Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 270, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37978532

RESUMEN

BACKGROUND: Subarachnoid hemorrhage (SAH) is an uncommon type of potentially fatal stroke. The pathophysiological mechanisms of brain injury remain unclear, which hinders the development of drugs for SAH. We aimed to investigate the pathophysiological mechanisms of SAH and to elucidate the cellular and molecular biological response to SAH-induced injury. METHODS: A cross-species (human and mouse) multiomics approach combining high-throughput data and bioinformatic analysis was used to explore the key pathophysiological processes and cells involved in SAH-induced brain injury. Patient data were collected from the hospital (n = 712). SAH was established in adult male mice via endovascular perforation, and flow cytometry, a bone marrow chimera model, qPCR, and microglial depletion experiments were conducted to explore the origin and chemotaxis mechanism of the immune cells. To investigate cell effects on SAH prognosis, murine neurological function was evaluated based on a modified Garcia score, pole test, and rotarod test. RESULTS: The bioinformatics analysis confirmed that inflammatory and immune responses were the key pathophysiological processes after SAH. Significant increases in the monocyte levels were observed in both the mouse brains and the peripheral blood of patients after SAH. Ly6C-high monocytes originated in the bone marrow, and the skull bone marrow contribute a higher proportion of these monocytes than neutrophils. The mRNA level of Ccl2 was significantly upregulated after SAH and was greater in CD11b-positive than CD11b-negative cells. Microglial depletion, microglial inhibition, and CCL2 blockade reduced the numbers of Ly6C-high monocytes after SAH. With CCR2 antagonization, the neurological function of the mice exhibited a slow recovery. Three days post-SAH, the monocyte-derived dendritic cell (moDC) population had a higher proportion of TNF-α-positive cells and a lower proportion of IL-10-positive cells than the macrophage population. The ratio of moDCs to macrophages was higher on day 3 than on day 5 post-SAH. CONCLUSIONS: Inflammatory and immune responses are significantly involved in SAH-induced brain injury. Ly6C-high monocytes derived from the bone marrow, including the skull bone marrow, infiltrated into mouse brains via CCL2 secreted from microglia. Moreover, Ly6C-high monocytes alleviated neurological dysfunction after SAH.


Asunto(s)
Lesiones Encefálicas , Accidente Cerebrovascular , Hemorragia Subaracnoidea , Humanos , Ratones , Masculino , Animales , Monocitos , Hemorragia Subaracnoidea/complicaciones , Lesiones Encefálicas/etiología , Macrófagos , Ratones Endogámicos C57BL
2.
Int J Mol Sci ; 24(3)2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36768908

RESUMEN

Hydrocephalus has been observed in rats with spontaneous hypertension (SHRs). It has been demonstrated that activation of the oxidative stress related protein retinoic acid receptor alpha (RARα) has neuroprotective impacts. Our investigation aims to determine the potential role and mechanism of RARα in hydrocephalus. The RARα-specific agonist (Am80) and RARα inhibitor (AGN196996) were used to investigate the role of RARα in cerebrospinal fluid (CSF) secretion in the choroid plexus of SHRs. Evaluations of CSF secretion, ventricular volume, Western blotting, and immunofluorescent staining were performed. Hydrocephalus and CSF hypersecretion were identified in SHRs but not in Wistar-Kyoto rats, occurring at the age of 7 weeks. The RARα/MAFB/MSR1 pathway was also activated in SHRs. Therapy with Am80 beginning in week 5 decreased CSF hypersecretion, hydrocephalus development, and pathological changes in choroid plexus alterations by week 7. AGN196996 abolished the effect of Am80. In conclusion, activation of the RARα attenuated CSF hypersecretion to inhibit hydrocephalus development via regulating the MAFB/MSR1 pathway. RARα may act as a possible therapeutic target for hydrocephalus.


Asunto(s)
Hidrocefalia , Hipertensión , Animales , Ratas , Plexo Coroideo/metabolismo , Hidrocefalia/metabolismo , Hipertensión/metabolismo , Factor de Transcripción MafB/metabolismo , Proteínas Oncogénicas/metabolismo , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores Depuradores de Clase A/metabolismo
3.
J Stroke Cerebrovasc Dis ; 31(11): 106803, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36174325

RESUMEN

OBJECTIVES: The Semaphorin 6D (SEMA6D) shows important roles in cell guidance and lipid metabolism, but the effects and mechanisms of SEMA6D on tissue repair, white matter injury and the recovery of neurological function after intracerebral hemorrhage have not been well studied. MATERIALS AND METHODS: In this study, the autologous whole blood injection model of intracerebral hemorrhage was established in C57 male mice. SEMA6D knockout CRISPR utilized in the study. Assessments included neurological score evaluation and immunofluorescence. RESULTS: SEMA6D increased and peaked at 7d after intracerebral hemorrhage, and mainly located in neurons, microglia and astrocytes. SEMA6D knockout CRISPR aggravated neurological function and showed signs of poorer corralling and hematoma resolution, with more compartments of well-established physical barrier and more extensive GFAP positive astrocytic border. Furthermore, SEMA6D can prevent the decrease of NF-H in the peri-hematoma region, while SEMA6D knockout aggravated WMI. CONCLUSIONS: Our study suggested that SEMA6D could influence the recovery of neurological function by regulating the corralling, hematoma compaction and WMI in mice after intracerebral hemorrhage.


Asunto(s)
Hemorragia Cerebral , Hematoma , Semaforinas , Sustancia Blanca , Animales , Masculino , Ratones , Hemorragia Cerebral/diagnóstico por imagen , Hemorragia Cerebral/metabolismo , Hematoma/diagnóstico por imagen , Semaforinas/genética , Semaforinas/metabolismo , Sustancia Blanca/diagnóstico por imagen
4.
J Stroke Cerebrovasc Dis ; 31(9): 106659, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35901587

RESUMEN

BACKGROUND: Polarization of microglia/macrophages toward the pro-inflammatory phenotype is a crucial contributor to neuroinflammation after subarachnoid hemorrhage (SAH). Mer belongs to the TAM receptor tyrosine kinases family, which is known to play a significant role in the resolution of inflammation. However, the effect and mechanism of Mer after SAH remain unclear. In this study, we explored the effect of Mer on modulating the microglia/macrophage phenotype and neuroinflammation and possible potential mechanism after SAH. METHOD: Endovascular perforation model of SAH was performed. There are 3 parts in this study. Firstly, the time course of Mer expression was determined within 72 hours after SAH. Secondly, the effect of Mer downregulation on brain water content, neurological function, and microglial polarization was evaluated at 24 h after SAH. Thirdly, the neuroprotective effects of pharmacological Mer agonist were assessed. RESULT: The expression of Mer increased after SAH, and was prominently localized in microglia/macrophages. Treatment with Mer siRNA increased pro-inflammatory phenotype and decreased anti-inflammatory phenotype of microglia/macrophage, thus resulted in exacerbation of neurological deficits and brain edema after SAH. Mechanistically, the downregulation of Mer inhibited the downstream anti-inflammatory signals, SOCS1/SOCS3, by decreasing phosphorylated STATs. CONCLUSION: Mer is involved in the microglia/macrophage polarization and inflammation resolution after SAH, and that mechanism, at least in part, may contribute to the involvement of the STATs/SOCSs pathway.


Asunto(s)
Lesiones Encefálicas , Hemorragia Subaracnoidea , Animales , Antiinflamatorios/farmacología , Modelos Animales de Enfermedad , Inflamación/tratamiento farmacológico , Macrófagos/metabolismo , Ratones , Microglía/metabolismo , Fenotipo , Transducción de Señal , Hemorragia Subaracnoidea/tratamiento farmacológico
5.
J Neuroinflammation ; 18(1): 210, 2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34530854

RESUMEN

BACKGROUND: Neuroinflammation and oxidative stress plays an important role in the pathogenesis of early brain injury (EBI) after subarachnoid hemorrhage (SAH). This study is the first to show that activation of autophagy protein nuclear receptor binding factor 2 (NRBF2) could reduce endoplasmic reticulum stress (ERS)-associated inflammation and oxidative stress after SAH. METHODS: Male C57BL/6J mice were subjected to endovascular perforation to establish a model of SAH. NRBF2 overexpression adeno-associated virus (AAV), NRBF2 small interfering RNAs (siRNA), lysosomal inhibitor-chloroquine (CQ), and late endosome GTPase Rab7 receptor antagonist-CID1067700 (CID) were used to investigate the role of NRBF2 in EBI after SAH. Neurological tests, brain water content, western blotting and immunofluorescence staining were evaluated. RESULTS: Our study found that the level of NRBF2 was increased after SAH and peaked at 24 h after SAH. In addition, we found that the overexpression of NRBF2 significantly improved neurobehavioral scores and reduced ERS, oxidative stress, and neuroinflammation in SAH, whereas the inhibition of NRBF2 exacerbated these phenotypes. In terms of mechanism, NRBF2 overexpression significantly promoted autophagosome maturation, with the downregulation of CHOP, Romo-1, TXNIP, NLRP3, TNF-α, and IL-1ß expression through interaction with Rab7. The protective effect of NRBF2 on ERS-associated neuroinflammation and oxidative stress after SAH was eliminated by treatment with CQ. Meanwhile, it was also reversed by intraperitoneal injection of CID. Moreover, the MIT domain of NRBF2 was identified as a critical binding site that interacts with Rab7 and thereby promotes autophagosome maturation. CONCLUSION: Our data provide evidence that the autophagy protein NRBF2 has a protective effect on endoplasmic reticulum stress-associated neuroinflammation and oxidative stress by promoting autophagosome maturation through interactions with Rab7 after SAH.


Asunto(s)
Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Estrés Oxidativo/fisiología , Hemorragia Subaracnoidea/metabolismo , Transactivadores/metabolismo , Proteínas de Unión a GTP rab7/metabolismo , Animales , Autofagia/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/prevención & control
6.
J Neuroinflammation ; 18(1): 184, 2021 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-34425835

RESUMEN

BACKGROUND: White matter injury (WMI) is a major neuropathological event associated with intracerebral hemorrhage (ICH). P2X purinoreceptor 4 (P2X4R) is a member of the P2X purine receptor family, which plays a crucial role in regulating WMI and neuroinflammation in central nervous system (CNS) diseases. Our study investigated the role of P2X4R in the WMI and the inflammatory response in mice, as well as the possible mechanism of action after ICH. METHODS: ICH was induced in mice via collagenase injection. Mice were treated with 5-BDBD and ANA-12 to inhibit P2X4R and tropomyosin-related kinase receptor B (TrkB), respectively. Immunostaining and quantitative polymerase chain reaction (qPCR) were performed to detect microglial phenotypes after the inhibition of P2X4R. Western blots (WB) and immunostaining were used to examine WMI and the underlying molecular mechanisms. Cylinder, corner turn, wire hanging, and forelimb placement tests were conducted to evaluate neurobehavioral function. RESULTS: After ICH, the protein levels of P2X4R were upregulated, especially on day 7 after ICH, and were mainly located in the microglia. The inhibition of P2X4R via 5-BDBD promoted neurofunctional recovery after ICH as well as the transformation of the pro-inflammatory microglia induced by ICH into an anti-inflammatory phenotype, and attenuated ICH-induced WMI. Furthermore, we found that TrkB blockage can reverse the protective effects of WMI as well as neuroprotection after 5-BDBD treatment. This result indicates that P2X4R plays a crucial role in regulating WMI and neuroinflammation and that P2X4R inhibition may benefit patients with ICH. CONCLUSIONS: Our results demonstrated that P2X4R contributes to WMI by polarizing microglia into a pro-inflammatory phenotype after ICH. Furthermore, the inhibition of P2X4R promoted pro-inflammatory microglia polarization into an anti-inflammatory phenotype, enhanced brain-derived neurotrophic factor (BDNF) production, and through the BDNF/TrkB pathway, attenuated WMI and improved neurological function. Therefore, the regulation of P2X4R activation may be beneficial for the reducing of ICH-induced brain injury.


Asunto(s)
Hemorragia Cerebral/patología , Microglía/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Sustancia Blanca/efectos de los fármacos , Animales , Benzodiazepinonas/farmacología , Hemorragia Cerebral/metabolismo , Modelos Animales de Enfermedad , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Microglía/metabolismo , Microglía/patología , Proteínas Tirosina Quinasas/metabolismo , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
7.
J Neuroinflammation ; 17(1): 165, 2020 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-32450897

RESUMEN

BACKGROUND: Neuroinflammation is closely associated with the poor prognosis in subarachnoid hemorrhage (SAH) patients. This study was aimed to determine the role of stimulator of IFN genes (STING), an essential regulator to innate immunity, in the context of SAH. METHODS: A total of 344 male C57BL/6 J mice were subjected to endovascular perforation to develop a model of SAH. Selective STING antagonist C-176 and STING agonist CMA were administered at 30 min or 1 h post-modeling separately. To investigate the underlying mechanism, the AMPK inhibitor compound C was administered intracerebroventricularly at 30 min before surgery. Post-SAH assessments included SAH grade, neurological test, brain water content, western blotting, RT-PCR, and immunofluorescence. Oxygenated hemoglobin was introduced into BV2 cells to establish a SAH model in vitro. RESULTS: STING was mainly distributed in microglia, and microglial STING expression was significantly increased after SAH. Administration of C-176 substantially attenuated SAH-induced brain edema and neuronal injury. More importantly, C-176 significantly alleviated both short-term and persistent neurological dysfunction after SAH. Meanwhile, STING agonist CMA remarkably exacerbated neuronal injury and deteriorated neurological impairments. Mechanically, STING activation aggravated neuroinflammation via promoting microglial activation and polarizing into M1 phenotype, evidenced by microglial morphological changes, as well as the increased level of microglial M1 markers including IL-1ß, iNOS, IL-6, TNF-α, MCP-1, and NLRP3 inflammasome, while C-176 conferred a robust anti-inflammatory effect. However, all the mentioned beneficial effects of C-176 including alleviated neuroinflammation, attenuated neuronal injury and the improved neurological function were reversed by AMPK inhibitor compound C. Meanwhile, the critical role of AMPK signal in C-176 mediated anti-inflammatory effect was also confirmed in vitro. CONCLUSION: Microglial STING yielded neuroinflammation after SAH, while pharmacologic inhibition of STING could attenuate SAH-induced inflammatory injury at least partly by activating AMPK signal. These data supported the notion that STING might be a potential therapeutic target for SAH.


Asunto(s)
Inflamación/patología , Proteínas de la Membrana/metabolismo , Hemorragia Subaracnoidea/patología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Modelos Animales de Enfermedad , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Hemorragia Subaracnoidea/inmunología , Hemorragia Subaracnoidea/metabolismo
8.
Cell Mol Neurobiol ; 39(1): 1-10, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30361892

RESUMEN

Germinal matrix hemorrhage (GMH) refers to bleeding that derives from the subependymal (or periventricular) germinal region of the premature brain. GMH can induce severe and irreversible damage attributing to the vulnerable structure of germinal matrix and deleterious circumstances. Molecular mechanisms remain obscure so far. In this review, we summarized the newest preclinical discoveries recent years about GMH to distill a deeper understanding of the neuropathology, and then discuss the potential diagnostic or therapeutic targets among these pathways. GMH studies mostly in recent 5 years were sorted out and the authors generalized the newest discoveries and ideas into four parts of this essay. Intrinsic fragile structure of preterm germinal matrix is the fundamental cause leading to GMH. Many molecules have been found effective in the pathophysiological courses. Some of these molecules like minocycline are suggested active to reduce the damage in animal GMH model. However, researchers are still trying to find efficient diagnostic methods and remedies that are available in preterm infants to rehabilitate or cure the sequent injury. Merits have been obtained in the last several years on molecular pathways of GMH, but more work is required to further unravel the whole pathophysiology.


Asunto(s)
Investigación Biomédica , Hemorragia Cerebral/patología , Animales , Encéfalo/patología , Hemorragia Cerebral/prevención & control , Hemorragia Cerebral/terapia , Humanos , Modelos Biológicos
9.
Stroke Vasc Neurol ; 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38485231

RESUMEN

BACKGROUND: Astrocytes regulate blood-brain barrier (BBB) integrity, whereas subarachnoid haemorrhage (SAH) results in astrocyte dysregulation and BBB disruption. Here, we explored the involvement of tissue inhibitor of matrix metalloprotease-1 (TIMP1) in astrocyte-mediated BBB protection during SAH, along with its underlying mechanisms. METHODS: C57BL/6J mice were used to establish a model of SAH. The effects of TIMP1 on SAH outcomes were analysed by intraperitoneal injection of recombinant mouse TIMP1 protein (rm-TIMP1; 250 µg/kg). The roles of TIMP1 and its effector ß1-integrin on astrocytes were observed by in vivo transduction with astrocyte-targeted adeno-associated virus carrying TIMP1 overexpression plasmid or ß1-integrin RNAi. The molecular mechanisms underlying TIMP1 and ß1-integrin interactions were explored in primary cultured astrocytes stimulated with red blood cells (RBCs). RESULTS: TIMP1 was upregulated after SAH. Administration of rm-TIMP1 mitigated SAH-induced early brain injury (EBI) in male and female mice. TIMP1 was primarily expressed in astrocytes; its overexpression in astrocytes led to increased ß1-integrin expression in astrocytes, along with the preservation of astrocytic endfoot attachment to the endothelium and subsequent recovery of endothelial tight junctions. All of these effects were reversed by the knockdown of ß1-integrin in astrocytes. Molecular analysis showed that TIMP1 overexpression decreased the RBC-induced ubiquitination of ß1-integrin; this effect was partially achieved by inhibiting the interaction between ß1-integrin and the E3 ubiquitin ligase Trim21. CONCLUSION: TIMP1 inhibits the interaction between ß1-integrin and Trim21 in astrocytes, thereby rescuing the ubiquitination of astrocytic ß1-integrin. It subsequently restores interactions between astrocytic endfeet and the endothelium, as well as BBB integrity, eventually mitigating SAH-induced EBI.

10.
Transl Stroke Res ; 2023 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-36913120

RESUMEN

Intracerebral hemorrhage (ICH) is a severe cerebrovascular disease, which impairs patients' white matter even after timely clinical interventions. Indicated by studies in the past decade, ICH-induced white matter injury (WMI) is closely related to neurological deficits; however, its underlying mechanism and pertinent treatment are yet insufficient. We gathered two datasets (GSE24265 and GSE125512), and by taking an intersection among interesting genes identified by weighted gene co-expression networks analysis, we determined target genes after differentially expressing genes in two datasets. Additional single-cell RNA-seq analysis (GSE167593) helped locate the gene in cell types. Furthermore, we established ICH mice models induced by autologous blood or collagenase. Basic medical experiments and diffusion tensor imaging were applied to verify the function of target genes in WMI after ICH. Through intersection and enrichment analysis, gene SLC45A3 was identified as the target one, which plays a key role in the regulation of oligodendrocyte differentiation involving in fatty acid metabolic process, etc. after ICH, and single-cell RNA-seq analysis also shows that it mainly locates in oligodendrocytes. Further experiments verified overexpression of SLC45A3 ameliorated brain injury after ICH. Therefore, SLC45A3 might serve as a candidate therapeutic biomarker for ICH-induced WMI, and overexpression of it may be a potential approach for injury attenuation.

11.
Front Immunol ; 14: 1213510, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37841241

RESUMEN

Thrombocytopenia, characterized by a decrease in platelet count, is commonly observed in sepsis and COVID-19. In sepsis, thrombocytopenia can result from various mechanisms, including impaired platelet production in the bone marrow, accelerated platelet destruction due to increased inflammation, sequestration of platelets in the spleen, immune-mediated platelet destruction, or dysregulated host responses. Similarly, thrombocytopenia has been reported in COVID-19 patients, but the immune-related mechanisms underlying this association remain unclear. Notably, interventions targeting thrombocytopenia have shown potential for improving outcomes in both sepsis and COVID-19 patients. Understanding these mechanisms is crucial for developing effective treatments.


Asunto(s)
Anemia , COVID-19 , Sepsis , Trombocitopenia , Humanos , Trombocitopenia/etiología , Trombocitopenia/terapia , Plaquetas , Recuento de Plaquetas , Sepsis/complicaciones , Sepsis/terapia
12.
J Clin Med ; 12(22)2023 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-38002653

RESUMEN

Aneurysmal subarachnoid hemorrhage (aSAH) frequently causes long-term disability, but predicting outcomes remains challenging. Routine parameters such as demographics, admission status, CT findings, and blood tests can be used to predict aSAH outcomes. The aim of this study was to compare the performance of traditional logistic regression with several machine learning algorithms using readily available indicators and to generate a practical prognostic scorecard based on machine learning. Eighteen routinely available indicators were collected as outcome predictors for individuals with aSAH. Logistic regression (LR), random forest (RF), support vector machines (SVMs), and fully connected neural networks (FCNNs) were compared. A scorecard system was established based on predictor weights. The results show that machine learning models and a scorecard achieved 0.75~0.8 area under the curve (AUC) predicting aSAH outcomes (LR 0.739, RF 0.749, SVM 0.762~0.793, scorecard 0.794). FCNNs performed best (~0.95) but lacked interpretability. The scorecard model used only five factors, generating a clinically useful tool with a total cutoff score of ≥5, indicating poor prognosis. We developed and validated machine learning models proven to predict outcomes more accurately in individuals with aSAH. The parameters found to be the most strongly predictive of outcomes were NLR, lymphocyte count, monocyte count, hypertension status, and SEBES. The scorecard system provides a simplified means of applying predictive analytics at the bedside using a few key indicators.

13.
Biosens Bioelectron ; 236: 115415, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37245459

RESUMEN

Cancer development is driven by diverse processes, and metabolic alterations are among the primary characteristics. Multiscale imaging of aberrant metabolites in cancer is critical to understand the pathology and identify new targets for treatment. While peroxynitrite (ONOO-) is reported being enriched in some tumors and plays important tumorigenic roles, whether it is upregulated in gliomas remains unexplored. To determine the levels and roles of ONOO- in gliomas, efficient tools especially those with desirable blood-brain barrier (BBB) permeability and can realize the in situ imaging of ONOO- in multiscale glioma-related samples are indispensable. Herein, we proposed a strategy of physicochemical property-guided probe design, which resulted in the development of a fluorogenic probe NOSTracker for smartly tracking ONOO-. The probe showed sufficient BBB permeability. ONOO- triggered oxidation of its arylboronate group was automatically followed by a self-immolative cleavage of a fluorescence-masking group, liberating its fluorescence signal. The probe was not only highly sensitive and selective towards ONOO-, but its fluorescence favored desirable stability in various complex biological milieus. Guaranteed by these properties, multiscale imaging of ONOO- was realized in vitro in patient-derived primary glioma cells, ex vivo in clinical glioma slices, and in vivo in the glioma of live mice. The results showed the upregulation of ONOO- in gliomas. Furthermore, a specific ONOO- scavenger uric acid (UA) was pharmaceutically used to downregulate ONOO- in glioma cell lines, and an anti-proliferative effect was observed. These results taken together imply the potential of ONOO- as a biomarker and target for glioma treatment, and propose NOSTracker as a reliable tool to further explore the role of ONOO- in glioma development.


Asunto(s)
Técnicas Biosensibles , Glioma , Ratones , Animales , Barrera Hematoencefálica/metabolismo , Ácido Peroxinitroso , Colorantes Fluorescentes/química , Glioma/diagnóstico por imagen , Glioma/tratamiento farmacológico , Biomarcadores , Imagen Óptica
14.
J Clin Med ; 11(15)2022 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-35955969

RESUMEN

It has been reported that neutrophil extracellular traps (NETs) involve inflammation, coagulation and cell death. Acute lung injury is also considered to be connected with NETs. Deoxyribonuclease I (DNase-1), a clinical medication for the respiratory system, has been reported to degrade cell-free DNA (cfDNA), which is the main component of NETs. Herein, we did research to clarify the therapeutic value of DNase-1 in NPE after SAH. In this model, we found that the treatment of DNase-1 remarkably decreased lung water, neutrophilic infiltration and inflammation. In addition, DNase-1 inhibited the NETs and proinflammatory subtype transition of the macrophages. Moreover, the depletion of neutrophil also verified the role of NETs in NPE. Our results suggest that DNase-1 has the potential to effectively relieve the NPE after SAH and to be a clinical drug for use after SAH.

15.
Front Neurol ; 13: 854671, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35401398

RESUMEN

Aim: Previous studies have proved that inhibiting inflammasome activation provides neuroprotection against early brain injury (EBI) after subarachnoid hemorrhage (SAH), which is mainly focused on the microglial inflammatory response, but the potential role of neuronal inflammasome activation in EBI has not been clearly identified. This study examined whether the pannexin-1 channel inhibitor probenecid could reduce EBI after SAH by inhibiting neuronal AIM2 inflammasome activation. Methods: There are in vivo and in vitro parts in this study. First, adult male SD rats were subjected to the endovascular perforation mode of SAH. The time course of pannexin-1 and AIM2 expressions were determined after SAH in 72 h. Brain water content, neurological function, AIM2 inflammasome activation, and inflammatory response were evaluated at 24 h after SAH in sham, SAH, and SAH + probenecid groups. In the in vitro part, HT22 cell treated with hemin was applied to mimic SAH. The expression of AIM2 inflammasome was detected by immunofluorescence staining. Neuronal death and mitochondrial dysfunction were determined by the LDH assay kit and JC-1 staining. Results: The pannexin-1 and AIM2 protein levels were upregulated after SAH. Pannexin-1 channel inhibitor probenecid attenuated brain edema and improved neurological dysfunction by reducing AIM2 inflammasome activation and reactive oxygen species (ROS) generation after SAH in rats. Treating HT22 cells with hemin for 12 h resulted in AIM2 and caspase-1 upregulation and increased mitochondrial dysfunction and neuronal cell death. Probenecid significantly attenuated the hemin-induced AIM2 inflammasome activation and neuronal death. Conclusions: AIM2 inflammasome is activated in neurons after SAH. Pharmacological inhibition of the pannexin-1 channel by probenecid attenuated SAH-induced AIM2 inflammasome activation and EBI in vivo and hemin-induced AIM2 inflammasome activation and neuronal death in vitro.

16.
J Clin Med ; 11(13)2022 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-35807002

RESUMEN

DJ-1 has been shown to play essential roles in neuronal protection and anti-inflammation in nervous system diseases. This study aimed to explore how DJ-1 regulates neuroinflammation after traumatic spinal cord injury (t-SCI). The rat model of spinal cord injury was established by the clamping method. The Basso, Beattie, Bresnahan (BBB) score and the inclined plane test (IPT) were used to evaluate neurological function. Western blot was then applied to test the levels of DJ-1, NLRP3, SOCS1, and related proinflammatory factors (cleaved caspase 1, IL-1ß and IL-18); ROS level was also examined. The distribution of DJ-1 was assessed by immunofluorescence staining (IF). BSCB integrity was assessed by the level of MMP-9 and tight junction proteins (Claudin-5, Occludin and ZO-1). We found that DJ-1 became significantly elevated after t-SCI and was mainly located in neurons. Knockdown of DJ-1 with specific siRNA aggravated NLRP3 inflammasome-related neuroinflammation and strengthened the disruption of BSCB integrity. However, the upregulation of DJ-1 by Sodium benzoate (SB) reversed these effects and improved neurological function. Furthermore, SOCS1-siRNA attenuated the neuroprotective effects of DJ-1 and increased the ROS, Rac1 and NLRP3. In conclusion, DJ-1 may alleviate neuroinflammation and the related BSCB destruction after t-SCI by suppressing NLRP3 inflammasome activation by SOCS1/Rac1/ROS pathways. DJ-1 shows potential as a feasible target for mediating neuroinflammation after t-SCI.

17.
Front Immunol ; 13: 919444, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36189326

RESUMEN

Aim: The complement cascade is activated and may play an important pathophysiologic role in brain injury after experimental intracerebral hemorrhage (ICH). However, the exact mechanism of specific complement components has not been well studied. This study determined the role of complement C1q/C3-CR3 signaling in brain injury after ICH in mice. The effect of minocycline on C1q/C3-CR3 signaling-induced brain damage was also examined. Methods: There were three parts to the study. First, the natural time course of C1q and CR3 expression was determined within 7 days after ICH. Second, mice had an ICH with CR3 agonists, LA-1 or vehicle. Behavioral score, neuronal cell death, hematoma volume, and oxidative stress response were assessed at 7 days after ICH. Third, the effect of minocycline on C1q/C3-CR3 signaling and brain damage was examined. Results: There were increased numbers of C1q-positive and CR3-positive cells after ICH. Almost all perihematomal C1q-positive and CR3-positive cells were microglia/macrophages. CR3 agonist LA-1 aggravated neurological dysfunction, neuronal cell death, and oxidative stress response on day 7 after ICH, as well as enhancing the expression of the CD163/HO-1 pathway and accelerating hematoma resolution. Minocycline treatment exerted neuroprotective effects on brain injury following ICH, partly due to the inhibition of C1q/C3-CR3 signaling, and that could be reversed by LA-1. Conclusions: The complement C1q/C3-CR3 signaling is upregulated after ICH. The activation of C1q/C3-CR3 signaling by LA-1 aggravates brain injury following ICH. The neuroprotection of minocycline, at least partly, is involved with the repression of the C1q/C3-CR3 signaling pathway.


Asunto(s)
Lesiones Encefálicas , Fármacos Neuroprotectores , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/etiología , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Complemento C1q , Hematoma , Ratones , Minociclina/farmacología , Fármacos Neuroprotectores/farmacología , Transducción de Señal
18.
Front Immunol ; 13: 962760, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36016932

RESUMEN

Background: The relationship between neutrophil to lymphocyte ratio (NLR) and poor outcome of aneurysmal subarachnoid hemorrhage (aSAH) is controversial. We aim to evaluate the relationship between NLR on admission and the poor outcome after aSAH. Method: Part I: Retrospective analysis of aSAH patients in our center. Baseline characteristics of patients were collected and compared. Multivariate analysis was used to evaluate parameters independently related to poor outcome. Receiver operating characteristic (ROC) curve analysis was used to determine the best cut-off value of NLR. Part II: Systematic review and meta-analysis of relevant literature. Related literature was selected through the database. The pooled odds ratio (OR) and corresponding 95% confidence interval (CI) were calculated to evaluate the correlation between NLR and outcome measures. Results: Part I: A total of 240 patients with aSAH were enrolled, and 52 patients had a poor outcome. Patients with poor outcome at 3 months had a higher admission NLR, Hunt & Hess score, Barrow Neurological Institute (BNI) scale score, Subarachnoid Hemorrhage Early Brain Edema Score (SEBES), and proportion of hypertension history. After adjustment, NLR at admission remained an independent predictor of poor outcome in aSAH patients (OR 0.76, 95% CI 0.69-0.83; P < 0.001). The best cut-off value of NLR in ROC analysis is 12.03 (area under the curve 0.805, 95% CI 0.735 - 0.875; P < 0.001). Part II: A total of 16 literature were included. Pooled results showed that elevated NLR was significantly associated with poor outcome (OR 1.31, 95% CI 1.14-1.49; P < 0.0001) and delayed cerebral ischemia (DCI) occurrence (OR 1.32, 95% CI 1.11-1.56; P = 0.002). The results are more reliable in large sample sizes, low NLR cut-off value, multicenter, or prospective studies. Conclusion: Elevated NLR is an independent predictor of poor outcome and DCI occurrence in aSAH.


Asunto(s)
Isquemia Encefálica , Hemorragia Subaracnoidea , Humanos , Linfocitos , Estudios Multicéntricos como Asunto , Neutrófilos , Estudios Prospectivos , Estudios Retrospectivos
19.
Transl Stroke Res ; 13(1): 112-131, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33852132

RESUMEN

Neuroinflammation is closely associated with poor prognosis in patients with subarachnoid hemorrhage (SAH). The purpose of this study was to investigate the role of neutrophil extracellular traps (NETs), which are important regulators of sterile inflammation, in SAH. In this study, markers of NET formation, quantified by the level of citrullinated histone H3 (CitH3), were significantly increased after SAH and correlated with SAH severity. CitH3 peaked at 12 h in peripheral blood and at 24 h in the brain. Administration of the peptidyl arginine deiminase 4 (PAD4) selective antagonist GSK484 substantially attenuated SAH-induced brain edema and neuronal injury. Moreover, the benefit of NET inhibition was also confirmed by DNAse I treatment and neutrophil depletion. Mechanistically, NETs markedly exacerbated microglial inflammation in vitro. NET formation aggravated neuroinflammation by promoting microglial activation and increased the levels of TNF-α, IL-1ß, and IL-6, while inhibiting NETs demonstrated anti-inflammatory effects by decreasing the levels of these proinflammatory factors. Moreover, neurogenic pulmonary edema (NPE), a severe nonneurological complication after SAH, is associated with a high level of NET formation. However, GSK484 effectively inhibited the formation of NETs in the lungs of NPE mice, thereby preventing the diffusion of neutrophilic infiltration and attenuating the swelling of the alveolar interstitium. In conclusion, NETs promoted neuroinflammation after SAH, while pharmacological inhibition of PAD4-NETs could reduce the inflammatory damage caused by SAH. These results supported the idea that NETs might be potential therapeutic targets for SAH.


Asunto(s)
Lesiones Encefálicas , Trampas Extracelulares , Hemorragia Subaracnoidea , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Humanos , Inflamación/complicaciones , Ratones , Microglía , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/tratamiento farmacológico
20.
Oxid Med Cell Longev ; 2021: 9931742, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336122

RESUMEN

Neutrophil extracellular traps (NETs) are complexes of decondensed DNA fibers and antimicrobial peptides that are released by neutrophils and play important roles in many noninfectious diseases, such as cystic fibrosis, systemic lupus erythematosus, diabetes, and cancer. Recently, the formation of NETs has been detected in many central nervous system diseases and is thought to play different roles in the occurrence and development of these diseases. Researchers have detected NETs in acute ischemic stroke thrombi, and these NETs are thought to promote coagulation and thrombosis. NETs in ischemic brain parenchyma were identified as the cause of secondary nerve damage. High levels of NETs were also detected in grade IV glioma tissues, where NETs were involved in the proliferation and invasion of glioma cells by activating a signaling pathway. Extracellular web-like structures have also recently been observed in mice with traumatic brain injury (TBI), and it was hypothesized that NETs contribute to the development of edema after TBI. This article reviews the effect of NETs on multiple diseases that affect the CNS and explores their clinical application prospects.


Asunto(s)
Enfermedades del Sistema Nervioso Central/fisiopatología , Trampas Extracelulares/genética , Neutrófilos/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA