Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
J Neuroinflammation ; 21(1): 36, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38287311

RESUMO

BACKGROUND: Sepsis-associated encephalopathy (SAE) is an acute cerebral dysfunction caused by sepsis. Neuroinflammation induced by sepsis is considered a potential mechanism of SAE; however, very little is known about the role of the meningeal lymphatic system in SAE. METHODS: Sepsis was established in male C57BL/6J mice by intraperitoneal injection of 5 mg/kg lipopolysaccharide, and the function of meningeal lymphatic drainage was assessed. Adeno-associated virus 1-vascular endothelial growth factor C (AAV1-VEGF-C) was injected into the cisterna magna to induce meningeal lymphangiogenesis. Ligation of deep cervical lymph nodes (dCLNs) was performed to induce pre-existing meningeal lymphatic dysfunction. Cognitive function was evaluated by a fear conditioning test, and inflammatory factors were detected by enzyme-linked immunosorbent assay. RESULTS: The aged mice with SAE showed a significant decrease in the drainage of OVA-647 into the dCLNs and the coverage of the Lyve-1 in the meningeal lymphatic, indicating that sepsis impaired meningeal lymphatic drainage and morphology. The meningeal lymphatic function of aged mice was more vulnerable to sepsis in comparison to young mice. Sepsis also decreased the protein levels of caspase-3 and PSD95, which was accompanied by reductions in the activity of hippocampal neurons. Microglia were significantly activated in the hippocampus of SAE mice, which was accompanied by an increase in neuroinflammation, as indicated by increases in interleukin-1 beta, interleukin-6 and Iba1 expression. Cognitive function was impaired in aged mice with SAE. However, the injection of AAV1-VEGF-C significantly increased coverage in the lymphatic system and tracer dye uptake in dCLNs, suggesting that AAV1-VEGF-C promotes meningeal lymphangiogenesis and drainage. Furthermore, AAV1-VEGF-C reduced microglial activation and neuroinflammation and improved cognitive dysfunction. Improvement of meningeal lymphatics also reduced sepsis-induced expression of disease-associated genes in aged mice. Pre-existing lymphatic dysfunction by ligating bilateral dCLNs aggravated sepsis-induced neuroinflammation and cognitive impairment. CONCLUSION: The meningeal lymphatic drainage is damaged in sepsis, and pre-existing defects in this drainage system exacerbate SAE-induced neuroinflammation and cognitive dysfunction. Promoting meningeal lymphatic drainage improves SAE. Manipulation of meningeal lymphangiogenesis could be a new strategy for the treatment of SAE.


Assuntos
Lesões Encefálicas , Disfunção Cognitiva , Encefalopatia Associada a Sepse , Sepse , Camundongos , Masculino , Animais , Fator C de Crescimento do Endotélio Vascular , Lipopolissacarídeos , Doenças Neuroinflamatórias , Camundongos Endogâmicos C57BL , Sepse/complicações , Lesões Encefálicas/complicações
2.
J Neurointerv Surg ; 15(5): 473-477, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-35459713

RESUMO

BACKGROUND: Trigeminocardiac reflex (TCR) is a brainstem reflex that can lead to hemodynamic instability manifested as bradycardia, decrease/increase of mean arterial pressure (MAP) and, in the worst case scenario, asystole during surgery. The effective intraoperative management of recurrent and profound TCR has yet to be established. This randomized paired study was performed to identify the effect of a prophylactic intra-arterial injection of lidocaine to prevent TCR caused by Onyx embolization during cerebrovascular intervention surgery. METHODS: A total of 136 patients who received Onyx embolization under general anesthesia were assigned to a control group pretreated with intra-arterial saline injection or a lidocaine group pretreated with an intra-arterial injection of 20 mg lidocaine. Heart rate (HR) and MAP were closely monitored during the embolization procedures and the incidence of TCR, mainly characterized by a decrease in HR of ≥20%, and perioperative adverse events was recorded. RESULTS: During dimethyl sulfoxide (DMSO)/Onyx injection, HR was much slower in the control group than in the lidocaine group (p<0.05). TCR occurred in 12 patients (17.6%) in the control group (cardiac arrest in 3 patients) with decreased (7 cases) or increased (5 cases) MAP, whereas no TCR was observed in the lidocaine group. Notably, most TCR episodes occurred in patients with dural arteriovenous fistula and middle meningeal artery being affected. The composite adverse events were significantly higher in the control group than in the lidocaine group (p<0.05). CONCLUSION: This prospective study shows that a prophylactic intra-arterial injection of 20 mg lidocaine could be recommended as a novel strategy to effectively and safely prevent TCR during endovascular embolization.


Assuntos
Malformações Vasculares do Sistema Nervoso Central , Embolização Terapêutica , Parada Cardíaca , Reflexo Trigêmino-Cardíaco , Humanos , Malformações Vasculares do Sistema Nervoso Central/terapia , Dimetil Sulfóxido , Embolização Terapêutica/efeitos adversos , Embolização Terapêutica/métodos , Parada Cardíaca/etiologia , Injeções Intra-Arteriais , Lidocaína/farmacologia , Lidocaína/uso terapêutico , Polivinil/efeitos adversos , Estudos Prospectivos , Reflexo Trigêmino-Cardíaco/fisiologia , Resultado do Tratamento
3.
J Mol Neurosci ; 72(9): 1875-1901, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35792980

RESUMO

Postoperative cognitive dysfunction (POCD) is a cognitive deterioration and dementia that arise after a surgical procedure, affecting up to 40% of surgery patients over the age of 60. The precise etiology and molecular mechanisms underlying POCD remain uncovered. These reasons led us to employ integrative bioinformatics and machine learning methodologies to identify several biological signaling pathways involved and molecular signatures to better understand the pathophysiology of POCD. A total of 223 differentially expressed genes (DEGs) comprising 156 upregulated and 67 downregulated genes were identified from the circRNA microarray dataset by comparing POCD and non-POCD samples. Gene ontology (GO) analyses of DEGs were significantly involved in neurogenesis, autophagy regulation, translation in the postsynapse, modulating synaptic transmission, regulation of the cellular catabolic process, macromolecule modification, and chromatin remodeling. Pathway enrichment analysis indicated some key molecular pathways, including mTOR signaling pathway, AKT phosphorylation of cytosolic targets, MAPK and NF-κB signaling pathway, PI3K/AKT signaling pathway, nitric oxide signaling pathway, chaperones that modulate interferon signaling pathway, apoptosis signaling pathway, VEGF signaling pathway, cellular senescence, RANKL/RARK signaling pathway, and AGE/RAGE pathway. Furthermore, seven hub genes were identified from the PPI network and also determined transcription factors and protein kinases. Finally, we identified a new predictive drug for the treatment of SCZ using the LINCS L1000, GCP, and P100 databases. Together, our results bring a new era of the pathogenesis of a deeper understanding of POCD, identified novel therapeutic targets, and predicted drug inhibitors in POCD.


Assuntos
Complicações Cognitivas Pós-Operatórias , RNA Circular , Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Ontologia Genética , Humanos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais
4.
Front Cell Neurosci ; 16: 771156, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35221919

RESUMO

Background: The pathophysiological mechanisms underlying postoperative cognitive dysfunction (POCD) remain unclear over the years. Neuroinflammation caused by surgery has been recognized as an important element in the development of POCD. Many studies also suggest that the vagus nerve plays an important role in transmitting peripheral injury signals to the central nervous system (CNS) and the resultant neuroinflammation. Previously, we have demonstrated that brain mast cells (BMCs), as the "first responders", play a vital role in neuroinflammation and POCD. However, how the vagus nerve communicates with BMCs in POCD has not yet been clarified. Methods: In the current study, we highlighted the role of the vagus nerve as a conduction highway in surgery-induced neuroinflammation for the first time. In our model, we tested if mice underwent unilateral cervical vagotomy (VGX) had less neuroinflammation compared to the shams after laparotomy (LP) at an early stage. To further investigate the roles of mast cells and glutamate in the process, we employed KitW-sh mice and primary bone marrow-derived MCs to verify the glutamate-NR2B axis on MCs once again. Results: Our results demonstrated that there were higher levels of glutamate and BMCs activation as early as 4 h after LP. Meanwhile, vagotomy could partially block the increases and reduce neuroinflammation caused by peripheral inflammation during the acute phase. Excitingly, inhibition of NR2B receptor and knockout of mast cells can attenuateneuroinflammation induced by glutamate. Conclusion: Taken together, our findings indicate that the vagus is a high-speed pathway in the transmission of peripheral inflammation to the CNS. Activation of BMCs triggered a neuroinflammatory cascade. Inhibition of NR2B receptor on BMCs can reduce glutamate-induced BMCs activation, neuroinflammation, and memory impairment, suggesting a novel treatment strategy for POCD.

5.
BMC Anesthesiol ; 21(1): 285, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34781892

RESUMO

BACKGROUND: Radiofrequency catheter ablation (RFCA) as a safe and effective method has been widely used in ventricular tachycardia (VT) patients, and with which anesthesiologists frequently manage their perioperative care. The aim of this study was to investigate the effects of different anesthetic depths on perioperative RFCA and recurrence in patients who with intractable VT and could not tolerate an awake procedure. METHODS: We reviewed electronic medical records of patients with VT who underwent RFCA by general anesthesia from January 2014 to March 2019. According to intraoperative VT induction, they were divided into two groups: non-inducible group (group N) and inducible group (group I). We constructed several multivariable regression models, in which covariates included patient characteristics, comorbidities, protopathy and bispectral index (BIS) value. RESULTS: One hundred one patients were analyzed. Twenty-nine patients (28.7%) experienced VT no induction, and 26 patients (25.7%) relapsed within 1 year. Compared with group I, the proportion of patients with arrhythmogenic right ventricular cardiomyopathy in group N were higher (P < 0.05), and the recurrence rate of VT was significantly higher (51.7% vs 15.3%) (P < 0.05). The BIS value in group N was significantly lower (P < 0.01), in addition, the BIS < 40 was associated with elevated odds of VT no induction compared with a BIS > 50 (odds ratio, 6.92; 95% confidence interval, 1.47-32.56; P = 0.01). VT no induction was an independent predictor of recurrence after RFCA (odds ratio, 5.01; 95% confidence interval, 1.88-13.83; P < 0.01). CONCLUSION: Lower BIS value during VT induction in RFCA operation was associated with high risk of VT no induction, which affects postoperative outcomes. We proposed that appropriate depth of anesthesia should be maintained during the process of VT induction.


Assuntos
Anestesia Geral/métodos , Ablação por Cateter/métodos , Taquicardia Ventricular/cirurgia , Adulto , Idoso , Monitores de Consciência , Registros Eletrônicos de Saúde , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva , Estudos Retrospectivos , Resultado do Tratamento
6.
Brain Res Bull ; 172: 61-78, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33892083

RESUMO

Although the global incidence of neurodegenerative diseases has been steadily increasing, especially in adults, there are no effective therapeutic interventions. Neurodegeneration is a heterogeneous group of disorders that is characterized by the activation of immune cells in the central nervous system (CNS) (e.g., mast cells and microglia) and subsequent neuroinflammation. Mast cells are found in the brain and the gastrointestinal tract and play a role in "tuning" neuroimmune responses. The complex bidirectional communication between mast cells and gut microbiota coordinates various dynamic neuro-cellular responses, which propagates neuronal impulses from the gastrointestinal tract into the CNS. Numerous inflammatory mediators from degranulated mast cells alter intestinal gut permeability and disrupt blood-brain barrier, which results in the promotion of neuroinflammatory processes leading to neurological disorders, thereby offsetting the balance in immune-surveillance. Emerging evidence supports the hypothesis that gut-microbiota exert a pivotal role in inflammatory signaling through the activation of immune and inflammatory cells. Communication between inflammatory cytokines and neurocircuits via the gut-brain axis (GBA) affects behavioral responses, activates mast cells and microglia that causes neuroinflammation, which is associated with neurological diseases. In this comprehensive review, we focus on what is currently known about mast cells and the gut-brain axis relationship, and how this relationship is connected to neurodegenerative diseases. We hope that further elucidating the bidirectional communication between mast cells and the GBA will not only stimulate future research on neurodegenerative diseases but will also identify new opportunities for therapeutic interventions.


Assuntos
Eixo Encéfalo-Intestino/fisiologia , Comunicação Celular/fisiologia , Mastócitos/metabolismo , Doenças Neurodegenerativas/metabolismo , Transdução de Sinais/fisiologia , Animais , Encéfalo/imunologia , Humanos , Mastócitos/imunologia , Doenças Neurodegenerativas/imunologia
7.
Neural Regen Res ; 16(11): 2184-2197, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33818491

RESUMO

Corticotropin-releasing hormone is a critical component of the hypothalamic-pituitary-adrenal axis, which plays a major role in the body's immune response to stress. Mast cells are both sensors and effectors in the interaction between the nervous and immune systems. As first responders to stress, mast cells can initiate, amplify and prolong neuroimmune responses upon activation. Corticotropin-releasing hormone plays a pivotal role in triggering stress responses and related diseases by acting on its receptors in mast cells. Corticotropin-releasing hormone can stimulate mast cell activation, influence the activation of immune cells by peripheral nerves and modulate neuroimmune interactions. The latest evidence shows that the release of corticotropin-releasing hormone induces the degranulation of mast cells under stress conditions, leading to disruption of the blood-brain barrier, which plays an important role in neurological diseases, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, autism spectrum disorder and amyotrophic lateral sclerosis. Recent studies suggest that stress increases intestinal permeability and disrupts the blood-brain barrier through corticotropin-releasing hormone-mediated activation of mast cells, providing new insight into the complex interplay between the brain and gastrointestinal tract. The neuroimmune target of mast cells is the site at which the corticotropin-releasing hormone directly participates in the inflammatory responses of nerve terminals. In this review, we focus on the neuroimmune connections between corticotropin-releasing hormone and mast cells, with the aim of providing novel potential therapeutic targets for inflammatory, autoimmune and nervous system diseases.

8.
J Neurosci ; 41(18): 4131-4140, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33664132

RESUMO

Ketamine is known to have a rapid and lasting antidepressant effect. Recent studies have shown that ketamine exerts it rapid antidepressant effect by blocking burst firing in the lateral habenula (LHb). Whether the sustained antidepressant effect of ketamine occurs through the same mechanism has not been explored. Here, using male rats, we found that local infusion of (R,S)-ketamine into the LHb resulted in a rapid antidepressant-like effect 1 h after infusion, which almost returned to baseline levels after 24 h. Intra-LHb injection of (S)-ketamine also showed a significant antidepressant-like effect 1 h after injection, which recovered at 24 h. No significant antidepressant-like effect was found at 1 or 24 h after the administration of (R)-ketamine into the LHb. Injection of (2R,6R)-hydroxynorketamine, a ketamine metabolite, into the LHb did not result in any obvious antidepressant-like effect 1 or 24 h after injection. Systemic administration of (R,S)-ketamine (intraperitoneally) significantly suppressed LHb bursting activity at 1 h, but the inhibitory effect was reversed 24 h after injection. No significant effect of (R,S)-ketamine on miniature excitatory postsynaptic potentials of LHb neurons was found at 1 or 24 h after systemic application. Our study demonstrated that the sustained antidepressant-like effect of ketamine may not depend on burst firing of LHb neurons.SIGNIFICANCE STATEMENT Ketamine exerts it rapid antidepressant effect by blocking burst firing in the lateral habenula (LHb). However, whether the sustained antidepressant effect of ketamine occurs through the same mechanism has not been explored. In the present study, we demonstrated that the sustained antidepressant effect of ketamine may not depend on the burst firing of LHb neurons. This finding may lead to a novel perspective on LHb in the antidepressant effect of ketamine.

9.
J Neuroinflammation ; 18(1): 68, 2021 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-33750404

RESUMO

OBJECTIVE: Neuroinflammation plays a critical role in central nervous system diseases. Exosomal miRNAs released from various cells are implicated in cell-to-cell communication. Prior studies have placed substantial emphasis on the role of cytokines in mast cell-microglia interactions during neuroinflammation. However, it has never been clearly determined whether exosomal miRNAs participate in the interaction between mast cells and microglia and thus mediate neuroinflammation. METHODS: The characteristics of exosomes isolated from cell culture supernatants were confirmed by transmission electron microscopy (TEM), nanoparticle-tracking analysis (NTA) and Western blot. The transfer of PKH67-labelled exosomes and Cy3-labelled miR-409-3p was observed by fluorescence microscopy. Migration and activation of murine BV-2 microglial cells were evaluated through Transwell assays and immunofluorescence staining for Iba1 and CD68. CD86, IL-1ß, IL-6 and TNF-α were assessed via qRT-PCR and ELISA. MiR-409-3p was detected by qRT-PCR. Nr4a2 and NF-κB levels were measured by western blot. Regulatory effects were identified by luciferase reporter assays. RESULTS: Lipopolysaccharide (LPS)-stimulated murine P815 mast cells secreted exosomes that were efficiently taken up by murine BV-2 cells, which promoted murine BV-2 cell migration and activation. LPS-P815 exosomes increased the CD86, IL-1ß, IL-6 and TNF-α levels in murine BV-2 microglia. Furthermore, activated mast cells delivered exosomal miR-409-3p to murine BV-2 microglia. Upregulated miR-409-3p promoted murine BV-2 microglial migration, activation and neuroinflammation by targeting Nr4a2 to activate the NF-κB pathway. CONCLUSION: Exosomal miR-409-3p secreted from activated mast cells promotes microglial migration, activation and neuroinflammation by targeting Nr4a2 to activate the NF-κB pathway, which provides evidence that not only cytokines but also exosomal miRNAs participate in neuroinflammation. In the future, targeting exosomal miRNAs may provide new insights into neuroinflammation.


Assuntos
Encefalite/patologia , Exossomos/patologia , Mastócitos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Microglia/patologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Animais , Linhagem Celular , Movimento Celular , Células Cultivadas , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos , Camundongos , NF-kappa B/efeitos dos fármacos , Nanopartículas , Transdução de Sinais/efeitos dos fármacos
10.
Behav Brain Res ; 402: 113113, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33412227

RESUMO

Early-life stress (ELS) is a high-risk factor for the development of chronic visceral pain in adulthood. Emerging evidence suggests that mast cells play a key role in the development of visceral hypersensitivity through interaction with neurons. The sensitization of corticotropin-releasing factor (CRF) neurons in the hypothalamic paraventricular nucleus (PVN) plays a pivotal role in the pathogenesis of visceral pain. However, the precise mechanism by which mast cells and CRF neurons interact in the PVN in the pathogenesis of visceral hypersensitivity remains elusive. In the present study, we used neonatal maternal separation (MS), an ELS model, and observed that neonatal MS induced visceral hypersensitivity and triggered PVN mast cell activation in adult rats, which was repressed by intra-PVN infusion of the mast cell stabilizer disodium cromoglycate (cromolyn). Wild-type (WT) mice but not mast cell-deficient KitW-sh/W-sh mice that had experienced neonatal MS exhibited chronic visceral hypersensitivity. MS was associated with an increase in the expression of proinflammatory mediators, the number of CRF+ cells and CRF protein in the PVN, which was prevented by intra-PVN infusion of cromolyn. Furthermore, we demonstrated that intra-PVN infusion of the mast degranulator compound 48/80 significantly induced mast cell activation, resulting in proinflammatory mediator release, CRF neuronal sensitization, and visceral hypersensitivity, which was suppressed by cromolyn. Overall, our findings demonstrated that neonatal MS induces the activation of PVN mast cells, which secrete numerous proinflammatory mediators that may participate in neighboring CRF neuronal activity, ultimately directly inducing visceral hypersensitivity in adulthood.


Assuntos
Hiperalgesia , Mastócitos , Privação Materna , Núcleo Hipotalâmico Paraventricular , Estresse Psicológico , Dor Visceral , Animais , Masculino , Camundongos , Ratos , Animais Recém-Nascidos , Modelos Animais de Doenças , Hiperalgesia/etiologia , Hiperalgesia/imunologia , Hiperalgesia/metabolismo , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos Transgênicos , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/imunologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos Sprague-Dawley , Dor Visceral/imunologia , Dor Visceral/metabolismo
12.
Neurochem Int ; 140: 104818, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32758588

RESUMO

Although type 2 diabetes is an important predictor of perioperative neurocognitive disorder (PND), little is currently known about its mechanism of action. Adult male db/db and db/m mice were subjected to four different treatments, including either sham or tibial fracture surgery as well as intraperitoneal injection of vehicle or TAK-242 (the selective inhibitor of TLR4) at 1, 24, and 48 h after surgery. The fear conditioning test was performed to detect cognitive impairment on post-operative day (POD) 3. The hippocampus was collected on POD 1 for western-blots and on POD 3 for western-blots, transmission electron microscopy, and electrophysiological experiments. Toll-like receptor 4 (TLR4) inhibition reversed more profound decline in the freezing behavior of db/db mice on POD 3. The surgery reduced the slope of hippocampal field excitatory postsynaptic potentials, and induced blood-brain barrier (BBB) damage in db/db mice on POD 3. The surgery also increased protein levels of TLR4, tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, albumin, matrix metalloproteinase (MMP)-2, and MMP-9, and decreased protein levels of claudin-5, occludin, tissue inhibitor of matrix metalloproteinase (TIMP)-1, and TIMP-2 in the hippocampus of db/db and db/m mice. These changes were all reversed by TAK-242 treatment. At last, compared with those in post-operative db/m mice, the surgery increased protein levels of TLR4, TNF-α, and IL-1ß, decreased protein levels of claudin-5 and occludin, and sustained the MMP/TIMP imbalance in the hippocampus of db/db mice on POD 3. Our results suggest that TLR4-mediated aggravated hippocampal MMP/TIMP imbalance, BBB disruption, sustained inflammatory cytokine release, and impairment of long-term potentiation play a key role in tibial fracture surgery-induced persistent PND in db/db mice.


Assuntos
Hipocampo/metabolismo , Metaloproteinases da Matriz/metabolismo , Transtornos Neurocognitivos/metabolismo , Complicações Cognitivas Pós-Operatórias/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos Neurocognitivos/etiologia , Complicações Cognitivas Pós-Operatórias/etiologia , Sulfonamidas/farmacologia , Fraturas da Tíbia/metabolismo , Fraturas da Tíbia/cirurgia , Receptor 4 Toll-Like/antagonistas & inibidores
13.
Mediators Inflamm ; 2020: 1921826, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32801993

RESUMO

Neuroinflammation plays a key role in the occurrence and development of neurodegenerative diseases. Microglia, the resident immune cells in the brain, have been recognized to contribute to neuroinflammation. Previous studies have shown that activated mast cells may be involved in surgery-induced neuroinflammation and neuronal apoptosis by using pharmacological methods. This study is aimed at ascertaining the exactly role of mast cells on neuroinflammation with the mast cell-deficient mice. Adult male C57BL6/J wild-type (WT) and mast cell-deficient (C57BL6/J KitWsh/Wsh (Wsh)) mice underwent tibial fracture surgery. Blood-brain barrier (BBB) breakdown, microglial activation, and neuroinflammatory levels were examined at 1 day after surgery. Surgery-induced BBB breakdown, microglial activation, and neuroinflammatory levels were significantly, pharmacologically reduced using a mast cell stabilizer, cromolyn sodium in WT mice (P < 0.05). These results were reproduced with mast cell deficiency. WT mice administered intraventricularly with cromolyn exhibited reduced BBB breakdown, microglial activation, and neuroinflammatory levels versus vehicle (P < 0.05). But there was no effect of cromolyn versus vehicle in Wsh mice, clarifying the specificity of cromolyn on brain mast cells. These findings demonstrated that activated mast cells promote surgery-induced BBB breakdown and neuroinflammation in mice, and open up a new therapeutic target for neuroinflammation-related diseases.


Assuntos
Mastócitos/metabolismo , Microglia/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Neurodegenerativas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
15.
J Neuroinflammation ; 17(1): 217, 2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32698899

RESUMO

BACKGROUND: Microglia, the principal sentinel immune cells of the central nervous system (CNS), play an extensively vital role in neuroinflammation and perioperative neurocognitive disorders (PND). Histamine, a potent mediator of inflammation, can both promote and prevent microglia-related neuroinflammation by activating different histamine receptors. Rat microglia express four histamine receptors (H1R, H2R, H3R, and H4R), among which the histamine 1 and 4 receptors can promote microglia activation, whereas the role and cellular mechanism of the histamine 2 and 3 receptors have not been elucidated. Therefore, we evaluated the effects and potential cellular mechanisms of histamine 2/3 receptors in microglia-mediated inflammation and PND. METHODS: This study investigated the role of histamine 2/3 receptors in microglia-induced inflammation and PND both in vivo and in vitro. In the in vivo experiments, rats were injected with histamine 2/3 receptor agonists in the right lateral ventricle and were then subjected to exploratory laparotomy. In the in vitro experiments, primary microglia were pretreated with histamine 2/3 receptor agonists before stimulation with lipopolysaccharide (LPS). Cognitive function, microglia activation, proinflammatory cytokine production, NF-κb expression, M1/M2 phenotypes, cell migration, and Toll-like receptor-4 (TLR4) expression were assessed. RESULTS: In our study, the histamine 2/3 receptor agonists inhibited exploratory laparotomy- or LPS-induced cognitive decline, microglia activation, proinflammatory cytokine production, NF-κb expression, M1/M2 phenotype transformation, cell migration, and TLR4 expression through the PI3K/AKT/FoxO1 pathway. CONCLUSION: Based on our findings, we conclude that histamine 2/3 receptors ameliorate PND by inhibiting microglia activation through the PI3K/AKT/FoxO1 pathway. Our results highlight histamine 2/3 receptors as potential therapeutic targets to treat neurological conditions associated with PND.


Assuntos
Agonistas dos Receptores Histamínicos/farmacologia , Microglia/efeitos dos fármacos , Complicações Cognitivas Pós-Operatórias/imunologia , Complicações Cognitivas Pós-Operatórias/metabolismo , Envelhecimento , Animais , Método Duplo-Cego , Proteína Forkhead Box O1/efeitos dos fármacos , Injeções Intraventriculares , Masculino , Metilistaminas/farmacologia , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Histamínicos , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia
16.
Mediators Inflamm ; 2020: 8098439, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32184702

RESUMO

Neuroinflammation contributes to or even causes central nervous system (CNS) diseases, and its regulation is thus crucial for brain disorders. Mast cells (MCs) and microglia, two resident immune cells in the brain, together with astrocytes, play critical roles in the progression of neuroinflammation-related diseases. MCs have been demonstrated as one of the fastest responders, and they release prestored and newly synthesized mediators including histamine, ß-tryptase, and heparin. However, temporal changes in MC activation in this inflammation process remain unclear. This study demonstrated that MC activation began at 2 h and peaked at 4 h after lipopolysaccharide (LPS) administration. The number of activated MCs remained elevated until 24 h after LPS administration. In addition, the levels of histamine and ß-tryptase in the hippocampus markedly and rapidly increased within 6 h and remained higher than the baseline level within 24 h after LPS challenge. Furthermore, mast cell-deficient KitW-sh/W-sh mice were used to investigate the effects of MCs on microglial and astrocytic activation and blood-brain barrier (BBB) permeability at 4 h after LPS stimulation. Notably, LPS-induced proinflammatory cytokine secretion, microglial activation, and BBB damage were inhibited in KitW-sh/W-sh mice. However, no detectable astrocytic changes were found in WT and KitW-sh/W-sh mice at 4 h after LPS stimulation. Our findings indicate that MC activation precedes CNS inflammation and suggest that MCs are among the earliest participants in the neuroinflammation-initiating events.


Assuntos
Barreira Hematoencefálica/metabolismo , Lipopolissacarídeos/farmacologia , Mastócitos/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Western Blotting , Ensaio de Imunoadsorção Enzimática , Heparina/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Histamina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Triptases/metabolismo
17.
J Neurochem ; 151(5): 595-607, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31520526

RESUMO

High-mobility group box-1 (HMGB-1) acts as a pro-inflammatory cytokine contributing to the occurrence of many central inflammatory and infectious disorders. Brain mast cells (MCs) are the first responders to peripheral inflammatory stimulation because of their rapid response to external stimuli coupled with their release of preformed and newly synthesized reactive chemicals. Little is known about the involvement of brain MCs in the pro-inflammatory effects of HMGB-1 on the central nervous system (CNS). Thus, we investigated the activation process of MCs by HMGB-1 and explored whether this process is involved in the pro-inflammatory effects of HMGB-1 on the CNS. In this study, we used P815 cells to study the activating role of HMGB-1 on MCs and to explore its potential mechanism in vitro. In an in vivo study, adult male Sprague-Dawley rats received i.c.v. injection of sterile saline or cromoglycate (stabilizer of MCs) 30 min prior to i.p. injection of HMGB-1. Increased levels of tumor necrosis factor and IL-1ß were observed in the P815 cells, as well as in the rats' brains, after HMGB-1 treatment. Pretreatment with the receptor of advanced glycation endproducts (RAGE)-siRNA inhibited the HMGB-1-induced inflammatory process in the P815 cells. Activation of the RAGE/nuclear factor-κB (NF-κB) pathway was observed in both the P815 cells and rats' brains. In addition, HMGB-1 induced the accumulation of brain MCs in the hippocampal CA1 region, and the blood-brain barrier was disrupted. Pretreatment with cromoglycate, a stabilizer of MCs, mitigated these HMGB-1-induced pro-inflammatory processes in rats. These findings indicate that brain MCs are involved in the pro-inflammatory effect of HMGB-1 on the CNS, probably via activating the RAGE/NF-κB pathway.


Assuntos
Encéfalo/imunologia , Proteína HMGB1/imunologia , Mastócitos/imunologia , Transdução de Sinais/imunologia , Animais , Encéfalo/metabolismo , Proteína HMGB1/metabolismo , Masculino , Mastócitos/metabolismo , Camundongos , NF-kappa B/imunologia , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor para Produtos Finais de Glicação Avançada/imunologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo
18.
Front Cell Neurosci ; 13: 191, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31130850

RESUMO

BACKGROUND: The functional aspects of mast cell-microglia interactions are important in neuroinflammation. Our previous studies have demonstrated that mast cell degranulation can directly induce microglia activation. However, the role of mast cells in Lipopolysaccharide (LPS)-induced microglia activation, neuroinflammation and cognitive impairment has not been clarified. METHODS: This study investigated the interaction between brain microglia and mast cells in vivo through site-directed injection of cromolyn into rat right hypothalamus using stereotaxic techniques. Cognitive function was subsequently assessed using trace fear conditioning and Y maze tests. Mast cells in rat brain were stained with toluidine blue and counted using Cell D software. Microglia activation was assessed by Iba1 immunohistochemistry both in rat brain and in mast cell-deficient KitW-sh/W-sh mice. Receptor expression in rat microglia was determined using flow cytometry analysis. Cytokine levels in rat brain tissue and cell supernatant were measured using high-throughput ELISA. Western blotting was used to analyze Cell signaling proteins. RESULTS: In this study, intraperitoneal injection of 1 mg/kg LPS induced mast cell activation in hypothalamus and cognitive dysfunction in rats, and that this process can be repressed by the mast cell stabilizer cromolyn (200 µg). Meanwhile, in mice, LPS IP injection induced significant microglia activation 24 h later in the hypothalamus of wild-type (WT) mice, but had little effect in KitW-sh/W-sh mice. The stabilization of mast cells in rats inhibited LPS-induced microglia activation, inflammatory factors release, and the activation of MAPK, AKT, and NF-κB signaling pathways. We also found that LPS selectively provokes upregulation of H1R, H4R, PAR2, and TLR4, but downregulation of H2R and H3R, in ipsilateral hypothalamus microglia; these effects were partially inhibited by cromolyn. In addition, LPS was also found to induce activation of P815 cells in vitro, consistent with findings from in vivo experiments. These activated P815 cells also induced cytokine release from microglia, which was mediated by the MAPK signaling pathway. CONCLUSION: Taken together, our results demonstrate that stabilization of mast cells can inhibit LPS-induced neuroinflammation and memory impairment, suggesting a novel treatment strategy for neuroinflammation-related diseases.

19.
Mediators Inflamm ; 2019: 8301725, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31011286

RESUMO

Postoperative cognitive dysfunction (POCD) is defined as new cognitive impairment (memory impairment and impaired performance) after surgery, especially in aged patients. Sleep disturbance is a common phenomenon before surgery that has been increasingly thought to affect patient recovery. However, little is known about the functional impact of preoperative sleep disturbance on POCD. Here, we showed that tibial fracture surgery induced cognitive deficit and production of proinflammatory cytokines interleukin-6 (IL-6) and IL-1ß, along with microglia and astrocyte activation, neuronal damage, and blood-brain barrier (BBB) disruption. Preoperative sleep disturbance enhanced the surgery-induced neuroinflammation, neuronal damage, BBB disruption, and memory impairment 24 h after surgery. Taken together, these results demonstrated that preoperative sleep disturbance aggravated postoperative cognitive function in aged mice and the mechanism may be related to central nervous system (CNS) inflammation and neuronal damage.


Assuntos
Transtornos Cognitivos/etiologia , Transtornos Cognitivos/fisiopatologia , Inflamação/etiologia , Animais , Western Blotting , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/fisiopatologia , Ensaio de Imunoadsorção Enzimática , Hipocampo/metabolismo , Imuno-Histoquímica , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Neurônios/citologia , Neurônios/fisiologia , Complicações Pós-Operatórias , Transtornos do Sono-Vigília
20.
Medicine (Baltimore) ; 98(7): e14362, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30762735

RESUMO

BACKGROUND: Appropriate postoperative pain management can improve outcomes in patients with esophageal cancer (EC). OBJECTIVE: To compare different combinations of anesthesia and analgesia techniques in patients with EC undergoing open thoracotomy. METHODS: This randomized, controlled, open-label trial enrolled 100 patients with EC (aged 40-65 years; American Society of Anesthesiologists [ASA] grade I/II) receiving elective surgery at Jiangsu Province Hospital (China) between July 2016 and December 2017. Patients were randomized to 4 groups (n = 25 per group): total intravenous general anesthesia plus patient-controlled intravenous analgesia (TIVA/PCIA); TIVA plus patient-controlled epidural analgesia (TIVA/PCEA); thoracic epidural anesthesia with intravenous general anesthesia plus PCIA (TEA-IVA/PCIA); and TEA-IVA/PCEA (TEA-IVA plus PCEA). Primary outcomes were plasma cortisol level (measured at baseline, 2 h after skin incision, surgery completion, and 24 and 48 h post-surgery) and pain (assessed at 24, 48, and 72 hours post-surgery using a visual analog scale). Secondary outcomes included time to first flatus, hospital stay and treatment costs. Postoperative adverse events (AEs) were analyzed. RESULTS: Baseline and operative characteristics were similar between the 4 groups. Plasma cortisol level increased (P <.05 vs baseline) earlier in the TIVA groups (2 h after skin incision) than in the TEA-IVA groups (24 h after surgery). At 48 hours after surgery, plasma cortisol had returned to baseline levels in the PCEA groups but not in the PCIA groups. VAS pain scores at rest and during coughing were lower in the PCEA groups than in the PCIA groups (P <.05). Compared with the PCIA groups, the PCEA groups had shorter time to first flatus and shorter hospital stay, while use of TEA-IVA lowered the costs of intraoperative anesthesia (P <.05). However, the PCEA groups had a higher incidence of nausea, vomiting, and pruritus. CONCLUSION: Thoracic epidural anesthesia/analgesia can reduce the stress response, improve postoperative recovery and reduce hospital stay and costs for patients with EC.


Assuntos
Neoplasias Esofágicas/cirurgia , Manejo da Dor/métodos , Dor Pós-Operatória/tratamento farmacológico , Procedimentos Cirúrgicos Torácicos/métodos , Adulto , Idoso , Analgesia Epidural/métodos , Analgesia Controlada pelo Paciente/métodos , Analgésicos Opioides/administração & dosagem , Anestesia Epidural/métodos , Anestesia Geral/métodos , China , Terapia Combinada , Feminino , Preços Hospitalares/estatística & dados numéricos , Humanos , Hidrocortisona/sangue , Tempo de Internação/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Medição da Dor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA