Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 253
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 2023 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-36914810

RESUMO

Recent studies based on animal models of various neurological disorders have indicated that mitophagy, a selective autophagy that eliminates damaged and superfluous mitochondria through autophagic degradation, may be involved in various neurological diseases. As an important mechanism of cellular stress response, much less is known about the role of mitophagy in stress-related mood disorders. Here, we found that tumor necrosis factor-α (TNF-α), an inflammation cytokine that plays a particular role in stress responses, impaired the mitophagy in the medial prefrontal cortex (mPFC) via triggering degradation of an outer mitochondrial membrane protein, NIP3-like protein X (NIX). The deficits in the NIX-mediated mitophagy by TNF-α led to the accumulation of damaged mitochondria, which triggered synaptic defects and behavioral abnormalities. Genetic ablation of NIX in the excitatory neurons of mPFC caused passive coping behaviors to stress, and overexpression of NIX in the mPFC improved TNF-α-induced synaptic and behavioral abnormalities. Notably, ketamine, a rapid on-set and long-lasting antidepressant, reversed the TNF-α-induced behavioral abnormalities through activation of NIX-mediated mitophagy. Furthermore, the downregulation of NIX level was also observed in the blood of major depressive disorder patients and the mPFC tissue of animal models. Infliximab, a clinically used TNF-α antagonist, alleviated both chronic stress- and inflammation-induced behavioral abnormalities via restoring NIX level. Taken together, these results suggest that NIX-mediated mitophagy links inflammation signaling to passive coping behaviors to stress, which underlies the pathophysiology of stress-related emotional disorders.

2.
J Neuroinflammation ; 20(1): 157, 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37391731

RESUMO

BACKGROUND: Neuroinflammation and microglia play critical roles in the development of depression. Cluster of differentiation 200 (CD200) is an anti-inflammatory glycoprotein that is mainly expressed in neurons, and its receptor CD200R1 is primarily in microglia. Although the CD200-CD200R1 pathway is necessary for microglial activation, its role in the pathophysiology of depression remains unknown. METHODS: The chronic social defeat stress (CSDS) with behavioral tests were performed to investigate the effect of CD200 on the depressive-like behaviors. Viral vectors were used to overexpress or knockdown of CD200. The levels of CD200 and inflammatory cytokines were tested with molecular biological techniques. The status of microglia, the expression of BDNF and neurogenesis were detected with immunofluorescence imaging. RESULTS: We found that the expression of CD200 was decreased in the dentate gyrus (DG) region of mice experienced CSDS. Overexpression of CD200 alleviated the depressive-like behaviors of stressed mice and inhibition of CD200 facilitated the susceptibility to stress. When CD200R1 receptors on microglia were knocked down, CD200 was unable to exert its role in alleviating depressive-like behavior. Microglia in the DG brain region were morphologically activated after exposure to CSDS. In contrast, exogenous administration of CD200 inhibited microglia hyperactivation, alleviated neuroinflammatory response in hippocampus, and increased the expression of BDNF, which in turn ameliorated adult hippocampal neurogenesis impairment in the DG induced by CSDS. CONCLUSIONS: Taken together, these results suggest that CD200-mediated alleviation of microglia hyperactivation contributes to the antidepressant effect of neurogenesis in dentate gyrus in mice.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Microglia , Animais , Camundongos , Hipocampo , Neurogênese , Giro Denteado
3.
Brain Behav Immun ; 108: 204-220, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36496170

RESUMO

Increasing evidence supports the pathogenic role of neuroinflammation in psychiatric diseases, including major depressive disorder (MDD) and neuropsychiatric symptoms of Coronavirus disease 2019 (COVID-19); however, the precise mechanism and therapeutic strategy are poorly understood. Here, we report that myeloid differentiation factor 88 (MyD88), a pivotal adaptor that bridges toll-like receptors to their downstream signaling by recruiting the signaling complex called 'myddosome', was up-regulated in the medial prefrontal cortex (mPFC) after exposure to chronic social defeat stress (CSDS) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. The inducible expression of MyD88 in the mPFC primed neuroinflammation and conferred stress susceptibility via amplifying immune danger signals, such as high-mobility group box 1 and SARS-CoV-2 spike protein. Overexpression of MyD88 aggravated, whereas knockout or pharmacological inhibition of MyD88 ameliorated CSDS-induced depressive-like behavior. Notably, TJ-M2010-5, a novel synthesized targeting inhibitor of MyD88 dimerization, alleviated both CSDS- and SARS-CoV-2 spike protein-induced depressive-like behavior. Taken together, our findings indicate that inhibiting MyD88 signaling represents a promising therapeutic strategy for stress-related mental disorders, such as MDD and COVID-19-related neuropsychiatric symptoms.


Assuntos
COVID-19 , Transtorno Depressivo Maior , Fator 88 de Diferenciação Mieloide , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , COVID-19/metabolismo , COVID-19/psicologia , Fator 88 de Diferenciação Mieloide/metabolismo , Doenças Neuroinflamatórias , SARS-CoV-2/metabolismo
4.
Brain Behav Immun ; 109: 23-36, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36581303

RESUMO

Synapse loss in medial prefrontal cortex (mPFC) has been implicated in stress-related mood disorders, such as depression. However, the exact effect of synapse elimination in the depression and how it is triggered are largely unknown. Through repeated longitudinal imaging of mPFC in the living brain, we found both presynaptic and postsynaptic components were declined, together with the impairment of synapse remodeling and cross-synaptic signal transmission in the mPFC during chronic stress. Meanwhile, chronic stress also induced excessive microglia phagocytosis, leading to engulfment of excitatory synapses. Further investigation revealed that the elevated complement C3 during the stress acted as the tag of synapses to be eliminated by microglia. Besides, chronic stress induced a reduction of the connectivity between the mPFC and neighbor regions. C3 knockout mice displayed significant reduction of synaptic pruning and alleviation of disrupted functional connectivity in mPFC, resulting in more resilience to chronic stress. These results indicate that complement-mediated excessive microglia phagocytosis in adulthood induces synaptic dysfunction and cortical hypo-connectivity, leading to stress-related behavioral abnormality.


Assuntos
Microglia , Derrota Social , Camundongos , Animais , Sinapses , Camundongos Knockout , Plasticidade Neuronal
5.
Acta Pharmacol Sin ; 44(8): 1576-1588, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37012493

RESUMO

Emerging evidence demonstrates the vital role of synaptic transmission and structural remodeling in major depressive disorder. Activation of melanocortin receptors facilitates stress-induced emotional behavior. Prolylcarboxypeptidase (PRCP) is a serine protease, which splits the C-terminal amino acid of α-MSH and inactivates it. In this study, we asked whether PRCP, the endogenous enzyme of melanocortin system, might play a role in stress susceptibility via regulating synaptic adaptations. Mice were subjected to chronic social defeat stress (CSDS) or subthreshold social defeat stress (SSDS). Depressive-like behavior was assessed in SIT, SPT, TST and FST. Based on to behavioral assessments, mice were divided into the susceptible (SUS) and resilient (RES) groups. After social defeat stress, drug infusion or viral expression and behavioral tests, morphological and electrophysiological analysis were conducted in PFX-fixed and fresh brain slices containing the nucleus accumbens shell (NAcsh). We showed that PRCP was downregulated in NAcsh of susceptible mice. Administration of fluoxetine (20 mg·kg-1·d-1, i.p., for 2 weeks) ameliorated the depressive-like behavior, and restored the expression levels of PRCP in NAcsh of susceptible mice. Pharmacological or genetic inhibition of PRCP in NAcsh by microinjection of N-benzyloxycarbonyl-L-prolyl-L-prolinal (ZPP) or LV-shPRCP enhanced the excitatory synaptic transmission in NAcsh, facilitating stress susceptibility via central melanocortin receptors. On the contrary, overexpression of PRCP in NAcsh by microinjection of AAV-PRCP alleviated the depressive-like behavior and reversed the enhanced excitatory synaptic transmission, abnormal dendritogenesis and spinogenesis in NAcsh induced by chronic stress. Furthermore, chronic stress increased the level of CaMKIIα, a kinase closely related to synaptic plasticity, in NAcsh. The elevated level of CaMKIIα was reversed by overexpression of PRCP in NAcsh. Pharmacological inhibition of CaMKIIα in NAcsh alleviated stress susceptibility induced by PRCP knockdown. This study has revealed the essential role of PRCP in relieving stress susceptibility through melanocortin signaling-mediated synaptic plasticity in NAcsh.


Assuntos
Transtorno Depressivo Maior , Núcleo Accumbens , Camundongos , Animais , Núcleo Accumbens/metabolismo , alfa-MSH/metabolismo , Plasticidade Neuronal/fisiologia , Receptores de Melanocortina/metabolismo , Estresse Psicológico
6.
EMBO Rep ; 21(9): e51235, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32789986

RESUMO

Response to comments on Cui Q-Q et al: "Hippocampal CD 39/ENTPD 1 promotes mouse depression-like behavior through hydrolyzing extracellular ATP".


Assuntos
Depressão , Hipocampo , Trifosfato de Adenosina , Animais , Camundongos
7.
EMBO Rep ; 21(4): e47857, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32133764

RESUMO

Emerging evidence implicates that low levels of ATP in the extracellular space may contribute to the pathophysiology of major depressive disorder (MDD). The concentration of extracellular ATP is regulated by its hydrolase ectonucleotide tri(di)phosphohydrolase (ENTPD). However, the role of ENTPD in depression remains poorly understood. Here we examine the role of CD39 (known as ENTPD1) in mouse depression-like behavior induced by chronic social defeat stress (CSDS). We demonstrate that CSDS enhances the expression and activity of CD39 in hippocampus. The CD39 functional analog apyrase also induces depression-like behavior, which can be ameliorated by ATP replenishment. Pharmacological inhibition and genetic silencing of CD39 has an antidepressant-like effect via increasing hippocampal extracellular ATP concentration, accompanied with an increase in hippocampal neurogenesis and dendritic spine numbers in defeated mice. These results suggest that hippocampal CD39 contributes to CSDS-induced depression-like behavior via hydrolyzing extracellular ATP, indicating that CD39 may be a promising new target for the treatment of depression.


Assuntos
Trifosfato de Adenosina/metabolismo , Apirase , Transtorno Depressivo Maior , Animais , Apirase/genética , Apirase/metabolismo , Depressão/genética , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/genética , Hipocampo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
8.
Eur J Nutr ; 61(4): 1823-1836, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34997266

RESUMO

PURPOSE: Diabetic cardiomyopathy (DCM), a common complication of diabetes mellitus and is characterized by myocardial hypertrophy and myocardial fibrosis. Pyrroloquinoline quinone (PQQ), a natural nutrient, exerts strong protection against various myocardial diseases. Pyroptosis, a type of inflammation-related programmed cell death, is vital to the development of DCM. However, the protective effects of PQQ against DCM and the associated mechanisms are not clear. This study aimed to investigate whether PQQ protected against DCM and to determine the underlying molecular mechanism. METHODS: Diabetes was induced in mice by intraperitoneal injection of streptozotocin, after which the mice were administered PQQ orally (10, 20, or 40 mg/kg body weight/day) for 12 weeks. AC16 human myocardial cells were divided into the following groups and treated accordingly: control (5.5 mmol/L glucose), high glucose (35 mmol/L glucose), and HG + PQQ groups (1 and 10 nmol/L PQQ). Cells were treated for 24 h. RESULTS: PQQ reduced myocardial hypertrophy and the area of myocardial fibrosis, which was accompanied by an increase in antioxidant function and a decrease in inflammatory cytokine levels. Moreover, myocardial hypertrophy-(ANP and BNP), myocardial fibrosis-(collagen I and TGF-ß1), and pyroptosis-related protein levels decreased in the PQQ treatment groups. Furthermore, PQQ abolished mitochondrial dysfunction and the activation of NF-κB/IκB, and decreased NLRP3 inflammation-mediated pyroptosis in AC16 cells under high-glucose conditions. CONCLUSION: PQQ improved DCM in diabetic mice by inhibiting NF-κB/NLRP3 inflammasome-mediated cell pyroptosis. Long-term dietary supplementation with PQQ may be greatly beneficial for the treatment of DCM. Diagram of the underlying mechanism of the effects of PQQ on DCM. PQQ inhibits ROS generation and NF-κB activation, which stimulates activation of the NLRP3 inflammasome and regulates the expression of caspase-1, IL-1ß, and IL-18. The up-regulated inflammatory cytokines trigger myocardial hypertrophy and cardiac fibrosis and promote the pathological process of DCM.


Assuntos
Diabetes Mellitus Experimental , Cardiomiopatias Diabéticas , Animais , Cardiomegalia , Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/tratamento farmacológico , Cardiomiopatias Diabéticas/etiologia , Cardiomiopatias Diabéticas/metabolismo , Fibrose , Glucose , Inflamassomos/metabolismo , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Cofator PQQ/metabolismo , Cofator PQQ/farmacologia , Cofator PQQ/uso terapêutico , Piroptose , Transdução de Sinais
9.
Acta Pharmacol Sin ; 43(2): 260-272, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33927360

RESUMO

Individual differences in the development of uncontrollable fear in response to traumatic stressors have been observed in clinic, but the underlying mechanisms remain unknown. In the present study we first conducted a meta-analysis of published clinical data and found that malondialdehyde, an oxidative stress biomarker, was significantly elevated in the blood of patients with fear-related anxiety disorders. We then carried out experimental study in rats subjected to fear conditioning. We showed that reestablishing redox homeostasis in basolateral amygdale (BLA) after exposure to fear stressors determined the capacity of learned fear inhibition. Intra-BLA infusion of buthionine sulfoximine (BSO) to deplete the most important endogenous antioxidant glutathione (GSH) blocked fear extinction, whereas intra-BLA infusion of dithiothreitol or N-acetylcysteine (a precursor of GSH) facilitated extinction. In electrophysiological studies conducted on transverse slices, we showed that fear stressors induced redox-dependent inhibition of NMDAR-mediated synaptic function, which was rescued by extinction learning or reducing agents. Our results reveal a novel pharmacological strategy for reversing impaired fear inhibition and highlight the role of GSH in the treatment of psychiatric disorders.


Assuntos
Acetilcisteína/farmacologia , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Glutationa/metabolismo , Memória/efeitos dos fármacos , Animais , Complexo Nuclear Basolateral da Amígdala/metabolismo , Complexo Nuclear Basolateral da Amígdala/fisiologia , Butionina Sulfoximina/farmacologia , Condicionamento Clássico , Sinais (Psicologia) , Ditiotreitol/farmacologia , Glutationa/fisiologia , Homeostase/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
10.
Neoplasma ; 69(5): 1129-1137, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36131607

RESUMO

Tumor cells show deregulated metabolism leading to an enrichment of lactate in the tumor microenvironment (TME). This lactate-rich environment has been reported to impair effector T cells. However, T-regulatory cells (Tregs) show metabolic advantages in lactate-rich TME that maintain a strong suppression of effector T cells, which leads to tumor immune evasion. Therefore, the glycolytic process of tumors could represent a therapeutic target, and agents that modify the energy metabolism of tumor cells have therapeutic potential. Resveratrol is a naturally occurring polyphenol that has been confirmed to suppress tumor cells' glycolytic metabolism. In this study, we show that resveratrol induces metabolic reprogramming in ovarian cancer cells. Resveratrol increases oxidative and decreases glycolysis, in association with decreased lactate production both in vitro and in vivo. Lactate reduction in TME weakens the suppressive function of Tregs, and subsequently restores anti-tumor immunity. Significantly, combined resveratrol and PD-1 blockade promote anti-tumor efficacy. These data suggest that resveratrol's anti-tumor actions in ovarian cancer could be explained, in part, through modification of the anti-tumor immunity, and indicate a novel treatment strategy for improving immune checkpoint blockade therapy using resveratrol.


Assuntos
Neoplasias , Neoplasias Ovarianas , Carcinoma Epitelial do Ovário , Feminino , Humanos , Inibidores de Checkpoint Imunológico , Ácido Láctico , Neoplasias/tratamento farmacológico , Neoplasias Ovarianas/patologia , Polifenóis , Receptor de Morte Celular Programada 1 , Resveratrol/farmacologia , Microambiente Tumoral
11.
Chem Res Toxicol ; 34(4): 1183-1196, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33793228

RESUMO

Outdoor air pollution, a spatially and temporally complex mixture, is a human carcinogen. However, ambient measurements may not reflect subject-level exposures, personal monitors do not assess internal dose, and spot assessments of urinary biomarkers may not recapitulate chronic exposures. Nucleophilic sites in serum albumin-particularly the free thiol at Cys34-form adducts with electrophiles. Due to the 4-week lifetime of albumin in circulation, accumulating adducts can serve as intermediate- to long-residence biomarkers of chronic exposure and implicate potential biological effects. Employing nanoflow liquid chromatography-high-resolution mass spectrometry (nLC-HRMS) and parallel reaction monitoring (PRM), we have developed and validated a novel targeted albumin adductomics platform capable of simultaneously monitoring dozens of Cys34 adducts per sample in only 2.5 µL of serum, with on-column limits of detection in the low-femtomolar range. Using this platform, we characterized the magnitude and impact of ambient outdoor air pollution exposures with three repeated measurements over 84 days in n = 26 nonsmoking women (n = 78 total samples) from Qidong, China, an area with a rising burden of lung cancer incidence. In concordance with seasonally rising ambient concentrations of NO2, SO2, and PM10 measured at stationary monitors, we observed elevations in concentrations of Cys34 adducts of benzoquinone (p < 0.05), benzene diol epoxide (BDE; p < 0.05), crotonaldehyde (p < 0.01), and oxidation (p < 0.001). Regression analysis revealed significant elevations in oxidation and BDE adduct concentrations of 300% to nearly 700% per doubling of ambient airborne pollutant levels (p < 0.05). Notably, the ratio of irreversibly oxidized to reduced Cys34 rose more than 3-fold during the 84-day period, revealing a dramatic perturbation of serum redox balance and potentially serving as a portent of increased pollution-related mortality risk. Our targeted albumin adductomics assay represents a novel and flexible approach for sensitive and multiplexed internal dosimetry of environmental exposures, providing a new strategy for personalized biomonitoring and prevention.


Assuntos
Poluentes Atmosféricos/análise , Monitoramento Biológico , Carcinógenos/análise , Albumina Sérica/análise , Humanos , Estrutura Molecular
12.
Pharmacol Res ; 163: 105355, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33285230

RESUMO

Adaptive responses to stress are critical to enhance physical and mental well-being, but excessive or prolonged stress may cause inadaptability and increase the risks of psychiatric disorders, such as depression. GABABR signaling is fundamental to brain function and has been identified in neuropsychiatric disorders. KCTD12 is a critical auxiliary subunit in GABABR signaling, but its role in mental disorders, such as depression is unclear. In the present study, we used a well-validated mice model, chronic social defeat stress (CSDS) to investigate behavioral responses to stress and explore the role of Kctd12 in stress response, as well as the relevant mechanisms. We found that CSDS increased the expression of Kctd12 in the dentate gyrus (DG), a subregion of hippocampus. Overexpression of Kctd12 in DG induced higher responsiveness to acute stress and increased vulnerability to social stress in mice, whereas knock-down of Kctd12 in DG prevented the social avoidance. Furthermore, an increased expression of GABAB receptor 2 (GB2) in the DG of CSDS-treated mice was observed, and CGP35348, an antagonist of GABABR, improved the stress-induced behavior responses along with suppressing the excess expression of Kctd12. In addition, Kctd12 regulated the excitability of granule cell in DG, and the stimulation of neuronal activity by silencing Kctd12 contributed to the antidepressant-like effect of fluoxetine. These findings identify that the Kctd12 in DG works as a critical mediator of stress responses, providing a promising therapeutic target in stress-related psychiatric disorders, including depression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Comportamento Animal , Derrota Social , Estresse Psicológico/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Antidepressivos/farmacologia , Giro Denteado/metabolismo , Depressão/metabolismo , Modelos Animais de Doenças , Fluoxetina/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Subunidades Proteicas , RNA Interferente Pequeno/genética , Receptores de GABA-B
13.
Cereb Cortex ; 29(4): 1509-1519, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29522177

RESUMO

ß-Catenin has been implicated in major depressive disorder (MDD), which is associated with synaptic plasticity and dendritic arborization. MicroRNAs (miRNA) are small noncoding RNAs containing about 22 nucleotides and involved in a variety of physiological and pathophysiological process, but their roles in MDD remain largely unknown. Here, we investigated the expression and function of miRNAs in the mouse model of chronic social defeat stress (CSDS). The regulation of ß-catenin by selected miRNA was validated by silico prediction, target gene luciferase reporter assay, and transfection experiment in neurons. We demonstrated that the levels of miR-214-3p, which targets ß-catenin transcripts were significantly increased in the medial prefrontal cortex (mPFC) of CSDS mice. Antagomir-214-3p, a neutralizing inhibitor of miR-214-3p, increased the levels of ß-catenin and reversed the depressive-like behavior in CSDS mice. Meanwhile, antagomir-214-3p increased the amplitude of miniature excitatory postsynaptic current (mEPSC) and the number of dendritic spines in mPFC of CSDS mice, which may be related to the elevated expression of cldn1. Furthermore, intranasal administered antagomir-214-3p also significantly increased the level of ß-catenin and reversed the depressive-like behaviors in CSDS mice. These results may represent a new therapeutic target for MDD.


Assuntos
Depressão/fisiopatologia , MicroRNAs/fisiologia , Estresse Psicológico/fisiopatologia , beta Catenina/fisiologia , Administração Intranasal , Animais , Antagomirs/administração & dosagem , Claudina-1/genética , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/fisiologia , Depressão/etiologia , Depressão/genética , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Regulação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiopatologia , Estresse Psicológico/genética , beta Catenina/genética
14.
Addict Biol ; 25(2): e12736, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-30788886

RESUMO

Cocaine is a strong central nervous system stimulant, which can induce drug addiction. Previous studies have reported that cocaine-induced autophagy is involved in neuroinflammation and cell death. However, the role of autophagy in psychomotor sensitivity to cocaine has not been explored. Here, we reported that D1 receptor -CaMKII-AMPK-FoxO3a signaling pathway was involved in acute cocaine application-induced autophagy in the nucleus accumbens (NAc) both in vitro and in vivo. Furthermore, we found that knockdown of the ATG5 gene in the NAc augmented behavioral response to cocaine, and induction of autophagy in the NAc with rapamycin attenuated cocaine-induced behavioral response, which was coincident with the alterations of dendritic spine density in neurons of NAc. These results suggest that cocaine exposure leads to the induction of autophagy, which is a protective mechanism against behavioral response to cocaine of male mice.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Transtornos Relacionados ao Uso de Cocaína/prevenção & controle , Cocaína/farmacologia , Núcleo Accumbens/metabolismo , Animais , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Modelos Animais de Doenças , Inibidores da Captação de Dopamina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Accumbens/efeitos dos fármacos
15.
Addict Biol ; 25(2): e12739, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31056833

RESUMO

Cocaine is a common abused drug that can induce abnormal synaptic and immune responses in the central nervous system (CNS). High mobility group box 1 (HMGB1) is one kind of inflammatory molecules that is expressed both on neurons and immune cells. Previous studies of HMGB1 in the CNS have largely focused on immune function, and the role of HMGB1 in neurons and cocaine addiction remains unknown. Here, we show that cocaine exposure induced the translocation and release of HMGB1 in the nucleus accumbens (NAc) neurons. Gain and loss of HMGB1 in the NAc bidirectionally regulate cocaine-induced conditioned place preference. From the nucleus to the cytosol, HMGB1 binds to glutamate receptor subunits (GluA2/GluN2B) on the membrane, which regulates cocaine-induced synaptic adaptation and the formation of cocaine-related memory. These data unveil the role of HMGB1 in neurons and provide the evidence for the HMGB1 involvement in drug addiction.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/genética , Proteína HMGB1/genética , Memória/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Recompensa , Animais , Cocaína/farmacologia , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Modelos Animais de Doenças , Masculino , Núcleo Accumbens/fisiopatologia , Ratos , Ratos Sprague-Dawley
16.
Zhonghua Nan Ke Xue ; 25(10): 874-882, 2019 Oct.
Artigo em Zh | MEDLINE | ID: mdl-32233217

RESUMO

OBJECTIVE: To investigate the improving effect of S1P3 silencing on the erectile function of spontaneous hypertension rats (SHR). METHODS: Five 12-week-old healthy male WKY rats were included in group A and another 15 12-week-old healthy male SHRs equally randomized into groups B1 (intracavernously injected with 20 µl S1P3 siRNA lentiviral vectors ï¼»2 ×108TU/mlï¼½), B2 (intracavernously injected with 20 µl GFP lentiviral vectors ï¼»2 ×108TU/mlï¼½), and C (control). At 1 week after transfection, the ratio of the maximum intracavernous pressure/mean arterial pressure (ICPmax/MAP) of the rats was measured, and the expressions of S1P3, ROCK1, ROCK2 and eNOS in the corpus cavernosal tissue were determined by immunohistochemistry, Western blot and RT-qPCR. RESULTS: There were no statistically significant differences in the body weight and serum T level among the three groups of rats. Under 0 V, 3 V and 5 V electrical stimulation, the ratios of ICPmax/MAP were 0.16 ± 0.01, 0.55 ± 0.03 and 0.82 ± 0.02 in group A, 0.15 ± 0.01, 0.55 ± 0.01 and 0.79 ± 0.03 in group B1, 0.10 ± 0.00, 0.22 ± 0.01 and 0.43 ± 0.01 in group B2, and 0.11 ± 0.01, 0.22 ± 0.01 and 0.42 ± 0.02 in group C, significantly lower in the latter two than in the former two groups (P < 0.05). The protein expressions of S1P3, ROCK1 and ROCK2 in the corpus cavernosum were remarkably down-regulated while that of eNOS up-regulated in groups A and B1 compared with those in groups B2 and C (P < 0.05). Groups A and B1, in comparison with B2 and C, also showed markedly decreased mRNA expressions of S1P3 (0.72 ± 0.04 and 0.71 ± 0.07 vs 1.00 ± 0.06 and 1.00 ± 0.10, P < 0.05), ROCK1 (0.99 ± 0.05 and 1.08 ± 0.16 vs 1.85 ± 0.44 and 2.02 ± 0.38, P < 0.05) and ROCK2 (1.00 ± 0.03 and 1.08 ± 0.16 vs 2.16 ± 0.78 and 2.46 ± 0.69, P < 0.05), but increased eNOS (1.04 ± 0.15 and 0.81 ± 0.23 vs 0.32 ± 0.08 and 0.32 ± 0.04, P < 0.05). CONCLUSIONS: S1P3 siRNA can improve the erectile function of SHRs by inhibiting the expression of the S1P3 gene and down-regulating the RhoA/Rho kinase signaling pathway in the corpus cavernosum.


Assuntos
Disfunção Erétil/terapia , Inativação Gênica , Hipertensão/complicações , Ereção Peniana , Receptores de Esfingosina-1-Fosfato/genética , Animais , Disfunção Erétil/genética , Vetores Genéticos , Lentivirus , Masculino , Óxido Nítrico Sintase Tipo III/metabolismo , Pênis , Distribuição Aleatória , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transdução de Sinais , Quinases Associadas a rho/metabolismo
17.
Mol Ther ; 25(3): 752-764, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28202389

RESUMO

Histone deacetylase 2 (HDAC2) plays a major role in the epigenetic regulation of gene expression. Previous studies have shown that HDAC2 expression is strongly increased in Alzheimer's disease (AD), a major neurodegenerative disorder and the most common form of dementia. Moreover, previous studies have linked HDAC2 to Aß overproduction in AD; however, its involvement in tau pathology and other memory-related functions remains unclear. Here, we show that increased HDAC2 levels strongly correlate with phosphorylated tau in a mouse model of AD. HDAC2 overexpression induced AD-like tau hyperphosphorylation and aggregation, which were accompanied by a loss of dendritic complexity and spine density. The ectopic expression of HDAC2 resulted in the deacetylation of the hepatocyte nuclear factor 4α (HNF-4A) transcription factor, which disrupted its binding to the miR-101b promoter. The suppression of miR-101b caused an upregulation of its target, AMP-activated protein kinase (AMPK). The introduction of miR-101b mimics or small interfering RNAs (siRNAs) against AMPK blocked HDAC2-induced tauopathy and dendritic impairments in vitro. Correspondingly, miR-101b mimics or AMPK siRNAs rescued tau pathology, dendritic abnormalities, and memory deficits in AD mice. Taken together, the current findings implicate the HDAC2/miR-101/AMPK pathway as a critical mediator of AD pathogenesis. These studies also highlight the importance of epigenetics in AD and provide novel therapeutic targets.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Histona Desacetilase 2/metabolismo , MicroRNAs/genética , Tauopatias/genética , Tauopatias/metabolismo , Doença de Alzheimer/patologia , Animais , Sítios de Ligação , Sequência Consenso , Dendritos/metabolismo , Dendritos/patologia , Modelos Animais de Doenças , Expressão Gênica , Regulação da Expressão Gênica , Inativação Gênica , Fator 4 Nuclear de Hepatócito/genética , Histona Desacetilase 2/genética , Transtornos da Memória/genética , Camundongos , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Células Piramidais/metabolismo , Células Piramidais/patologia , Tauopatias/patologia , Proteínas tau/metabolismo
18.
Zhonghua Nan Ke Xue ; 24(2): 172-175, 2018 Feb.
Artigo em Zh | MEDLINE | ID: mdl-30156080

RESUMO

Penile erectile dysfunction (ED) is ascribed to the contraction-relaxation imbalance of smooth muscle cells (SMC), the weakening of their diastolic function and the strengthening of their systolic function. The contraction-related signaling pathways, cell membrane ion channels and SMC phenotypes all participate in the regulation of their contraction and its malfunction may cause a variety of SMC-related diseases. The signaling pathways RhoA/Rock and Raf/MEK/ERK1/2 interact with each other, suppressing the expression of the RhoA protein or reducing the level of Rock2 phosphorylation, which may contribute to the treatment of ED. The poor performance of VDCC or TRPC is reckoned to be an important cause of hypertension- or diabetes-related ED. The expressions of CaV1.2, TRPC1 and TRPC4 can be upregulated by many pathological factors, which may enhance the contraction of SMCs. The pathogenesis of ED may be associated with the differentiation of the phenotypes corpus cavernosal SMCs. This review focuses on the recent progress in the studies of the relationship between SMC contraction and ED.


Assuntos
Disfunção Erétil/fisiopatologia , Contração Muscular/fisiologia , Miócitos de Músculo Liso/fisiologia , Ereção Peniana/fisiologia , Animais , Complicações do Diabetes/etiologia , Complicações do Diabetes/fisiopatologia , Disfunção Erétil/etiologia , Humanos , Hipertensão/complicações , Canais Iônicos/metabolismo , Masculino , Proteína Quinase 3 Ativada por Mitógeno , Fosforilação , Transdução de Sinais/fisiologia
19.
Neurochem Res ; 41(11): 2890-2903, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27426946

RESUMO

Sulfhydryl compounds such as dithiothreitol (DTT) and ß-mercaptoethanol (ß-ME) are widely used as redox agents. Previous studies in our group and other laboratory have reported the effect of sulfhydryl compounds on the function of glutamate receptor, including plasticity. Most of these findings have focused on the N-methyl-D-aspartic acid receptor, in contrast, very little is known about the effect of sulfhydryl compounds on α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR). Here, we observed that DTT (100 µM), ß-ME (200 µM) and L-cysteine (200 µM) significantly elevated the surface expression of AMPARs via reducing their palmitoylation in rat hippocampal slices in vitro. Increased surface stability of AMPARs was not be correlated with the altered redox status, because the chemical entities containing mercapto group such as penicillamine (200 µM) and 2-mercapto-1-methylimidazole (200 µM) exhibited little effects on the surface expression of AMPARs. Computing results of Asp-His-His-Cys (DHHC) 3, the main enzyme for palmitoylation of AMPARs, indicated that only the alkyl mercaptans with chain-like configuration, such as DTT and ß-ME, can enter the pocket of DHHC3 and disrupt the catalytic activity via inhibiting DHHC3 auto-palmitoylation. Collectively, our findings indicate a novel redox-independent mechanism underlay the multiple effects of thiol reductants on synaptic function.


Assuntos
Hipocampo/efeitos dos fármacos , N-Metilaspartato/metabolismo , Receptores de AMPA/metabolismo , Compostos de Sulfidrila/metabolismo , Compostos de Sulfidrila/farmacologia , Animais , Cisteína/metabolismo , Hipocampo/metabolismo , Lipoilação/fisiologia , Masculino , Oxirredução/efeitos dos fármacos , Ratos Sprague-Dawley , Receptores de Glutamato/metabolismo
20.
Clin Exp Pharmacol Physiol ; 43(2): 193-202, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26510178

RESUMO

Orofacial pain is a common clinical symptom that is accompanied by tooth pain, migraine and gingivitis. Accumulating evidence suggests that acid-sensing ion channels (ASICs), especially ASIC3, can profoundly affect the physiological properties of nociception in peripheral sensory neurons. The aim of this study is to examine the contribution of ASICs in trigeminal ganglion (TG) neurons to orofacial inflammatory pain. A Western blot (WB), immunofluorescence assay of labelled trigeminal ganglion neurons, orofacial formalin test, cell preparation and electrophysiological experiments are performed. This study demonstrated that ASIC1, ASIC2a and ASIC3 are highly expressed in TG neurons innervating the orofacial region of rats. The amplitude of ASIC currents in these neurons increased 119.72% (for ASIC1-like current) and 230.59% (for ASIC3-like current) in the formalin-induced orofacial inflammatory pain model. In addition, WB and immunofluorescence assay demonstrated a significantly augmented expression of ASICs in orofacial TG neurons during orofacial inflammation compared with the control group. The relative protein density of ASIC1, ASIC2a and ASIC3 also increased 58.82 ± 8.92%, 45.30 ± 11.42% and 55.32 ± 14.71%, respectively, compared with the control group. Furthermore, pharmacological blockade of ASICs and genetic deletion of ASIC1 attenuated the inflammation response. These findings indicate that peripheral inflammation can induce the upregulation of ASICs in TG neurons, causing orofacial inflammatory pain. Additionally, the specific inhibitor of ASICs may have a significant analgesic effect on orofacial inflammatory pain.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Dor Facial/metabolismo , Dor Facial/patologia , Neurônios/metabolismo , Gânglio Trigeminal/patologia , Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/deficiência , Canais Iônicos Sensíveis a Ácido/genética , Animais , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Dor Facial/induzido quimicamente , Dor Facial/fisiopatologia , Formaldeído/efeitos adversos , Técnicas de Inativação de Genes , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Inflamação/fisiopatologia , Camundongos , Neurônios/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA