Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 671: 87-95, 2023 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-37300945

RESUMO

Stroke is the leading cause of death and long-term disability worldwide. But treatments are not available to promote functional recovery, and efficient therapies need to be investigated. Stem cell-based therapies hold great promise as potential technologies to restore function in brain disorders. Loss of GABAergic interneurons after stroke may result in sensorimotor defects. Here, by transplanting human brain organoids resembling the MGE domain (human MGE organoids, hMGEOs) derived from human induced pluripotent stem cells (hiPSCs) into the infarcted cortex of stroke mice, we found that grafted hMGEOs survived well and primarily differentiated into GABAergic interneurons and significantly restored the sensorimotor deficits of stroke mice for a long time. Our study offers the feasibility of stem cell replacement therapeutics strategy for stroke.


Assuntos
Células-Tronco Pluripotentes Induzidas , Acidente Vascular Cerebral , Humanos , Camundongos , Animais , Células-Tronco Pluripotentes Induzidas/fisiologia , Acidente Vascular Cerebral/terapia , Encéfalo , Interneurônios , Diferenciação Celular
2.
NPJ Regen Med ; 8(1): 27, 2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-37253754

RESUMO

Stroke usually causes prolonged or lifelong disability, owing to the permanent loss of infarcted tissue. Although a variety of stem cell transplantation has been explored to improve neuronal defect behavior by enhancing neuroplasticity, it remains unknown whether the infarcted tissue can be reconstructed. We here cultured human cerebral organoids derived from human pluripotent stem cells (hPSCs) and transplanted them into the junction of the infarct core and the peri-infarct zone of NOD-SCID mice subjected to stroke. Months later, we found that the grafted organoids survived well in the infarcted core, differentiated into target neurons, repaired infarcted tissue, sent axons to distant brain targets, and integrated into the host neural circuit and thereby eliminated sensorimotor defect behaviors of stroke mice, whereas transplantation of dissociated single cells from organoids failed to repair the infarcted tissue. Our study offers a new strategy for reconstructing infarcted tissue via organoids transplantation thereby reversing stroke-induced disability.

3.
Neurosci Bull ; 39(9): 1439-1453, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37074530

RESUMO

In the central nervous system, nitric oxide (NO), a free gas with multitudinous bioactivities, is mainly produced from the oxidation of L-arginine by neuronal nitric oxide synthase (nNOS). In the past 20 years, the studies in our group and other laboratories have suggested a significant involvement of nNOS in a variety of neurological and neuropsychiatric disorders. In particular, the interactions between the PDZ domain of nNOS and its adaptor proteins, including post-synaptic density 95, the carboxy-terminal PDZ ligand of nNOS, and the serotonin transporter, significantly influence the subcellular localization and functions of nNOS in the brain. The nNOS-mediated protein-protein interactions provide new attractive targets and guide the discovery of therapeutic drugs for neurological and neuropsychiatric disorders. Here, we summarize the work on the roles of nNOS and its association with multiple adaptor proteins on neurological and neuropsychiatric disorders.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Doenças do Sistema Nervoso , Humanos , Óxido Nítrico Sintase Tipo I/metabolismo , Encéfalo/metabolismo
4.
J Affect Disord ; 333: 181-192, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37080493

RESUMO

BACKGROUND: The basolateral amygdala (BLA) neurons are primarily glutamatergic and have been associated with emotion regulation. However, little is known about the roles of BLA neurons expressing neuronal nitric oxide synthase (nNOS, Nos1) in the regulation of emotional behaviors. METHODS: Using Nos1-cre mice and chemogenetic and optogenetic manipulations, we specifically silenced or activated Nos1+ or Nos1- neurons in the BLA, or silenced their projections to the anterdorsal bed nucleus of the stria terminalis (adBNST) and ventral hippocampus (vHPC). We measured anxiety behaviors in elevated plus maze (EPM) and open-field test (OFT), and measured depression behaviors in forced swimming test (FST) and tail suspension test (TST). RESULTS: BLA Nos1+ neurons were predominantly glutamatergic, and glutamatergic but not GABAergic Nos1+ neurons were involved in controlling anxiety- and depression-related behaviors. Interestingly, by selectively manipulating the activities of BLA Nos1+ and Nos1- excitatory neurons, we found that they had opposing effects on anxiety- and depression-related behaviors. BLA Nos1+ excitatory neurons projected to the adBNST, this BLA-adBNST circuit controlled the expression of anxiety- and depression-related behaviors, while BLA Nos1- excitatory neurons projected to vHPC, this BLA-vHPC circuit contributed to the expression of anxiety- and depression-related behaviors. Moreover, excitatory vHPC-adBNST circuit antagonized the role of BLA-adBNST circuit in regulating anxiety- and depression-related behaviors. CONCLUSIONS: BLA Nos1+ and Nos1- excitatory neuron subpopulations exert different effects on anxiety- and depression-related behaviors through distinct projection circuits, providing a new insight of BLA excitatory neurons in emotional regulation. LIMITATIONS: We did not perform retrograde labeling from adBNST and vHPC regions.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Camundongos , Animais , Complexo Nuclear Basolateral da Amígdala/metabolismo , Depressão , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Ansiedade , Neurônios/metabolismo
5.
Cell Rep ; 42(4): 112294, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-36947544

RESUMO

Stroke is a leading cause of adult disability worldwide, and better drugs are needed to promote functional recovery after stroke. Growing evidence suggests the critical role of network excitability during the repair phase for stroke recovery. Here, we show that ß-hydroxybutyrate (ß-HB), an essential ketone body (KB) component, is positively correlated with improved outcomes in patients with stroke and promotes functional recovery in rodents with stroke during the repair phase. These beneficial effects of ß-HB depend on HDAC2/HDAC3-GABA transporter 1 (GAT-1) signaling-mediated enhancement of excitability and phasic GABA inhibition in the peri-infarct cortex and structural and functional plasticity in the ipsilateral cortex, the contralateral cortex, and the corticospinal tract. Together with available clinical approaches to elevate KB levels, our results offer a clinically translatable means to promote stroke recovery. Furthermore, GAT-1 can serve as a pharmacological target for developing drugs to promote functional recovery after stroke.


Assuntos
Corpos Cetônicos , Acidente Vascular Cerebral , Humanos , Proteínas da Membrana Plasmática de Transporte de GABA
6.
Acta Pharmacol Sin ; 44(5): 954-968, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36460834

RESUMO

Chronic pain patients often have anxiety disorders, and some of them suffer from anxiety even after analgesic administration. In this study, we investigated the role of AMPAR-mediated synaptic transmission in the ventromedial prefrontal cortex (vmPFC) in chronic pain-induced persistent anxiety in mice and explored potential drug targets. Chronic inflammatory pain was induced in mice by bilateral injection of complete Freund's adjuvant (CFA) into the planta of the hind paws; anxiety-like behaviours were assessed with behavioural tests; S-nitrosylation and AMPAR-mediated synaptic transmission were examined using biochemical assays and electrophysiological recordings, respectively. We found that CFA induced persistent upregulation of AMPAR membrane expression and function in the vmPFC of anxious mice but not in the vmPFC of non-anxious mice. The anxious mice exhibited higher S-nitrosylation of stargazin (an AMPAR-interacting protein) in the vmPFC. Inhibition of S-nitrosylation by bilaterally infusing an exogenous stargazin (C302S) mutant into the vmPFC rescued the surface expression of GluA1 and AMPAR-mediated synaptic transmission as well as the anxiety-like behaviours in CFA-injected mice, even after ibuprofen treatment. Moreover, administration of ZL006, a small molecular inhibitor disrupting the interaction of nNOS and PSD-95 (20 mg·kg-1·d-1, for 5 days, i.p.), significantly reduced nitric oxide production and S-nitrosylation of AMPAR-interacting proteins in the vmPFC, resulting in anxiolytic-like effects in anxious mice after ibuprofen treatment. We conclude that S-nitrosylation is necessary for AMPAR trafficking and function in the vmPFC under chronic inflammatory pain-induced persistent anxiety conditions, and nNOS-PSD-95 inhibitors could be potential anxiolytics specific for chronic inflammatory pain-induced persistent anxiety after analgesic treatment.


Assuntos
Ansiedade , Dor Crônica , Córtex Pré-Frontal , Receptores de Glutamato , Animais , Camundongos , Ansiedade/etiologia , Ansiedade/metabolismo , Transtornos de Ansiedade , Dor Crônica/complicações , Dor Crônica/metabolismo , Ibuprofeno , Córtex Pré-Frontal/metabolismo , Transmissão Sináptica , Receptores de Glutamato/química , Receptores de Glutamato/metabolismo , Inflamação/complicações , Inflamação/metabolismo
7.
Science ; 378(6618): 390-398, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36302033

RESUMO

Major depressive disorder (MDD) is one of the most common mental disorders. We designed a fast-onset antidepressant that works by disrupting the interaction between the serotonin transporter (SERT) and neuronal nitric oxide synthase (nNOS) in the dorsal raphe nucleus (DRN). Chronic unpredictable mild stress (CMS) selectively increased the SERT-nNOS complex in the DRN in mice. Augmentation of SERT-nNOS interactions in the DRN caused a depression-like phenotype and accounted for the CMS-induced depressive behaviors. Disrupting the SERT-nNOS interaction produced a fast-onset antidepressant effect by enhancing serotonin signaling in forebrain circuits. We discovered a small-molecule compound, ZZL-7, that elicited an antidepressant effect 2 hours after treatment without undesirable side effects. This compound, or analogous reagents, may serve as a new, rapidly acting treatment for MDD.


Assuntos
Antidepressivos , Transtorno Depressivo Maior , Núcleo Dorsal da Rafe , Desenho de Fármacos , Óxido Nítrico Sintase Tipo I , Proteínas da Membrana Plasmática de Transporte de Serotonina , Animais , Camundongos , Antidepressivos/química , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Transtorno Depressivo Maior/tratamento farmacológico , Núcleo Dorsal da Rafe/efeitos dos fármacos , Núcleo Dorsal da Rafe/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo
8.
Cell Death Dis ; 13(5): 462, 2022 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-35568723

RESUMO

Blood-brain barrier (BBB) dysfunction has been recognized as an early pathological feature and contributing factor in multiple sclerosis. Endothelial-to-mesenchymal transition is a process associated with endothelial dysfunction leading to the disruption of vessel stability and barrier function, yet its functional consequence in multiple sclerosis remains unclear. Here, we demonstrated that endothelial-to-mesenchymal transition accompanied the blood-brain barrier dysfunction in several neurological disorders, especially in multiple sclerosis. The activity of transcription factor ETS1, which is highly expressed in endothelial cells (ECs) and responded to an inflammatory condition, is suppressed in the central nervous system (CNS) ECs in MS and its animal model experimental autoimmune encephalomyelitis. We identify ETS1 as a central regulator of endothelial-to-mesenchymal transition (EndMT) associated with the compromise of barrier integrity. These phenotypical and functional alterations can further induce high permeability, immune infiltration, and organ fibrosis in multiple sclerosis, thus promoting disease progression. Together, these results demonstrate a functional role of EndMT in blood-brain barrier dysfunction and propose ETS1 as a potential transcriptional switch of EndMT to target the development of multiple sclerosis.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Animais , Barreira Hematoencefálica/patologia , Encefalomielite Autoimune Experimental/patologia , Células Endoteliais/patologia , Endotélio/patologia , Esclerose Múltipla/patologia
9.
Biochem Biophys Res Commun ; 585: 139-145, 2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-34801934

RESUMO

The pathogenesis of post-traumatic stress disorder (PTSD) remains largely unclear. A large body of evidence suggests that the abnormal level of serotonin (5-HT) is closely related to the onset of PTSD. Several reports reveal that nitric oxide (NO) affects extracellular 5-HT levels in various brain regions, but no consistent direction of change was found and the underlying mechanisms remain unknown. The most of serotonergic neurons in dorsal raphe nucleus (DRN), a major source of serotonergic input to the forebrain, co-expresses neuronal nitric oxide synthase (nNOS), a synthase derived nitric oxide (NO) in the central nervous system. Here, we found that the excessive expression of nNOS and thereby the high concentration of NO followed by single-prolonged stress (SPS) caused suppression of the activity of DRN 5-HT neurons, inducing PTSD-like phenotype including increased anxiety-like behaviors, enhanced contextual fear memory, and fear generalization. Our study uncovered an important role of DRN nNOS-NO pathway in the pathology of PTSD, which may contribute to new understanding of the molecular mechanism of PTSD.


Assuntos
Comportamento Animal/fisiologia , Núcleo Dorsal da Rafe/fisiopatologia , Óxido Nítrico Sintase Tipo I/metabolismo , Neurônios Serotoninérgicos/metabolismo , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Estresse Psicológico/fisiopatologia , Animais , Ansiedade/fisiopatologia , Ansiedade/psicologia , Núcleo Dorsal da Rafe/enzimologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia , Óxido Nítrico/metabolismo , Neurônios Serotoninérgicos/citologia , Serotonina/metabolismo , Transtornos de Estresse Pós-Traumáticos/metabolismo , Estresse Psicológico/psicologia
10.
Mol Psychiatry ; 26(11): 6506-6519, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33931732

RESUMO

Exposure therapy based on the extinction of fear memory is first-line treatment for post-traumatic stress disorder (PTSD). However, fear extinction is relatively easy to learn but difficult to remember, extinguished fear often relapses under a number of circumstances. Here, we report that extinction learning-induced association of neuronal nitric oxide synthase (nNOS) with its carboxy-terminal PDZ ligand (CAPON) in the infralimbic (IL) subregion of medial prefrontal cortex negatively regulates extinction memory and dissociating nNOS-CAPON can prevent the return of extinguished fear in mice. Extinction training significantly increases nNOS-CAPON association in the IL. Disruptors of nNOS-CAPON increase extracellular signal-regulated kinase (ERK) phosphorylation and facilitate the retention of extinction memory in an ERK2-dependent manner. More importantly, dissociating nNOS-CAPON after extinction training enhances long-term potentiation and excitatory synaptic transmission, increases spine density in the IL, and prevents spontaneous recovery, renewal and reinstatement of remote fear of mice. Moreover, nNOS-CAPON disruptors do not affect other types of learning. Thus, nNOS-CAPON can serve as a new target for treating PTSD.


Assuntos
Extinção Psicológica , Medo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Ligantes , Camundongos , Óxido Nítrico Sintase Tipo I/metabolismo
11.
Theranostics ; 11(12): 5970-5985, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33897893

RESUMO

Overactivation of N-methyl-D-aspartate receptor (NMDAR) in the spinal cord dorsal horn (SDH) in the setting of injury represents a key mechanism of neuropathic pain. However, directly blocking NMDAR or its downstream signaling, interaction between postsynaptic density-95 (PSD-95) and neuronal nitric oxide synthase (nNOS), causes analgesic tolerance, mainly due to GABAergic disinhibition. The aim of this study is to explore the possibility of preventing analgesic tolerance through co-targeting NMDAR downstream signaling and γ-aminobutyric acid type A receptors (GABAARs). Methods: Mechanical/thermal hyperalgesia were quantified to assess analgesic effects. Miniature postsynaptic currents were tested by patch-clamp recording to evaluate synaptic transmission in the SDH. GABA-evoked currents were tested on HEK293 cells expressing different subtypes of recombinant GABAARs to assess the selectivity of (+)-borneol and ZL006-05. The expression of α2 and α3 subunits of GABAARs and BDNF, and nNOS-PSD-95 complex levels were analyzed by western blotting and coimmunoprecipitation respectively. Open field test, rotarod test and Morris water maze task were conducted to evaluate the side-effect of ZL006-05. Results: (+)-Borneol selectively potentiated α2- and α3-containing GABAARs and prevented the disinhibition of laminae I excitatory neurons in the SDH and analgesic tolerance caused by chronic use of ZL006, a nNOS-PSD-95 blocker. A dual-target compound ZL006-05 produced by linking ZL006 and (+)-borneol through an ester bond blocked nNOS-PSD-95 interaction and potentiated α2-containing GABAAR selectively. Chronic use of ZL006-05 did not produce analgesic tolerance and unwanted side effects. Conclusion: By targeting nNOS-PSD-95 interaction and α2-containing GABAAR simultaneously, chronic use of ZL006-05 can avoid analgesic tolerance and unwanted side effects. Therefore, we offer a novel candidate drug without analgesic tolerance for treating neuropathic pain.


Assuntos
Analgésicos/farmacologia , Proteína 4 Homóloga a Disks-Large/metabolismo , Neuralgia/tratamento farmacológico , Óxido Nítrico Sintase Tipo I/metabolismo , Receptores de GABA-A/metabolismo , Ácidos Aminossalicílicos/farmacologia , Animais , Benzilaminas/farmacologia , Linhagem Celular , Células HEK293 , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Manejo da Dor/métodos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Ácido gama-Aminobutírico/metabolismo
12.
J Neurosci ; 41(11): 2523-2539, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33500273

RESUMO

Stress-induced depression is common worldwide. NAc, a "reward" center, is recently reported to be critical to confer the susceptibility to chronic social defeat stress (CSDS) and the depression-related outcome. However, the underlying molecular mechanisms have not been well characterized. In this study, we induced depression-like behaviors with CSDS and chronic mild stress in male mice to mimic social and environmental factors, respectively, and observed animal behaviors with social interaction test, tail suspension test, and sucrose preference test. To determine the role of neuronal nitric oxide synthase (nNOS) and its product nitric oxide (NO), we used brain region-specifically nNOS overexpression and stereotaxic injection of NO inhibitor or donor. Moreover, the downstream molecular cyclin-dependent kinase 5 (CDK5) was explored by conditional KO and gene mutation. We demonstrate that nNOS-implicated mechanisms in NAc shell (NAcSh), including increased cell number, increased protein expression levels, and increased specific enzyme activity, contribute the susceptibility to social defeat and the following depression-like behaviors. NAcSh nNOS does not directly respond to chronic mild stress but facilitates the depression-like behaviors. The increased NAcSh nNOS expression after CSDS leads to the social avoidance and depression-like behaviors in defeated mice, which is dependent on the nNOS enzyme activity and NO production. Moreover, we identify the downstream signal in NAcSh. S-nitrosylation of CDK5 by NO contributes to enhanced CDK5 activity, leading to depression-related behaviors in susceptible mice. Therefore, NAcSh nNOS mediates susceptibility to social defeat stress and the depression-like behaviors through CDK5.SIGNIFICANCE STATEMENT Stress-induced depression is common worldwide, and chronic exposure to social and psychological stressors is important cause of human depression. Our study conducted with chronic social defeat stress mice models demonstrates that nNOS in NAcSh is crucial to regulate the susceptibility to social defeat stress and the following depression-like behaviors, indicating NAcSh nNOS as the responding molecule to social factors of depression. Moreover, we discover the downstream mechanism of NAcSh nNOS in mediating the susceptibility is NO and S-nitrosylation of CDK5. Thus, NAcSh nNOS mediates susceptibility to social defeat stress through CDK5 is a potential mechanism for depression, which may interpret how the brain transduces social stress exposure into depression.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Núcleo Accumbens/metabolismo , Derrota Social , Estresse Psicológico/metabolismo , Animais , Masculino , Camundongos
13.
Cereb Cortex ; 31(3): 1707-1718, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33188393

RESUMO

Posttraumatic stress disorder subjects usually show impaired recall of extinction memory, leading to extinguished fear relapses. However, little is known about the neural mechanisms underlying the impaired recall of extinction memory. We show here that the activity of dorsal hippocampus (dHPC) to infralimbic (IL) cortex circuit is essential for the recall of fear extinction memory in male mice. There were functional neural projections from the dHPC to IL. Using optogenetic manipulations, we observed that silencing the activity of dHPC-IL circuit inhibited recall of extinction memory while stimulating the activity of dHPC-IL circuit facilitated recall of extinction memory. "Impairment of extinction consolidation caused by" conditional deletion of extracellular signal-regulated kinase 2 (ERK2) in the IL prevented the dHPC-IL circuit-mediated recall of extinction memory. Moreover, silencing the dHPC-IL circuit abolished the effect of intra-IL microinjection of ERK enhancer on the recall of extinction memory. Together, we identify a dHPC to IL circuit that mediates the recall of extinction memory, and our data suggest that the dysfunction of dHPC-IL circuit and/or impaired extinction consolidation may contribute to extinguished fear relapses.


Assuntos
Extinção Psicológica/fisiologia , Hipocampo/fisiologia , Memória/fisiologia , Vias Neurais/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Condicionamento Clássico , Masculino , Camundongos Endogâmicos C57BL , Transtornos de Estresse Pós-Traumáticos/fisiopatologia
14.
Neurosci Bull ; 37(2): 229-241, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33180308

RESUMO

The paraventricular nucleus of the thalamus (PVT), which serves as a hub, receives dense projections from the medial prefrontal cortex (mPFC) and projects to the lateral division of central amygdala (CeL). The infralimbic (IL) cortex plays a crucial role in encoding and recalling fear extinction memory. Here, we found that neurons in the PVT and IL were strongly activated during fear extinction retrieval. Silencing PVT neurons inhibited extinction retrieval at recent time point (24 h after extinction), while activating them promoted extinction retrieval at remote time point (7 d after extinction), suggesting a critical role of the PVT in extinction retrieval. In the mPFC-PVT circuit, projections from IL rather than prelimbic cortex to the PVT were dominant, and disrupting the IL-PVT projection suppressed extinction retrieval. Moreover, the axons of PVT neurons preferentially projected to the CeL. Silencing the PVT-CeL circuit also suppressed extinction retrieval. Together, our findings reveal a new neural circuit for fear extinction retrieval outside the classical IL-amygdala circuit.


Assuntos
Núcleo Central da Amígdala , Medo , Extinção Psicológica , Córtex Pré-Frontal , Tálamo
15.
Neurotherapeutics ; 17(3): 1016-1030, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32632774

RESUMO

Neuropathic pain is usually persistent due to maladaptive neuroplasticity-induced central sensitization and, therefore, necessitates long-term treatment. N-methyl-D-aspartate receptor (NMDAR)-mediated hypersensitivity in the spinal dorsal horn represents key mechanisms of central sensitization. Short-term use of NMDAR antagonists produces antinociceptive efficacy in animal pain models and in clinical practice by reducing central sensitization. However, how prolonged use of NMDAR antagonists affects central sensitization remains unknown. Surprisingly, we find that prolonged blockage of NMDARs does not prevent but aggravate nerve injury-induced central sensitization and produce analgesic tolerance, mainly due to reduced synaptic inhibition. The disinhibition that results from the continuous decrease in the production of nitric oxide from neuronal nitric oxide synthase, downstream signal of NMDARs, leads to the reduction of GABAergic inhibitory synaptic transmission by upregulating brain-derived neurotrophic factor expression and inhibiting the expression and function of potassium-chloride cotransporter. Together, our findings suggest that chronic blockage of NMDARs develops analgesic tolerance through the neuronal nitric oxide synthase-brain-derived neurotrophic factor-potassium-chloride cotransporter pathway. Thus, preventing the GABAergic disinhibition induced by nitric oxide reduction may be necessary for the long-term maintenance of the analgesic effect of NMDAR antagonists.


Assuntos
Analgésicos/administração & dosagem , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Neurônios GABAérgicos/metabolismo , Neuralgia/metabolismo , Óxido Nítrico Sintase Tipo I/deficiência , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Analgésicos/efeitos adversos , Animais , Maleato de Dizocilpina/administração & dosagem , Maleato de Dizocilpina/efeitos adversos , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/fisiologia , Antagonistas de Aminoácidos Excitatórios/efeitos adversos , Neurônios GABAérgicos/efeitos dos fármacos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Neuralgia/tratamento farmacológico , Neuralgia/genética , Óxido Nítrico/deficiência , Óxido Nítrico/genética , Óxido Nítrico Sintase Tipo I/genética , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley
16.
Nat Commun ; 11(1): 2501, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32427844

RESUMO

Anxiety is common in patients suffering from chronic pain. Here, we report anxiety-like behaviors in mouse models of chronic pain and reveal that nNOS-expressing neurons in ventromedial prefrontal cortex (vmPFC) are essential for pain-induced anxiety but not algesia, using optogenetic and chemogenetic strategies. Additionally, we determined that excitatory projections from the posterior subregion of paraventricular thalamic nucleus (pPVT) provide a neuronal input that drives the activation of vmPFC nNOS-expressing neurons in our chronic pain models. Our results suggest that the pain signal becomes an anxiety signal after activation of vmPFC nNOS-expressing neurons, which causes subsequent release of nitric oxide (NO). Finally, we show that the downstream molecular mechanisms of NO likely involve enhanced glutamate transmission in vmPFC CaMKIIα-expressing neurons through S-nitrosylation-induced AMPAR trafficking. Overall, our data suggest that pPVT excitatory neurons drive chronic pain-induced anxiety through activation of vmPFC nNOS-expressing neurons, resulting in NO-mediated AMPAR trafficking in vmPFC pyramidal neurons.


Assuntos
Dor Crônica/enzimologia , Dor Crônica/psicologia , Núcleos da Linha Média do Tálamo/enzimologia , Neurônios/enzimologia , Óxido Nítrico Sintase Tipo I/metabolismo , Córtex Pré-Frontal/enzimologia , Animais , Ansiedade , Comportamento Animal , Dor Crônica/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleos da Linha Média do Tálamo/citologia , Neurônios/citologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Córtex Pré-Frontal/citologia
17.
J Neurochem ; 155(6): 679-696, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32415988

RESUMO

Environmental enrichment (EE) is a generally accepted strategy to promote stroke recovery and its beneficial effect is positively correlated with neuroplasticity. However, the mechanisms underlying it remain elusive. Histone deacetylase 2 (HDAC2), a negative regulator of neuroplasticity, is up-regulated after stroke. Thus, we hypothesized that HDAC2 may participate in EE-mediated stroke recovery. In this study, focal stroke was induced by photothrombosis in male mice exposing to EE or standard housing (SH) conditions. Recombinant virus vectors, including Ad-HDAC2-Flag, AAV-CAG-EGFP-Cre, LV-shHDAC2, or their controls were microinjected into the motor cortex at 3 days before stroke. Grid-walking and cylinder tasks were conducted to assess motor function. Western blot and immunostaining were used to uncover the mechanisms underlying EE-mediated stroke recovery. We found that EE exposure reversed stroke-induced HDAC2 up-regulation, implicating HDAC2 in EE-mediated functional recovery. Importantly, EE-dependent stroke recovery was counteracted by over-expressing HDAC2, and HDAC2 knockdown promoted functional recovery from stroke to the similar extent as EE exposure. Moreover, the knockdown of HDAC2 epigenetically enhanced expressions of neurotrophins and neuroplasticity-related proteins, with similar effects as EE, and consequently, whole brain and corticospinal tract (CST) rewiring. Together, our findings indicate that HDAC2 is critical for EE-dependent functional restoration. Precisely targeting HDAC2 may mimic EE and serve as a novel therapeutic strategy for stroke recovery.


Assuntos
Meio Ambiente , Histona Desacetilase 2/metabolismo , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/enzimologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
18.
Br J Pharmacol ; 177(16): 3674-3690, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32343840

RESUMO

BACKGROUND AND PURPOSE: Anxiety disorder is a common mental health disorder. However, there are few safe and fast-acting anxiolytic drugs available that can treat anxiety disorder. We previously demonstrated that the interaction of neuronal NOS (nNOS) with its carboxy-terminal PDZ ligand (CAPON) is involved in regulating anxiety-related behaviours. Here, we further investigated the anxiolytic effects of nNOS-CAPON disruptors in chronic stress-induced anxiety in animals. EXPERIMENTAL APPROACH: Mice were intravenously treated with nNOS-CAPON disruptors, ZLc-002 or Tat-CAPON12C, at the last week of chronic mild stress (CMS) exposure. We also infused corticosterone (CORT) into the hippocampus of mice to model anxiety behaviours and also delivered ZLc-002 or Tat-CAPON12C on the last week of chronic CORT treatment via pre-implanted cannula. Anxiety-related behaviours were examined using elevated plus maze, open field, novelty-suppressed feeding and light-dark (LD) tests. The level of nNOS-CAPON interaction was determined by co-immunoprecipitation (CO-IP) and proximity ligation assay (PLA). The neural mechanisms underlying the behavioural effects of nNOS-CAPON uncoupling in anxiety animal models were assessed by western blot, immunofluorescence and Golgi-Cox staining. KEY RESULTS: ZLc-002 and Tat-CAPON12C reversed CMS- or CORT-induced anxiety-related behaviours. ZLc-002 and Tat-CAPON12C increased synaptogenesis along with improved dendritic remodelling in CMS mice or CORT-treated cultured neurons. Meanwhile, blocking nNOS-CAPON interaction significantly activated the cAMP response element-binding protein (CREB)-brain-derived neurotrophic factor (BDNF) pathway, which is associated with synaptic plasticity. CONCLUSION AND IMPLICATIONS: Collectively, these results provide evidence for the anxiolytic effects of nNOS-CAPON uncouplers and their underlying mechanisms in anxiety disorders.


Assuntos
Ansiolíticos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Ansiolíticos/farmacologia , Ansiedade/tratamento farmacológico , Transtornos de Ansiedade/tratamento farmacológico , Camundongos , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo
19.
Biochem Biophys Res Commun ; 525(2): 520-527, 2020 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-32113678

RESUMO

Extremely high relapse rate is the dramatic challenge of drug abuse at present. Environmental cues play an important role in relapse of drug abuse. However, the specific mechanism underlying relapse remains unclear. Using morphine conditioned place preference (CPP) model, we show that association of neuronal nitric oxide synthase (nNOS) with postsynaptic density-95 (PSD-95) plays a significant role in morphine priming-induced reinstatement. The nNOS-PSD-95 coupling and c-Fos expression in the medial prefrontal cortex (mPFC) was significantly increased after extinction of morphine CPP. Dissociation of nNOS-PSD-95 in the mPFC by ZL006 inhibited the reinstatement of morphine CPP induced by a priming dose of morphine. Significantly reduced phosphorylation of cAMP-response element binding protein (CREB) in the mPFC was observed in the mice exposed to morphine after the extinction training. Uncoupling nNOS-PSD-95 reversed the morphine-induced CREB dysfunction. Moreover, effects of ZL006 on the reinstatement of morphine CPP and CREB activation depended on nNOS-PSD-95 target. Together, our findings suggest that nNOS-PSD-95 in the mPFC contributes to reinstatement of morphine CPP, possibly through CREB dysfunction, offering a potential target to prevent relapse of drug abuse.


Assuntos
Proteína 4 Homóloga a Disks-Large/metabolismo , Morfina/farmacologia , Entorpecentes/farmacologia , Óxido Nítrico Sintase Tipo I/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Extinção Psicológica/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/metabolismo , Mapas de Interação de Proteínas/efeitos dos fármacos
20.
J Neurosci ; 39(29): 5728-5739, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31097621

RESUMO

Contextual fear memory becomes less context-specific over time, a phenomenon referred to as contextual fear generalization. Overgeneralization of contextual fear memory is a core symptom of post-traumatic stress disorder (PTSD), but circuit mechanisms underlying the generalization remain unclear. We show here that neural projections from the anterior cingulate cortex (ACC) to ventral hippocampus (vHPC) mediate contextual fear generalization in male mice. Retrieval of contextual fear in a novel context at a remote time point activated cells in the ACC and vHPC, as indicated by significantly increased C-fos+ cells. Using chemogenetic or photogenetic manipulations, we observed that silencing the activity of ACC or vHPC neurons reduced contextual fear generalization at the remote time point, whereas stimulating the activity of ACC or vHPC neurons facilitated contextual fear generalization at a recent time point. We found that ACC neurons projected to the vHPC unidirectionally, and importantly, silencing the activity of projection fibers from the ACC to vHPC inhibited contextual fear generalization at the remote time point. Together, our findings reveal an ACC to vHPC circuit that controls expression of fear generalization and may offer new strategies to prevent or reverse contextual fear generalization in subjects with anxiety disorders, especially in PTSD.SIGNIFICANCE STATEMENT Overgeneralization of contextual fear memory is a cardinal feature of PTSD, but circuit mechanisms underlying it remain unclear. Our study indicates that neural projections from the anterior cingulate cortex to ventral hippocampus control the expression of contextual fear generalization. Thus, manipulating the circuit may prevent or reverse fear overgeneralization in subjects with PTSD.


Assuntos
Condicionamento Psicológico/fisiologia , Medo/fisiologia , Medo/psicologia , Giro do Cíngulo/fisiologia , Hipocampo/fisiologia , Rede Nervosa/fisiologia , Animais , Giro do Cíngulo/química , Hipocampo/química , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...