Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.944
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 25(13)2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38999957

RESUMEN

Abnormalities in mucosal immunity are involved in the onset and progression of ulcerative colitis (UC), resulting in a high incidence of colorectal cancer (CRC). While high-mobility group box-1 (HMGB1) is overexpressed during colorectal carcinogenesis, its role in UC-related carcinogenesis remains unclear. In the present study, we investigated the role of HMGB1 in UC-related carcinogenesis and sporadic CRC. Both the azoxymethane colon carcinogenesis and dextran sulfate sodium colitis carcinogenesis models demonstrated temporal increases in mucosal HMGB1 levels. Activated CD8+ cells initially increased and then decreased, whereas exhausted CD8+ cells increased. Additionally, we observed increased regulatory CD8+ cells, decreased naïve CD8+ cells, and decreased mucosal epithelial differentiation. In the in vitro study, HMGB1 induced energy reprogramming from oxidative phosphorylation to glycolysis in CD8+ cells and intestinal epithelial cells. Furthermore, in UC dysplasia, UC-related CRC, and hyperplastic mucosa surrounding human sporadic CRC, we found increased mucosal HMGB1, decreased activated CD8+ cells, and suppressed mucosal epithelial differentiation. However, we observed increased activated CD8+ cells in active UC mucosa. These findings indicate that HMGB1 plays an important role in modulating mucosal immunity and epithelial dedifferentiation in both UC-related carcinogenesis and sporadic CRC.


Asunto(s)
Linfocitos T CD8-positivos , Diferenciación Celular , Colitis Ulcerosa , Proteína HMGB1 , Inmunidad Mucosa , Mucosa Intestinal , Proteína HMGB1/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Colitis Ulcerosa/patología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/inducido químicamente , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Ratones , Masculino , Células Epiteliales/metabolismo , Células Epiteliales/patología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/inmunología , Ratones Endogámicos C57BL , Carcinogénesis/inmunología , Carcinogénesis/patología , Carcinogénesis/metabolismo
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000167

RESUMEN

Skeletal muscle aging and sarcopenia result in similar changes in the levels of aging markers. However, few studies have examined cancer sarcopenia from the perspective of aging. Therefore, this study investigated aging in cancer sarcopenia and explored its causes in vitro and in vivo. In mouse aging, in vitro cachexia, and mouse cachexia models, skeletal muscles showed similar changes in aging markers including oxidative stress, fibrosis, reduced muscle differentiation potential, and telomere shortening. Furthermore, examination of mitochondrial DNA from skeletal muscle revealed a 5 kb deletion in the major arc; truncation of complexes I, IV, and V in the electron transport chain; and reduced oxidative phosphorylation (OXPHOS). The mouse cachexia model demonstrated high levels of high-mobility group box-1 (HMGB1) and tumor necrosis factor-α (TNFα) in cancer ascites. Continuous administration of neutralizing antibodies against HMGB1 and TNFα in this model reduced oxidative stress and abrogated mitochondrial DNA deletion. These results suggest that in cancer sarcopenia, mitochondrial oxidative stress caused by inflammatory cytokines leads to mitochondrial DNA damage, which in turn leads to decreased OXPHOS and the promotion of aging.


Asunto(s)
Envejecimiento , Daño del ADN , ADN Mitocondrial , Proteína HMGB1 , Músculo Esquelético , Estrés Oxidativo , Sarcopenia , Animales , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ratones , Envejecimiento/metabolismo , Envejecimiento/genética , Sarcopenia/metabolismo , Sarcopenia/patología , Sarcopenia/genética , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Caquexia/metabolismo , Caquexia/patología , Caquexia/genética , Caquexia/etiología , Fosforilación Oxidativa , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , Masculino , Ratones Endogámicos C57BL
3.
Int J Mol Sci ; 25(13)2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-39000266

RESUMEN

Liver resection (LR) is the primary treatment for hepatic tumors, yet posthepatectomy liver failure (PHLF) remains a significant concern. While the precise etiology of PHLF remains elusive, dysregulated inflammatory processes are pivotal. Therefore, we explored the theragnostic potential of extracellular high-mobility-group-box protein 1 (HMGB1), a key damage-associated molecular pattern (DAMP) released by hepatocytes, in liver recovery post LR in patients and animal models. Plasma from 96 LR patients and liver tissues from a subset of 24 LR patients were analyzed for HMGB1 levels, and associations with PHLF and liver injury markers were assessed. In a murine LR model, the HMGB1 inhibitor glycyrrhizin, was administered to assess its impact on liver regeneration. Furthermore, plasma levels of keratin-18 (K18) and cleaved cytokeratin-18 (ccK18) were quantified to assess suitability as predictive biomarkers for PHLF. Patients experiencing PHLF exhibited elevated levels of intrahepatic and circulating HMGB1, correlating with markers of liver injury. In a murine LR model, inhibition of HMGB1 improved liver function, reduced steatosis, enhanced regeneration and decreased hepatic cell death. Elevated levels of hepatic cell death markers K18 and ccK18 were detected in patients with PHLF and correlations with levels of circulating HMGB1 was observed. Our study underscores the therapeutic and predictive potential of HMGB1 in PHLF mitigation. Elevated HMGB1, K18, and ccK18 levels correlate with patient outcomes, highlighting their predictive significance. Targeting HMGB1 enhances liver regeneration in murine LR models, emphasizing its role in potential intervention and prediction strategies for liver surgery.


Asunto(s)
Proteína HMGB1 , Hepatectomía , Fallo Hepático , Proteína HMGB1/metabolismo , Proteína HMGB1/sangre , Animales , Humanos , Hepatectomía/efectos adversos , Ratones , Fallo Hepático/etiología , Fallo Hepático/metabolismo , Fallo Hepático/patología , Masculino , Femenino , Persona de Mediana Edad , Regeneración Hepática , Biomarcadores , Muerte Celular , Queratina-18/metabolismo , Queratina-18/sangre , Anciano , Hepatocitos/metabolismo , Hígado/metabolismo , Hígado/patología , Ácido Glicirrínico/farmacología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
4.
J Cell Mol Med ; 28(14): e18542, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39046369

RESUMEN

This study aims to investigate the relationship between toxoplasmosis and this pathway, which may be effective in the formation of epilepsy by acting through the HMGB1/RAGE/TLR4/NF-κB signalling pathway in patients with idiopathic epilepsy. In the study, four different experimental groups were formed by selecting Toxoplasma gondii IgG positive and negative patients with idiopathic epilepsy and healthy controls. Experimental groups were as follows: Group 1: Epilepsy+/Toxo- (E+, T-) (n = 10), Group 2: Epilepsy-/Toxo- (E-, T-) (n = 10), Group 3: Epilepsy-/Toxo+ (E-, T+) (n = 10), Group 4: Epilepsy+/Toxo+ (E+, T+) (n = 10). HMGB1, RAGE, TLR4, TLR1, TLR2, TLR3, IRAK1, IRAK2, IKBKB, IKBKG, BCL3, IL1ß, IL10, 1 L8 and TNFα mRNA expression levels in the HMGB/RAGE/TLR4/NF-κB signalling pathway were determined by quantitative simultaneous PCR (qRT-PCR) after collecting blood samples from all patients in the groups. Statistical analysis was performed by one-way ANOVA followed by LSD post-hoc tests, and p < 0.05 was considered to denote statistical significance. The gene expression levels of HMGB1, TLR4, IL10, IL1B, IL8, and TLR2 were significantly higher in the G1 group than in the other groups (p < 0.05). In the G3 group, RAGE and BCL3 gene expression levels were significantly higher than in the other groups (p < 0.05). In the G4 group, however, IRAK2, IKBKB, and IKBKG gene expression levels were significantly higher than in the other groups (p < 0.05). HMGB1, TLR4, IRAK2, IKBKB, IL10, IL1B, IL1B, and IL8 in this signalling pathway are highly expressed in epilepsy patients in G1 and seizures occur with the stimulation of excitatory mechanisms by acting through this pathway. The signalling pathway in epilepsy may be activated by HMGB1, TLR4, and TLR2, which are considered to increase the level of proinflammatory cytokines. In T. gondii, this pathway is activated by RAGE and BCL3.


Asunto(s)
Epilepsia , Proteína HMGB1 , FN-kappa B , Transducción de Señal , Receptor Toll-Like 4 , Toxoplasmosis , Humanos , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genética , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , FN-kappa B/metabolismo , FN-kappa B/genética , Masculino , Femenino , Epilepsia/metabolismo , Epilepsia/genética , Epilepsia/parasitología , Adulto , Toxoplasmosis/parasitología , Toxoplasmosis/metabolismo , Toxoplasmosis/complicaciones , Toxoplasmosis/sangre , Toxoplasmosis/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor para Productos Finales de Glicación Avanzada/genética , Estudios de Casos y Controles , Adulto Joven , Persona de Mediana Edad , Antígenos de Neoplasias , Proteínas Quinasas Activadas por Mitógenos
5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 481-487, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-38952086

RESUMEN

Objective To elucidate the role of chaperone-mediated autophagy (CMA) in alleviating emotional dysfunction in mice with sepsis-associated encephalopathy (SAE). Methods The SAE mouse model was established by cecal ligation and perforation (CLP). The severity of sepsis was assessed using the sepsis severity score (MSS). Emotional function in SAE mice was assessed by the open-field test and elevated plus-maze. The expression levels of cognitive heat shock cognate protein 70 (HSC70), lysosomal-associated membrane protein 2A (LAMP2A) and high mobility group box 1 protein B1 (HMGB1) were detected using Western blotting. Co-localization of LAMP2A in the hippocampal neurons was observed by immunofluorescence. The release of inflammatory factors interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) was measured using ELISA. Following 12 hours post-CLP, mice were orally administered resveratrol at a dose of 30 mg/kg once daily until day 14. Results The mortality rate of CLP mice was 45.83% 24 days post CLP, and all surviving mice exhibited emotional disturbances. 24 hours after CLP, a significant decrease in HSC70 and LAMP2A expression in hippocampal neurons was observed, indicating impaired CMA activity. Meanwhile, HMGB1 and inflammatory cytokines (IL-6 and TNF-α) levels increased. After resveratrol treatment, an increase of HSC70 and LAMP2A expression, and a decrease of HMGB1 expression and inflammatory cytokine release were observed, suggesting enhanced CMA activity and reduced neuroinflammation. Behavioral tests showed that emotional dysfunction was improved in SAE mice after resveratrol treatment. Conclusion CMA activity of hippocampal neurons in SAE mice is significantly reduced, leading to emotional dysfunction. Resveratrol can alleviate neuroinflammation and emotional dysfunction in SAE mice by promoting CMA and inhibiting the expression of HMGB1 and the release of inflammatory factors.


Asunto(s)
Autofagia Mediada por Chaperones , Proteína HMGB1 , Resveratrol , Encefalopatía Asociada a la Sepsis , Animales , Ratones , Encefalopatía Asociada a la Sepsis/tratamiento farmacológico , Encefalopatía Asociada a la Sepsis/fisiopatología , Encefalopatía Asociada a la Sepsis/metabolismo , Masculino , Resveratrol/farmacología , Proteína HMGB1/metabolismo , Autofagia Mediada por Chaperones/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/metabolismo , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Interleucina-6/metabolismo , Estilbenos/farmacología , Proteínas del Choque Térmico HSC70/metabolismo , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Sepsis/metabolismo , Sepsis/fisiopatología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
6.
Food Chem Toxicol ; 190: 114762, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38871110

RESUMEN

In recent years, carbonized silicon nanoparticles (SiC NPs) have found widespread scientific and engineering applications, raising concerns about potential human health risks. SiC NPs may induce pulmonary damage through sustained inflammatory responses and oxidative stress, with unclear toxicity mechanisms. This study uses an in vitro co-culture model of alveolar macrophages (NR8383) and alveolar epithelial cells (RLE-6TN) to simulate the interaction between airway epithelial cells and immune cells, providing initial insights into SiC NP-triggered inflammatory responses. The research reveals that increasing SiC NP exposure prompts NR8383 cells to release high mobility group box 1 protein (HMGB1), which migrates into RLE-6TN cells and activates the receptor for advanced glycation end-products (RAGE) and Toll-like receptor 4 (TLR4). RAGE and TLR4 synergistically activate the MyD88/NF-κB inflammatory pathway, ultimately inducing inflammatory responses and oxidative stress in RLE-6TN cells, characterized by excessive ROS generation and altered cytokine levels. Pretreatment with RAGE and TLR4 inhibitors attenuates SiC-induced HMGB1 expression and downstream pathway proteins, reducing inflammatory responses and oxidative damage. This highlights the pivotal role of RAGE-TLR4 crosstalk in SiC NP-induced pulmonary inflammation, providing insights into SiC NP cytotoxicity and nanomaterial safety guidelines.


Asunto(s)
Células Epiteliales , Proteína HMGB1 , Pulmón , Macrófagos Alveolares , Nanopartículas , Compuestos de Silicona , Animales , Humanos , Ratas , Compuestos Inorgánicos de Carbono/toxicidad , Línea Celular , Técnicas de Cocultivo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Proteína HMGB1/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Nanopartículas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Compuestos de Silicona/toxicidad , Receptor Toll-Like 4/metabolismo
7.
Hepatol Commun ; 8(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38934719

RESUMEN

BACKGROUND: MASH is a common clinical disease that can lead to advanced liver conditions, but no approved pharmacotherapies are available due to an incomplete understanding of its pathogenesis. Damaged DNA binding protein 1 (DDB1) participates in lipid metabolism. Nevertheless, the function of DDB1 in MASH is unclear. METHODS: Clinical liver samples were obtained from patients with MASH and control individuals by liver biopsy. Hepatocyte-specific Ddb1-knockout mice and liver Hmgb1 knockdown mice were fed with a methionine-and choline-deficient diet to induce MASH. RESULTS: We found that the expression of DDB1 in the liver was significantly decreased in MASH models. Hepatocyte-specific ablation of DDB1 markedly alleviated methionine-and choline-deficient diet-induced liver steatosis but unexpectedly exacerbated inflammation and fibrosis. Mechanistically, DDB1 deficiency attenuated hepatic steatosis by downregulating the expression of lipid synthesis and uptake genes. We identified high-mobility group box 1 as a key candidate target for DDB1-mediated liver injury. DDB1 deficiency upregulated the expression and extracellular release of high-mobility group box 1, which further increased macrophage infiltration and activated HSCs, ultimately leading to the exacerbation of liver inflammation and fibrosis. CONCLUSIONS: These data demonstrate the independent regulation of hepatic steatosis and injury in MASH. These findings have considerable clinical implications for the development of therapeutic strategies for MASH.


Asunto(s)
Proteínas de Unión al ADN , Hígado Graso , Proteína HMGB1 , Hepatocitos , Cirrosis Hepática , Ratones Noqueados , Animales , Ratones , Hepatocitos/metabolismo , Hepatocitos/patología , Cirrosis Hepática/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Proteínas de Unión al ADN/genética , Humanos , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Hígado Graso/patología , Hígado Graso/metabolismo , Hígado Graso/genética , Masculino , Deficiencia de Colina/complicaciones , Modelos Animales de Enfermedad , Metionina/deficiencia , Hígado/patología , Hígado/metabolismo , Metabolismo de los Lípidos
8.
Drug Dev Res ; 85(4): e22219, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38845211

RESUMEN

Sepsis is a life-threatening organ dysfunction that endangers patient lives and is caused by an imbalance in the host defense against infection. Sepsis continues to be a significant cause of morbidity and mortality in critically sick patients. Oxymatrine (OMT), a quinolizidine alkaloid derived from the traditional Chinese herb Sophora flavescens Aiton, has been shown to have anti-inflammatory effects on a number of inflammatory illnesses according to research. In this study, we aimed to evaluate the therapeutic effects of OMT on sepsis and explore the underlying mechanisms. We differentiated THP-1 cells into THP-1 macrophages and studied the anti-inflammatory mechanism of OMT in a lipopolysaccharide (LPS)-induced THP-1 macrophage sepsis model. Activation of the receptor for advanced glycation end products (RAGE), as well as NF-κB, was assessed by Western blot analysis and immunofluorescence staining. ELISA was used to measure the levels of inflammatory factors. We found that OMT significantly inhibited HMGB1-mediated RAGE/NF-κB activation and downstream inflammatory cytokine production in response to LPS stimulation. Finally, an in vivo experiment was performed on septic mice to further study the effect of OMT on injured organs. The animal experiments showed that OMT significantly inhibited HMGB1-mediated RAGE/NF-κB activation, protected against the inflammatory response and organ injury induced by CLP, and prolonged the survival rate of septic mice. Herein, we provide evidence that OMT exerts a significant therapeutic effect on sepsis by inhibiting the HMGB1/RAGE/NF-κB signaling pathway.


Asunto(s)
Alcaloides , Proteína HMGB1 , Inflamación , Lipopolisacáridos , FN-kappa B , Quinolizinas , Receptor para Productos Finales de Glicación Avanzada , Sepsis , Transducción de Señal , Alcaloides/farmacología , Alcaloides/uso terapéutico , Quinolizinas/farmacología , Quinolizinas/uso terapéutico , Animales , Sepsis/tratamiento farmacológico , Sepsis/complicaciones , Sepsis/metabolismo , FN-kappa B/metabolismo , Proteína HMGB1/metabolismo , Proteína HMGB1/antagonistas & inhibidores , Humanos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal/efectos de los fármacos , Ratones , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Células THP-1 , Ratones Endogámicos C57BL , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Matrinas
9.
Int Immunopharmacol ; 137: 112440, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38870882

RESUMEN

Yucca filamentosa (YF) is widely used in folk medicine for its anti-inflammatory effects. Our study aimed to evaluate the chemical profile of YF extracts. Additionally, the gastroprotective efficacy of its crude leaf extract and nano-cubosomal formulation was assessed in a rat model of ethanol-induced gastric injury by altering the HMGB-1/RAGE/TLR4/NF-κB pathway. The phytochemical composition of YF was investigated using FTIR spectroscopy and LC-MS/MS techniques. Standardization was further accomplished using HPLC. Rats were treated orally with yucca crude extract or its nano-cubosomal formulation at doses of 25, 50, and 100 mg/kg. Famotidine (50 mg/kg, IP) was used as a reference drug. After 1 h, rats were administered ethanol (1 ml, 95 %, orally). One hour later, the rats were sacrificed, and the serum was separated to determine TNF-α and IL-6 levels. Stomachs were excised for the calculation of the ulcer index and histopathological examinations. Stomach tissue homogenate was used to determine MDA and catalase levels. Additionally, the expression levels of HMGB-1/RAGE/TLR4/NF-κB were assessed. Phytochemical analysis confirmed the predominance of steroidal saponins, sucrose, organic and phenolic acids, and kaempferol. The nano-cubosomal formulation demonstrated enhanced gastroprotective, anti-oxidant, and anti-inflammatory efficacy compared to the crude extract at all tested doses. The most prominent effect was observed in rats pretreated with the YF nano-cubosomal formulation at a dose of 100 mg/kg, which was similar to normal control and famotidine-treated rats. Our results highlighted the enhanced gastroprotective impact of the yucca nano-cubosomal formulation in a dose-dependent manner. This suggests its potential use in preventing peptic ulcer recurrence.


Asunto(s)
Antiulcerosos , Etanol , Proteína HMGB1 , Extractos Vegetales , Hojas de la Planta , Úlcera Gástrica , Yucca , Animales , Extractos Vegetales/química , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Etanol/química , Hojas de la Planta/química , Masculino , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/tratamiento farmacológico , Úlcera Gástrica/patología , Proteína HMGB1/metabolismo , Ratas , Antiulcerosos/farmacología , Antiulcerosos/uso terapéutico , Antiulcerosos/química , Antiulcerosos/administración & dosificación , Yucca/química , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Ratas Wistar , Nanopartículas/química , Interleucina-6/metabolismo , Interleucina-6/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/sangre
10.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(6): 639-645, 2024 Jun 15.
Artículo en Chino | MEDLINE | ID: mdl-38926382

RESUMEN

OBJECTIVES: To explore the effects of iris xanthin on airway inflammation, airway remodeling, and the high mobility group box 1 protein (HMGB1)/Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) pathway in asthmatic young mice. METHODS: Sixty male BALB/c young mice were randomly assigned into six groups: a blank group, a model group, a dexamethasone group, and low, medium, and high dose groups of iris xanthin, with ten mice per group. Asthma models were induced through intraperitoneal injections of a sensitizing agent [ovalbumin (OVA) 20 µg + aluminum hydroxide gel 2 mg], followed by 4% OVA aerosol inhalation. Lung function was measured using a pulmonary function tester to determine lung volume (LV), resting ventilation per minute (VE), and airway reactivity (Penh value). Hematoxylin-eosin (HE) staining was employed to examine and analyze airway remodeling. The contents of interleukin (IL)-1ß, IL-6, and tumor necrosis factor alpha (TNF-α) in bronchoalveolar lavage fluid were quantified using ELISA. Real-time fluorescence quantitative polymerase chain reaction and Western blot analysis were used to assess the expression of HMGB1/TLR4/NF-κB pathway-related mRNA and proteins in lung tissues. RESULTS: Compared to the model group, the dexamethasone and iris xanthin-treated groups (low, medium, and high doses) exhibited significant increases in LV and VE (P<0.05), with incremental dose-dependent increases observed in the iris xanthin groups. Additionally, Penh values, IL-1ß, IL-6, TNF-α, and airway remodeling indicators, along with mRNA levels of HMGB1, TLR4, and NF-κB p65 and protein levels of HMGB1, TLR4, and p-NF-κB p65, were all reduced (P<0.05) in a dose-dependent manner. When compared to the dexamethasone group, the low and medium dose iris xanthin groups showed decreases in LV and VE (P<0.05), whereas Penh values, IL-1ß, IL-6, TNF-α, and airway remodeling indicators, along with mRNA levels of HMGB1, TLR4, NF-κB p65 and protein levels of HMGB1, TLR4, and p-NF-κB p65, were increased (P<0.05). No significant differences were noted in these indices between the high dose iris xanthin group and the dexamethasone group (P>0.05). CONCLUSIONS: Iris xanthin can effectively alleviates airway inflammation and inhibits airway remodeling in asthmatic young mice, possibly through the suppression of the HMGB1/TLR4/NF-κB pathway.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma , Proteína HMGB1 , Ratones Endogámicos BALB C , FN-kappa B , Transducción de Señal , Receptor Toll-Like 4 , Animales , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Asma/tratamiento farmacológico , Asma/metabolismo , Masculino , Ratones , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Int J Mol Sci ; 25(11)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38892076

RESUMEN

Epidural and subdural hematomas are commonly associated with traumatic brain injury. While surgical removal is the primary intervention for these hematomas, it is also critical to prevent and reduce complications such as post-traumatic epilepsy, which may result from inflammatory responses in the injured brain areas. In the present study, we observed that high mobility group box-1 (HMGB1) decreased in the injured brain area beneath the epidural hematoma (EDH) in rats, concurrent with elevated plasma levels of HMGB1. Anti-HMGB1 monoclonal antibody therapy strongly inhibited both HMGB1 release and the subsequent increase in plasma levels. Moreover, this treatment suppressed the up-regulation of inflammatory cytokines and related molecules such as interleukin-1-beta (IL-1ß), tumor necrosis factor-alpha (TNF-α), and inducible nitric oxide synthase (iNOS) in the injured areas. Our in vitro experiments using SH-SY5Y demonstrated that hematoma components-thrombin, heme, and ferrous ion- prompted HMGB1 translocation from the nuclei to the cytoplasm, a process inhibited by the addition of the anti-HMGB1 mAb. These findings suggest that anti-HMGB1 mAb treatment not only inhibits HMGB1 translocation but also curtails inflammation in injured areas, thereby protecting the neural tissue. Thus, anti-HMGB1 mAb therapy could serve as a complementary therapy for an EDH before/after surgery.


Asunto(s)
Anticuerpos Monoclonales , Proteína HMGB1 , Hematoma Epidural Craneal , Proteína HMGB1/metabolismo , Animales , Ratas , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Hematoma Epidural Craneal/tratamiento farmacológico , Masculino , Humanos , Ratas Sprague-Dawley , Interleucina-1beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Citocinas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Línea Celular Tumoral
12.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(3): 653-657, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-38926949

RESUMEN

OBJECTIVE: To explore the effect of UV radiation resistance-associated gene (UVRAG) on ferroptosis induced by sorafenib in leukemia K562 cells. METHODS: K562 cells were treated with 0, 0.625, 1.25, 2.5, 5, 10, and 20 µmol/L sorafenib for 24 or 48 hours, and the cell viability was detected by CCK-8 assay. Flow cytometry technology was used to detect the changes of reactive oxygen species (ROS) in K562 cells treated with 0, 5, and 10 µmol/L sorafenib for 24 hours. Western blot was used to detect the protein expression of GPX4 in K562 cells treated with 0, 5, and 10 µmol/L sorafenib and pretreatment with ferroptosis inhibitor. A recombinant lentiviral vector was used to construct UVRAG overexpression cell line in K562 cells. qPCR and Western blot were used to verify UVRAG gene overexpression, and Western blot detected the effect of UVRAG on the protein expression of GPX4 and HMGB1 after treatment with sorafenib. RESULTS: Different concentrations of sorafenib could significantly inhibit the proliferation of K562 cells, and the cell viability gradually decreased with the increase of concentration (r 24 h=-0.9841, r 48 h=-0.9970). The level of ROS was increased (When the concentration was 10 µmol/L, P <0.001), while the expression of GPX4 protein was decreased in the process of 0, 5, 10 µmol/L sorafenib-induced K562 cell death (P <0.05), and the decrease in GPX4 protein could be partially reversed by pretreatment with ferroptosis inhibitor (P <0.05). Compared with NC group and NC-Sorafenib group, the expression of GPX4 protein was significantly decreased (both P <0.05), while HMGB1 protein was significantly increased (both P <0.05). CONCLUSION: Sorafenib can induce ferroptosis in K562 cells, and this process can be promoted by UVRAG.


Asunto(s)
Ferroptosis , Especies Reactivas de Oxígeno , Sorafenib , Sorafenib/farmacología , Humanos , Células K562 , Especies Reactivas de Oxígeno/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Proliferación Celular , Supervivencia Celular , Proteína HMGB1/metabolismo , Proteína HMGB1/genética
13.
Clinics (Sao Paulo) ; 79: 100391, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38848634

RESUMEN

BACKGROUND: The newly discovered CircUBE2D2 has been shown to abnormally upregulate and promote cancer progression in a variety of cancers. The present study explored circUBE2D2 (hsa_circ_0005728) in Ovarian Cancer (OC) progression. METHODS: CircUBE2D2, miR-885-5p, and HMGB1 were examined by RT-qPCR or WB. SKOV-3 cell functions (including cell viability, apoptosis, migration, and invasion) were validated using the CCK-8, flow cytometry, scratch assay, and transwell assay, respectively. The direct relationship between miR-885-5p and circUBE2D2 or HMGB1 was confirmed by a dual-luciferase reporter and RNA pull-down analysis. circUBE2D2's role in vivo tumor xenograft experiment was further probed. RESULTS: OC tissue and cell lines had higher circUBE2D2 and HMGB1 and lower miR-885-5p. Mechanically, CircUBE2D2 shared a binding relation with miR-885-5p, while miR-885-5p can directly target HMGB1. Eliminating circUBE2D2 or miR-885-5p induction inhibited OC cell activities. However, these functions were relieved by down-regulating miR-885-5p or HMGB1 induction. Furthermore, circUBE2D2 knockout reduced tumor growth. CONCLUSION: CircUBE2D2 regulates the expression of HMGB1 by acting as a sponge of ceRNA as miR-885-5p, thereby promoting the control of OC cell proliferation and migration and inhibiting cell apoptosis. Targeting CircUBE2D2 could serve as a new potential treatment strategy for OC.


Asunto(s)
Apoptosis , Proteína HMGB1 , MicroARNs , Neoplasias Ováricas , ARN Circular , Animales , Femenino , Humanos , Ratones , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , ARN Circular/genética
14.
Eur J Pharmacol ; 978: 176769, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-38925287

RESUMEN

Preeclampsia (PE) is often associated with multiple organ damage that remains noticeable postnatally. Here, we tested the hypotheses that antenatal therapy with nonsteroidal antiinflammatory drugs (NSAIDs) refashions liver damage induced by PE in weaning rats and that the high mobility group box 1 (HMGB1) signaling modulates this interaction. PE was induced by pharmacologic nitric oxide deprivation during the last week of gestation (Nω-nitro-L-arginine methyl ester, L-NAME, 50 mg/kg/day, oral gavage). Compared with control rats, weaning PE rats revealed substantial rises in serum transaminases together with histopathological signs of hepatic cytoplasmic changes, portal inflammation, and central vein dilation. While gestational NSAIDs reversed the elevated transaminases, they had no effects (celecoxib, naproxen) or even worsened (diclofenac) the structural damage. Molecularly, celecoxib was the most effective NSAID in (i) reversing PE-evoked upregulation of hepatic HMGB1 gene expression and concomitant increments and decrements in mitogen-activated protein kinases MAPKERK and MAPKp38 expression, respectively, and (ii) elevating and suppressing serum interleukin-10 and tumor necrosis factor-α, respectively. Alternatively, rises in serum interleukin-1ß and shifts in macrophage polarization towards an inflammatory phenotype caused by PE were comparably diminished by all NSAIDs. The data disclose an advantageous therapeutic potential for gestational celecoxib over diclofenac or naproxen in controlling hepatic dysfunction and HMGB1-interrelated inflammatory and oxidative sequels of PE.


Asunto(s)
Celecoxib , Proteína HMGB1 , Hígado , Preeclampsia , Animales , Embarazo , Femenino , Proteína HMGB1/metabolismo , Celecoxib/farmacología , Celecoxib/uso terapéutico , Preeclampsia/tratamiento farmacológico , Ratas , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo
15.
Int J Mol Sci ; 25(12)2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38928193

RESUMEN

A central role for neuroinflammation in epileptogenesis has recently been suggested by several investigations. This systematic review explores the role of inflammatory mediators in epileptogenesis, its association with seizure severity, and its correlation with drug-resistant epilepsy (DRE). The study analysed articles published in JCR journals from 2019 to 2024. The search strategy comprised the MESH, free terms of "Neuroinflammation", and selective searches for the following single biomarkers that had previously been selected from the relevant literature: "High mobility group box 1/HMGB1", "Toll-Like-Receptor 4/TLR-4", "Interleukin-1/IL-1", "Interleukin-6/IL-6", "Transforming growth factor beta/TGF-ß", and "Tumour necrosis factor-alpha/TNF-α". These queries were all combined with the MESH terms "Epileptogenesis" and "Epilepsy". We found 243 articles related to epileptogenesis and neuroinflammation, with 356 articles from selective searches by biomarker type. After eliminating duplicates, 324 articles were evaluated, with 272 excluded and 55 evaluated by the authors. A total of 21 articles were included in the qualitative evaluation, including 18 case-control studies, 2 case series, and 1 prospective study. As conclusion, this systematic review provides acceptable support for five biomarkers, including TNF-α and some of its soluble receptors (sTNFr2), HMGB1, TLR-4, CCL2 and IL-33. Certain receptors, cytokines, and chemokines are examples of neuroinflammation-related biomarkers that may be crucial for the early diagnosis of refractory epilepsy or may be connected to the control of epileptic seizures. Their value will be better defined by future studies.


Asunto(s)
Biomarcadores , Proteína HMGB1 , Enfermedades Neuroinflamatorias , Humanos , Enfermedades Neuroinflamatorias/diagnóstico , Enfermedades Neuroinflamatorias/metabolismo , Proteína HMGB1/metabolismo , Epilepsia/diagnóstico , Epilepsia/metabolismo , Citocinas/metabolismo , Receptor Toll-Like 4/metabolismo , Epilepsia Refractaria/diagnóstico , Epilepsia Refractaria/metabolismo
16.
Turk J Med Sci ; 54(1): 33-41, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38812640

RESUMEN

Background/aim: Medication overuse is common among chronic migraine patients and nonsteroidal antiinflammatory drugs (NSAIDs) are the most frequently overused drugs. The pathophysiological mechanisms underlying medication overuse headache (MOH) are not completely understood. Intestinal hyperpermeability and leaky gut are reported in patients using NSAIDs. The aim of the study is to investigate the role of leaky gut and inflammation in an MOH model MOH model in male rats. Methods: The study was conducted in male Sprague Dawley rats. There were two experimental groups. The first group was the chronic NSAID group in which the rats received mefenamic acid (n = 8) for four weeks intraperitoneally (ip) and the second group was the vehicle group (n = 8) that received 5% dimethyl sulfoxide+sesame oil (ip) for 4 weeks. We assessed spontaneous pain-like behavior, periorbital mechanical withdrawal thresholds, and anxiety-like behavior using an elevated plus maze test. After behavioral testing, serum levels of occludin and lipopolysaccharide-binding protein (LBP) and brain levels of IL-17, IL-6, and high mobility group box 1 protein (HMGB1) were evaluated with ELISA.Results: Serum LBP and occludin levels and brain IL-17 and HMGB1 levels were significantly elevated in the chronic NSAID group compared to its vehicle (p = 0.006, p = 0.016, p = 0.016 and p = 0.016 respectively) while brain IL-6 levels were comparable (p = 0.67) between the groups. The chronic NSAID group showed pain-like and anxiety-like behavior in behavioral tests. Brain IL-17 level was positively correlated with number of head shakes (r = 0.64, p = 0.045), brain IL-6 level was negatively correlated with periorbital mechanical withdrawal thresholds (r = -0.71, p = 0.049), and serum occludin level was positively correlated with grooming duration (r = 0.73, p = 0.032) in chronic NSAID group. Conclusion: Elevated serum occludin and LBP levels and brain IL-17 and HMGB1 levels indicate a possible role of leaky gut and inflammation in an MOH model in male rats. Additionally, a significant correlation between pain behavior and markers of inflammation and intestinal hyperpermeability, supports the role of inflammation and leaky gut in MOH pathophysiology.


Asunto(s)
Antiinflamatorios no Esteroideos , Biomarcadores , Proteínas Portadoras , Modelos Animales de Enfermedad , Cefaleas Secundarias , Interleucina-17 , Ratas Sprague-Dawley , Animales , Masculino , Ratas , Biomarcadores/sangre , Cefaleas Secundarias/sangre , Interleucina-17/sangre , Interleucina-17/metabolismo , Proteínas Portadoras/sangre , Proteínas Portadoras/metabolismo , Ocludina/metabolismo , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/metabolismo , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Interleucina-6/sangre , Inflamación/sangre , Inflamación/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Proteínas de Fase Aguda
17.
J Affect Disord ; 358: 211-221, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38705530

RESUMEN

BACKGROUND: Neuroinflammation is involved in the advancement of depression. Du-moxibustion can treat depression. Here, we explored whether Du-moxibustion could alleviate neuroglia-associated neuro-inflammatory process in chronic unpredictable mild stress (CUMS) mice. METHODS: C57BL/6J mice were distributed into five groups. Except for the CON group, other four groups underwent CUMS for four consecutive weeks, and Du-moxibustion was given simultaneously after modeling. Behavioral tests were then carried out. Additionally, Western blot was conducted to measure the relative expression levels of high-mobility group box 1 (HMGB1), toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and nuclear factor-kappa B (NF-κB). Immunofluorescence was employed to evaluate the positive cells of ionized calcium binding adapter molecule 1 (Iba-1) and glial fibrillary acidic protein (GFAP). Furthermore, interleukin-1 beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α) were analyzed using an ELISA assay. RESULTS: We found that CUMS induced depression-like behaviors, such as reduced sucrose preference ratio, decreased locomotor and exploratory activity, decreased the time in open arms and prolonged immobility. Furthermore, versus the CON group, the expression of HMGB1, TLR4, MyD88, NF-κB, positive cells of Iba-1, IL-1ß and TNF-α were increased but positive cells of GFAP were decreased in CUMS group. However, the detrimental effects were ameliorated by treatment with CUMS+FLU and CUMS+DM. LIMITATIONS: A shortage of this study is that only CUMS model of depression were used, while other depression model were not included. CONCLUSIONS: Du-moxibustion alleviates depression-like behaviors in CUMS mice mainly by reducing neuroinflammation, which offers novel insights into the potential treatment of depression.


Asunto(s)
Depresión , Modelos Animales de Enfermedad , Proteína HMGB1 , Ratones Endogámicos C57BL , Moxibustión , Factor 88 de Diferenciación Mieloide , Enfermedades Neuroinflamatorias , Estrés Psicológico , Animales , Ratones , Estrés Psicológico/complicaciones , Depresión/tratamiento farmacológico , Masculino , Proteína HMGB1/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Receptor Toll-Like 4/metabolismo , Conducta Animal/efectos de los fármacos , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-1beta/metabolismo
18.
Biochim Biophys Acta Rev Cancer ; 1879(3): 189105, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38701938

RESUMEN

The present study explores the complex roles of High Mobility Group Box 1 (HMGB1) in the context of cancer development, emphasizing glioblastoma (GBM) and other central nervous system (CNS) cancers. HMGB1, primarily known for its involvement in inflammation and angiogenesis, emerges as a multifaceted player in the tumorigenesis of GBM. The overexpression of HMGB1 correlates with glioma malignancy, influencing key pathways like RAGE/MEK/ERK and RAGE/Rac1. Additionally, HMGB1 secretion is linked to the maintenance of glioma stem cells (GSCs) and contributes to the tumor microenvironment's (TME) vascular leakiness. Henceforth, our review discusses the bidirectional impact of HMGB1, acting as both a promoter of tumor progression and a mediator of anti-tumor immune responses. Notably, HMGB1 exhibits tumor-suppressive roles by inducing apoptosis, limiting cellular proliferation, and enhancing the sensitivity of GBM to therapeutic interventions. This dualistic nature of HMGB1 calls for a nuanced understanding of its implications in GBM pathogenesis, offering potential avenues for more effective and personalized treatment strategies. The findings underscore the need to explore HMGB1 as a prognostic marker, therapeutic target, and a promising tool for stimulating anti-tumor immunity in GBM.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Glioblastoma , Proteína HMGB1 , Microambiente Tumoral , Humanos , Proteína HMGB1/metabolismo , Glioblastoma/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/inmunología , Neoplasias del Sistema Nervioso Central/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Animales , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/metabolismo , Transducción de Señal , Proliferación Celular
19.
J Cancer Res Clin Oncol ; 150(5): 231, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38703241

RESUMEN

PURPOSE: Acute myeloid leukemia (AML) is a refractory hematologic malignancy that poses a serious threat to human health. Exploring alternative therapeutic strategies capable of inducing alternative modes of cell death, such as ferroptosis, holds great promise as a viable and effective intervention. METHODS: We analyzed online database data and collected clinical samples to verify the expression and function of BMAL1 in AML. We conducted experiments on AML cell proliferation, cell cycle, ferroptosis, and chemotherapy resistance by overexpressing/knocking down BMAL1 and using assays such as MDA detection and BODIPY 581/591 C11 staining. We validated the transcriptional regulation of HMGB1 by BMAL1 through ChIP assay, luciferase assay, RNA level detection, and western blotting. Finally, we confirmed the results of our cell experiments at the animal level. RESULTS: BMAL1 up-regulation is an observed phenomenon in AML patients. Furthermore, there existed a strong correlation between elevated levels of BMAL1 expression and inferior prognosis in individuals with AML. We found that knocking down BMAL1 inhibited AML cell growth by blocking the cell cycle. Conversely, overexpressing BMAL1 promoted AML cell proliferation. Moreover, our research results revealed that BMAL1 inhibited ferroptosis in AML cells through BMAL1-HMGB1-GPX4 pathway. Finally, knocking down BMAL1 can enhance the efficacy of certain first-line cancer therapeutic drugs, including venetoclax, dasatinib, and sorafenib. CONCLUSION: Our research results suggest that BMAL1 plays a crucial regulatory role in AML cell proliferation, drug resistance, and ferroptosis. BMAL1 could be a potential important therapeutic target for AML.


Asunto(s)
Factores de Transcripción ARNTL , Resistencia a Antineoplásicos , Ferroptosis , Proteína HMGB1 , Leucemia Mieloide Aguda , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Transducción de Señal , Animales , Femenino , Humanos , Masculino , Ratones , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/genética , Ratones Desnudos , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Pronóstico , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Clin Neurol Neurosurg ; 241: 108291, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38701547

RESUMEN

OBJECTIVE: Acute cerebral infarction (ACI) contributes to disability and death accross the globe. Remote ischemic preconditioning (RIPC) reduces cerebral infarct size and improves neurological function in ACI. We conducted this research to reveal the effects of RIPC intervention on serum levels of microRNA-582-5p (miR-582-5p)/high mobility group box-1 protein (HMGB1), inflammation, oxidative stress and neurological function in patients with ACI. METHODS: In this study, 158 patients with ACI were prospectively selected and randomized into the control (administered symptomatic medication alone) and the RIPC (underwent RIPC of the limbs based on medication) groups, with their clinical baseline data documented. Serum levels of miR-582-5p, and HMGB1 and inflammatory factors [tumor necrosis factor alpha (TNF-α)/interleukin-1beta (IL-1ß)/IL-10] were assessed by RT-qPCR/ELISA, followed by comparisons of oxidative stress indices [glutathione-peroxidase (GSH-Px)/catalase (CAT)/superoxide dismutase (SOD)] using a fully automatic biochemical analyzer. Correlations between serum miR-582-5p with serum HMGB1, and between their levels with TNF-α/IL-1ß/IL-10 were analyzed by Pearson analysis. The NIHSS score/Barthel Index scale were used to assess neurological function/daily living ability. Intervention safety for ACI patients was evaluated. RESULTS: RIPC intervention increased serum miR-582-5p levels and decreased serum HMGB1 levels in ACI patients. RIPC intervention significantly reduced inflammation (diminished TNF-α/IL-1ß levels, increased IL-10 level) and oxidative stress (elevated GSH-Px/CAT/SOD levels) in ACI patients. Serum miR-582-5p was negatively correlated with TNF-α and IL-1ß levels, while positively correlated with IL-10 level, while HMGB1 was positively correlated with TNF-α and IL-1ß levels, while negatively correlated with IL-10 level. miR-582-5p was negatively correlated with HMGB1. RIPC intervention improved neurological function (reduced NIHSS, increased Barthel scores) in ACI patients to some extent. RIPC had certain effectiveness and safety in the treatment of ACI. CONCLUSION: After RIPC intervention, serum miR-582-5p levels were increased, HMGB1 levels were decreased, and inflammation and oxidative stress were reduced in ACI patients, which mitigated neurological deficits, improved patients' ability to perform life activities, and exerted neuroprotective effects to some extent.


Asunto(s)
Infarto Cerebral , Proteína HMGB1 , Precondicionamiento Isquémico , MicroARNs , Estrés Oxidativo , Humanos , Masculino , Proteína HMGB1/sangre , Femenino , Precondicionamiento Isquémico/métodos , MicroARNs/sangre , Persona de Mediana Edad , Anciano , Infarto Cerebral/sangre , Infarto Cerebral/terapia , Estrés Oxidativo/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA