transmissão Sináptica/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Etanol/farmacologia , Glicina/metabolismo , Humanos , Dependência de Morfina/fisiopatologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Accumbens/fisiologia , Ratos , Receptores de N-Metil-D-Aspartato/biossíntese , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...