Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 271
Filtrar
1.
Immun Ageing ; 21(1): 29, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730291

RESUMEN

BACKGROUND: Quercetin is a flavonol compound widely distributed in plants that possesses diverse biological properties, including antioxidative, anti-inflammatory, anticancer, neuroprotective and senescent cell-clearing activities. It has been shown to effectively alleviate neurodegenerative diseases and enhance cognitive functions in various models. The immune system has been implicated in the regulation of brain function and cognitive abilities. However, it remains unclear whether quercetin enhances cognitive functions by interacting with the immune system. RESULTS: In this study, middle-aged female mice were administered quercetin via tail vein injection. Quercetin increased the proportion of NK cells, without affecting T or B cells, and improved cognitive performance. Depletion of NK cells significantly reduces cognitive ability in mice. RNA-seq analysis revealed that quercetin modulated the RNA profile of hippocampal tissues in aging animals towards a more youthful state. In vitro, quercetin significantly inhibited the differentiation of Lin-CD117+ hematopoietic stem cells into NK cells. Furthermore, quercetin promoted the proportion and maturation of NK cells by binding to the MYH9 protein. CONCLUSIONS: In summary, our findings suggest that quercetin promotes the proportion and maturation of NK cells by binding to the MYH9 protein, thereby improving cognitive performance in middle-aged mice.

5.
Foods ; 13(8)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38672840

RESUMEN

We investigated fourteen antibiotics, three illegal drugs, and two toxic elements in commercially available gastropods from southeast China. The data revealed high detection frequencies (DFs) for florfenicol (61.32%), florfenicol amine (47.33%), and thiamphenicol (39.88%), with maximum concentrations of 1110, 2222, and 136 µg/kg wet weight (ww), respectively. The DFs of illegal drugs were 3.54% for leucomalachite green and 0.3% for chloramphenicol. The average levels of Cd and As were 1.17 and 6.12 mg/kg ww, respectively. All chemicals presented diverse DFs in different sampling months. The highest DFs of florfenicol, florfenicol amine, and thiamphenicol were in July. The health risk assessment showed that targeted hazard quotients (THQs) of antibiotics, Cd, and As for children, teens, and adults were all less than one. Notably, the toxic elements (Cd and As) were identified as the primary health risk in gastropods, contributing to over 90% of the total THQs.

6.
Brain Res ; 1832: 148864, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38484924

RESUMEN

AIMS: Lysophosphatidylcholine acyltransferase 3 (LPCAT3) is known to play a pivotal role in lipid metabolism, but its role in the early brain injury (EBI) following subarachnoid hemorrhage (SAH) remains unclear. This study provides insights into LPCAT3 expression alterations and functional implications in EBI following SAH. METHODS: SAH models of adult male Sprague-Dawley (SD) rats were established by intravascular perforation. Lentivirus vectors were administered by intracerebroventricular injection (i.c.v.) to either induce LPCAT3 overexpression or knockdown 14 days before SAH induction. Western blot, immunofluorescence, Nissl staining, MDA detection, ROS detection, iron content detection, and short-term and long-term neurobehavioral tests were performed to investigate the effects of regulated-LPCAT3 after SAH. RESULTS: LPCAT3 levels were found to be significantly elevated in SAH. Suppression of LPCAT3 expression via shRNA improved oxidative stress, reduced brain edema, alleviated behavioral and cognitive deficits following SAH and decreased neuronal death, while upregulating LPCAT3 expression showed opposing effects. CONCLUSION: LPCAT3 is involved in SAH-induced EBI and associated with ferroptosis. Our findings provide a referential basis for potential therapeutic interventions aimed at alleviating EBI following SAH.


Asunto(s)
Lesiones Encefálicas , Ferroptosis , Hemorragia Subaracnoidea , Ratas , Masculino , Animales , Ratas Sprague-Dawley , Encéfalo/metabolismo , Hemorragia Subaracnoidea/metabolismo , Lesiones Encefálicas/metabolismo , Apoptosis
7.
CNS Neurosci Ther ; 30(3): e14679, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38528842

RESUMEN

AIMS: Intracerebral hemorrhage (ICH) is a disease with high rates of disability and mortality. The role of epidermal growth factor receptor 1 (ERBB1) in ICH was elucidated in this study. METHODS: ICH model was constructed by injecting autologous arterial blood into the right basal ganglia. The protein level of ERBB1 was detected by western blot analysis. To up- and downregulation of ERBB1 in rats, intraventricular injection of a lentivirus overexpression vector of ERBB1 and AG1478 (a specific inhibitor of ERBB1) was used. The cell apoptosis, neuronal loss, and pro-inflammatory cytokines were assessed by TUNEL, Nissl staining, and ELISA. Meanwhile, behavioral cognitive impairment of ICH rats was evaluated after ERBB1-targeted interventions. RESULTS: ERBB1 increased significantly in brain tissue of ICH rats. Overexpression of ERBB1 remarkably reduced cell apoptosis and neuronal loss induced by ICH, as well as pro-inflammatory cytokines and oxidative stress. Meanwhile, the behavioral and cognitive impairment of ICH rats were alleviated after upregulation of ERBB1; however, the secondary brain injury (SBI) was aggravated by AG1478 treatment. Furthermore, the upregulation of PLC-γ and PKC in ICH rats was reversed by AG1478 treatment. CONCLUSIONS: ERBB1 can improve SBI and has a neuroprotective effect in experimental ICH rats via PLC-γ/PKC pathway.


Asunto(s)
Lesiones Encefálicas , Hemorragia Cerebral , Receptores ErbB , Quinazolinas , Animales , Ratas , Apoptosis , Lesiones Encefálicas/metabolismo , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/metabolismo , Citocinas/metabolismo , Fosfolipasa C gamma/metabolismo , Ratas Sprague-Dawley , Tirfostinos , Receptores ErbB/metabolismo , Proteína Quinasa C/metabolismo
8.
Cancer Sci ; 115(5): 1388-1404, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38480275

RESUMEN

Glioblastoma (GBM) is the most common malignant diffuse glioma of the brain. Although immunotherapy with immune checkpoint inhibitors (ICIs), such as programmed cell death protein (PD)-1/PD ligand-1 inhibitors, has revolutionized the treatment of several cancers, the clinical benefit in GBM patients has been limited. Lymphocyte-activation gene 3 (LAG-3) binding to human leukocyte antigen-II (HLA-II) plays an essential role in triggering CD4+ T cell exhaustion and could interfere with the efficiency of anti-PD-1 treatment; however, the value of LAG-3-HLA-II interactions in ICI immunotherapy for GBM patients has not yet been analyzed. Therefore, we aimed to investigate the expression and regulation of HLA-II in human GBM samples and the correlation with LAG-3+CD4+ T cell infiltration. Human leukocyte antigen-II was highly expressed in GBM and correlated with increased LAG-3+CD4+ T cell infiltration in the stroma. Additionally, HLA-IIHighLAG-3High was associated with worse patient survival. Increased interleukin-10 (IL-10) expression was observed in GBM, which was correlated with high levels of HLA-II and LAG-3+ T cell infiltration in stroma. HLA-IIHighIL-10High GBM associated with LAG-3+ T cells infiltration synergistically showed shorter overall survival in patients. Combined anti-LAG-3 and anti-IL-10 treatment inhibited tumor growth in a mouse brain GL261 tumor model. In vitro, CD68+ macrophages upregulated HLA-II expression in GBM cells through tumor necrosis factor-α (TNF-α). Blocking TNF-α-dependent inflammation inhibited tumor growth in a mouse GBM model. In summary, T cell-tumor cell interactions, such as LAG-3-HLA-II, could confer an immunosuppressive environment in human GBM, leading to poor prognosis in patients. Therefore, targeting the LAG-3-HLA-II interaction could be beneficial in ICI immunotherapy to improve the clinical outcome of GBM patients.


Asunto(s)
Antígenos CD , Neoplasias Encefálicas , Linfocitos T CD4-Positivos , Glioblastoma , Proteína del Gen 3 de Activación de Linfocitos , Regulación hacia Arriba , Glioblastoma/inmunología , Glioblastoma/patología , Glioblastoma/metabolismo , Humanos , Animales , Ratones , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Antígenos CD/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Femenino , Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase II/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Interleucina-10/metabolismo , Microambiente Tumoral/inmunología , Persona de Mediana Edad
9.
CNS Neurosci Ther ; 30(2): e14400, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37614198

RESUMEN

AIM: Stromal cell-derived factor-1 (SDF-1) and CXC chemokine receptor 4 (CXCR4) have a substantial role in neuronal formation, differentiation, remodeling, and maturation and participate in multiple physiological and pathological events. In this study, we investigated the role of SDF-1/CXCR4 in neural functional injury and neuroprotection after intracerebral hemorrhage (ICH). METHODS: Western blot, immunofluorescence and immunoprecipitation were used to detect SDF-1/CXCR4 expression and combination respectively after ICH. TUNEL staining, Lactate dehydrogenase assay, Reactive oxygen species assay, and Enzyme-linked immunosorbent assay to study neuronal damage; Brain water content to assay brain edema, Neurological scores to assess short-term neurological deficits. Pharmacological inhibition and genetic intervention of SDF-1/CXCR4 signaling were also used in this study. RESULTS: ICH induced upregulation of SDF-1/CXCR4 and increased their complex formation, whereas AMD3100 significantly reduced it. The levels of TNF-α and IL-1ß were significantly reduced after AMD3100 treatment. Additionally, AMD3100 treatment can alleviate neurobehavioral dysfunction of ICH rats. Conversely, simultaneous SDF-1/CXCR4 overexpression induced the opposite effect. Moreover, immunoprecipitation confirmed that SDF-1/CXCR4 combined to initiate neurodamage effects. CONCLUSION: This study indicated that inhibition of SDF-1/CXCR4 complex formation can rescue the inflammatory response and alleviate neurobehavioral dysfunction after ICH. SDF-1/CXCR4 may have applications as a therapeutic target after ICH.


Asunto(s)
Bencilaminas , Ciclamas , Neuroprotección , Receptores CXCR4 , Animales , Ratas , Hemorragia Cerebral , Quimiocina CXCL12/metabolismo , Regulación hacia Abajo , Células del Estroma/metabolismo
10.
Neurosci Lett ; 818: 137553, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37949291

RESUMEN

Disruption of the blood-brain barrier (BBB) following cerebral ischemia-reperfusion injury (CIRI) is a major factor in the pathophysiology of stroke. Endothelial cell-cell communication is essential for maintaining BBB integrity. By analyzing GSE227651 data, we found that a decrease in endothelial cell-cell communication mediated by Sema3/Nrp1 may be due to the downregulation of Nrp1 transcription, which could contribute to BBB breakdown after CIRI. We confirmed this hypothesis by using western blot analysis to show a reduction in Nrp1 protein levels in penumbra endothelial cells after CIRI in mice. We then overexpressed Nrp1 specifically in brain endothelial cells using adeno-associated virus in mice. Furthermore, Nrp1 overexpression had a protective effect on BBB integrity, as evidenced by a decrease in IgG and albumin leakage caused by CIRI in mice. Finally, we found that Nrp1 overexpression also reduced brain cell death and neurological deficits induced by cerebral ischemia-reperfusion in mice. Our findings suggest that Nrp1 downregulation may be a key factor in the breakdown of endothelial cell-cell communication and subsequent BBB disruption following CIRI. Targeting Nrp1-mediated pathways may be a promising approach for mitigating BBB damage and alleviating neurological consequences in stroke patients.


Asunto(s)
Barrera Hematoencefálica , Isquemia Encefálica , Daño por Reperfusión , Accidente Cerebrovascular , Animales , Humanos , Ratones , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Isquemia Encefálica/metabolismo , Infarto Cerebral/metabolismo , Regulación hacia Abajo , Células Endoteliales/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Neuropilina-1/metabolismo , Reperfusión/efectos adversos , Daño por Reperfusión/metabolismo
11.
Ecotoxicol Environ Saf ; 270: 115868, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38142590

RESUMEN

Ochratoxin A (OTA) is a mycotoxin commonly found in several food commodities worldwide with potential nephrotoxic, hepatotoxic and carcinogenic effects. We previously showed for the first time that OTA treatment enhanced glycolysis in human gastric epithelium (GES-1) cells in vitro. Here, we found that OTA exposure activated inflammatory responses, evidenced by increasing of NF-κB signaling pathway-related protein (p-p65 and p-IκBα) expressions and elevating of inflammatory cytokine (IL-1ß and IL-6) mRNA expressions in GES-1 cells. To elucidate the role of glycolysis in inflammatory effects triggered by OTA, we pretreated GES-1 cells with glycolysis inhibitor (2-deoxy-D-glucose, 2-DG) before OTA exposure. The result showed that 2-DG reduced the protein expressions of p-p65 and p-IκBα and alleviated the mRNA expressions of inflammatory cytokines in OTA-treated GES-1 cells. Furthermore, OTA activated the mTOR/HIF-1α pathway by increasing the protein expressions of p-mTOR, p-eIF4E and HIF-1α, and inhibition of mTOR with rapamycin or silencing HIF-1α with siRNA significantly attenuated OTA-enhanced glycolysis by reducing glycolysis related genes and thereby decreasing inflammatory effects of GES-1 cells. These results demonstrate that OTA activates inflammatory responses in GES-1 cells and this is controlled by mTOR/HIF-1α pathway-mediated glycolysis enhancement. Our findings provide a novel mechanistic view into OTA-induced gastric cytotoxicity.


Asunto(s)
Ocratoxinas , Transducción de Señal , Serina-Treonina Quinasas TOR , Humanos , Inhibidor NF-kappaB alfa , Línea Celular , Serina-Treonina Quinasas TOR/genética , Glucólisis , ARN Mensajero , Epitelio
12.
BMC Genomics ; 24(1): 795, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38129780

RESUMEN

BACKGROUND: GDSL esterase/lipases (GELPs) play important roles in plant growth, development, and response to biotic and abiotic stresses. Presently, an extensive and in-depth analysis of GELP family genes in cotton is still not clear enough, which greatly limits the further understanding of cotton GELP function and regulatory mechanism. RESULTS: A total of 389 GELP family genes were identified in three cotton species of Gossypium hirsutum (193), G. arboreum (97), and G. raimondii (99). These GELPs could be classified into three groups and eight subgroups, with the GELPs in same group to have similar gene structures and conserved motifs. Evolutionary event analysis showed that the GELP family genes tend to be diversified at the spatial dimension and certain conservative at the time dimension, with a trend of potential continuous expansion in the future. The orthologous or paralogous GELPs among different genomes/subgenomes indicated the inheritance from genome-wide duplication during polyploidization, and the paralogous GELPs were derived from chromosomal segment duplication or tandem replication. GELP genes in the A/D subgenome underwent at least three large-scale replication events in the evolutionary process during the period of 0.6-3.2 MYA, with two large-scale evolutionary events between 0.6-1.8 MYA that were associated with tetraploidization, and the large-scale duplication between 2.6-9.1 MYA that occurred during diploidization. The cotton GELPs indicated diverse expression patterns in tissue development, ovule and fiber growth, and in response to biotic and abiotic stresses, combining the existing cis-elements in the promoter regions, suggesting the GELPs involvements of functions to be diversification and of the mechanisms to be a hormone-mediated manner. CONCLUSIONS: Our results provide a systematic and comprehensive understanding the function and regulatory mechanism of cotton GELP family, and offer an effective reference for in-depth genetic improvement utilization of cotton GELPs.


Asunto(s)
Esterasas , Lipasa , Esterasas/genética , Esterasas/metabolismo , Lipasa/genética , Lipasa/metabolismo , Gossypium/metabolismo , Genoma de Planta , Duplicación de Gen , Biología Computacional , Regulación de la Expresión Génica de las Plantas , Filogenia , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo
13.
Neuroscience ; 535: 108-123, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37913857

RESUMEN

Subarachnoid Hemorrhage (SAH) is a cerebrovascular disorder that has been found to have severe consequences, including a high mortality and disability rate. Research has indicated that neuronal death, particularly apoptosis, plays a major role in the neurological impairment that follows SAH. RNA-binding protein Pum2 can interfere with translation or other biological functions by connecting to the UGUAHAUA sequence on RNA. Noncoding RNA activated by DNA damage (Norad) contains some Pum2 recognition sequences, which may bind to Pum2 protein and affect its capacity to attach to target mRNA. The time course expression of Norad and Pum2 after SAH is analyzed by establishing a mouse SAH model. Subsequently, the purpose of this study is to investigate the potential role and mechanism of the Norad-Pum2 axis after SAH using lentivirus overexpression of Pum2 and knockdown of Norad. Analysis of Pum2 and Norad levels reveal that the former is significantly reduce and the latter is significantly increased in the SAH group compared to the sham group. Subsequent overexpression of Pum2 and Norad knockdown is found to reduce SAH-induced oxidative stress, neuronal apoptosis, and ultimately improve behavioral and cognitive changes in SAH mice. Our study indicates that Norad-Pum2 acts as a neuromodulator in SAH, and that by increasing Pum2 and decreasing Norad levels, SAH-induced neuronal apoptosis can be reduced and neurological deficits alleviated. Consequently, Norad-Pum2 may be a promising therapeutic target for SAH.


Asunto(s)
Hemorragia Subaracnoidea , Ratones , Animales , Hemorragia Subaracnoidea/metabolismo , Neuroprotección , Modelos Animales de Enfermedad , Apoptosis/fisiología , ARN no Traducido , Proteínas de Unión al ARN/genética
14.
Exp Neurol ; 368: 114508, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37598879

RESUMEN

BACKGROUND: Intracerebral hemorrhage (ICH) is one of the stroke subtypes with the highest mortality. Secondary brain injury is associated with neurological dysfunction and poor prognosis after ICH. Caveolin-1 (CAV1) is the key protein of Caveolae. Previous studies have shown that CAV1 plays an important role in central nervous system diseases, and pointed out that in a collagenase-induced ICH model in vivo, CAV1 is associated with neuroinflammatory activation and poor neurological prognosis. In this study, we explore the role and the molecular mechanism of CAV1 in brain injury via a rat autologous whole blood injection model and an in vitro model of ICH. METHODS: Adult male Sprague-Dawley rats ICH model was induced through autologous whole blood injecting into the right basal ganglia. The changes in protein levels of CAV1 in brain tissues of ICH rats were detected by western blot analysis. The immunofluorescent staining was used to explore the changes of CAV1 in microglia/macrophages (Iba1+ cells). Lentivirus vectors were administered by intracerebroventricular injection to induce CAV1 overexpression and knockdown respectively. The western blot analysis, immunofluorescence staining, enzyme-linked immunosorbent assay, terminal deoxynucleotidyl transferase dUTP nick end labeling and Nissl staining were performed to explore the role of CAV1 in secondary brain injury after ICH. Meanwhile, the rotarod test, foot fault test, adhesive-removal test, and Modified Garcia Test, as well as Morris Water Maze test, were performed to evaluate the behavioral cognitive impairment of ICH rats after genetic intervention. Additionally, BV-2 cells treated with oxygen hemoglobin for 24 h, were used as an in vitro model of ICH in this study to explore the molecular mechanism of CAV1 in brain injury; we performed western blot analysis after precise regulation of CAV1 in BV2 cells to observe changes in protein levels and phosphorylated levels of C-Src, IKK-ß, and NF-κB. RESULTS: The expression of CAV1 in microglia/macrophages (Iba1+ cells) was elevated and reached the peak at 24 h after ICH. CAV1 knockdown ameliorated ICH-induced neurological deficits, while CAV1 overexpression significantly worsened neurological dysfunction of ICH rats. CAV1 knockdown attenuated cellular apoptosis and promoted neuronal survival in brain tissues of ICH rats, while the ICH rats with CAV1 overexpression presented more cellular apoptosis and neuronal loss. Meanwhile, CAV1 knockdown inhibited the microglia activation and neuroinflammatory response, while CAV1 overexpression abolished these effects and aggravated neuroinflammation in brain tissues of ICH rats. Additionally, by inducing to CAV1 knockdown in BV2 cells in an in vitro model of ICH, the levels of p-C-Src, CAV-1, p-CAV-1, and p-IKK-ß in cytoplasm and the level of NF-κB p65 in nucleus of BV2 cells were significantly decreased, while they were increased by inducing to CAV1 overexpression. CONCLUSIONS: Our research revealed CAV1 aggravated neurological dysfunction in a rat ICH model. CAV1 knockdown exerted neuroprotective effect by suppressing microglia activation and neuroinflammation after ICH might via the C-Src/CAV1/IKK-ß/NF-κB signaling pathway.


Asunto(s)
Lesiones Encefálicas , Neoplasias Encefálicas , Animales , Masculino , Ratas , Caveolina 1 , Hemorragia Cerebral/complicaciones , Enfermedades Neuroinflamatorias , FN-kappa B , Ratas Sprague-Dawley
15.
Brain Res ; 1820: 148556, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37648093

RESUMEN

BACKGROUND: Ferroptosis is an important therapeutic target to alleviate early brain injury (EBI) after subarachnoid hemorrhage (SAH), yet the mechanism of neuronal ferroptosis after SAH remains unclear. System xc- dysfunction is one of the key pathways to induce ferroptosis. System xc- activity is mainly regulated by the expression of xCT. This study was designed to investigate the effect of xCT expression and System xc- activity on ferroptosis and EBI in an experimental SAH model both in vitro and in vivo. METHODS: SAH was induced in adult male Sprague-Dawley rats by injecting autologous blood into the prechiasmatic cistern. Primary neurons treated with oxyhemoglobin (10 µM) were used to mimic SAH in vitro. Plasmid transfection was used to induce xCT overexpression. Western blotting, immunofluorescence staining, measurement of cystine uptake, enzyme-linked immunosorbent assay, transmission electron microscopy, Nissl staining, and a series of neurobehavioral tests were conducted to explore the role of xCT and System xc- activity in ferroptosis and EBI after SAH. RESULTS: We found that System xc- dysfunction induced ferroptosis and exacerbated EBI after SAH in rats. xCT deficiency after SAH resulted in System xc- dysfunction, weakened neuronal antioxidant capacity and activated neuronal ferroptosis. xCT overexpression improved neuronal antioxidant capacity and inhibited neuronal ferroptosis by restoring System xc- activity. Rats with xCT overexpression after SAH presented with attenuated brain edema and inflammation, increased neuronal survival, and ameliorated neurological deficits. CONCLUSIONS: Our study revealed that restoring System xc- activity by xCT overexpression inhibited neuronal ferroptosis and EBI and improved neurological deficits after SAH.

16.
Int J Mol Sci ; 24(14)2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37511310

RESUMEN

As an essential constituent of the mitochondrial contact site and cristae organization system (MICOS), MIC19 plays a crucial role in maintaining the stability of mitochondrial function and microstructure. However, the mechanisms and functions of MIC19 in intracerebral hemorrhage (ICH) remain unknown and need to be investigated. Sprague Dawley (SD) rats injected with autologous blood obtained from the caudal artery, and cultured neurons exposed to oxygen hemoglobin (OxyHb) were used to establish and emulate the ICH model in vivo and in vitro. Lentiviral vector encoding MIC19 or MIC19 short hairpin ribonucleic acid (shRNA) was constructed and administered to rats by intracerebroventricular injection to overexpress or knock down MIC19, respectively. First, MIC19 protein levels were increased after ICH modeling. After virus transfection and subsequent ICH modeling, we observed that overexpression of MIC19 could mitigate cell apoptosis and neuronal death, as well as abnormalities in mitochondrial structure and function, oxidative stress within mitochondria, and neurobehavioral deficits in rats following ICH. Conversely, knockdown of MIC19 had the opposite effect. Moreover, we found that the connection between MIC19 and SAM50 was disrupted after ICH, which may be a reason for the impairment of the mitochondrial structure after ICH. In conclusion, MIC19 exerts a protective role in the subsequent injury induced by ICH. The investigation of MIC19 may offer clinicians novel therapeutic insights for patients afflicted with ICH.


Asunto(s)
Hemorragia Cerebral , Mitocondrias , Membranas Mitocondriales , Animales , Ratas , Apoptosis , Hemorragia Cerebral/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Ratas Sprague-Dawley
17.
J Chem Neuroanat ; 132: 102318, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37482144

RESUMEN

Semaphorin 4 C (SEMA4C) and its cognate receptor Plexin B2 are important regulators of axon guidance and are involved in many neurological diseases, in which SEMA4C acts not only as a ligand ("forward" mode) but also as a signaling receptor ("reverse" mode). However, the role of SEMA4C/Plexin B2 in intracerebral hemorrhage (ICH) remains unclear. In this study, ICH in adult male Sprague-Dawley rats was induced by autologous blood injection in the right basal ganglia. In vitro, cultured primary neurons were subjected to OxyHb to imitate ICH injury. Recombinant SEMA4C (rSEMA4C) and overexpressing lentiviruses encoding full-length SEMA4C or secretory SEMA4C (sSEMA4C) were administered to rats by intraventricular injection. First, we found that elevated levels of sSEMA4C in the cerebrospinal fluid (CSF) of clinical patients were associated with poor prognosis. Both SEMA4C and sSEMA4C were increased in brain tissue around the hematoma after ICH in rats. Overexpression of SEMA4C attenuated neuronal apoptosis, neurosis, and neurologic impairment after ICH. However, treatment with rSEMA4C or sSEMA4C overexpression exacerbated neuronal injury. In addition, when treated with SEMA4C overexpression, the forward mode downstream protein RhoA and the reverse mode downstream ID1/3 transcriptional factors of SEMA4C/Plexin B2 signaling were all activated. Nevertheless, when exposed to rSEMA4C or sSEMA4C overexpression, only the forward mode was activated. Thus, sSEMA4C may be a novel molecular biomarker to predict the prognosis of patients with ICH, and the prevention of SEMA4C cleavage is expected to be a promising therapeutic target.


Asunto(s)
Hemorragia Cerebral , Fármacos Neuroprotectores , Semaforinas , Animales , Masculino , Ratas , Hemorragia Cerebral/diagnóstico , Ratas Sprague-Dawley , Semaforinas/metabolismo
19.
Toxicology ; 490: 153506, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37028639

RESUMEN

Paraquat (PQ) poisoning can result in multiple organ dysfunction syndrome, mainly manifesting as acute lung injury and acute respiratory distress syndrome. No specific cure exists for PQ poisoning. However, by scavenging mitochondrial DNA (mtDNA), the damage-associated molecular pattern during PQ poisoning, mitophagy can ameliorate the downstream inflammatory pathways activated by mtDNA. Melatonin (MEL), however, can promote the expression of PINK1 and BNIP3, which are key proteins involved in mitophagy. In this study, we first explored whether MT could reduce PQ-induced acute lung injury by affecting mitophagy in animal models, and then, we studied the specific mechanism associated with this process through in vitro experiments. We also evaluated MEL intervention in the PQ group, while inhibiting the expression of PINK1 and BNIP3, to further determine whether the protective effects of MEL are associated with its effect on mitophagy. We found that when the expression of PINK1 and BNIP3 was inhibited, MEL intervention could not reduce mtDNA leakage and the release of inflammatory factors caused by PQ exposure, suggesting that the protective effect of MEL was blocked. These results suggest that by promoting the expression of PINK1 and BNIP3 and activating mitophagy, MEL can reduce mtDNA/TLR9-mediated acute lung injury during PQ poisoning. The results of this study could provide guidance for the clinical treatment of PQ poisoning to reduce associated mortality.


Asunto(s)
Lesión Pulmonar Aguda , Melatonina , Animales , Melatonina/farmacología , Paraquat/toxicidad , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/genética , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Pulmón
20.
Food Chem Toxicol ; 176: 113756, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36997055

RESUMEN

Aflatoxin G1 (AFG1), a member of the aflatoxin family with cytotoxic and carcinogenic properties, is one of the most common mycotoxins occurring in various agricultural products, animal feed, and human foods and drinks worldwide. Epithelial cells in the gastrointestinal tract are the first line of defense against ingested mycotoxins. However, the toxicity of AFG1 to gastric epithelial cells (GECs) remains unclear. In this study, we explored whether and how AFG1-induced gastric inflammation regulates cytochrome P450 to contribute to DNA damage in GECs. Oral administration of AFG1 induced gastric inflammation and DNA damage in mouse GECs associated with P450 2E1 (CYP2E1) upregulation. Treatment with the soluble TNF-α receptor sTNFR:Fc inhibited AFG1-induced gastric inflammation, and reversed CYP2E1 upregulation and DNA damage in mouse GECs. TNF-α-mediated inflammation plays an important role in AFG1-induced gastric cell damage. Using the human gastric cell line GES-1, AFG1 upregulated CYP2E1 through NF-κB, causing oxidative DNA damage in vitro. The cells were also treated with TNF-α and AFG1 to mimic AFG1-induced TNF-α-mediated inflammation. TNF-α activated the NF-κB/CYP2E1 pathway to promote AFG1 activation, which enhanced DNA cellular damage in vitro. In conclusion, AFG1 ingestion induces TNF-α-mediated gastric inflammation, which upregulates CYP2E1 to promote AFG1-induced DNA damage in GECs.


Asunto(s)
Aflatoxinas , Citocromo P-450 CYP2E1 , Ratones , Humanos , Animales , Citocromo P-450 CYP2E1/genética , Citocromo P-450 CYP2E1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , FN-kappa B/metabolismo , Células Epiteliales/metabolismo , Aflatoxinas/toxicidad , Estrés Oxidativo , Inflamación/inducido químicamente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...