Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 234
Filtrar
1.
ACS Chem Neurosci ; 15(8): 1712-1727, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38581382

RESUMO

Short-chain fatty acids (SCFAs) are gut microbial metabolic derivatives produced during the fermentation of ingested complex carbohydrates. SCFAs have been widely regarded to have a potent anti-inflammatory and neuro-protective role and have implications in several disease conditions, such as, inflammatory bowel disease, type-2 diabetes, and neurodegenerative disorders. Japanese encephalitis virus (JEV), a neurotropic flavivirus, is associated with life threatening neuro-inflammation and neurological sequelae in infected hosts. In this study, we hypothesize that SCFAs have potential in mitigating JEV pathogenesis. Postnatal day 10 BALB/c mice were intraperitoneally injected with either a SCFA mixture (acetate, propionate, and butyrate) or PBS for a period of 7 days, followed by JEV infection. All mice were observed for onset and progression of symptoms. The brain tissue was collected upon reaching terminal illness for further analysis. SCFA-supplemented JEV-infected mice (SCFA + JEV) showed a delayed onset of symptoms, lower hindlimb clasping score, and decreased weight loss and increased survival by 3 days (p < 0.0001) upon infection as opposed to the PBS-treated JEV-infected animals (JEV). Significant downregulation of inflammatory cytokines TNF-α, MCP-1, IL-6, and IFN-Υ in the SCFA + JEV group relative to the JEV-infected control group was observed. Inflammatory mediators, phospho-NF-kB (P-NF-kB) and iba1, showed 2.08 ± 0.1 and 3.132 ± 0.43-fold upregulation in JEV versus 1.19 ± 0.11 and 1.31 ± 0.11-fold in the SCFA + JEV group, respectively. Tissue section analysis exhibited reduced glial activation (JEV group─42 ± 2.15 microglia/ROI; SCFA + JEV group─27.07 ± 1.8 microglia/ROI) in animals that received SCFA supplementation prior to infection as seen from the astrocytic and microglial morphometric analysis. Caspase-3 immunoblotting showed 4.08 ± 1.3-fold upregulation in JEV as compared to 1.03 ± 0.14-fold in the SCFA + JEV group and TUNEL assay showed a reduced cellular death post-JEV infection (JEV-6.4 ± 1.5 cells/ROI and SCFA + JEV-3.7 ± 0.73 cells/ROI). Our study critically contributes to the increasing evidence in support of SCFAs as an anti-inflammatory and neuro-protective agent, we further expand its scope as a potential supplementary intervention in JEV-mediated neuroinflammation.


Assuntos
Encefalite Japonesa , Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Doenças Neuroinflamatórias , Microbioma Gastrointestinal/fisiologia , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/imunologia , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/microbiologia , Microglia/efeitos dos fármacos , Microglia/imunologia , Encefalite Japonesa/tratamento farmacológico , Encefalite Japonesa/imunologia , Encefalite Japonesa/microbiologia , Encefalite Japonesa/prevenção & controle , Encefalite Japonesa/virologia , Ácidos Graxos Voláteis/farmacologia , Ácidos Graxos Voláteis/uso terapêutico , Vírus da Encefalite Japonesa (Subgrupo)/efeitos dos fármacos , Vírus da Encefalite Japonesa (Subgrupo)/imunologia , Vírus da Encefalite Japonesa (Subgrupo)/patogenicidade , Análise de Sobrevida , Quimiocinas/imunologia , Quimiocinas/metabolismo , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/metabolismo , Síndrome da Liberação de Citocina/prevenção & controle , Humanos , Feminino , Animais , Camundongos , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/virologia , Carga Viral/efeitos dos fármacos , Fatores de Tempo
2.
J Neurosci Res ; 102(4): e25336, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38656664

RESUMO

Chronic neuroinflammation has been implicated in neurodegenerative disease pathogenesis. A key feature of neuroinflammation is neuronal loss and glial activation, including microglia and astrocytes. 4R-cembranoid (4R) is a natural compound that inhibits hippocampal pro-inflammatory cytokines and increases memory function in mice. We used the lipopolysaccharide (LPS) injection model to study the effect of 4R on neuronal density and microglia and astrocyte activation. C57BL/6J wild-type mice were injected with LPS (5 mg/kg) and 2 h later received either 4R (6 mg/kg) or vehicle. Mice were sacrificed after 72 h for analysis of brain pathology. Confocal images of brain sections immunostained for microglial, astrocyte, and neuronal markers were used to quantify cellular hippocampal phenotypes and neurons. Hippocampal lysates were used to measure the expression levels of neuronal nuclear protein (NeuN), inducible nitrous oxide synthase (iNOS), arginase-1, thrombospondin-1 (THBS1), glial cell-derived neurotrophic factor (GDNF), and orosomucoid-2 (ORM2) by western blot. iNOS and arginase-1 are widely used protein markers of pro- and anti-inflammatory microglia, respectively. GDNF promotes neuronal survival, and ORM2 and THBS1 are astrocytic proteins that regulate synaptic plasticity and inhibit microglial activation. 4R administration significantly reduced neuronal loss and the number of pro-inflammatory microglia 72 h after LPS injection. It also decreased the expression of the pro-inflammatory protein iNOS while increasing arginase-1 expression, supporting its anti-inflammatory role. The protein expression of THBS1, GDNF, and ORM2 was increased by 4R. Our data show that 4R preserves the integrity of hippocampal neurons against LPS-induced neuroinflammation in mice.


Assuntos
Hipocampo , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Neuroglia , Neurônios , Animais , Lipopolissacarídeos/toxicidade , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/patologia , Doenças Neuroinflamatórias/tratamento farmacológico , Fenótipo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia
3.
Nature ; 628(8006): 195-203, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38480879

RESUMO

Sustained smouldering, or low-grade activation, of myeloid cells is a common hallmark of several chronic neurological diseases, including multiple sclerosis1. Distinct metabolic and mitochondrial features guide the activation and the diverse functional states of myeloid cells2. However, how these metabolic features act to perpetuate inflammation of the central nervous system is unclear. Here, using a multiomics approach, we identify a molecular signature that sustains the activation of microglia through mitochondrial complex I activity driving reverse electron transport and the production of reactive oxygen species. Mechanistically, blocking complex I in pro-inflammatory microglia protects the central nervous system against neurotoxic damage and improves functional outcomes in an animal disease model in vivo. Complex I activity in microglia is a potential therapeutic target to foster neuroprotection in chronic inflammatory disorders of the central nervous system3.


Assuntos
Complexo I de Transporte de Elétrons , Inflamação , Microglia , Doenças Neuroinflamatórias , Animais , Feminino , Humanos , Masculino , Camundongos , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Transporte de Elétrons/efeitos dos fármacos , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Complexo I de Transporte de Elétrons/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Multiômica , Células Mieloides/metabolismo , Células Mieloides/patologia , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/patologia , Espécies Reativas de Oxigênio/metabolismo
4.
Int Immunopharmacol ; 130: 111693, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38428144

RESUMO

Oxidative stress and neuroinflammation are two major causes leading to early brain injury after subarachnoid hemorrhage (SAH). Nuclear factor E2-related factor 2 (Nrf2) is a critical transcription factor that contributes to antioxidant responses. Additionally, Nrf2 could inhibit transforming growth factor beta-activated kinase 1 (TAK1), which plays a vital role in microglial activation-mediated neuroinflammation. Neferine (NE) exhibits considerable protective effects in diverse disease models. However, the detailed effect and mechanism of NE on SAH remain unknown. Our data showed that NE treatment significantly reduced behavior and cognitive impairment, and brain edema in the early period after SAH. In addition, NE mitigated SAH-induced oxidative damage, neuroinflammation, and neural death. Moreover, NE inhibited M1 microglial polarization and enhanced M2 phenotype microglia both in vivo and in vitro. Further investigations revealed that NE enhanced the Nrf2-antioxidant response element (ARE) signaling pathway and suppressed TAK1-NF-κB signaling. In contrast, depletion of Nrf2 by ML385 suppressed Nrf2-ARE signaling, induced TAK1-NF-κB activation, and further promoted M1 microglial polarization. Additionally, ML385 abated the neuroprotective effects of NE against SAH. Notably, LPS also aggravated TAK1-NF-κB activation and reversed the beneficial effects of NE after SAH. In summary, NE provides protection after SAH by inhibiting oxidative stress and modulating microglial polarization through Nrf2 activation and TAK1-NF-κB suppression.


Assuntos
Benzilisoquinolinas , Microglia , Fator 2 Relacionado a NF-E2 , NF-kappa B , Doenças Neuroinflamatórias , Hemorragia Subaracnóidea , Masculino , Benzilisoquinolinas/farmacologia , Benzilisoquinolinas/uso terapêutico , Camundongos Endogâmicos C57BL , Microglia/patologia , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/etiologia , Fator 2 Relacionado a NF-E2/agonistas , NF-kappa B/metabolismo , Transdução de Sinais , Hemorragia Subaracnóidea/complicações , Modelos Animais de Doenças
5.
Brain Res Bull ; 209: 110918, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38432497

RESUMO

BACKGROUND: Traumatic brain injury (TBI) is a leading cause of high mortality and disability worldwide. Overactivation of astrocytes and overexpression of inflammatory responses in the injured brain are characteristic pathological features of TBI. Rosiglitazone (ROS) is a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist known for its anti-inflammatory activity. However, the relationship between the inflammatory response involved in ROS treatment and astrocyte A1 polarization remains unclear. OBJECTIVE: This study aimed to investigate whether ROS treatment improves dysfunction and astrocyte A1 polarization induced after TBI and to elucidate the underlying mechanisms of these functions. METHODS: SD rats were randomly divided into sham operation group, TBI group, TBI+ROS group, and TBI+ PPAR-γ antagonist group (GW9662 + TBI). The rat TBI injury model was prepared by the CCI method; brain water content test and wire grip test scores suggested the prognosis; FJB staining showed the changes of ROS on the morphology and number of neurons in the peripheral area of cortical injury; ELISA, immunofluorescence staining, and western blotting analysis revealed the effects of ROS on inflammatory response and astrocyte activation with the degree of A1 polarization after TBI. RESULTS: Brain water content, inflammatory factor expression, and astrocyte activation in the TBI group were higher than those in the sham-operated group (P < 0.05); compared with the TBI group, the expression of the above indexes in the ROS group was significantly lower (P < 0.05). Compared with the TBI group, PPAR-γ content was significantly higher and C3 content was considerably lower in the ROS group (P < 0.05); compared with the TBI group, PPAR-γ content was significantly lower and C3 content was substantially higher in the inhibitor group (P < 0.05). CONCLUSION: ROS can exert neuroprotective effects by inhibiting astrocyte A1 polarization through the PPAR-γ pathway based on the reduction of inflammatory factors and astrocyte activation in the brain after TBI.


Assuntos
Astrócitos , Lesões Encefálicas Traumáticas , Hipoglicemiantes , Doenças Neuroinflamatórias , Rosiglitazona , Animais , Ratos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/patologia , Doenças Neuroinflamatórias/tratamento farmacológico , PPAR gama/metabolismo , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Rosiglitazona/farmacologia , Rosiglitazona/uso terapêutico , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Masculino
6.
Nat Rev Neurosci ; 25(5): 334-350, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38531962

RESUMO

Epilepsy remains a major health concern as anti-seizure medications frequently fail, and there is currently no treatment to stop or prevent epileptogenesis, the process underlying the onset and progression of epilepsy. The identification of the pathological processes underlying epileptogenesis is instrumental to the development of drugs that may prevent the generation of seizures or control pharmaco-resistant seizures, which affect about 30% of patients. mTOR signalling and neuroinflammation have been recognized as critical pathways that are activated in brain cells in epilepsy. They represent a potential node of biological convergence in structural epilepsies with either a genetic or an acquired aetiology. Interventional studies in animal models and clinical studies give strong support to the involvement of each pathway in epilepsy. In this Review, we focus on available knowledge about the pathophysiological features of mTOR signalling and the neuroinflammatory brain response, and their interactions, in epilepsy. We discuss mitigation strategies for each pathway that display therapeutic effects in experimental and clinical epilepsy. A deeper understanding of these interconnected molecular cascades could enhance our strategies for managing epilepsy. This could pave the way for new treatments to fill the gaps in the development of preventative or disease-modifying drugs, thus overcoming the limitations of current symptomatic medications.


Assuntos
Progressão da Doença , Epilepsia , Doenças Neuroinflamatórias , Serina-Treonina Quinases TOR , Humanos , Epilepsia/tratamento farmacológico , Animais , Serina-Treonina Quinases TOR/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Transdução de Sinais/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Anticonvulsivantes/uso terapêutico , Anticonvulsivantes/farmacologia
7.
Eur J Pharmacol ; 964: 176267, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38072038

RESUMO

OBJECTIVE: The inflammatory response is acknowledged as a crucial pathological aspect of spinal cord injury (SCI). Tetramethylpyrazine (TMP) has been demonstrated to possess neuroprotective properties within the central nervous system via its anti-inflammatory mechanisms. This study aims to investigate the molecular mechanism by which TMP alleviates SCI from an anti-inflammatory standpoint. METHODS: The SCI model was established using the MASCIS impactor device. The Basso-Beattie-Bresnahan (BBB) locomotor rating scale was utilised to assess rat locomotion. Nissl and Golgi staining were used to observe neuron and dendritic spine morphology, respectively. A transmission electron microscope was used to observe the microcosmic morphology of the axon. ELISA kits were used to measure the concentrations of IL-1ß and IL-18 in the spinal cord. Immunofluorescence staining was used to detect P2X7R+/IBA-1+ cells, and Western blot and RT-qPCR were used to analyze the protein and mRNA expression of P2X7R in the spinal cord. Additionally, Western blot was used to detect NLRP3 and Cleaved-Caspase-1 (p20), the critical proteins in the NLRP3 inflammasome pathway. RESULTS: TMP ameliorated the microcosmic morphology of the axon and had an inhibitory effect on the concentrations of IL-1ß and IL-18 after SCI. Furthermore, TMP inhibited the expression of both P2X7R and critical proteins of the NLRP3 inflammasome pathway on microglia after SCI. The aforementioned effects of TMP exhibit similarities to those of BBG (P2X7R antagonist); however, they can be effectively reversed by BzATP (P2X7R activator). CONCLUSION: TMP alleviated SCI via reducing tissue damage, neuroinflammation, and the expression of P2X7R, NLRP3, IL-1ß, and IL-18.


Assuntos
Anti-Inflamatórios , Doenças Neuroinflamatórias , Fármacos Neuroprotetores , Pirazinas , Traumatismos da Medula Espinal , Animais , Ratos , Anti-Inflamatórios/uso terapêutico , Inflamassomos , Interleucina-18 , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/etiologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Proteína 3 que Contém Domínio de Pirina da Família NLR , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Ratos Sprague-Dawley , Medula Espinal , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo
8.
Brain Res ; 1822: 148612, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37778649

RESUMO

Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by neuroinflammation. Dexmedetomidine (Dex) is known for its neuroprotective properties in clinical settings. In this study, we investigated the potential of Dex in protecting against neuroinflammation in an AD mouse model induced by amyloid-beta (Aß) injection. First, in the AD mouse model, Aß injection were administered, and the model was confirmed through behavioral tests, including the Morris water maze and Y-maze. Neuroinflammatory states in Aß-injected mice were assessed using hematoxylin and eosin staining and enzyme-linked immunosorbent assay. Expression levels of microRNA (miR)-204-3p and F-box/LRR-repeat protein 7 (FBXL7) in mouse tissues were determined through real-time quantitative polymerase chain reaction and Western blot. The binding interaction between miR-204-3p and FBXL7 was elucidated using dual-luciferase analysis. Aß-injected mice exhibited cognitive impairment, neuroinflammation, and downregulated miR-204-3p. Upregulation of miR-204-3p reduced inflammatory infiltration and mitigated neuroinflammation in Aß-injected mice. Dex treatment reduced inflammation in hippocampal tissues of Aß-injected mice. Dex treatment upregulated miR-204-3p, leading to suppressed FBXL7 expression in tissues. Inhibition of miR-204-3p or overexpression of FBXL7 reversed the alleviating effect of Dex on neuroinflammation in Aß-injected mice. Overall, Dex increased miR-204-3p expression, resulting in the inhibition of FBXL7, and subsequently alleviated neuroinflammation in Aß-injected mice.


Assuntos
Doença de Alzheimer , Dexmedetomidina , MicroRNAs , Animais , Camundongos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Dexmedetomidina/farmacologia , MicroRNAs/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Proteínas F-Box/efeitos dos fármacos , Proteínas F-Box/metabolismo
9.
FASEB J ; 38(1): e23394, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38149910

RESUMO

Neutrophils and their production of neutrophil extracellular traps (NETs) significantly contribute to neuroinflammation and brain damage after intracerebral hemorrhage (ICH). Although Akebia saponin D (ASD) demonstrates strong anti-inflammatory activities and blood-brain barrier permeability, its role in regulating NETs formation and neuroinflammation following ICH is uncharted. Our research focused on unraveling the influence of ASD on neuroinflammation mediated by NETs and the mechanisms involved. We found that increased levels of peripheral blood neutrophils post-ICH are correlated with worse prognostic outcomes. Through network pharmacology, we identified ASD as a promising therapeutic target for ICH. ASD administration significantly improved neurobehavioral performance and decreased NETs production in neutrophils. Furthermore, ASD was shown to upregulate the membrane protein NTSR1 and activate the cAMP signaling pathway, confirmed through transcriptome sequencing, western blot, and immunofluorescence. Interestingly, the NTSR1 inhibitor SR48692 significantly nullified ASD's anti-NETs effects and dampened cAMP pathway activation. Mechanistically, suppression of PKAc via H89 negated ASD's anti-NETs effects but did not affect NTSR1. Our study suggests that ASD may reduce NETs formation and neuroinflammation, potentially involving the NTSR1/PKAc/PAD4 pathway post-ICH, underlining the potential of ASD in mitigating neuroinflammation through its anti-NETs properties.


Assuntos
Hemorragia Cerebral , Armadilhas Extracelulares , Doenças Neuroinflamatórias , Saponinas , Farmacologia em Rede , Perfilação da Expressão Gênica , Saponinas/farmacologia , Armadilhas Extracelulares/efeitos dos fármacos , Doenças Neuroinflamatórias/tratamento farmacológico , Hemorragia Cerebral/tratamento farmacológico , Humanos , Animais , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Receptores de Neurotensina/metabolismo , Proteína-Arginina Desiminase do Tipo 4/metabolismo
10.
Parkinsonism Relat Disord ; 117: 105906, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37924806

RESUMO

INTRODUCTION: Parkinson's disease represents a neurodegenerative condition characterized by the progressive loss of dopaminergic neurons within the Substantia Nigra pars compacta (SNpc), resulting in diminished dopamine levels in the striatum (STR) and chronic neuroinflammation. Recent investigations have proposed the neuroprotective potential of the endocannabinoid system in neurodegenerative disorders. ß-caryophyllene (BCP) is recognized for its antioxidant and anti-inflammatory properties, attributed to its activation of the type 2 cannabinoid receptor. This study aimed to assess the neuroprotective impact of BCP on dopaminergic neurons, with a particular focus on inhibiting the NLRP3 inflammasome. METHODS: A model of hemiparkinsonism, induced by 6-hydroxydopamine (6-OHDA), served as the experimental framework. Motor function was evaluated using the cylinder test, and inflammasome inhibition was determined by assessing the expression of NLRP3, caspase-1, and the pro-inflammatory cytokine IL-1ß in both the SNpc and STR through ELISA analysis. Furthermore, the evaluation of oxidative stress was facilitated by quantifying malondialdehyde (MDA) levels in the same regions. RESULTS: BCP treatment demonstrated significant improvements in motor dysfunction, as assessed by the cylinder test (p=0.0011) and exhibited a neuroprotective effect on dopaminergic neurons within the SNpc (p=0.0017), as well as nerve fibers in the STR (p=0.0399). In terms of its ability to inhibit the inflammasome, BCP led to decreased expression levels of NLRP3 (p=0.0401 in STR and p = 0.0139 in SNpc), caspase-1 (p=0.0004 in STR), and MDA (p=0.0085 in STR and p=0.0414 in SNpc). CONCLUSION: These results point to BCP's potential in mitigating the motor deficit, inhibiting NLRP3 inflammasome activation, and attenuating lipid peroxidation induced by 6-OHDA.


Assuntos
Doenças Neuroinflamatórias , Fármacos Neuroprotetores , Humanos , Caspases/metabolismo , Caspases/farmacologia , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Inflamassomos/metabolismo , Camundongos Endogâmicos C57BL , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Neuroproteção , Fármacos Neuroprotetores/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Oxidopamina , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Camundongos , Animais
11.
Cell Commun Signal ; 21(1): 264, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37770901

RESUMO

BACKGROUND: The poor prognosis of subarachnoid hemorrhage (SAH) is often attributed to neuroinflammation. The cGAS-STING axis, a cytoplasmic pathway responsible for detecting dsDNA, plays a significant role in mediating neuroinflammation in neurological diseases. However, the effects of inhibiting cGAS with the selective small molecule inhibitor RU.521 on brain injury and the underlying mechanisms after SAH are still unclear. METHODS: The expression and microglial localization of cGAS following SAH were investigated with western blot analysis and immunofluorescent double-staining, respectively. RU.521 was administered after SAH. 2'3'-cGAMP, a second messenger converted by activated cGAS, was used to activate cGAS-STING. The assessments were carried out by adopting various techniques including neurological function scores, brain water content, blood-brain barrier permeability, western blot analysis, TUNEL staining, Nissl staining, immunofluorescence, morphological analysis, Morris water maze test, Golgi staining, CCK8, flow cytometry in the in vivo and in vitro settings. RESULTS: Following SAH, there was an observed increase in the expression levels of cGAS in rat brain tissue, with peak levels observed at 24 h post-SAH. RU.521 resulted in a reduction of brain water content and blood-brain barrier permeability, leading to an improvement in neurological deficits after SAH. RU.521 had beneficial effects on neuronal apoptosis and microglia activation, as well as improvements in microglial morphology. Additionally, RU.521 prompted a shift in microglial phenotype from M1 to M2. We also noted a decrease in the production of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-6, and an increase in the level of the anti-inflammatory cytokine IL-10. Finally, RU.521 treatment was associated with improvements in cognitive function and an increase in the number of dendritic spines in the hippocampus. The therapeutic effects were mediated by the cGAS/STING/NF-κB pathway and were found to be abolished by 2'3'-cGAMP. In vitro, RU.521 significantly reduced apoptosis and neuroinflammation. CONCLUSION: The study showed that SAH leads to neuroinflammation caused by microglial activation, which contributes to early brain injury. RU.521 improved neurological outcomes and reduced neuroinflammation by regulating microglial polarization through the cGAS/STING/NF-κB pathway in early brain injury after SAH. RU.521 may be a promising candidate for the treatment of neuroinflammatory pathology after SAH. Video Abstract.


Assuntos
Lesões Encefálicas , Hemorragia Subaracnóidea , Animais , Ratos , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Microglia/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/patologia
12.
ACS Chem Neurosci ; 14(20): 3869-3882, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37775304

RESUMO

A substantial body of evidence demonstrates an association between a malfunction in the resolution of acute inflammation and the development of chronic inflammation. Recently, in this context, the importance of formyl peptide receptor 2 (FPR2) has been underlined. FPR2 activity is modulated by a wide range of endogenous ligands, including specialized pro-resolving mediators (SPMs) (e.g., LXA4 and AT-LXA4) and synthetic ligands. Since SPMs have unfavorable pharmacokinetic properties, we aimed to evaluate the protective and pro-resolving effects of a new potent FPR2 agonist, compound CMC23, in organotypic hippocampal cultures (OHCs) stimulated with lipopolysaccharide (LPS). The protective activity of CMC23 limited the lactate dehydrogenase release in LPS-stimulated cultures. This activity was mediated by the interaction with FPR2 as pretreatment with the FPR2 selective antagonist WRW4 abolished CMC23-induced protection. Furthermore, decreased levels of pro-inflammatory IL-1ß and IL-6 were observed after CMC23 administration in LPS-treated OHCs. CMC23 also diminished the LPS-induced increase in IL-17A and both IL-23 subunits p19 and p40 in OHCs. Finally, we demonstrated that CMC23 exerts its beneficial impact via the STAT3/SOCS3 signaling pathway since it attenuated the level of phospho-STAT3 and maintained the LPS-induced SOCS3 levels in OHCs. Collectively, our research implies that the new FPR2 agonist CMC23 has beneficial protective and anti-inflammatory properties in nanomolar doses and FPR2 represents a promising target for the enhancement of inflammation resolution.


Assuntos
Doenças Neuroinflamatórias , Receptores de Formil Peptídeo , Humanos , Endotoxinas , Hipocampo/metabolismo , Lipopolissacarídeos/toxicidade , Receptores de Formil Peptídeo/agonistas , Doenças Neuroinflamatórias/induzido quimicamente , Doenças Neuroinflamatórias/tratamento farmacológico
13.
Neurochem Int ; 170: 105603, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37633650

RESUMO

Intracerebral hemorrhage (ICH), which has high mortality and disability rate is associated with microglial pyroptosis and neuroinflammation, and the effective treatment methods are limited Epigallocatechin-3-gallate (EGCG) has been found to play a cytoprotective role by regulating the anti-inflammatory response to pyroptosis in other systemic diseases. However, the role of EGCG in microglial pyroptosis and neuroinflammation after ICH remains unclear. In this study, we investigated the effects of EGCG pretreatment on neuroinflammation-mediated neuronal pyroptosis and the underlying neuroprotective mechanisms in experimental ICH. EGCG pretreatment was found to remarkably improved neurobehavioral performance, and decreased the hematoma volume and cerebral edema in mice. We found that EGCG pretreatment attenuated the release of hemin-induced inflammatory cytokines (IL-1ß, IL-18, and TNF-α). EGCG significantly upregulated the expression of heme oxygenase-1 (HO-1), and downregulated the levels of pyroptotic molecules and inflammatory cytokines including Caspase-1, GSDMD, NLRP3, mature IL-1ß, and IL-18. EGCG pretreatment also decreased the number of Caspase-1-positive microglia and GSDMD along with NLRP3-positive microglia after ICH. Conversely, an HO-1-specific inhibitor (ZnPP), significantly inhibited the anti-pyroptosis and anti-neuroinflammation effects of EGCG. Therefore, EGCG pretreatment alleviated microglial pyroptosis and neuroinflammation, at least in part through the Caspase-1/GSDMD/NLRP3 pathway by upregulating HO-1 expression after ICH. In addition, EGCG pretreatment promoted the polarization of microglia from the M1 phenotype to M2 phenotype after ICH. The results suggest that EGCG is a potential agent to attenuate neuroinflammation via its anti-pyroptosis effect after ICH.


Assuntos
Hemorragia Cerebral , Heme Oxigenase-1 , Microglia , Doenças Neuroinflamatórias , Fármacos Neuroprotetores , Animais , Camundongos , Caspases/metabolismo , Caspases/farmacologia , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/genética , Hemorragia Cerebral/metabolismo , Citocinas/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Interleucina-18/metabolismo , Interleucina-18/farmacologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/efeitos dos fármacos , Piroptose/genética , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico
14.
Nature ; 620(7976): 1071-1079, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37587343

RESUMO

Identifying therapeutics to delay, and potentially reverse, age-related cognitive decline is critical in light of the increased incidence of dementia-related disorders forecasted in the growing older population1. Here we show that platelet factors transfer the benefits of young blood to the ageing brain. Systemic exposure of aged male mice to a fraction of blood plasma from young mice containing platelets decreased neuroinflammation in the hippocampus at the transcriptional and cellular level and ameliorated hippocampal-dependent cognitive impairments. Circulating levels of the platelet-derived chemokine platelet factor 4 (PF4) (also known as CXCL4) were elevated in blood plasma preparations of young mice and humans relative to older individuals. Systemic administration of exogenous PF4 attenuated age-related hippocampal neuroinflammation, elicited synaptic-plasticity-related molecular changes and improved cognition in aged mice. We implicate decreased levels of circulating pro-ageing immune factors and restoration of the ageing peripheral immune system in the beneficial effects of systemic PF4 on the aged brain. Mechanistically, we identified CXCR3 as a chemokine receptor that, in part, mediates the cellular, molecular and cognitive benefits of systemic PF4 on the aged brain. Together, our data identify platelet-derived factors as potential therapeutic targets to abate inflammation and rescue cognition in old age.


Assuntos
Envelhecimento , Cognição , Disfunção Cognitiva , Doenças Neuroinflamatórias , Nootrópicos , Fator Plaquetário 4 , Animais , Masculino , Camundongos , Envelhecimento/sangue , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Doenças Neuroinflamatórias/sangue , Doenças Neuroinflamatórias/complicações , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/prevenção & controle , Fator Plaquetário 4/sangue , Fator Plaquetário 4/metabolismo , Fator Plaquetário 4/farmacologia , Fator Plaquetário 4/uso terapêutico , Nootrópicos/sangue , Nootrópicos/metabolismo , Nootrópicos/farmacologia , Nootrópicos/uso terapêutico , Plasma/química , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Disfunção Cognitiva/sangue , Disfunção Cognitiva/complicações , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/prevenção & controle , Transcrição Gênica/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos
15.
Environ Toxicol ; 38(11): 2691-2704, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37483155

RESUMO

OBJECTIVE: The most prevalent brain-specific microRNA, MicroRNA-124, exhibits anti-inflammatory properties. Luteolin nano-formulation with Zn oxide in the form of L/ZnO NPs may boost anti-diabetic properties; however, its beneficial effect on miRNAs is yet unknown in diabetes. The effectiveness of L/ZnONPs supplements in preventing diabetic neurodegeneration by modulating inflammatory responses in a diabetic model was investigated. METHODS: A diabetic rat model was induced by a high-fat diet and streptozotocin (30 mg/kg I.P.). Plasma glucose, insulin, and HOMR-IR levels, as well as cytokines, lipid peroxidation, GSH/GSSG, and glucose transporter 1, were determined along with the tight junction proteins occludin (OCLN) and zona occludens 1 (ZO-1). Moreover, the expressions of brain CCAAT/enhancer-binding protein (C/EBPA mRNA), miR-124, glial fibrillary acidic protein (GFAP), and NF-kBp65 were measured alongside the histological investigation. RESULTS: The results revealed that L/ZnO NPs were able to diminish lipid peroxidation, increase the activity of antioxidant enzymes, and reduce inflammation under oxidative stress. Consequently, it was able to reduce hyperglycemia, elevate insulin levels, and improve insulin resistance. Besides, L/ZnO NPs upregulate miR-124, reduce C/EBPA mRNA, increase BCl-2, and inhibit apoptosis. The results indicate that diabetes raises BBB permeability via tight junction protein decline, which is restored following L/ZnO NPs treatment. Luteolin/ZnO NPs regulate miR-124 and microglia polarization by targeting C/EBPA and are expected to alleviate inflammatory injury via modulation of the redox-sensitive signal transduction pathways. Luteolin/ZnO NPs have a novel target for the protection of the BBB and the prevention of neurological complications in diabetes.


Assuntos
Diabetes Mellitus Experimental , Insulinas , MicroRNAs , Doenças Neuroinflamatórias , Óxido de Zinco , Animais , Ratos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/induzido quimicamente , Insulinas/farmacologia , Luteolina/farmacologia , MicroRNAs/genética , Nanopartículas , Doenças Neuroinflamatórias/tratamento farmacológico , Estresse Oxidativo , RNA Mensageiro , Proteínas de Junções Íntimas , Óxido de Zinco/farmacologia
16.
Biomed Pharmacother ; 165: 115026, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37336148

RESUMO

This study sought to investigate the anti-amyloid ß (Aß) and anti-neuroinflammatory effects of catalpol in an Alzheimer's disease (AD) mouse model. METHODS: The effects of catalpol on Aß formation were investigated by thioflavin T assay. The effect of catalpol on generating inflammatory cytokines from microglial cells and the cytotoxicity of microglial cells on HT22 hippocampal cells were assessed by real-time quantitative PCR, ELISA, redox reactions, and cell viability. APPswe/PS1ΔE9 mice were treated with catalpol, and their cognitive ability was investigated using the water maze and novel object recognition tests. Immunohistochemistry and immunofluorescence were used to probe for protein markers of microglia and astrocyte, Aß deposits, and NF-κB pathway activity. Aß peptides, neuroinflammation, and nitric oxide production were examined using ELISA and redox reactions. RESULTS: Catalpol potently inhibited Aß fibril and oligomer formation. In microglial cells stimulated by Aß, catalpol alleviated the expression of the proinflammatory cytokines tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and inducible nitric oxide synthase (iNOS) but promoted the expression of the anti-inflammatory cytokine IL-10. Catalpol alleviated the cytotoxic effects of Aß-exposed microglia on HT22 cells. Treatment with catalpol in APPswe/PS1ΔE9 mice downregulated neuroinflammation production, decreased Aß deposits in the brains and alleviated cognitive impairment. Catalpol treatment decreased the number of IBA-positive microglia and GFAP-positive astrocytes and their activities of the NF-κB pathway in the hippocampus of APPswe/PS1ΔE9 mice. CONCLUSION: The administration of catalpol protected neurons by preventing neuroinflammation and Aß deposits in an AD mouse model. Therefore, catalpol may be a promising strategy for treating AD.


Assuntos
Peptídeos beta-Amiloides , Disfunção Cognitiva , Medicamentos de Ervas Chinesas , Glucosídeos Iridoides , Doenças Neuroinflamatórias , Fármacos Neuroprotetores , Placa Amiloide , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Glucosídeos Iridoides/farmacologia , Glucosídeos Iridoides/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Placa Amiloide/tratamento farmacológico , Doenças Neuroinflamatórias/tratamento farmacológico , Animais , Camundongos , Modelos Animais de Doenças , Citocinas/metabolismo , Linhagem Celular , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Peptídeos beta-Amiloides/antagonistas & inibidores , Camundongos Endogâmicos C57BL , Masculino , Feminino , Camundongos Transgênicos
17.
ACS Chem Neurosci ; 14(6): 1045-1062, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36868874

RESUMO

The gut-brain axis (GBA) is the umbrella term to include all bidirectional communication between the brain and gastrointestinal (GI) tract in the mammalian body. Evidence from over two centuries describes a significant role of GI microbiome in health and disease states of the host organism. Short-chain fatty acids (SCFAs), mainly acetate, butyrate, and propionate that are the physiological forms of acetic acid, butyric acid, and propionic acid respectively, are GI bacteria derived metabolites. SCFAs have been reported to influence cellular function in multiple neurodegenerative diseases (NDDs). In addition, the inflammation modulating properties of SCFAs make them suitable therapeutic candidates in neuroinflammatory conditions. This review provides a historical background of the GBA and current knowledge of the GI microbiome and role of individual SCFAs in central nervous system (CNS) disorders. Recently, a few reports have also identified the effects of GI metabolites in the case of viral infections. Among these viruses, the flaviviridae family is associated with neuroinflammation and deterioration of CNS functions. In this context, we additionally introduce SCFA based mechanisms in different viral pathogenesis to understand the former's potential as agents against flaviviral disease.


Assuntos
Eixo Encéfalo-Intestino , Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Doenças Neurodegenerativas , Doenças Neuroinflamatórias , Viroses , Animais , Humanos , Eixo Encéfalo-Intestino/fisiologia , Ácido Butírico/metabolismo , Ácido Butírico/uso terapêutico , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/uso terapêutico , Mamíferos/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Viroses/tratamento farmacológico , Viroses/metabolismo , Microbioma Gastrointestinal/fisiologia , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo
18.
CNS Neurosci Ther ; 29(4): 1000-1011, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36601662

RESUMO

INTRODUCTION: Sciatica causes intense pain. No satisfactory therapeutic drugs exist to treat sciatica. This study aimed to probe the potential mechanism of ferulic acid in sciatica treatment. METHODS: Thirty-two SD rats were randomly divided into 4 groups: sham operation, chronic constriction injury (CCI), mecobalamin, and ferulic acid. We conducted RNA sequencing, behavioral tests, ELISA, PCR, western blotting, and immunofluorescence analysis. TAK-242 and JSH23 were administered to RSC96 and GMI-R1 cells to explore whether ferulic acid can inhibit apoptosis and alleviate inflammation. RESULTS: RNA sequencing showed that TLR4/NF-κB pathway is involved in the mechanism of sciatica. CCI induced cold and mechanical hyperalgesia; destroyed the sciatic nerve structure; increased IL-1ß, IL-6, TNF-α, IL-8, and TGF-ß protein levels and IL-1ß, IL-6, TNF-α, TGF-ß, TLR4, and IBA-1 mRNA levels; and decreased IL-10 and INF-γ protein levels and IL-4 mRNA levels. Immunohistochemistry showed that IBA-1, CD32, IL-1ß, iNOS, nNOS, COX2, and TLR4 expression was increased while S100ß and Arg-1 decreased. CCI increased TLR4, IBA-1, IL-1ß, iNOS, Myd88, p-NF-κB, and p-p38MAPK protein levels. Treatment with mecobalamin and ferulic acid reversed these trends. Lipopolysaccharide (LPS) induced RSC96 cell apoptosis by reducing Bcl-2 and Bcl-xl protein and mRNA levels and increasing Bax and Bad mRNA and IL-1ß, TLR4, Myd88, p-NF-κB, and p-p38MAPK protein levels, while ferulic acid inhibited cell apoptosis by decreasing IL-1ß, TLR4, Myd88, p-NF-κB, and p-p38MAPK levels and increasing Bcl-2 and Bcl-xl levels. In GMI-R1 cells, Ferulic acid attenuated LPS-induced M1 polarization by decreasing the M1 polarization markers IL-1ß, IL-6, iNOS, and CD32 and increasing the M2 polarization markers CD206, IL-4, IL-10 and Arg-1. After LPS treatment, IL-1ß, iNOS, TLR4, Myd88, p-p38MAPK, and p-NF-κB levels were obviously increased, and Arg-1 expression was reduced, while ferulic acid reversed these changes. CONCLUSION: Ferulic acid can promote injured sciatic nerve repair by reducing neuronal cell apoptosis and inflammatory infiltration though the TLR4/NF-κB pathway.


Assuntos
Ácidos Cumáricos , NF-kappa B , Ciática , Receptor 4 Toll-Like , Animais , Ratos , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Fator 88 de Diferenciação Mieloide/metabolismo , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , NF-kappa B/metabolismo , Ratos Sprague-Dawley , RNA Mensageiro , Ciática/tratamento farmacológico , Ciática/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Ácidos Cumáricos/farmacologia , Ácidos Cumáricos/uso terapêutico
19.
CNS Neurosci Ther ; 29(4): 1094-1108, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36627822

RESUMO

BACKGROUND: A growing body of research shows that drug monomers from traditional Chinese herbal medicines have antineuroinflammatory and neuroprotective effects that can significantly improve the recovery of motor function after spinal cord injury (SCI). Here, we explore the role and molecular mechanisms of Alpinetin on activating microglia-mediated neuroinflammation and neuronal apoptosis after SCI. METHODS: Stimulation of microglia with lipopolysaccharide (LPS) to simulate neuroinflammation models in vitro, the effect of Alpinetin on the release of pro-inflammatory mediators in LPS-induced microglia and its mechanism were detected. In addition, a co-culture system of microglia and neuronal cells was constructed to assess the effect of Alpinetin on activating microglia-mediated neuronal apoptosis. Finally, rat spinal cord injury models were used to study the effects on inflammation, neuronal apoptosis, axonal regeneration, and motor function recovery in Alpinetin. RESULTS: Alpinetin inhibits microglia-mediated neuroinflammation and activity of the JAK2/STAT3 pathway. Alpinetin can also reverse activated microglia-mediated reactive oxygen species (ROS) production and decrease of mitochondrial membrane potential (MMP) in PC12 neuronal cells. In addition, in vivo Alpinetin significantly inhibits the inflammatory response and neuronal apoptosis, improves axonal regeneration, and recovery of motor function. CONCLUSION: Alpinetin can be used to treat neurodegenerative diseases and is a novel drug candidate for the treatment of microglia-mediated neuroinflammation.


Assuntos
Flavonas , Doenças Neuroinflamatórias , Traumatismos da Medula Espinal , Animais , Ratos , Apoptose/efeitos dos fármacos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Janus Quinase 2/efeitos dos fármacos , Janus Quinase 2/metabolismo , Lipopolissacarídeos , Microglia , Doenças Neuroinflamatórias/tratamento farmacológico , Ratos Sprague-Dawley , Transdução de Sinais , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo , Flavonas/farmacologia , Flavonas/uso terapêutico , Fator de Transcrição STAT3/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo
20.
J Neuroinflammation ; 19(1): 312, 2022 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-36566220

RESUMO

BACKGROUND: Neuroinflammation and neuronal apoptosis are closely associated with a poor prognosis in patients with subarachnoid hemorrhage (SAH). We investigated the role of C-C motif chemokine receptor 2 (CCR2) in SAH. METHODS: Pre-processed RNA-seq transcriptome datasets GSE167110 and GSE79416 from the Gene Expression Omnibus (GEO) database were screened for genes differentially expressed between mice with SAH and control mice, using bioinformatics analysis. The endovascular perforation model was performed to establish SAH. RS504393 (a CCR2 antagonist) and LY294002 (PI3K inhibitor) were administered to explore the mechanism of neuroinflammation after SAH. SAH grading, neurological scoring, brain water content and blood-brain barrier (BBB) permeability determination, enzyme-linked immunosorbent assay (ELISA), western blotting, and immunofluorescence were performed. An in vitro model of SAH was induced in H22 cells by hemin treatment. The protective mechanism of CCR2 inhibition was studied by adding RS504393 and LY294002. Clinical cerebrospinal fluid (CST) samples were detected by ELISA. RESULTS: Expression of CCR2 was upregulated in both datasets and was identified as a hub gene. CCR2 expression was significantly upregulated in the cytoplasm of neurons after SAH, both in vitro and in vivo. RS significantly reduced the brain water content and blood-brain barrier permeability, alleviated neuroinflammation, and reduced neuronal apoptosis after SAH. Additionally, the protective effects of CCR2 inhibition were abolished by LY treatment. Finally, the levels of CCR2, inflammatory factors, and apoptotic factors were elevated in the CSF of patients with SAH. CCR2 levels were associated with patient outcomes at the 6-month follow-up. CONCLUSION: CCR2 expression was upregulated in both in vitro and in vivo SAH models. Additionally, inhibition of CCR2, at least partly through the PI3K/AKT pathway, alleviated neuroinflammation and neuronal apoptosis in vivo and in vitro. CCR2 levels in the CSF have a moderate diagnostic value for 6-month outcome prediction in patients with SAH.


Assuntos
Apoptose , Doenças Neuroinflamatórias , Proteínas Proto-Oncogênicas c-akt , Receptores CCR2 , Hemorragia Subaracnóidea , Animais , Camundongos , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/etiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores CCR2/antagonistas & inibidores , Transdução de Sinais , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...