Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
1.
China Pharmacy ; (12): 166-171, 2024.
Artigo em Chinês | WPRIM | ID: wpr-1006173

RESUMO

OBJECTIVE To investigate the improvement effects of glycyrrhizin (GL) on Helicobacter pylori (HP)-associated gastritis in rats and its mechanism. METHODS HP-associated gastritis rat model was induced by inoculating with 1×109 cfu/mL HP. The model rats were randomly divided into model group, positive control group (HP standard quadruple group), GL low-dose, medium-dose and high-dose groups (5, 20, 50 mg/kg), with 12 rats in each group. Another 12 healthy rats were selected as normal control group. Except the normal control group and model group were given constant volume of normal saline intragastrically, the other groups were given corresponding drugs intragastrically, once a day, for 30 consecutive days. After administration, rats received 13C urea breath test, and delta-over-baseline (DOB) was recorded; the pathological and cellular morphological changes of gastric mucosa in rats were observed, and pathological scoring was performed; the levels of interleukin-8 (IL-8), IL-1β, tumor necrosis factor-α (TNF-α), reactive oxygen species (ROS) and malondialdehyde (MDA) were detected in gastric mucosa of rats; mRNA expressions of high mobility group box-1 protein (HMGB1) and nuclear factor-κ-B (NF-κB), relative expressions of nitric oxide synthases (iNOS) and HMGB1, the phosphorylation level of NF- κBp65 were also detected in rats. RESULTS Compared with normal control group, the DOB value, histopathological score of gastric mucosa, the levels of IL-8, IL-1β, TNF-α, ROS and MDA, relative expressions of HMGB1 and NF- κB mRNA, relative expressions of iNOS and HMGB1 protein and the phosphorylation level of NF-κB p65 were all increased significantly in model group (P<0.05); the epithelial cells of gastric mucosa in rats were incomplete in structure and decreased in the number, with an increase in cell fragments and vacuoles, and significant cell pyknosis. Compared with model group, the changes of the above indexes in GL groups and positive control group were significantly reversed (P<0.05); the changes in the above indicators in the GL high-dose group were more significant than GL low-dose and medium-dose groups (P<0.05); the pathological changes of gastric mucosal cells in rats had all improved. CONCLUSIONS GL may inhibit inflammation and oxidative stress by inhibiting the activation of HMGB1/NF-κB pathway, thus relieving HP-induced gastric mucosal injury.

2.
Artigo em Chinês | WPRIM | ID: wpr-1018839

RESUMO

Objective To investigate the clinical efficacy of CT-guided pulsed radiofrequency combined with continuous nerve block in the treatment of refractory postherpetic neuralgia(PHN).Methods A total of 208 patients with refractory PHN,who were admitted to the Hengshui Municipal People's Hospital of China between January 2021 and January 2023,were selected as the subjects of study.Using random number table method,the patients were divided into combination group and control group,with 104 patients in each group.The patients of control group received CT-guided pulsed radiofrequency therapy,and the patients of combination group received additional continuous nerve block therapy on the basis of the treatment of control group.The pain degree at different time point,clinical effective rate,number of analgesia remedy times,quality of sleep,and the levels of serum high mobility group box 1(HMGB1),interleukin-1 β(IL-1β)and interleukin-10(IL-10)were compared between the two groups.Results During the follow-up period,4 patients were lost in touch.Finally,103 patients were included in the combination group and 101 patients were included in the control group.The total treatment response rate in the combination group was 89.32%,which was significantly higher than 78.22%in the control group(P<0.05).There were statistically significant differences in visual analogue scale(V AS)scores and Athens insomnia scale(AIS)scores including the time effect,inter-group effect and time-group interaction effect,between the two groups(P<0.05).The postoperative one-week,2-week,4-week VAS scores and AIS scores in the combination group were remarkably lower than those in the control group(P<0.05).The number of analgesia remedy times in the combination group was smaller than that in the control group,and the used dosage of tramadol in the combination group was lower than that in the control group(P<0.05).Four weeks after treatment,the serum levels of HMGB1,IL-1β and IL-10 in the combination group were lower than those in the control group(P<0.05).Conclusion For the treatment of refractory PHN,CT-guided pulsed radiofrequency combined with continuous nerve block can effectively alleviate neural inflammatory damage,and improve pain symptoms and sleep quality,besides,its analgesic effect and clinical efficacy are superior to CT-guided pulsed radiofrequency alone.(J Intervent Radiol,2024,33:264-268)

3.
Artigo em Chinês | WPRIM | ID: wpr-1031767

RESUMO

Objective@#To investigate the effect of fluid flow shear stress (FFSS) on the fluid mechanic threshold of high-mobility group box 1 (HMGB1) release by synovial cells and chondrocytes. Moreover, the mechanism of chondrocyte and synovial cell damage induced by abnormal mechanical force was investigated to provide an experimental basis for exploring the pathogenesis and pathology of temporomandibular joint osteoarthritis.@*Methods@#With the approval of the Ethics Committee for Animal Experiments of the hospital, synovial tissue and cartilage tissue blocks were obtained from the knee joints of Sprague-Dawley (SD) rats, and synovial cells and chondrocytes were cultured and digested for subsequent experiments. Synovial cells and chondrocytes of 3-4 generations were acquired, and FFSS was applied to synovial and cartilage cells using a fluid shear mechanical device. The cells were divided according to the FFSS values of different sizes. Synovial cells were stimulated for 1 h with 1, 3, 5, or 10 dyn/cm2 of FFSS, and chondrocytes were stimulated for 1 h with 4, 8, 12, or 16 dyn/cm2 of FFSS. Resting cultures (0 dyn/cm2) were used as the control group. Changes in the morphology of the cells were observed. The expression and distribution of HMGB1 and interleukin-1β (IL-1β) were observed by immunohistochemistry. The expression of HMGB1 and IL-1β in the supernatant was analyzed by ELISA. The protein expression levels of intracellular HMGB1 and IL-1β were detected by Western blot.@*Results@#With increasing FFSS, the synovial cells and chondrocytes gradually swelled and ruptured, and the number of cells decreased. With increasing FFSS, the localizationof HMGB1 and IL-1β gradually shifted from the nucleus to the cytoplasm. In synovial cells, compared with those in the control group, the expression levels of HMGB1 and IL-1β were increased both in the supernatant and cells in the 1, 3, 5 and 10 dyn/cm2 intervention groups (P<0.01). In chondrocytes, compared with those in the control group, the expression levels of HMGB1 in the supernatant were increased in the 4, 12 and 16 dyn/cm2 intervention groups (P<0.05), and the protein expression levels of HMGB1 were significantly increased (P<0.01). The expression levels of HMGB1 in the supernatant were significantly increased in the 8 dyn/cm2 intervention groups (P<0.01); however, the protein expression levels of HMGB1 were significantly decreased. Compared with those in the control group, the expression levels of IL-1β in the supernatant gradually increased in the 4, 8, 12 and 16 dyn/cm2 intervention groups (P<0.01). With the exception of those in the 4 dyn/cm2 group, the protein expression levels of IL-1β gradually increased with increasing FFSS (P<0.05).@*Conclusion@#With increasing FFSS, synovial cells and chondrocytes gradually swelled and burst, and the hydromechanical thresholds of HMGB1 release were 1 dyn/cm2 and 8 dyn/cm2, respectively. Therefore, upon stimulation with a mechanical force, synovial damage was damaged before chondrocytes.

4.
Chinese Journal of Neuromedicine ; (12): 181-185, 2024.
Artigo em Chinês | WPRIM | ID: wpr-1035978

RESUMO

Chronic subdural hematoma (CSDH) is a collection of blood, blood clots and their degradation products, encapsulated by membrane and located within dural border cell layer. Pathophysiological processes such as inflammatory responses within hematoma cavity, coagulation abnormalities, and abnormalities in neovascularization play significant roles in CSDH development. High mobility group box 1 (HMGB1) can mediate processes such as inflammation, angiogenesis, and hemostasis, while thrombomodulin (TM) can bind with HMGB1 and rely on thrombin to degrade HMGB1. Current research has confirmed that the expressions of TM, HMGB1, and their downstream related factors are abnormally increased in the hematoma fluid of CSDH; however, the role of TM-thrombin-HMGB1 pathway in CSDH development is not fully clear. This article reviews the role of TM-thrombin-HMGB1 pathway in CSDH development, aiming to provide some references for pathogenesis and new therapeutic targets of CSDH.

5.
Artigo em Chinês | WPRIM | ID: wpr-1036487

RESUMO

Objective In this study, inbred BALB/c mice infected with the pneumonia virus of mice (PVM) were used to establish an animal model of viral pneumonia, and the changes in the pro-inflammatory alarmin molecule, high mobility group box 1 protein (HMGB1), during PVM infection were observed, as well as the in vivo intervention effects of the HMGB1 inhibitor, glycyrrhizic acid (GA), on PVM-induced lung injury. Methods Three-week-old female BALB/c mice were randomly divided into three groups, each consisting of 6 mice. One group, uninfected by PVM, served as the control group (Control). The other two groups were inoculated intranasally with PVM at a dose of 1×104 50% tissue culture infective dose (TCID50)/25 μL, and subsequently treated with GA saline solution (GA group) or plain saline solution (normal saline, NS group) via gavage for 15 consecutive days. During this period, changes in body weight and appearance were monitored in each group. At the end of the experiment, lung tissue samples were collected from all groups. The distribution of PVM and HMGB1 proteins in the lung tissues was analyzed using hematoxylin-eosin staining and immunohistochemistry. The expression levels of HMGB1 and its Toll-like receptor 4 (TLR-4), advanced glycosylation end-product-specific receptor (AGER), and inflammatory cytokines such as interleukin (IL)-1β, IL-2, and tumor necrosis factor-α (TNF-α) in lung tissues of mice were measured using real time fluorescence quantitative PCR. Results Compared with the Control group, the NS group showed a significant weight loss after 6 days (P<0.05). Histopathological tests revealed pronounced inflammatory lesions in their lungs. Immunohistochemistry results showed that HMGB1 was released from the nucleus to the cytoplasm, and real time fluorescence quantitative PCR results indicated that the expression levels of HMGB1, IL-1β, and IL-2 were significantly upregulated (P<0.05). In the GA group, there was no significant change in the clinical symptoms or body weight. However, compared with the NS group, the pathological damages of lung tissues in the GA group were significantly reduced, and the expression levels of HMGB1, IL-1β, IL-2, and interferon-γ (IFN-γ) in lung tissues were also significantly decreased (P<0.05), although the expression level of AGER was significantly increased (P<0.05).ConclusionPVM infection can cause significant inflammatory pathological lung damages in mice, and GA can effectively alleviate the damages. Its therapeutic effect may be related to the activation of HMGB1 signaling pathway.

6.
International Eye Science ; (12): 18-23, 2024.
Artigo em Chinês | WPRIM | ID: wpr-1003499

RESUMO

AIM: To explore the dynamic expression of high mobility group box 1(HMGB1)in scar tissues after glaucoma drainage valve implantation, and to further reveal the role and possible mechanism of HMGB1 in scarring after glaucoma surgery.METHODS: A total of 60 New Zealand white rabbits were randomly divided into control group(n=20), model group(n=20, silicone implantation under conjunctival sac)and model with drug administration group(n=20, silicone implantation under conjunctival sac combined with 5-fluorouracil injection). The conjunctival tissues were collected at 4 and 8 wk after surgery. HE staining and Masson staining were used to detect the proliferation and distribution of fibroblasts and collagen fibers in conjunctival tissues. Immunohistochemistry was utilized to detect the distribution and changes of HMGB1, transforming growth factor(TGF)-β1, Smad3 and α-smooth muscle actin(SMA)in conjunctival tissues. RT-PCR and Western blot were adopted to detect the mRNA and protein expression of HMGB1, TGF-β1, Smad3 and α-SMA in conjunctival tissues.RESULTS: HE staining and Masson staining showed that the proliferation of inflammatory cells, fibroblasts and collagen fibers in the model group was significantly higher than that in the control group at both 4 and 8 wk. Meanwhile, the proliferation of fibroblasts and collagen fibers in the model with drug administration group was significantly lower than that in the model group. Immunohistochemical staining showed that the expression of HMGB1, TGF-β1, Smad3 and α-SMA protein was observed in the conjunctival tissues of the model group both 4 and 8 wk, with brown and significantly deeper staining of the model group at 8 wk. Meanwhile, the positive staining in the model with drug administration group at both 4 and 8 wk was significantly lower than that in the model group. There was positive correlations between the number of fibroblasts stained with HE and the expression of HMGB1 in the conjunctival tissue of the model group at both 4 and 8 wk(r=0.602, 0.703, all P&#x0026;#x003C;0.05). RT-PCR and Western blot revealed that the mRNA and protein expression levels of HMGB1, TGF-β1, Smad3 and α-SMA in the model group were significantly higher than those in the control group at both 4 and 8 wk(all P&#x0026;#x003C;0.05). Meanwhile, the mRNA and protein expression levels of HMGB1, TGF-β1, Smad3 and α-SMA in the model with drug administration group were significantly lower than those in the model group(all P&#x0026;#x003C;0.05). There was positive correlations between mRNA expressions of HMGB1 and TGF-β1, Smad3 in the model group and the model with drug administration group(all P&#x0026;#x003C;0.05).CONCLUSION: The expression of HMGB1 increased at a time-dependent manner after glaucoma valve implantation. HMGB1 acts an indispensable role in the initiation and progression of scar formation after glaucoma surgery, which may be involved in the regulation of TGF-β/Smad signaling pathway.

7.
Artigo em Chinês | WPRIM | ID: wpr-984577

RESUMO

ObjectiveTo explore the mechanism of Dahuang Mudantang in alleviating the intestinal injury in the rat model of acute pancreatitis via the high-mobility group box 1 (HMGB1)/receptor for advanced glycation endproduct (RAGE)/nuclear factor-κB (NF-κB) signaling pathway. MethodOne hundred and twenty SPF-grade Wistar rats received retrograde injection of 5% sodium taurocholate into the biliopancreatic duct for the modeling of intestinal injury in acute pancreatitis. The rats were randomized into blank, model, low-, medium-, and high-dose (3.5, 7, 14 g·kg-1, administrated by gavage) Dahuang Mudantang, and octreotide (1×10-5 g·kg-1, subcutaneous injection) groups (n=20). The rats in blank and model groups received equal volume of distilled water by gavage. Drugs were administered 1 h before and every 12 h after modeling, and samples were collected 24 h after modeling. The general status of the rats was observed. The biochemical methods were employed to measure the levels of amylase (AMS) and C-reactive protein (CRP) in the serum. The enzyme-linked immunosorbent assay (ELISA) was employed to measure the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 in the colon tissue. The morphological changes of pancreatic and colon tissues were observed by hematoxylin-eosin (HE) staining. Real-time fluorescence quantitative polymerase chain reaction (Real-time PCR) and Western blot were employed to measure the expression levels of HMGB1, RAGE, inhibitor of NF-κB kinase (IKK), and NF-κB suppressor protein α(IκBα)in the colon tissue. ResultThe rats in the model group showed poor general survival, writhing response, reduced frequency of defecation, and dry stool. The symptoms of rats in the model group were mitigated in each treatment group, and the high-dose Dahuang Mudantang showed the most significant effect. Compared with the normal group, the model group had elevated AMS and CRP levels (P<0.05), which were lowered by Dahuang Mudantang (P<0.05), especially that at the high dose (P<0.05). Compared with the normal group, the modeling elevated that levels of TNF-α, IL-1β, and IL-6 (P<0.05). Such elevations were lowered by Dahuang Mudantang (P<0.05), and the high-dose group and the octreotide group showed better performance (P<0.05). The modeling caused necrotic, congested, and destructed pancreatic and colonic tissues, which were ameliorated by the drugs, especially high-dose Dahuang Mudantang. Compared with the normal group, the modeling up-regulated the mRNA levels of HMGB1, RAGE, IKK, IκBα, and NF-κB (P<0.05). Compared with the model group, Dahuang Mudantang and octreotide down-regulated the mRNA levels of HMGB1, RAGE, IKK, IκBα, and NF-κB (P<0.05), and the high-dose Dahuang Mudantang demonstrated the best performance (P<0.05). Western blot results showed a trend consistent with the results of Real-time PCR. ConclusionDahuang Mudantang can improved the general status, reduce inflammation, and alleviate histopathological changes in the pancreatic and colon tissues in the rat model of acute pancreatitis by inhibiting the HMGB1/RAGE/NF-κB signaling pathway.

8.
Artigo em Chinês | WPRIM | ID: wpr-981436

RESUMO

This paper aimed to investigate the effects of high mobility group box 1(HMGB1)-mediated pulmonary artery smooth muscle cell pyroptosis and immune imbalance on chronic obstructive pulmonary disease-associated pulmonary hypertension(COPD-PH) in rats and the intervening mechanism of Compound Tinglizi Decoction. Ninety rats were randomly divided into a normal group, a model group, low-dose, medium-dose, and high-dose Compound Tinglizi Decoction groups, and a simvastatin group. The rat model of COPD-PH was established by fumigation combined with lipopolysaccharide(LPS) intravascular infusion, which lasted 60 days. Rats in the low, medium, and high-dose Compound Tinglizi Decoction groups were given 4.93, 9.87, and 19.74 g·kg~(-1) Compound Tinglizi Decoction by gavage, respectively. Rats in the simvastatin group were given 1.50 mg·kg~(-1) simvastatin by gavage. After 14 days, the lung function, mean pulmonary artery pressure, and arterial blood gas of rats were analyzed. Lung tissues of rats were collected for hematoxylin-eosin(HE) staining to observe the pathological changes. Real-time fluorescent quantitative polymerase chain reaction(qRT-PCR) was used to determine the expression of related mRNA in lung tissues, Western blot(WB) was used to determine the expression of related proteins in lung tissues, and enzyme linked immunosorbent assay(ELISA) was used to determine the levels of inflammatory factors in the lung tissues of rats. The ultrastructure of lung cells was observed by transmission electron microscope. The forced vital capacity(FVC), forced expiratory volume in 0.3 second(FEV_(0.3)), FEV_(0.3)/FVC, peek expiratory flow(PEF), respiratory dynamic compliance(Cdyn), arterial partial pressure of oxygen(PaO_2), and arterial oxygen saturation(SaO_2) were increased, and resistance of expiration(Re), mean pulmonary arterial pressure(mPAP), right ventricular hypertrophy index(RVHI), and arterial partial pressure of carbon dioxide(PaCO_2) were decreased by Compound Tinglizi Decoction in rats with COPD-PH. Compound Tinglizi Decoction inhibited the protein expression of HMGB1, receptor for advanced glycation end products(RAGE), pro caspase-8, cleaved caspase-8, and gasdermin D(GSDMD) in lung tissues of rats with COPD-PH, as well as the mRNA expression of HMGB1, RAGE, and caspase-8. Pulmonary artery smooth muscle cell pyroptosis was inhibited by Compound Tinglizi Decoction. Interferon-γ(IFN-γ) and interleukin-17(IL-17) were reduced, and interleukin-4(IL-4) and interleukin-10(IL-10) were incresead by Compound Tinglizi Decoction in lung tissues of rats with COPD-PH. In addition, the lesion degree of trachea, alveoli, and pulmonary artery in lung tissues of rats with COPD-PH was improved by Compound Tinglizi Decoction. Compound Tinglizi Decoction had dose-dependent effects. The lung function, pulmonary artery pressure, arterial blood gas, inflammation, trachea, alveoli, and pulmonary artery disease have been improved by Compound Tinglizi Decoction, and its mechanism is related to HMGB1-mediated pulmonary artery smooth muscle cell pyroptosis and helper T cell 1(Th1)/helper T cell 2(Th2), helper T cell 17(Th17)/regulatory T cell(Treg) imbalance.


Assuntos
Animais , Ratos , Caspase 8 , Piroptose , Proteína HMGB1/genética , Hipertensão Pulmonar/etiologia , Doença Pulmonar Obstrutiva Crônica/genética
9.
Artigo em Chinês | WPRIM | ID: wpr-992771

RESUMO

Objective:To investigate the role and underlying mechanisms of inhibiting high mobility group box-1 (HMGB1) in the expression of matrix metalloproteinase-9 (MMP-9) in spinal cord astrocytes (AS) in rats after spinal cord injury (SCI).Methods:After an SCI model was established in Sprague-Dawley (SD) rats using a modified Allen's Weight-Dropping method and ethyl pyruvate (EP) or glycyrrhizin (GL) was used to inhibit the effect of HMGB1, the rats were divided into a sham group, an SCI group, an SCI+EP (50 mg/kg) group, and an SCI+GL (100 mg/kg) group. The expression levels of glial fibrillary acid protein (GFAP) and MMP-9 in spinal cord AS were observed. After the spinal cord AS in SD rats was cultured and incubated by the oxygen-glucose deprivation/reoxygenation (OGD/R) procedure, the expression of MMP-9 protein was detected at 6 h/R 6 h, 12 h, 24 h, and 48 h after OGD. The time point with the highest expression was chosen in the subsequent experiments as an OGD/R group. HMGB1 was inhibited by HMGB1 shRNA or EP to observe the effect of HMGB1 on the expression of MMP-9 protein in AS treated with OGD/R. Then, toll-like receptor 4 (TLR4) inhibitor, TIR-domain-containing adaptor inducing interferon- β (TRIF) inhibitor, and nuclear factor-kappa B (NF- κB) inhibitor were used to investigate the effects of TLR4/TRIF/NF- κB signaling pathway during the regulation of HMGB1 on MMP-9 in vitro. Results:Western blot showed that the expression of MMP-9 protein in the spinal cord was significantly increased in rats at 1 d after SCI, and the expression of MMP-9 protein in the SCI+EP group and the SCI+GL group was significantly lower than that in the SCI group ( P<0.001). Immunofluorescence showed that GFAP and MMP-9 proteins were co-localized in the spinal cord after SCI, and the expression of GFAP and MMP-9 proteins in the SCI+EP and SCI+GL groups was significantly lower than that in the SCI group ( P<0.05). Since the expression of MMP-9 protein in the spinal cord AS cultured in vitro was significantly higher in the OGD 6h/R 12h group than that in the normal group and the OGD 6h/R 6h, 24, and 48 h groups, the OGD 6h/R 12h was taken as the OGD/R group. The MMP-9 protein expression in AS in the OGD/R+HMGB1 shRNA group and the OGD/R+EP group was significantly lower than that in the OGD/R group ( P<0.001). In the cultured AS, moreover, inhibiting TLR4, TRIF, and NF- κB reduced MMP-9 protein expression after OGD 6 h/R 12 h when compared with that in the OGD/R group ( P<0.001). Conclusions:HMGB1 inhibition may result in a reduction in MMP-9 expression both in the spinal cord AS in SCI rats and in AS after OGD/R treatment in vitro. HMGB1 may regulate MMP-9 protein expression in AS after OGD/R treatment via the TLR4/TRIF/NF- κB signal pathway.

10.
Artigo em Chinês | WPRIM | ID: wpr-1031751

RESUMO

@#Objective To explore the mechanism of the gut microbiota metabolite trimethylamine-N-oxide (TMAO) acting on high mobility group box 1 (HMGB1)/NOD-like receptor protein 3 (NLRP3) to regulate inflammatory response after cerebral ischemia/reperfusion (I/R) injury in mice. Methods A mouse atherosclerosis model was established by feeding on TMAO and high-fat diet.A mouse model of middle cerebral artery occlusion (MCAO) was prepared using the suture method.Mice were randomly divided into sham group,2-week-TMAO-feeding group,and 6-week-TMAO-feeding group.After I/R injury,we measured the protein levels of NLRP3,HMGB1,cleaved interleukin-1β (Cle-IL-1β),and zonula occludens-1 (ZO-1) by Western blotting.After six weeks of TMAO feeding,mice were randomly divided into sham group,I/R group,TMAO+I/R group,and TMAO+DMB+I/R group.For each group,we scored neurological function,determined NLRP3,HMGB1,Cle-IL-1β,and ZO-1 protein expression by Western blot,measured brain infarct volume with TTC staining,and measured the water content of brain tissue. Results Compared with those of the sham group,NLRP3,HMGB1,and Cle-IL-1β protein expression increased significantly and ZO-1 decreased significantly with the length of TMAO feeding (all P<0.05).Compared with the I/R group,the TMAO+I/R group showed a significant decline in neurological scores,significantly increased NLRP3,HMGB1,and Cle-IL-1β expression,significantly decreased ZO-1 expression,a significant increase in brain infarct volume,and a significant decrease in the water content of brain tissue (all P<0.05).With DMB lowering TMAO,the TMAO+DMB+I/R group had significantly better neurological scores,significantly lower NLRP3,HMGB1,and Cle-IL-1β levels,a significantly increased ZO-1 level,and significantly reduced brain infarct volume and brain tissue water content,as compared with the TMAO+I/R group (all P<0.05). Conclusion The gut microbiota metabolite TMAO can upregulate HMGB1/NLRP3-mediated inflammatory response in mice with cerebral I/R injury,worsening the breakdown of the blood-brain barrier to exacerbate brain tissue damage.

11.
Chinese Journal of School Health ; (12): 1883-1888, 2023.
Artigo em Chinês | WPRIM | ID: wpr-1004912

RESUMO

Objective@#To investigate the role of miR-142-3p in alleviation of house dust mite induced allergic airway inflammation among children, so as to provide insights into unraveling the pathogenesis of allergic airway inflammation.@*Methods@#Serum samples were collected from 15 patients with house dust mite induced allergic asthma and 15 healthy children in Jiangnan University Medical Center from September to November 2022, and serum miR-142-3p expression was quantified using a fluorescent quantitative real time PCR (qPCR) assay. The levels of interleukin 6 (IL 6) and tumor necrosis factor α (TNF α) were measured in the cell culture supernatant using enzyme linked immunosorbent assay (ELISA), and the expression of high mobility group box 1 (HMGB1) was detected at transcriptional and translational lvels using qPCR and Western blotting assays. The negative regulation of the HMGB1 gene by miR 142 3p was identified using a dual luciferase gene reporter assay, and the expression of downstream regulatory proteins was determined in human normal lung epithelial cells (BEAS 2B) cells transfected with miR 142 3p using Western blotting. In addition, female C57BL/6 mice at ages of 6-8 weeks were randomly assigned to the phosphate buffer saline (PBS) group, house dust mite sensitized airway inflammation group and house dust mite sensitized airway inflammation + miR 142 3p intervention group. Mouse airway inflammation was evaluated using hematoxylin eosin staining, and the expression of inflammatory cells and inflammatory cytokines were detected in mouse bronchoalveolar lavage fluid (BALF) using Giemsa staining and ELISA.@*Results@#Lower serum miR-142-3p expression was quantified among children with house dust mite induced allergic asthma than among healthy controls (1.33±0.21 vs. 4.74±0.62, t=5.22, P <0.05). Stimulation with dermatophagoides farinae extract (DFE) resulted in a reduction in miR-142-3p expression in BEAS-2B cells (0.82±0.25), while transfection with miR-142-3p mimics resulted in a rise in miR-142-3p expression in BEAS-2B cells (0.55±0.14)( t=3.31, 3.94, P <0.05). Pre treatment with miR-142-3p reduced the expression of IL 6(2.25±0.46)and TNF α(6.58±1.95) ( t=4.86, 3.38, P <0.05) in BEAS 2B cells stimulated with DFE, and treatment with miR-142-3p mimics resulted in a reduction in TLR4 and NF-κB expression in BEAS-2B cells via negative regulation of the HMGB1 expression. In addition, treatment with miR-142-3p was found to alleviate inflammatory cell infiltration in lung tissues of house dust mite sensitized mice, and results in a reduction in interleukin 4 (IL-4)[(107.60±10.43)pg/mL], interleukin 5 (IL 5)[(95.78±13.14)pg/mL] and HMGB1[(2.52±0.87)pg/mL] expression in BALF ( t=10.32, 7.29, 2.90, P <0.05).@*Conclusion@#miR-142-3p alleviates house dust mite induced allergic airway inflammation among children via negative regulation of the HMGB1/TLR4/NF-κB pathway.

12.
Artigo em Chinês | WPRIM | ID: wpr-1005748

RESUMO

【Objective】 To observe the reactive change of cortical perivascular cells after craniocerebral injury and explore its mechanism. 【Methods】 The controllable cortical impact animal model was used to simulate craniocerebral injury, the expressions of cortical pericyte markers at different time points after trauma were studied by Western blotting, and the biological behavior of vascular pericytes after craniocerebral injury was determined by transmission electron microscopy. Post-traumatic high mobility group box 1 (HMGB1), receptor for advanced glycation end product (RAGE), and nuclear factor κB (NF-κB) were detected by Western blotting. The experimental animals were divided into FPS-ZM1 (a specific RAGE receptor blocker) injection group and wild-type group. Wet and dry brain weight and transmission electron microscopy were used to study the post-traumatic effects of HMGB1-RAGE on pericytes. The primary mouse brain microvascular pericytes were cultured and supplemented with HMGB1 recombinant protein; the cultured pericytes supplemented with FPS-ZM1 were used as the control to explore the effect of HMGB1-RAGE pathway on vascular pericytes in vitro. 【Results】 The expression levels of early post-traumatic cortical pericyte markers platelet-derived growth factor receptor beta (PDGFR-β) and NG2 proteoglycan (NG2) decreased (PDGFR-β, Control vs. CCI 3D P<0.05; NG2, Control vs. CCI 6H P<0.05; Control vs. CCI 1D P<0.05). We found that pericytes were detached from blood vessels, accompanied by local blood-brain barrier opening. The expression of HMGB1-RAGE-NF-κB signaling pathway was increased in the early cortex after trauma (HMGB1, Control vs. CCI 6H P<0.05, Control vs. CCI 1D P<0.05; RAGE, Control vs. CCI 6H P<0.05, Control vs. CCI 1D P<0.05, Control vs. CCI 3D P<0.05, Control vs. CCI 5D P<0.05, Control vs. CCI 7D P<0.05; NF-κB, Control vs. CCI 6H P<0.05, Control vs. CCI 1D P<0.05). After blocking the binding of RAGE with the ligand, cortical edema was reduced (CCI 6H P<0.05, CCI 1D P<0.05), and neurovascular unit damage was reduced. HMGB1 recombinant protein could increase the migration ability of cultured pericytes (Control vs. HMGB1 P<0.05, Control vs. HMGB1+FPS-ZM1 P<0.05), and could be reversed by FPS-ZM1 (HMGB1 vs. HMGB1+FPS-ZM1 P<0.05). 【Conclusion】 High-level HMGB1 after traumatic brain injury mediates pericytes’ detachment from blood vessels through RAGE on pericytes and leads to the occurrence of local cerebral edema.

13.
Artigo em Chinês | WPRIM | ID: wpr-1011065

RESUMO

Objective:To investigate the expression level and regulatory mechanism of 15-hydroxyprostaglandin dehydrogenase(HPGD) in chronic rhinosinusitis with nasal polyps(CRSwNP). Methods:The expression pattern and level of HPGD in CRSwNP and control was observed using immunofluorescence, and western blot was used for analysis of HPGD expression in nasal polyp tissues. The effect of recombinant human high mobility group box-1(HMGB1) on HPGD expression in primary human nasal epithelial cells was observed, and the potential blocking effect of RAGE neutralizing antibody on HMGB1-induced HPGD expression was investigated. Results:The expression of HPGD was elevated in CRSwNP patients compared to the control, while the protein mainly localized at CD68-positive cells and epithelial cells. Recombinant human HMGB1 stimulated an increase in HPGD expression in primary human nasal mucosal epithelial cells at a time-dependent manner. Additionally, increased phosphorylation levels of MEK and elevated RAGE expression were also observed at 12 hours, but decreased at 24 hours after the incubation of HMGB1. The increase in the expression of HPGD induced by HMGB1 in primary human nasal epithelial cells was partly inhibited with RAGE neutralizing antibody. Conclusion:Elevated HPGD expression is observed in CRSwNP, predominantly in macrophages and epithelial cells. HMGB1 regulates HPGD expression through the RAGE-MEK signaling pathway, potentially providing a new target for future regulation of PGE2levels in CRSwNP.


Assuntos
Humanos , Anticorpos Neutralizantes/metabolismo , Doença Crônica , Proteína HMGB1/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mucosa Nasal/metabolismo , Pólipos Nasais/metabolismo , Rinite
14.
Artigo em Chinês | WPRIM | ID: wpr-961682

RESUMO

ObjectiveTo study the effect of modified Erchentang on the expression of key molecules in the high mobility group Box 1 protein (HMGB1)/receptor for advanced glycation endproduct (RAGE)/nuclear factor-κB (NF-κB) signaling pathway in bronchioles of rats with chronic obstructive pulmonary disease (COPD), to explore the mechanism of modified Erchentang against bronchiolar inflammation of COPD rats via HMGB1/RAGE/NF-κB signaling pathway. MethodSixty SD rats were randomly divided into normal group, model group, modified Erchentang low-, medium- and high-dose groups (5, 10, 20 g·kg-1·d-1) and ethyl pyruvate (HMGB1 inhibitor) group, with 10 in each group. The COPD rat model was prepared by cigarette smoke combined with tracheal injection of lipopolysaccharide (LPS). After modeling, the modified Erchentang groups were given corresponding drugs (ig) and Ringer's solution (4 mL, ip), while the EP group was treated with equal volume of normal saline (ig) and EP (0.04 g·kg-1·d-1, ip). The normal group and the model group received equal volume of normal saline (ig) and Ringer's solution (ip) for 21 consecutive days. The contents of HMGB1, chemokine (C-X-C motif) ligand 1 (CXCL1), CXCL2 and monocyte chemotactic protein-1 (MCP-1) in bronchoalveolar lavage fluid (BALF) were detected by enzyme-linked immunosorbent assay (ELISA). The mRNA expressions of HMGB1, RAGE and NF-κB p65 were determined by Real-time polymerase chain reaction (Real-time PCR), and the protein expressions of HMGB1, RAGE, p-NF-κB p65, and alpha-smooth muscle actin (α-SMA) in bronchioles tissue of rats were determined by immunohistochemistry (IHC). ResultCompared with the conditions in the normal group, the forced vital capacity (FVC), forced expiratory volume in the first second (FEV1) and FEV1/FVC in the model group were decreased (P<0.01) while the contents of HMGB1, CXCL1, CXCL2 and MCP-1 in BALF were increased (P<0.01). And the model group presented higher mRNA expressions of HMGB1, RAGE and NF-κB p65 (P<0.01) and protein expressions of HMGB1, RAGE, p-NF-κB p65 and α-SMA (P<0.05, P<0.01) than the normal group. Compared with the model group, the modified Erchentang medium- and high-dose groups had increased FEV1/FVC (P<0.05, P<0.01), lowered contents of HMGB1, CXCL1, CXCL2 and MCP-1 in BALF (P<0.05, P<0.05), and reduced mRNA expressions of HMGB1, RAGE and NF-κB p65 (P<0.05, P<0.01) and protein expressions of HMGB1, RAGE, p-NF-κB p65 and α-SMA (P<0.05, P<0.01). ConclusionModified Erchentang can resist bronchiolar inflammation of COPD rats. The mechanism may be related to down-regulating the mRNA expressiona of HMGB1 and RAGE, inhibiting the activity of NF-κB, and reducing the release of HMGB1, CXCL1, CXCL2 and MCP-1, thus suppressing the inflammatory injury and abnormal repair of bronchioles.

15.
Chinese Journal of Oncology ; (12): 919-925, 2023.
Artigo em Chinês | WPRIM | ID: wpr-1045822

RESUMO

Objective: To investigate the molecular mechanism of how lactate induces high mobility group box 1 (HMGB1) release. Methods: Gastric cancer HGC-27 cells were divided into the control group and the lactate group (The cells were treated with lactate for 6 h). The level of HMGB1 in the cell culture medium was detected by enzyme-linked immunosorbent assay (ELISA), the localization of HMGB1 was detected using laser confocal microscopy, and the nuclear translocation of HMGB1 was detected using the nucleoplasmic separation assay. The phosphorylation and acetylation levels of HMGB1 were determined by co-immunoprecipitation, and Western blot was used to measure the phosphorylation of Akt and protein kinase C (PKC). HGC-27 cells were first treated with lactate and LY294002, the inhibitor of Akt, and then the phosphorylation of HMGB1 and Akt was analyzed by co-immunoprecipitation and Western blot, respectively. The localization of HMGB1 in cells was detected by laser confocal microscopy. EdU and Transwell assays were used to detect the proliferation and migration abilities of HGC-27 cells, respectively. HGC-27 cells were then injected into the BALB/C null mice for subcutaneous tumor implantation. Mice in the lactate group were intraperitoneally injected with lactate (0.2 g/kg/2 d), while those in the control group were intraperitoneally injected with an equal amount of PBS for 20 consecutive days. ELISA was used to detect the HMGB1 levels in the blood samples taken from the medial canthus vein of the mice, while co-immunoprecipitation and Western blot were used to detect the phosphorylation of HMGB1 and Akt in tumor tissue proteins, respectively. Results: The release levels of HMGB1 in the lactate group were (2 995.00±660.91) pg/ml and (696.33±22.03) pg/ml, after lactate treatment for 6 h and 12 h, respectively, both higher than those in the control group (485.00±105.83) pg/ml (P<0.001 and P=0.028, respectively). After lactate treatment for 6 h, the relative expression of HMGB1 protein in the cytoplasm of HGC-27 cells was 1.13±0.09, higher than that of the control group (0.83±0.07, P=0.001), while the relative expression of HMGB1 in the nucleus was 0.79±0.06, lower than that of the control group (1.07±0.06, P=0.007). The phosphorylation level of HMGB1 reached 1.41±0.09, which was higher than that of the control group (0.97±0.10, P=0.031). The phosphorylation level of Akt was 11.16±0.06, higher than that of the control group (0.91±0.022, P=0.002). The phosphorylation level and nuclear translocation of HMGB1 induced by lactate decreased obviously after Akt inhibition; the proliferation and migration abilities induced by lactate were also obviously inhibited after Akt inhibition. In vivo, the HMGB1 level in the peripheral blood was (1 280.70±389.66) pg/ml in the lactate group, which was obviously higher than that in the control group (595.11±44.75) pg/ml (P=0.008), and the phosphorylation levels of HMGB1 and Akt in tumor tissues in the lactate group were obviously enhanced compared with the control group. Conclusion: Lactate induces HMGB1 release through enhancing HMGB1 phosphorylation via the Akt signaling pathway.


Assuntos
Camundongos , Animais , Neoplasias Gástricas/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína HMGB1/metabolismo , Fosforilação , Ácido Láctico , Camundongos Endogâmicos BALB C , Transdução de Sinais
16.
Chinese Journal of Oncology ; (12): 919-925, 2023.
Artigo em Chinês | WPRIM | ID: wpr-1046145

RESUMO

Objective: To investigate the molecular mechanism of how lactate induces high mobility group box 1 (HMGB1) release. Methods: Gastric cancer HGC-27 cells were divided into the control group and the lactate group (The cells were treated with lactate for 6 h). The level of HMGB1 in the cell culture medium was detected by enzyme-linked immunosorbent assay (ELISA), the localization of HMGB1 was detected using laser confocal microscopy, and the nuclear translocation of HMGB1 was detected using the nucleoplasmic separation assay. The phosphorylation and acetylation levels of HMGB1 were determined by co-immunoprecipitation, and Western blot was used to measure the phosphorylation of Akt and protein kinase C (PKC). HGC-27 cells were first treated with lactate and LY294002, the inhibitor of Akt, and then the phosphorylation of HMGB1 and Akt was analyzed by co-immunoprecipitation and Western blot, respectively. The localization of HMGB1 in cells was detected by laser confocal microscopy. EdU and Transwell assays were used to detect the proliferation and migration abilities of HGC-27 cells, respectively. HGC-27 cells were then injected into the BALB/C null mice for subcutaneous tumor implantation. Mice in the lactate group were intraperitoneally injected with lactate (0.2 g/kg/2 d), while those in the control group were intraperitoneally injected with an equal amount of PBS for 20 consecutive days. ELISA was used to detect the HMGB1 levels in the blood samples taken from the medial canthus vein of the mice, while co-immunoprecipitation and Western blot were used to detect the phosphorylation of HMGB1 and Akt in tumor tissue proteins, respectively. Results: The release levels of HMGB1 in the lactate group were (2 995.00±660.91) pg/ml and (696.33±22.03) pg/ml, after lactate treatment for 6 h and 12 h, respectively, both higher than those in the control group (485.00±105.83) pg/ml (P<0.001 and P=0.028, respectively). After lactate treatment for 6 h, the relative expression of HMGB1 protein in the cytoplasm of HGC-27 cells was 1.13±0.09, higher than that of the control group (0.83±0.07, P=0.001), while the relative expression of HMGB1 in the nucleus was 0.79±0.06, lower than that of the control group (1.07±0.06, P=0.007). The phosphorylation level of HMGB1 reached 1.41±0.09, which was higher than that of the control group (0.97±0.10, P=0.031). The phosphorylation level of Akt was 11.16±0.06, higher than that of the control group (0.91±0.022, P=0.002). The phosphorylation level and nuclear translocation of HMGB1 induced by lactate decreased obviously after Akt inhibition; the proliferation and migration abilities induced by lactate were also obviously inhibited after Akt inhibition. In vivo, the HMGB1 level in the peripheral blood was (1 280.70±389.66) pg/ml in the lactate group, which was obviously higher than that in the control group (595.11±44.75) pg/ml (P=0.008), and the phosphorylation levels of HMGB1 and Akt in tumor tissues in the lactate group were obviously enhanced compared with the control group. Conclusion: Lactate induces HMGB1 release through enhancing HMGB1 phosphorylation via the Akt signaling pathway.


Assuntos
Camundongos , Animais , Neoplasias Gástricas/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína HMGB1/metabolismo , Fosforilação , Ácido Láctico , Camundongos Endogâmicos BALB C , Transdução de Sinais
17.
Organ Transplantation ; (6): 397-2023.
Artigo em Chinês | WPRIM | ID: wpr-972930

RESUMO

Objective To unravel the possible mechanism of the role of recombinant human high mobility group box 1 (rhHMGB1) protein in regulating the angiogenesis of endothelial cells. Methods Endothelial cells were divided into the control group, bone marrow mesenchymal stem cells (MSC) supernatant group and rhHMGB1 group. The proliferation and survival of endothelial cells were detected by cell counting kit(CCK)-8 assay. The relative expression levels of vascular endothelial growth factor (VEGF), Yes-associated protein (YAP), CD31 and hypoxia inducible factor (HIF)-1α proteins were determined by Western blot. The relative expression levels of VEGF, YAP, CD31 and HIF-1α messenger RNA (mRNA) were detected by real-time fluorescent quantitative polymerase chain reaction (RT-qPCR). The migration ability of endothelial cells was assessed by Transwell chamber test. The localization of YAP was detected by immunofluorescence staining. Results Compared with the control group, the migration rate of endothelial cells was increased in the rhHMGB1 group (P < 0.05), and the cell migration rate was enhanced over time. Compared with the control group, the relative expression levels of VEGF and p-YAP proteins were up-regulated in the MSC supernatant group, and the differences were statistically significant (both P < 0.05). Compared with the control group, the relative expression levels of VEGF and HIF-1α proteins, VEGF and CD31 mRNA and YAP and p-YAP proteins were up-regulated, and YAP/p-YAP ratio was increased in the rhHMGB1 group, and the differences were statistically significant (all P < 0.05). Compared with the MSC supernatant group, the relative expression levels of CD31 mRNA and YAP protein were up-regulated, and the YAP/p-YAP ratio was increased in the rhHMGB1 group, and the differences were statistically significant (all P < 0.05). Conclusions Exogenous high-concentration rhHMGB1 may promote the migration ability of endothelial cells and up-regulate the expression levels of angiogenesis-related proteins by regulating the recruitment of YAP to the nucleus.

18.
Artigo em Inglês | WPRIM | ID: wpr-928995

RESUMO

High-mobility group box 1 (HMGB1) is a non-histone nuclear protein in most eukaryocytes. Inside the nucleus, HMGB1 plays an important role in several DNA events such as DNA repair, transcription, telomere maintenance, and genome stability. While outside the nucleus, it fulfils more complicated functions, including promoting cell proliferation, inflammation, angiogenesis, immune tolerance and immune escape, which may play a pro-tumoral role.Meanwhile, HMGB1 acts as an anti-tumoral protein by regulating immune cell recruitment and inducing immunogenic cell death (ICD) during the carcinogenesis process. Therefore, abnormal expression of HMGB1 is associated with oncogenesis, development, and metastasis of cancer, which may play a dual role of pro-tumor and anti-tumor.


Assuntos
Humanos , Carcinogênese , Proliferação de Células , Proteína HMGB1/metabolismo , Neoplasias/patologia , Neovascularização Patológica
19.
Artigo em Chinês | WPRIM | ID: wpr-957616

RESUMO

Objective:To investigate the effects of metformin on the expression of high mobility group box 1 (HMGB1) and receptor for advanced glycation end product(RAGE) around implants in type 2 diabetic rats and underlying mechanism on bone bonding. To investigate the effect of metformin on osseointegration in non-diabetic rats.Methods:Forty male SD rats aged 6 to 8 weeks were randomly divided into 4 groups by random number table with 10 rats each: normal group (T0 group), normal+ metformin group (T1 group), diabetic group (T2 group), and diabetic+ metformin group (T3 group). After type 2 diabetes model was established in T2 and T3 groups, pure titanium implants were implanted in bilateral tibial epiphyseal of all rats. On the same day, T1 and T3 groups were given 300 mg·kg -1·d -1 metformin, and other groups were gavaged with the same amount of normal saline. At the 4th and 8th week after surgery, 5 rats in each group were randomly sacrificed, and bone structure was examined using HE staining and micro-computed tomography (micro-CT). The expression of related factors was detected with immunohistochemistry and enzyme linked immunosorbent assay. Results:At the 4th and 8th week after surgery, the trabecular bone structure, new bone formation quality, and bone microparameters in T3 group were better than those in T2 group. Compared with T2 group, the expression of HMGB1 and RAGE was decreased, the content of osteocalcin was increased, and the expression of tumor necrosis factor-α was decreased (all P<0.01). 4 weeks after operation, bone volume/tissue volume and trabecular number in T1 group was higher than that in T0 group [(0.569±0.013)% vs (0.523±0.030)%, P=0.014; (1.695±0.059)/mm vs (1.569±0.050)/mm, P=0.007]. 8 weeks after operation, trabecular number in T1 group was higher than that in T0 group [(2.324±0.337)/mm vs (1.882±0.057)/mm, P=0.042]. Compared with T0 group, the content of RAGE in T1 group was significantly decreased [(35.49±2.77)ng/L vs (44.92±7.99)ng/L, P=0.005]. The osteocalcin content in T1 group was significantly higher than that in T0 group [(1.32±0.19)ng/L vs (0.89±0.26)ng/L, P=0.001]. Conclusions:Metformin can reduce the expression of HMGB1 and RAGE in type 2 diabetic rats, which may be one of the mechanisms promoting implant bone binding. Metformin has bone protective effect on non-diabetic rats.

20.
Artigo em Chinês | WPRIM | ID: wpr-941040

RESUMO

OBJECTIVE@#To investigate the effect of suppressing high-mobility group box 1 (HMGB1) on neuronal autophagy and apoptosis in rats after intracerebral hemorrhage (ICH) in rats.@*METHODS@#Rat models of ICH induced by intracerebral striatum injection of 0.2 U/mL collagenase Ⅳ were treated with 1 mg/kg anti-HMGB1 mAb or a control anti-IgG mAb injected via the tail immediately and at 6 h after the operation (n=5). The rats in the sham-operated group (with intracranial injection of 2 μL normal saline) and ICH model group (n=5) were treated with PBS in the same manner after the operation. The neurological deficits of the rats were evaluated using modified neurological severity score (mNSS). TUNEL staining was used to detect apoptosis of the striatal neurons, and the expressions of HMGB1, autophagy-related proteins (Beclin-1, LC3-Ⅱ and LC3-Ⅰ) and apoptosis-related proteins (Bcl-2, Bax and cleaved caspase-3) in the brain tissues surrounding the hematoma were detected using Western blotting. The expression of HMGB1 in the striatum was detected by immunohistochemistry, and serum level of HMGB1 was detected with ELISA.@*RESULTS@#The rat models of ICH showed significantly increased mNSS (P < 0.05), which was markedly lowered after treatment with anti- HMGB1 mAb (P < 0.05). ICH caused a significant increase of apoptosis of the striatal neurons (P < 0.05), enhanced the expressions of beclin-1, LC3-Ⅱ, Bax and cleaved caspase-3 (P < 0.05), lowered the expressions of LC3-Ⅰ and Bcl-2 (P < 0.05), and increased the content of HMGB1 (P < 0.05). Treatment with anti-HMGB1 mAb obviously lowered the apoptosis rate of the striatal neurons (P < 0.05), decreased the expressions of Beclin-1, LC3-Ⅱ, Bax and cleaved caspase-3 (P < 0.05), increased the expressions of LC3-Ⅰ and Bcl-2 (P < 0.05), and reduced the content of HMGB1 in ICH rats (P < 0.05).@*CONCLUSION@#Down- regulation of HMGB1 by anti-HMGB1 improves neurological functions of rats after ICH possibly by inhibiting autophagy and apoptosis of the neurons.


Assuntos
Animais , Ratos , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Proteína Beclina-1 , Caspase 3/metabolismo , Hemorragia Cerebral/terapia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA