Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 61(2): 1119-1139, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37688710

RESUMO

Although uncoupling protein 4 (UCP4) is the most abundant protein reported in the brain, the biological function of UCP4 in cerebellum and pathological outcome of UCP4 deficiency in cerebellum remain obscure. To evaluate the role of Ucp4 in the cerebellar Purkinje cells (PCs), we generated the conditional knockdown of Ucp4 in PCs (Pcp2cre;Ucp4fl/fl mice) by breeding Ucp4fl/fl mice with Pcp2cre mice. Series results by Western blot, immunofluorescent staining, and triple RNAscope in situ hybridization confirmed the specific ablation of Ucp4 in PCs in Pcp2cre;Ucp4fl/fl mice, but did not affect the expression of Ucp2, the analog of Ucp4. Combined behavioral tests showed that Pcp2cre;Ucp4fl/fl mice displayed a characteristic bradykinesia in the spontaneous movements. The electromyogram recordings detection excluded the possibility of hypotonia in Pcp2cre;Ucp4fl/fl mice. And the electrical patch clamp recordings showed the altered properties of PCs in Pcp2cre;Ucp4fl/fl mice. Moreover, transmission electron microscope (TEM) results showed the increased mitochondrial circularity in PCs; ROS probe imaging showed the increased ROS generation in molecular layer; and finally, microplate reader assay showed the significant changes of mitochondrial functions, including ROS, ATP, and MMP in the isolated cerebellum tissue. The results suggested that the specific knockdown of mitochondrial protein Ucp4 could damage PCs possibly by attacking their mitochondrial function. The present study is the first to report a close relationship between UCP4 deletion with PCs impairment, and suggests the importance of UCP4 in the substantial support of mitochondrial function homeostasis in bradykinesia. UCP4 might be a therapeutic target for the cerebellar-related movement disorder.


Assuntos
Hipocinesia , Células de Purkinje , Animais , Camundongos , Encéfalo , Cerebelo , Hipocinesia/metabolismo , Células de Purkinje/metabolismo , Espécies Reativas de Oxigênio/metabolismo
2.
J Affect Disord ; 348: 283-296, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38159656

RESUMO

AIMS: To assess the effect of the translocator protein 18 kDa (TSPO) on postpartum depression and explore its mechanism. METHODS: Postpartum depression (PPD) mouse model was established, and flow cytometry, immunofluorescence, Western blot analysis, real-time quantitative PCR, adeno-associated virus (AAV), co-immunoprecipitation-mass spectrometry and immunofluorescence co-staining were used to detect the effect of TSPO ligand ZBD-2 on PPD mice. RESULTS: ZBD-2 inhibits the overactivation of microglia in the hippocampus and amygdala of PPD model mice. ZBD-2 not only inhibited the inflammation but also repressed the burst of reactive oxygen species (ROS) and mitochondrial ROS (mtROS). Meanwhile, ZBD-2 protects mitochondria from LPS-induced damages through inhibiting the influx of calcium. ZBD-2 modulated the calcium influx by increasing the level of translocase of the outer mitochondrial membrane 40 (TOM40) and reducing the interaction of TSPO and TOM40. In addition, the effect of ZBD-2 was partially dependent on anti-oxidative process. Knockdown of TOM40 by adeno-associated virus (AAV) in the hippocampus or amygdala dramatically reduced the effect of ZBD-2 on PPD, indicating that TOM40 mediates the effect of ZBD-2 on PPD. CONCLUSIONS: TOM40 is required for the effect of ZBD-2 on treating anxiety and depression in PPD mice. This study reveals the role of microglia TSPO in PPD development and provides the new therapeutic strategy for PPD.


Assuntos
Depressão Pós-Parto , Microglia , Animais , Feminino , Camundongos , Cálcio/metabolismo , Proteínas de Transporte , Depressão Pós-Parto/tratamento farmacológico , Depressão Pós-Parto/metabolismo , Homeostase , Microglia/metabolismo , Membranas Mitocondriais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de GABA/metabolismo
3.
Brain Res Bull ; 202: 110734, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37586426

RESUMO

Abnormalities in hippocampal synaptic plasticity contribute to the pathogenesis of post-traumatic stress disorder (PTSD). The Wnt/ß-catenin signaling pathway is critical for the regulation of synaptic plasticity. PTSD symptoms can be alleviated by correcting impaired neural plasticity in the hippocampus (Hipp). Electroacupuncture (EA) has a therapeutic effect by relieving PTSD-like behaviors. However, little is known about whether the Wnt/ß-catenin pathway is involved in EA-mediated improvements of PTSD symptoms. In this study, we found that enhanced single prolonged stress (ESPS)-induced PTSD led to abnormal neural plasticity, characterized by the decline of dendritic spines, the expression of postsynaptic density 95 (PSD95), and synaptophysin (Syn) in the stressed Hipp along with the reduction of Wnt3a and ß-catenin, and increased GSK-3ß. EA significantly alleviated PTSD-like behaviors, as assessed by the open field test, elevated platform maze test and conditioning fear test. This was paralleled by correcting abnormal neural plasticity by promoting the expression of PSD95 and Syn, as well as the number of dendritic spines in the Hipp. Importantly, EA exerted anti-PTSD effects by augmenting the expression levels of Wnt3a and ß-catenin, and decreasing that of GSK-3ß. The effects mediated by EA were abolished by XAV939, an inhibitor of the Wnt/ß-catenin pathway. This suggests that EA relieved ESPS-induced PTSD-like behaviors, which can largely be ascribed to impaired neural plasticity in the Hipp. These findings provide new insights into possible mechanisms linking neural plasticity in the Hipp as potential novel targets for PTSD treatment in EA therapy.


Assuntos
Eletroacupuntura , Transtornos de Estresse Pós-Traumáticos , Animais , beta Catenina/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipocampo/metabolismo , Plasticidade Neuronal , Transtornos de Estresse Pós-Traumáticos/terapia , Transtornos de Estresse Pós-Traumáticos/metabolismo , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt , Camundongos
4.
Mol Neurobiol ; 60(11): 6410-6423, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37453994

RESUMO

Fragile X syndrome (FXS) is an inherited human mental retardation that arises from expansion of a CGG repeat in the Fmr1 gene, causing loss of the fragile X mental retardation protein (FMRP). It is reported that N-methyl-D-aspartate receptor (NMDAR)-mediated facilitation of long-term potentiation (LTP) and fear memory are impaired in Fmr1 knockout (KO) mice. In this study, biological, pharmacological, and electrophysiological techniques were performed to determine the roles of D-aspartate (D-Asp), a modulator of NMDAR, and its metabolizing enzyme D-aspartate oxidase (DDO) in Fmr1 KO mice. Levels of D-Asp were decreased in the medial prefrontal cortex (mPFC ); however, the levels of its metabolizing enzyme DDO were increased. Electrophysiological recordings indicated that oral drinking of D-Asp recovered LTP induction in mPFC from Fmr1 KO mice. Moreover, chronic oral administration of D-Asp reversed behavioral deficits of cognition and locomotor coordination in Fmr1 KO mice. The therapeutic action of D-Asp was partially through regulating functions of NMDARs and mGluR5/mTOR/4E-BP signaling pathways. In conclusion, supplement of D-Asp may benefit for synaptic plasticity and behaviors in Fmr1 KO mice and offer a potential therapeutic strategy for FXS.


Assuntos
Ácido D-Aspártico , Síndrome do Cromossomo X Frágil , Camundongos , Animais , Humanos , Síndrome do Cromossomo X Frágil/tratamento farmacológico , Síndrome do Cromossomo X Frágil/metabolismo , Aprendizagem , Potenciação de Longa Duração/fisiologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Camundongos Knockout , Encéfalo/metabolismo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
5.
Phytother Res ; 37(10): 4838-4850, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37458182

RESUMO

Diabetic encephalopathy is a common consequence of diabetes mellitus that causes cognitive dysfunction and neuropsychiatric disorders. Praeruptorin C (Pra-C) from the traditional Chinese medicinal herb Peucedanum praeruptorum Dunn. is a potential antioxidant and neuroprotective agent. This study was conducted to investigate the molecular mechanisms underlying the effect of Pra-C on diabetic cognitive impairment. A novel object recognition test and the Morris water maze test were performed to assess the behavioral performance of mice. Electrophysiological recordings were made to monitor synaptic plasticity in the hippocampus. A protein-protein interaction network of putative Pra-C targets was constructed, and molecular docking simulations were performed to predict the potential mechanisms of the action of Pra-C. Protein expression levels were detected by western blotting. Pra-C administration significantly lowered body weight and fasting blood glucose levels and alleviated learning and memory deficits in type 2 diabetic mice. Network pharmacology and molecular docking results suggested that Pra-C affects the PI3K/AKT/GSK3ß signaling pathway. Western blot analysis confirmed significant increases in phosphorylated PI3K, AKT, and GSK3ß levels in vivo and in vitro upon Pra-C administration. Pra-C alleviated cognitive impairment in type 2 diabetic mice by activating PI3K/AKT/GSK3ß pathway.

6.
Mol Pain ; 19: 17448069231177634, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37207346

RESUMO

Chronic pain, along with comorbid psychiatric disorders, is a common problem worldwide. A growing number of studies have focused on non-opioid-based medicines, and billions of funds have been put into digging new analgesic mechanisms. Peripheral inflammation is one of the critical causes of chronic pain, and drugs with anti-inflammatory effects usually alleviate pain hypersensitivity. Sophoridine (SRI), one of the most abundant alkaloids in Chinese herbs, has been proved to exert antitumor, antivirus and anti-inflammation effects. Here, we evaluated the analgesic effect of SRI in an inflammatory pain mouse model induced by complete Freund's adjuvant (CFA) injection. SRI treatment significantly decreased pro-inflammatory factors release after LPS stimuli in microglia. Three days of SRI treatment relieved CFA-induced mechanical hypersensitivity and anxiety-like behavior, and recovered abnormal neuroplasticity in the anterior cingulate cortex of mice. Therefore, SRI may be a candidate compound for the treatment of chronic inflammatory pain and may serve as a structural basis for the development of new drugs.


Assuntos
Dor Crônica , Hiperalgesia , Camundongos , Animais , Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Hiperalgesia/induzido quimicamente , Adjuvante de Freund/toxicidade , Matrinas , Dor Crônica/tratamento farmacológico , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Ansiedade/complicações , Ansiedade/tratamento farmacológico
7.
Mol Neurobiol ; 60(6): 3379-3395, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36854997

RESUMO

Fragile X syndrome (FXS) is one of the most common inherited mental retardation diseases and is caused by the loss of fragile X mental retardation protein (FMRP) expression. The metabotropic glutamate receptor (mGluR) theory of FXS states that enhanced mGluR-dependent long-term depression (LTD) due to FMRP loss is involved in aberrant synaptic plasticity and autistic-like behaviors, but little is known about the underlying molecular mechanism. Here, we found that only hippocampal mGluR-LTD was exaggerated in adolescent Fmr1 KO mice, while N-methyl-D-aspartate receptor (NMDAR)-LTD was intact in mice of all ages. This development-dependent alteration was related to the differential expression of caveolin-1 (Cav1), which is essential for caveolae formation. Knockdown of Cav1 restored the enhanced mGluR-LTD in Fmr1 KO mice. Moreover, hippocampal Cav1 expression in Fmr1 KO mice induced excessive endocytosis of the α-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate (AMPA) receptor subunit GluA2. This process relied on mGluR1/5 activation rather than NMDAR. Interference with Cav1 expression reversed these changes. Furthermore, massive cholesterol accumulation contributed to redundant caveolae formation, which provided the platform for mGluR-triggered Cav1 coupling to GluA2. Importantly, injection of the cholesterol scavenger methyl-ß-cyclodextrin (Mß-CD) recovered AMPA receptor trafficking and markedly alleviated hyperactivity, hippocampus-dependent fear memory, and spatial memory defects in Fmr1 KO mice. Together, our findings elucidate the important role of Cav1 in mediating mGluR-LTD enhancement and further inducing AMPA receptor endocytosis and suggest that cholesterol depletion by Mß-CD during caveolae formation may be a novel and safe strategy to treat FXS.


Assuntos
Síndrome do Cromossomo X Frágil , Receptores de Glutamato Metabotrópico , Camundongos , Animais , Camundongos Knockout , Caveolina 1/metabolismo , Receptores de AMPA/metabolismo , Depressão , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Hipocampo/metabolismo , Plasticidade Neuronal , Síndrome do Cromossomo X Frágil/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Cognição , Camundongos Endogâmicos C57BL
8.
Neuroscience ; 498: 289-299, 2022 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-35872251

RESUMO

The beneficial effects of exercise on human brain function have been demonstrated in previous studies. Myokines secreted by muscle have attracted increasing attention because of their bridging role between exercise and brain health. Regulated by PPARγ coactivator 1α, fibronectin type III domain-containing protein 5 releases irisin after proteolytic cleavage. Irisin, a type of myokine, is secreted during exercise, which induces white adipose tissue browning and relates to energy metabolism. Recently, irisin has been shown to exert a protective effect on the central nervous system. Irisin secretion triggers an increase in brain-derived neurotrophic factor levels in the hippocampus, contributing to the amelioration of cognition impairments. Irisin also plays an important role in the survival, differentiation, growth, and development of neurons. This review summarizes the role of irisin in neurodegenerative diseases and other neurological disorders. As a novel positive mediator of exercise in the brain, irisin may effectively prevent or decelerate the progress of neurodegenerative diseases in models and also improve cognitive functions. We place emphasis herein on the potential of irisin for prevention rather than treatment in neurodegenerative diseases. In ischemic diseases, irisin can alleviate the pathophysiological processes associated with stroke. Meanwhile, irisin has anxiolytic and antidepressant effects. The potential therapeutic effects of irisin in epilepsy and pain have been initially revealed. Due to the pleiotropic and beneficial properties of irisin, the possibility of irisin treating other neurological diseases could be gradually explored in the future.


Assuntos
Fibronectinas , Doenças Neurodegenerativas , Exercício Físico , Hipocampo , Humanos , Músculo Esquelético , Fatores de Transcrição
9.
Phytother Res ; 36(10): 3932-3948, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35801985

RESUMO

Posttraumatic stress disorder (PTSD) is one of the most common psychiatric diseases, which is characterized by the typical symptoms such as re-experience, avoidance, and hyperarousal. However, there are few drugs for PTSD treatment. In this study, conditioned fear and single-prolonged stress were employed to establish PTSD mouse model, and we investigated the effects of Tanshinone IIA (TanIIA), a natural product isolated from traditional Chinese herbal Salvia miltiorrhiza, as well as the underlying mechanisms in mice. The results showed that the double stress exposure induced obvious PTSD-like symptoms, and TanIIA administration significantly decreased freezing time in contextual fear test and relieved anxiety-like behavior in open field and elevated plus maze tests. Moreover, TanIIA increased the spine density and upregulated synaptic plasticity-related proteins as well as activated CREB/BDNF/TrkB signaling pathway in the hippocampus. Blockage of CREB remarkably abolished the effects of TanIIA in PTSD model mice and reversed the upregulations of p-CREB, BDNF, TrkB, and synaptic plasticity-related protein induced by TanIIA. The molecular docking simulation indicated that TanIIA could interact with the CREB-binding protein. These findings indicate that TanIIA ameliorates PTSD-like behaviors in mice by activating the CREB/BDNF/TrkB pathway, which provides a basis for PTSD treatment.


Assuntos
Produtos Biológicos , Fator Neurotrófico Derivado do Encéfalo , Abietanos , Animais , Ansiedade/tratamento farmacológico , Produtos Biológicos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína de Ligação a CREB/metabolismo , Proteína de Ligação a CREB/farmacologia , Medo , Hipocampo/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Transdução de Sinais
10.
Brain Res Bull ; 188: 11-20, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35850187

RESUMO

Exercise not only builds up our body but also improves cognitive function. Skeletal muscle secretes myokine during exercise as a large reservoir of signaling molecules, which can be considered as a medium between exercise and brain health. Irisin is a circulating myokine derived from the Fibronectin type III domain-containing protein 5 (FNDC5). Irisin regulates energy metabolism because it can stimulate the "Browning" of white adipose tissue. It has been reported that irisin can cross the blood-brain barrier and increase the expression of a brain-derived neurotrophic factor (BDNF) in the hippocampus, which improves learning and memory. In addition, the neuroprotective effect of irisin has been verified in various disease models. Therefore, this review summarizes how irisin plays a neuroprotective role, including its signal pathway and mechanism. In addition, we will briefly discuss the therapeutic potential of irisin for neurological diseases.


Assuntos
Fibronectinas , Fármacos Neuroprotetores , Encéfalo/metabolismo , Exercício Físico/fisiologia , Fibronectinas/metabolismo , Músculo Esquelético/metabolismo , Fármacos Neuroprotetores/metabolismo , Fatores de Transcrição/metabolismo
11.
Front Endocrinol (Lausanne) ; 13: 887238, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35712239

RESUMO

Background: Chronic pain is defined as pain that persists typically for a period of over six months. Chronic pain is often accompanied by an anxiety disorder, and these two tend to exacerbate each other. This can make the treatment of these conditions more difficult. Glucose-dependent insulinotropic polypeptide (GIP) is a member of the incretin hormone family and plays a critical role in glucose metabolism. Previous research has demonstrated the multiple roles of GIP in both physiological and pathological processes. In the central nervous system (CNS), studies of GIP are mainly focused on neurodegenerative diseases; hence, little is known about the functions of GIP in chronic pain and pain-related anxiety disorders. Methods: The chronic inflammatory pain model was established by hind paw injection with complete Freund's adjuvant (CFA) in C57BL/6 mice. GIP receptor (GIPR) agonist (D-Ala2-GIP) and antagonist (Pro3-GIP) were given by intraperitoneal injection or anterior cingulate cortex (ACC) local microinjection. Von Frey filaments and radiant heat were employed to assess the mechanical and thermal hypersensitivity. Anxiety-like behaviors were detected by open field and elevated plus maze tests. The underlying mechanisms in the peripheral nervous system and CNS were explored by GIPR shRNA knockdown in the ACC, enzyme-linked immunosorbent assay, western blot analysis, whole-cell patch-clamp recording, immunofluorescence staining and quantitative real-time PCR. Results: In the present study, we found that hind paw injection with CFA induced pain sensitization and anxiety-like behaviors in mice. The expression of GIPR in the ACC was significantly higher in CFA-injected mice. D-Ala2-GIP administration by intraperitoneal or ACC local microinjection produced analgesic and anxiolytic effects; these were blocked by Pro3-GIP and GIPR shRNA knockdown in the ACC. Activation of GIPR inhibited neuroinflammation and activation of microglia, reversed the upregulation of NMDA and AMPA receptors, and suppressed the enhancement of excitatory neurotransmission in the ACC of model mice. Conclusions: GIPR activation was found to produce analgesic and anxiolytic effects, which were partially due to attenuation of neuroinflammation and inhibition of excitatory transmission in the ACC. GIPR may be a suitable target for treatment of chronic inflammatory pain and pain-related anxiety.


Assuntos
Dor Crônica , Receptores dos Hormônios Gastrointestinais , Animais , Dor Crônica/tratamento farmacológico , Dor Crônica/metabolismo , Adjuvante de Freund , Polipeptídeo Inibidor Gástrico/fisiologia , Giro do Cíngulo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno , Receptores dos Hormônios Gastrointestinais/agonistas , Receptores dos Hormônios Gastrointestinais/antagonistas & inibidores , Receptores dos Hormônios Gastrointestinais/metabolismo
12.
J Neurol ; 269(9): 4939-4954, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35486215

RESUMO

Sleep-related hypermotor epilepsy (SHE) is a focal epilepsy syndrome. The underlying pathophysiology is presumed to be closely related with disruption of GABAergic neurotransmission, which is mainly medicated by γ-aminobutyric acid type A receptor (GABAAR). Thus, it is reasonable to assume that rare GABAAR variants might contribute to the pathogenesis of SHE. To test this hypothesis, we performed next-generation sequencing in 58 SHE patients and analyzed the functional effects of the identified variants in both neuronal and non-neuronal cells using a combination of electrophysiology recordings, western blot, flow cytometry, and confocal microscopy. In our study, we detected three rare variants (NM_198904.2: c.269C > T, p.T90M; NM_198904.2: c.950C > A, p.T317N and NM_198903.2: c.649C > T, p.Q217X) in GABRG2 (MIM:137,164, encoding GABAAR γ2 subunit) in three unrelated patients. Two of the three rare variants were transmitted unaffected maternally (T90M) or unaffected paternally (Q217X), whereas the T317N variant arose de novo. The mother of proband carrying the T90M variant was unaffected and being mosaicism for this variant. Functional analysis showed that T90M and T317N variants decreased GABA-evoked current amplitudes by diverse mechanisms including impaired surface expression, endoplasmic reticulum retention, and channel gating defects. And Q217X variant reduced synaptic clustering and distribution of GABAAR. While a causal role of these variants cannot be established directly from these results, the functional assessment together with the genetic sequencing suggests that these rare GABRG2 variants may constitute genetic risk factors for SHE. Our study further expands the GABRG2 phenotypic spectrum and supports the view that GABAergic neurotransmission participates in the epileptogenesis of SHE.


Assuntos
Epilepsia , Receptores de GABA-A , Humanos , Neurônios/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Sono , Ácido gama-Aminobutírico
13.
Brain Res Bull ; 181: 77-86, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35093468

RESUMO

Hormone therapy (HT) has failed to improve learning and memory in postmenopausal women according to recent clinical studies; however, the reason for failure of HT in improving cognitive performance is unknown. In our research, we found cognitive flexibility was improved by 17ß-Estradiol (E2) in mice 1 week after ovariectomy (OVXST), but not in mice 3 months after ovariectomy (OVXLT). Isobaric tags for relative and absolute quantitation (iTRAQ) revealed increased cannabinoid receptor interacting protein 1 (CNRIP1) in E2-treated OVXLT mice compared with E2-treated OVXST mice. Adeno-associated virus 2/9 (AAV2/9) delivery of Cnrip1 short-hairpin small interfering RNA (Cnrip1-shRNA) rescued the impaired cognitive flexibility in E2 treated OVXLT mice. This effect is dependent on CB1 function, which could be blocked by AM251-a CB1 antagonist. Our results indicated a new method to increasing cognitive flexibility in women receiving HT by disrupting CNRIP1.


Assuntos
Antagonistas de Receptores de Canabinoides/farmacologia , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Estradiol/farmacologia , Terapia de Reposição Hormonal , Córtex Pré-Frontal/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Ovariectomia , Piperidinas/farmacologia , Pós-Menopausa , Pirazóis/farmacologia , RNA Interferente Pequeno , Receptor CB1 de Canabinoide/antagonistas & inibidores
14.
Neurosci Lett ; 765: 136284, 2021 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-34624394

RESUMO

Depression is a kind of common mental disorder associated with neuroinflammation, and astrocytes play a vital role in regulating and mediating neuroinflammation in central nervous system. Scutellarin has significant anti-inflammatory and neuroprotective effects. However, whether scutellarin exerts antidepressant effect remains unknown. In present study, it was found that scutellarin suppressed LPS-induced neuroinflammation in the hippocampus and alleviated depression-like behaviors in mice. In addition, scutellarin inhibited LPS-induced elevation of TNFα, IL-1ß, IL-6 and iNOS, and reversed the downregulation of IL-4 and BDNF in astrocytes in vitro. Furthermore, the activated TLR4/NF-κB pathway in LPS-treated astrocytes was suppressed by scutellarin. Collectively, these results suggest that scutellarin ameliorates depression-like behaviors induced by neuroinflammation partially through inhibiting the TLR4/NF-κB pathway in astrocytes.


Assuntos
Apigenina/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Depressão/imunologia , Glucuronatos/farmacologia , Doenças Neuroinflamatórias/imunologia , Animais , Anti-Inflamatórios/farmacologia , Hipocampo/efeitos dos fármacos , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
Mol Neurobiol ; 58(10): 5272-5288, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34278533

RESUMO

Sleep deprivation (SD) leads to cognitive impairment due to neuroinflammation associated with impaired hippocampal neuronal plasticity and memory processes. Liver X receptors (LXRs), including LXRα and LXRß isoforms, are crucial for synaptic plasticity and neuroinflammation. However, the potential roles of LXRs in the pathogenesis of cognitive impairment induced by SD remain unclear. We revealed that SD resulted in LXRß reduction in the hippocampus, which was associated with upregulated expression of high mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4)/NF-κB p65, and knockdown of hippocampal LXRß by shRNA (shLXRß) led to cognitive impairment. GW3965, a dual agonist for both LXRα and LXRß, ameliorated SD-induced cognitive impairment by inhibiting microglia activation, suppressing HMGB1/TLR4/NF-κB p65 pathway, and ultimately affecting the hippocampal expression of inflammatory cytokines in SD mice. LXRß knockdown by shLXRß abrogated the GW3965-mediated inhibition of the HMGB1/TLR4/NF-κB p65 pathway, therefore, abolishing the cognitive improvement. Moreover, inhibition of HMGB1 by glycyrrhizin (GLY) synergistic promoted GW3965-mediated anti-inflammation in activated microglia after lipopolysaccharide (LPS)/ATP stimulation and facilitated the cognitive improvement after GW administration by activating LXRß. All the data suggested that GW3965 ameliorated impaired cognition in SD mice by suppressing the HMGB1/TLR4/NF-κB p65 pathway followed LXRß activation. This study correlates a deficit of LXRß in cognitive dysfunction in SD associated with HMGB1 inflammatory pathway in hippocampus, and LXRs may serve as a potential therapeutic target for cognitive impairment with anti-inflammation.


Assuntos
Disfunção Cognitiva/metabolismo , Hipocampo/metabolismo , Receptores X do Fígado/metabolismo , Doenças Neuroinflamatórias/metabolismo , Privação do Sono/metabolismo , Animais , Anti-Inflamatórios/administração & dosagem , Benzoatos/administração & dosagem , Benzilaminas/administração & dosagem , Disfunção Cognitiva/tratamento farmacológico , Relação Dose-Resposta a Droga , Ácido Glicirrízico/administração & dosagem , Hipocampo/efeitos dos fármacos , Receptores X do Fígado/agonistas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microinjeções , Doenças Neuroinflamatórias/tratamento farmacológico , Distribuição Aleatória , Privação do Sono/tratamento farmacológico
16.
Phytother Res ; 35(7): 3936-3944, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33856723

RESUMO

Anxiety disorders are a common frequently psychiatric symptom in patients that lead to disruption of daily life. Scutellarin (Scu) is the main component of Erigeron breviscapus, which has been used as a neuroprotective agent against glutamate-induced excitotoxicity. However, the potential effect of Scu on the stress-related neuropsychological disorders has not been clarified. In this study, Anxiety-like behavior was induced by acute restraint stress in mice. Scu were injected intraperitoneally (twice daily, 3 days). Results showed that Scu exhibited good protective activity on mice by decreasing transmitter release levels. Restraint stress caused significant anxiety like behavior in mice. Treatment of Scu could significantly improve the moving time of open arms in Elevated Plus Maze and central time on open field test. Scu treatment suppressed action potential firing frequency, restored excessive presynaptic quantal release, and down-regulated glutamatergic receptor expression levels in the prefrontal cortex (PFC) of stressed mice. GABAA Rα1 and GABAA γ2 expression in the brain PFC tissues of mice were nearly abrogated by Scu treatment. In stress-induced anxiety mice, stress can increase the frequency of mini excitatory postsynaptic currents (mEPSC), which can be reversed by Scu treatment. Therefore, Scu has a potent anxiolytic activity and may be valuable for the treatment of stress-induced anxiety disorders.


Assuntos
Ansiedade , Apigenina , Glucuronatos , Neurotransmissores/fisiologia , Animais , Ansiedade/tratamento farmacológico , Apigenina/farmacologia , Glucuronatos/farmacologia , Camundongos
17.
Brain Res Bull ; 172: 22-30, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33848615

RESUMO

Neuroexcitotoxicity is a common feature in neuronal damage and neurodegenerative diseases. Our previous studies have confirmed that neuronal and astrocytic G­protein-coupled receptor 30 (GPR30) play a key role in neuroprotection in vivo and in vitro. Microglia are considered as immune cells in the central nervous system. However, the role of microglial GPR30 in neuroprotection against neuroexcitotoxicity remained unclear. In this study, MTT, Western blot, immunocytochemical staining, phagocytosis assay and wound healing assay were employed to detect the effect of GPR30 in N9 microglial cells after exposure to glutamate. We found that the treatment of GPR30 specific agonist G1 inhibited glutamate-induced proliferation and activation in N9 microglial cells. G1 inhibited M1 polarization, facilitated M2 polarization, and decreased over-phagocytosis but had no effect on migration ability in microglia. The result of neurons and microglia co-culture showed that the activation of microglial GPR30 protected neurons from excitotoxicity through the NF-κB/MAPK signaling pathways. Our findings suggested a key role of microglial GPR30 in excitatory neuronal damage and neurodegenerative diseases.


Assuntos
Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Técnicas de Cocultura , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos , Microglia/metabolismo , NF-kappa B/metabolismo , Neurônios/metabolismo , Fagocitose/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
18.
Mol Pain ; 16: 1744806920972241, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33243040

RESUMO

Visceral pain is a common clinical symptom, which is caused by mechanical stretch, spasm, ischemia and inflammation. Fragile X syndrome (FXS) with lack of fragile X mental retardation protein (FMRP) protein is an inherited disorder that is characterized by moderate or severe intellectual and developmental disabilities. Previous studies reported that FXS patients have self-injurious behavior, which may be associated with deficits in nociceptive sensitization. However, the role of FMRP in visceral pain is still unclear. In this study, the FMR1 knock out (KO) mice and SH-SY5Y cell line were employed to demonstrate the role of FMRP in the regulation of visceral pain. The data showed that FMR1 KO mice were insensitive to zymosan treatment. Recording in the anterior cingulate cortex (ACC), a structure involved in pain process, showed less presynaptic glutamate release and postsynaptic responses in the FMR1 KO mice as compared to the wild type (WT) mice after zymosan injection. Zymosan treatment caused enhancements of adenylyl cyclase 1 (AC1), a pain-related enzyme, and NMDA GluN2B receptor in the ACC. However, these up-regulations were attenuated in the ACC of FMR1 KO mice. Last, we found that zymosan treatment led to increase of FMRP levels in the ACC. These results were further confirmed in SH-SY5Y cells in vitro. Our findings demonstrate that FMRP is required for NMDA GluN2B and AC1 upregulation, and GluN2B/AC1/FMRP forms a positive feedback loop to modulate visceral pain.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , Dor Visceral/metabolismo , Adenilil Ciclases/metabolismo , Animais , Comportamento Animal , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Masculino , Camundongos Knockout , Fosforilação , Terminações Pré-Sinápticas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Regulação para Cima
20.
Glia ; 68(1): 27-43, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31429156

RESUMO

Ischemic stroke leads to neuronal damage induced by excitotoxicity, inflammation, and oxidative stress. Astrocytes play diverse roles in stroke and ischemia-induced inflammation, and autophagy is critical for maintaining astrocytic functions. Our previous studies showed that the activation of G protein-coupled receptor 30 (GPR30), an estrogen membrane receptor, protected neurons from excitotoxicity. However, the role of astrocytic GPR30 in maintaining autophagy and neuroprotection remained unclear. In this study, we found that the neuroprotection induced by G1 (GPR30 agonist) in wild-type mice after a middle cerebral artery occlusion was completely blocked in GPR30 conventional knockout (KO) mice but partially attenuated in astrocytic or neuronal GPR30 KO mice. In cultured primary astrocytes, glutamate exposure induced astrocyte proliferation and decreased astrocyte autophagy by activating mammalian target of rapamycin (mTOR) and c-Jun N-terminal kinase (JNK) and inhibiting p38 mitogen-activated protein kinase (MAPK) pathway. G1 treatment restored autophagy to its basal level by regulating the p38 pathway but not the mTOR and JNK signaling pathways. Our findings revealed a key role of GPR30 in neuroprotection via the regulation of astrocyte autophagy and support astrocytic GPR30 as a potential drug target against ischemic brain damage.


Assuntos
Astrócitos/metabolismo , Autofagia/fisiologia , Fármacos Neuroprotetores/farmacologia , Quinolinas/farmacologia , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fármacos Neuroprotetores/uso terapêutico , Quinolinas/uso terapêutico , Receptores Acoplados a Proteínas G/agonistas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...