Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros

Medicinas Complementares
País/Região como assunto
Intervalo de ano de publicação
1.
Chin J Integr Med ; 30(10): 917-926, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38532152

RESUMO

OBJECTIVE: To explore the protective effect and the underlying mechanism of silibinin (SIB), one of the active compounds from Silybum marianum (L.) Gaertn in endotoxemia. METHODS: Mouse peritoneal macrophage were isolated via intraperitoneally injection of BALB/c mice with thioglycolate medium. Cell viability was assessed using the cell counting kit-8, while cytotoxicity was determined through lactate dehydrogenase cytotoxicity assay. The protein expressions of interleukin (IL)-1 α, IL-1 ß, and IL-18 were determined by enzyme-linked immunosorbent assay. Intracellular lipopolysaccharide (LPS) levels were measured by employing both the limulus amoebocyte lysate assay and flow cytometry. Additionally, proximity ligation assay was employed for the LPS and caspase-11 interaction. Mice were divided into 4 groups: the control, LPS, high-dose-SIB (100 mg/kg), and low-dose-SIB (100 mg/kg) groups (n=8). Zebrafish were divided into 4 groups: the control, LPS, high-dose-SIB (200 εmol/L), and low-dose-SIB (100 εmol/L) groups (n=30 for survival experiment and n=10 for gene expression analysis). The expression of caspase-11, gasdermin D (GSDMD), and N-GSDMD was determined by Western blot and the expressions of caspy2, gsdmeb, and IL-1 ß were detected using quantitative real-time PCR. Histopathological observation was performed through hematoxylineosin staining, and protein levels in bronchoalveolar lavage fluid were quantified using the bicinchoninicacid protein assay. RESULTS: SIB noticeably decreased caspase-11 and GSDMD-mediated pyroptosis and suppressed the secretion of IL-1 α, IL-1 ß, and IL-18 induced by LPS (P<0.05). Moreover, SIB inhibited the translocation of LPS into the cytoplasm and the binding of caspase-11 and intracellular LPS (P<0.05). SIB also attenuated the expression of caspase-11 and N-terminal fragments of GSDMD, inhibited the relative cytokines, prolonged the survival time, and up-regulated the survival rate in the endotoxemia models (P<0.05). CONCLUSIONS: SIB can inhibit pyroptosis in the LPS-mediated endotoxemia model, at least in part, by inhibiting the caspase-11-mediated cleavage of GSDMD. Additionally, SIB inhibits the interaction of LPS and caspase-11 and inhibits the LPS-mediated up-regulation of caspase-11 expression, which relieves caspase-11-dependent cell pyroptosis and consequently attenuates LPS-mediated lethality.


Assuntos
Endotoxemia , Lipopolissacarídeos , Camundongos Endogâmicos BALB C , Piroptose , Silibina , Piroptose/efeitos dos fármacos , Endotoxemia/tratamento farmacológico , Endotoxemia/induzido quimicamente , Animais , Silibina/farmacologia , Caspases Iniciadoras/metabolismo , Peixe-Zebra , Camundongos , Masculino , Substâncias Protetoras/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo
2.
Artif Cells Nanomed Biotechnol ; 51(1): 33-40, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36656591

RESUMO

Sepsis is a devastating complication of infection and injury that, through widespread endothelial dysfunction, can cause perfusion deficits and multi-organ failure. To address the recognised need for therapeutics targetting the endothelial barrier, a topical formulation (CUR; VASCEPTOR™; Vascarta Inc, Summit, NJ) was developed to transdermally deliver bio-active concentrations of curcumin-an anti-inflammatory and nitric oxide promoter. Male, Sprague Dawley rats were treated daily with lipopolysaccharide (LPS, 10 mg/kg, IP) to induce endotoxemia, and topical applications of Vehicle Control (LPS + VC; N = 7) or Curcumin (LPS + CUR; N = 7). A third group received neither LPS nor treatment (No-LPS; N = 8). After 72 h, animals were surgically prepared for measurements of physiology and endothelial dysfunction in the exteriorised spinotrapezius muscle through the extravasation of 67 kDa TRITC-BSA (albumin) and 500 kDa FITC-dextran (dextran). At 72 h, LPS + VC saw weight loss, and increases to pulse pressure, lactate, pCO2, CXCL5 (vs No-LPS) and IL-6 (vs 0 h; p < 0.05). LPS + CUR was similar to No-LPS, but with hypotension. Phenylephrine response was increased in LPS + CUR. Regarding endothelial function, LPS + CUR albumin and dextran extravasation were significantly reduced versus LPS + VC suggesting that Curcumin mitigated endotoxemic endothelial dysfunction. The speculated mechanisms are nitric oxide modulation of the endothelium and/or an indirect anti-inflammatory effect.


Assuntos
Curcumina , Endotoxemia , Animais , Masculino , Ratos , Albuminas , Anti-Inflamatórios , Curcumina/farmacologia , Dextranos , Endotélio , Endotoxemia/induzido quimicamente , Endotoxemia/tratamento farmacológico , Lipopolissacarídeos , Óxido Nítrico , Ratos Sprague-Dawley
3.
Oxid Med Cell Longev ; 2023: 1464853, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36647427

RESUMO

Background and Aims: Endotoxemia (ET) is a common critical illness in patients receiving intensive care and is associated with high mortality and prolonged hospital stay. The intestinal epithelial cell dysfunction is regarded as the "engine" of deteriorated ET. Although electroacupuncture (EA) can mitigate endotoxin-induced intestinal epithelial cell dysfunction in ET, the mechanism through which EA improves endotoxin-induced intestinal injury for preventing ET deterioration needs further investigation. Methods: An in vivo ET model was developed by injecting lipopolysaccharide (LPS) in wild-type and PINK1-knockout mice. An in vitro model was also established by incubating epithelial cells in the serum samples obtained from both groups of mice. Hemin and zinc protoporphyrin IX (ZnPP) were applied to activate or inhibit heme oxygenase 1 (HO-1) production. EA treatment was performed for 30 min consecutively for 5 days before LPS injection, and on the day of the experiment, EA was performed throughout the process. Samples were harvested at 6 h after LPS induction for analyzing tissue injury, oxidative stress, ATP production, activity of diamine oxidase (DAO), and changes in the levels of HO-1, PTEN-induced putative kinase 1 (PINK1), mitochondrial fusion and fission marker gene, caspase-1, and interleukin 1 beta (IL-1ß). Results: In the wild-type models (both in vivo and vitro), EA alleviated LPS-induced intestinal injury and mitochondrial dysfunction, as indicated by decreased reactive oxygen species (ROS) production and oxygen consumption rate (OCR) and reduced levels of mitochondrial fission proteins. EA treatment also boosted histopathological morphology, ATP levels, DAO activity, and levels of mitochondrial fusion proteins in vivo and vitro. The effect of EA was enhanced by hemin but suppressed by Znpp. However, EA + AP, Znpp, or hemin had no effects on the LPS-induced, PINK1-knocked out mouse models. Conclusion: EA may improve the HO-1/PINK1 pathway-mediated mitochondrial dynamic balance to protect the intestinal barrier in patients with ET.


Assuntos
Eletroacupuntura , Endotoxemia , Heme Oxigenase-1 , Proteínas Quinases , Animais , Camundongos , Trifosfato de Adenosina , Endotoxemia/induzido quimicamente , Endotoxemia/terapia , Endotoxinas , Heme Oxigenase-1/metabolismo , Hemina/farmacologia , Lipopolissacarídeos/toxicidade , Dinâmica Mitocondrial
4.
J Ethnopharmacol ; 302(Pt B): 115922, 2023 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-36414212

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Qingdu Decoction (QDT) is a traditional Chinese medicine (TCM) that was derived from Xiaochengqi Decoction, a famous decoction documented in the book of Treatise on Exogenous Febrile Disease in the Eastern Han Dynasty. According to our years of clinical application, QDT showed satisfactory efficacy in the treatment of endotoxemia in acute-on-chronic liver failure (ACLF). However, the underlying molecular mechanisms remain largely unknown. AIM OF STUDY: In this study, we aimed to systematically evaluate the intervention effect of QDT on endotoxemia in rats and further clarify its potential regulatory mechanism. MATERIALS AND METHODS: The rat model of ACLF endotoxemia was induced by TAA and LPS + D-Gal. Then the rats were treated with clinical doses of QDT and lactulose. The rats were divided into four groups: CG, MG, QG and LG. The target microRNA was screened by high-throughput sequencing. The rat weight, liver index, hepatointestinal phenotype, serum biochemical indexes, mast cell activity, and hepatointestinal histopathology were used to evaluate the intervention effect. Western blot analysis was used to detect the expression levels of MAZ and its downstream genes ZO-1 and Occludin, and the expression levels of Zonulin and its downstream gene EGFR in colon. Finally, the expression of the miR-34c, MAZ, ZO-1, Occludin, miR-122a, Zonulin, and EGFR in colon was detected by qRT-PCR to further confirm the mechanism of the miR-34c/MAZ/TJs pathway and the miR-122a/Zonulin/EGFR pathway. RESULTS: The rat weight, liver index, liver and colon phenotype, and serum biochemical indexes showed that QDT could significantly reduce liver and intestine injury and inhibit the progress of ACLF and endotoxemia. Toluidine blue staining and cytokine indexes showed that QDT could inhibit the activity of MCs and reduce the release of inflammatory factors. Mechanistically, QDT can inhibit the activity of MCs, activate miR-34c/MAZ/TJs pathway and miR-122a/Zonulin/EGFR pathway in colon, promote the recovery of intestinal barrier homeostasis, reduce and restore the damage of endotoxemia. CONCLUSION: Our results suggested that QDT can significantly reduce rat ACLF endotoxemia by regulating the miR-34c/MAZ/TJs pathway and the miR-122a/Zonulin/EGFR pathway in colon.


Assuntos
Insuficiência Hepática Crônica Agudizada , Medicamentos de Ervas Chinesas , Endotoxemia , MicroRNAs , Animais , Ratos , Endotoxemia/induzido quimicamente , Endotoxemia/tratamento farmacológico , Receptores ErbB , Lipopolissacarídeos , MicroRNAs/genética , Ocludina , Transdução de Sinais , Medicamentos de Ervas Chinesas/farmacologia
5.
Front Immunol ; 13: 825171, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35281027

RESUMO

Sepsis, a systemic inflammatory response to pathogenic factors, is a difficult to treat life-threatening condition associated with cytokine and eicosanoid storms and multi-organ damage. Omega-3 polyunsaturated fatty acids, such as eicosapentaenoic (EPA) and docosahexaenoic acid, are the precursors of potent anti-inflammatory lipid mediators, including 17,18-epoxyeicosatetraenoic acid (17,18-EEQ), the main metabolite of EPA generated by cytochrome P450 epoxygenases. Searching for novel therapeutic or preventative agents in sepsis, we tested a metabolically robust synthetic analog of 17,18-EEQ (EEQ-A) for its ability to reduce mortality, organ damage, and pro-inflammatory cytokine transcript level in a mouse model of lipopolysaccharide (LPS)-induced endotoxemia, which is closely related to sepsis. Overall survival significantly improved following preventative EEQ-A administration along with decreased transcript level of pro-inflammatory cytokines. On the other hand, the therapeutic protocol was effective in improving survival at 48 hours but insignificant at 72 hours. Histopathological analyses showed significant reductions in hemorrhagic and necrotic damage and infiltration in the liver. In vitro studies with THP-1 and U937 cells showed EEQ-A mediated repression of LPS-induced M1 polarization and enhancement of IL-4-induced M2 polarization of macrophages. Moreover, EEQ-A attenuated the LPS-induced decline of mitochondrial function in THP-1 cells, as indicated by increased basal respiration and ATP production as well as reduction of the metabolic shift to glycolysis. Taken together, these data demonstrate that EEQ-A has potent anti-inflammatory and immunomodulatory properties that may support therapeutic strategies for ameliorating the endotoxemia.


Assuntos
Endotoxemia , Ácidos Graxos Ômega-3 , Animais , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/farmacologia , Citocinas , Eicosanoides , Endotoxemia/induzido quimicamente , Endotoxemia/tratamento farmacológico , Ácidos Graxos Ômega-3/uso terapêutico , Lipopolissacarídeos/toxicidade , Camundongos
6.
Food Funct ; 13(2): 781-794, 2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-34981106

RESUMO

Consumption of high fat diets (HFD) and the associated metabolic endotoxemia can initiate liver inflammation and lipid deposition that with time can progress to non-alcoholic fatty liver disease (NAFLD). We previously observed that 14 weeks supplementation with the anthocyanidins cyanidin and delphinidin mitigated HFD-induced metabolic endotoxemia and liver insulin resistance, steatosis, inflammation and oxidative stress. This work investigated if a 4-week supplementation of mice with a cyanidin- and delphinidin-rich extract (CDRE) could mitigate or reverse HFD (60% calories from lard fat)-induced liver steatosis and inflammation. After a first 4-weeks period on the HFD, mice showed increased endotoxemia and activation of liver proinflammatory signaling cascades. Supplementation with CDRE between weeks 4 and 8 did not mitigate liver steatosis or the altered lipid and glucose plasma levels. However, CDRE supplementation reverted HFD-induced metabolic endotoxemia, in parallel with the mitigation of the overexpression of hepatic TLR2 and TLR4, and of the activation of: (i) NF-κB, (ii) AP-1 and upstream mitogen-activated kinases p38 and ERK1/2, and (iii) HIF-1. Thus, even a short-term consumption of cyanidin and delphinidin could help mitigate the adverse consequences, i.e. metabolic endotoxemia and associated liver inflammation, triggered by the regular consumption of diets rich in fat.


Assuntos
Antocianinas/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Endotoxemia/tratamento farmacológico , Inflamação/tratamento farmacológico , Ração Animal , Animais , Suplementos Nutricionais , Endotoxemia/induzido quimicamente , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Camundongos , NF-kappa B , Estresse Oxidativo , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
7.
Biomed Pharmacother ; 144: 112345, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34678721

RESUMO

Potentilla discolor Bunge (PD) is a traditional Chinese medicine which has been widely used for the treatment of various inflammatory diseases (e.g., diarrhea, fever and furuncle). However, few studies focused on its effect on classical inflammation. This study aimed to investigate the anti-inflammatory effect and potential mechanism of the ethanol extract of the whole herbs of PD (EPD) in lipopolysaccharide (LPS)-induced inflammatory models. The obtained results showed that EPD decreased supernatant NO, tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) in LPS-activated RAW264.7 cells and mouse peritoneal macrophages. Moreover, its effect on NO was attributed to the suppression of iNOS expression rather than its activity. At the transcriptional level, EPD suppressed iNOS, TNF-α and MCP-1 mRNA expressions in LPS-stimulated RAW264.7 cells. Further study showed that EPD didn't affect the phosphorylation and degradation of IκBα, but yet impeded the nuclear translocation of p65 to inhibit NF-κB activation. Meanwhile, it also prevented JNK, ERK1/2 and p38 phosphorylation to dampen the activation of AP-1. In endotoxemia mouse model, EPD not only decreased interleukin-6, TNF-α and MCP-1 levels in serum, but also potently ameliorated diarrhea. These findings provide the theoretical basis for PD to treat inflammatory diseases, especially intestinal inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Endotoxemia/prevenção & controle , Inflamação/prevenção & controle , Macrófagos/efeitos dos fármacos , NF-kappa B/metabolismo , Extratos Vegetais/farmacologia , Potentilla , Fator de Transcrição AP-1/metabolismo , Animais , Anti-Inflamatórios/isolamento & purificação , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Diarreia/induzido quimicamente , Diarreia/imunologia , Diarreia/metabolismo , Diarreia/prevenção & controle , Modelos Animais de Doenças , Endotoxemia/induzido quimicamente , Endotoxemia/imunologia , Endotoxemia/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Lipopolissacarídeos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Inibidor de NF-kappaB alfa/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , Extratos Vegetais/isolamento & purificação , Potentilla/química , Células RAW 264.7 , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
8.
J Food Biochem ; 45(7): e13805, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34096077

RESUMO

High mobility group box 1 (HMGB1) is a well-defined mediator involved in the pathophysiologic response to endotoxemia and sepsis. However, the mechanisms and therapeutic agents that could prevent its release are not fully elucidated. Here, the present study demonstrates that the ginseng leaf extract (GLE) regulates lipopolysaccharide (LPS)-triggered release of HMGB1 in macrophages and endotoxemic animal model. Treatment of RAW264.7 macrophages with GLE significantly inhibited the release of HMGB1 stimulated by LPS. GLE also suppressed the generation of nitric oxide (NO) and expression of inducible NO synthase (iNOS) in a dose-dependent manner. These effects of GLE were accompanied by inhibition of HMGB1 release stimulated by LPS, indicating a potential mechanism by which GLE regulates HMGB1 release through NO signaling. Furthermore, induction of suppressor of cytokine signaling 1 by GLE-mediated GLE-dependent suppression of HMGB1 release and NO/iNOS induction by inhibiting Janus kinase 2/signal transducer and activator of transcription 1 signal in RAW 264.7 cells exposed to LPS. Finally, administration of the GLE ameliorated the survival rate of LPS-injected endotoxemic mice in a NO-dependent manner. Thus, GLE may block the LPS-stimulated release of HMGB1 by regulating cellular signal networks, thereby providing a therapeutic strategy for endotoxemia as a functional food. PRACTICAL APPLICATIONS: High mobility group box 1 (HMGB1) is released into the extracellular milieu when immune cells are exposed to pathogen-related molecules such as lipopolysaccharide (LPS), in which it acts as a critical mediator of lethality in sepsis and endotoxemia. The extract of ginseng leaf, which is a part that can be easily thrown away, ameliorated the survival rate of endotoxemic mice by inhibiting HMGB1 secretion in a NO-dependent manner. Thus, this study suggests that ginseng leaf can be used as a functional food by resolving the immune responses in the pathology of endotoxemia.


Assuntos
Endotoxemia , Proteína HMGB1 , Panax , Animais , Endotoxemia/induzido quimicamente , Endotoxemia/tratamento farmacológico , Camundongos , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Células RAW 264.7
9.
Mol Nutr Food Res ; 64(17): e2000532, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32729948

RESUMO

SCOPE: Soy protein is a high-quality protein and its consumption has been associated with a reduction of serum cholesterol and triglycerides and an improvement in insulin resistance. However, it is not known whether the effects of soy protein are mediated by the gut microbiota. Thus, the aim of this study is to assess whether using antibiotics to partially eradicate the gut microbiota can prevent the beneficial effects of soy protein in rats. METHODS AND RESULTS: Thus, rats are fed one of the following diets for 16 weeks: casein control, soy protein control, high-fat casein, and high-fat soy protein. The rats are then treated for 4 weeks with antibiotics. Body weight and composition, energy expenditure, glucose tolerance test, metabolic endotoxemia, and gut microbiota are measured before and after treatment with antibiotic. The results show that soy protein consumption decreases weight gain, body fat, metabolic endotoxemia, and increases energy expenditure and glucose tolerance. Antibiotic treatment suppresses all these metabolic effects. These changes are accompanied by modifying the diversity and taxonomy of the gut microbiota. CONCLUSION: In conclusion, the evidence suggests that the health benefits of soy protein are partly dependent of the gut microbiota.


Assuntos
Antibacterianos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Proteínas de Soja/farmacologia , Tecido Adiposo/efeitos dos fármacos , Ampicilina/efeitos adversos , Ampicilina/farmacologia , Animais , Antibacterianos/efeitos adversos , Biomarcadores/metabolismo , Composição Corporal/efeitos dos fármacos , Caseínas/farmacologia , Dieta Hiperlipídica/efeitos adversos , Endotoxemia/induzido quimicamente , Metabolismo Energético/efeitos dos fármacos , Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal/fisiologia , Inflamação/genética , Inflamação/metabolismo , Masculino , Neomicina/efeitos adversos , Neomicina/farmacologia , Ratos Wistar , Aumento de Peso/efeitos dos fármacos
10.
J Ethnopharmacol ; 259: 112882, 2020 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-32325181

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Jiao Mei Gu (JMG), Cayratia albifolia C.L.Li, is a type of Dong plant widely growing in Dong autonomous counties, Hunan province, China. As a type of traditional herbal medicine, the root of JMG plant has been used to treat inflammatory-related diseases such as arthritis because of its prominent anti-inflammatory effects in Dong medicine. AIM OF THE STUDY: This work investigated the anti-inflammatory effects and mechanisms of the water extract from the root of JMG on lipopolysaccharide (LPS)-induced inflammatory models. METHODS: Endotoxemia was induced in C57BL/6 mice by intraperitoneal injection of LPS (20 mg/kg), meanwhile intraperitoneal administration of safe doses of JMG. The survival curve of mice was determined. Serum inflammatory cytokines were detected by the Bio-Plex Mouse Cytokine 23-Plex Panel Kit and enzyme-linked immunosorbent assay (ELISA) at 6 h after drug treatment. Hematoxylin-eosin (HE) staining of important organs was completed at 24 h after treatment. The mechanism of inflammatory action was investigated in vitro on LPS-stimulated macrophages. Macrophage inflammation was then induced using 10 µg/mL LPS. The anti-inflammatory effect of JMG was investigated by the quantitative polymerase chain reaction (qPCR) and ELISA. The anti-inflammatory mechanism was determined using western blotting, the electrophoretic mobility shift assay, and immunocytochemistry. Finally, the antimicrobial activity of JMG was verified by survival experiments in vivo and by bacterial culture experiments in vitro. RESULTS: A 200 mg/kg water extract of JMG was safe for mice and had a significant protective effect on LPS-induced sepsis. Organ damage of heart, liver, lung and kidney was also significantly reduced at 24 h in the JMG group, when compared with the LPS group. The serum MIP-1α (CCL-3), MIP-1ß (CCL-4), IL-1ß, and TNFα cytokines were significantly decreased at 6 h in the JMG group, when compared with the LPS group. In a similar manner, 0.2µg/ml JMG significantly reduced mRNA and protein levels of MIP-1α (CCL-3), MIP-1ß (CCL-4), IL-1ß, and TNFα in LPS-stimulated macrophage. JMG treatment inhibited the phosphorylation of NF-κB p65 and reduced nuclear transduction, thus reducing transcriptional activity. At the same time, we showed that JMG had no protective effect on Escherichia coli-induced sepsis, as well as no antimicrobial activity. CONCLUSIONS: Our results showed that a water-soluble extract of JMG inhibited LPS-induced inflammation via attenuating the NF-κB signaling pathway, which provides an important rationale for the treatment of inflammation-related diseases.


Assuntos
Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Endotoxemia/tratamento farmacológico , NF-kappa B/metabolismo , Animais , Citocinas/sangue , Endotoxemia/induzido quimicamente , Escherichia coli , Proteínas I-kappa B/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
J Biochem Mol Toxicol ; 33(12): e22406, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31593353

RESUMO

Soybean Bowman-Birk protease inhibitor (BBI) and genistein, two biological compounds from soybean, are well-known for their anti-inflammatory, antioxidant, and anticancer activities. The aim of this study was designing a BBI-genistein conjugate and then investigating its protective effect on lipopolysaccharide (LPS)-induced inflammation in BALB/c mice, compared with the effects of combination of BBI and genistein. BBI was purified from soybean and the BBI-genistein conjugate was synthesized. The BALB/c mice were intraperitoneally treated 2 hours before LPS induction. Our results showed that treatment with the combination of BBI and genistein greatly led to more reduced serum levels of tumor necrosis factor (TNF)-α and interferon (IFN)-γ compared with the treatments of BBI alone, the BBI-genistein conjugate, and genistein alone, respectively. Moreover, the expression of TNF-α and IFN-γ in the splenocytes was significantly downregulated along with improving host survival against the LPS-induced lethal endotoxemia in the same way. Our data support a new combined therapy using BBI and genistein, as natural anti-inflammatory agents, to develop a new drug for inflammatory diseases.


Assuntos
Anti-Inflamatórios/uso terapêutico , Endotoxemia/tratamento farmacológico , Genisteína/uso terapêutico , Glycine max/química , Extratos Vegetais/uso terapêutico , Inibidor da Tripsina de Soja de Bowman-Birk/uso terapêutico , Animais , Combinação de Medicamentos , Endotoxemia/induzido quimicamente , Genisteína/administração & dosagem , Inflamação/metabolismo , Injeções Intraperitoneais , Interferon gama/antagonistas & inibidores , Interferon gama/sangue , Estimativa de Kaplan-Meier , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/antagonistas & inibidores , Baço/patologia , Taxa de Sobrevida , Resultado do Tratamento , Inibidor da Tripsina de Soja de Bowman-Birk/administração & dosagem , Inibidor da Tripsina de Soja de Bowman-Birk/isolamento & purificação , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/sangue
12.
Cell Rep ; 28(1): 218-230.e7, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31269442

RESUMO

Classical activation of macrophages (M(LPS+IFNγ)) elicits the expression of inducible nitric oxide synthase (iNOS), generating large amounts of NO and inhibiting mitochondrial respiration. Upregulation of glycolysis and a disrupted tricarboxylic acid (TCA) cycle underpin this switch to a pro-inflammatory phenotype. We show that the NOS cofactor tetrahydrobiopterin (BH4) modulates IL-1ß production and key aspects of metabolic remodeling in activated murine macrophages via NO production. Using two complementary genetic models, we reveal that NO modulates levels of the essential TCA cycle metabolites citrate and succinate, as well as the inflammatory mediator itaconate. Furthermore, NO regulates macrophage respiratory function via changes in the abundance of critical N-module subunits in Complex I. However, NO-deficient cells can still upregulate glycolysis despite changes in the abundance of glycolytic intermediates and proteins involved in glucose metabolism. Our findings reveal a fundamental role for iNOS-derived NO in regulating metabolic remodeling and cytokine production in the pro-inflammatory macrophage.


Assuntos
Ciclo do Ácido Cítrico , Inflamação/metabolismo , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Succinatos/metabolismo , Animais , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Ciclo do Ácido Cítrico/efeitos dos fármacos , Transporte de Elétrons/efeitos dos fármacos , Endotoxemia/induzido quimicamente , Endotoxemia/metabolismo , GTP Cicloidrolase/genética , GTP Cicloidrolase/metabolismo , Glicólise/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-1beta/metabolismo , Isocitrato Desidrogenase/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Infecções por Mycobacterium/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteoma/genética , Proteoma/metabolismo , Ácido Succínico/metabolismo , Espectrometria de Massas em Tandem
13.
Int J Biol Macromol ; 116: 753-764, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29775719

RESUMO

The selenium polysaccharides (SPS) by Oudemansiella radicata were isolated, and two hydrolysates (enzymatic-SPS (ESPS) and acidic-SPS (ASPS)) were obtained by enzymolysis and acidolysis. The endo-toxemic-induced lung and kidney damages were successfully established by the lipopolysaccharides (LPS) injection (5 mg/kg/d). The in vivo mice experiments indicated that the ESPS showed superior antioxidant and protective effects against the LPS-toxicities by increasing the antioxidant activities, reducing lipid peroxidation, improving the inflammatory response and elevating the kidney and lung functions than SPS and ASPS. Furthermore, the selenium contents and structure of SPS, ESPS and ASPS were investigated through FAAS, GC, UV, HPGPC, FT-IR, NMR and the results indicated that SPS, ESPS and ASPS were homogeneous heteropolysaccharide with an average molecular weight of 3.12 × 104, 1.77 × 104, 1.41 × 104 Da, respectively. The conclusions demonstrated that the polysaccharides by O. radicata might be suitable for functional foods and natural drugs in preventing the endo-toxemia and its complications, and providing references for exploitation and utilization of natural substances.


Assuntos
Lesão Pulmonar Aguda , Agaricales/química , Endotoxemia , Polissacarídeos Fúngicos , Lipopolissacarídeos/toxicidade , Selênio , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Endotoxemia/induzido quimicamente , Endotoxemia/tratamento farmacológico , Endotoxemia/metabolismo , Endotoxemia/patologia , Polissacarídeos Fúngicos/química , Polissacarídeos Fúngicos/farmacologia , Masculino , Camundongos , Selênio/química , Selênio/farmacologia
14.
Sci Rep ; 7(1): 4716, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28680065

RESUMO

Current efforts are directed to reducing the gut dysbiosis and inflammation produced by obesity. The purpose of this study was to investigate whether consuming nopal, a vegetable rich in dietary fibre, vitamin C, and polyphenols can reduce the metabolic consequences of obesity by modifying the gut microbiota and preventing metabolic endotoxemia in rats fed a high fat and sucrose diet. With this aim, rats were fed a high fat diet with 5% sucrose in the drinking water (HFS) for 7 months and then were fed for 1 month with HFS + 5% nopal (HFS + N). The composition of gut microbiota was assessed by sequencing the 16S rRNA gene. Nopal modified gut microbiota and increased intestinal occludin-1 in the HFS + N group. This was associated with a decrease in metabolic endotoxemia, glucose insulinotropic peptide, glucose intolerance, lipogenesis, and metabolic inflexibility. These changes were accompanied by reduced hepatic steatosis and oxidative stress in adipose tissue and brain, and improved cognitive function, associated with an increase in B. fragilis. This study supports the use of nopal as a functional food and prebiotic for its ability to modify gut microbiota and to reduce metabolic endotoxemia and other obesity-related biochemical abnormalities.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Endotoxemia/prevenção & controle , Microbioma Gastrointestinal/efeitos dos fármacos , Obesidade/metabolismo , Opuntia/química , Preparações de Plantas/administração & dosagem , Sacarose/efeitos adversos , Animais , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/isolamento & purificação , DNA Bacteriano/genética , DNA Ribossômico/genética , Endotoxemia/induzido quimicamente , Proteínas Substratos do Receptor de Insulina/metabolismo , Fígado/metabolismo , Masculino , Preparações de Plantas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Ribossômico 16S/genética , Ratos , Análise de Sequência de DNA/métodos
15.
BMC Complement Altern Med ; 17(1): 212, 2017 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-28403838

RESUMO

BACKGROUND: Dalbergia odorifera T. Chen (Leguminosae) is an indigenous medicinal herb that is widely used as a popular remedy in northern and eastern Asia. However, the cellular mechanisms underlying the biological activity of D. odorifera are not fully elucidated. METHODS: Anti-inflammatory effect of D. odorifera extract (DOE) was determined through intraperitoneal injection in a mouse model of endotoxemia induced by lipopolysaccharide (LPS). RAW 264.7 cells, a murine macrophage, were also treated with LPS to generate a cellular model of inflammation, and investigated the anti-inflammatory activity and underlying mechanisms of DOE and its constituent isoliquiritigenin. RESULTS: DOE dose-dependently inhibited LPS-induced release of high mobility group box 1 (HMGB1), a late proinflammatory cytokine, and decreased cytosolic translocation of HMGB1 in RAW264.7 cells. This inhibitory effect of DOE on HMGB1 release was observed in cells treated with DOE before or after LPS treatment, suggesting that DOE is effective for both treatment and prevention. In addition, DOE significantly inhibited LPS-induced formation of nitric oxide (NO) and expression of inducible NO synthase (iNOS) in a dose-dependent manner. These effects of DOE were accompanied by suppression of HMGB1 release triggered by LPS, suggesting a possible mechanism by which DOE modulates HMGB1 release through NO signaling. Isoriquiritigenin, a constituent of DOE, also attenuated LPS-triggered NO formation and HMGB1 release in RAW264.7 cells, indicating that isoriquiritigenin is an indexing molecule for the anti-inflammatory properties of DOE. Furthermore, c-Jun N-terminal kinase, but not extracellular signal-regulated kinase and p38, mediated DOE-dependent inhibition of HMGB1 release and NO/iNOS induction in RAW 264.7 cells exposed to LPS. Notably, administration of DOE ameliorated survival rates in a mouse model of endotoxemia induced by LPS, where decreased level of circulating HMGB1 was observed. CONCLUSION: These results suggest that DOE confers resistance to LPS-triggered inflammation through NO-mediated inhibitory effects on HMGB1 release.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Dalbergia/química , Endotoxemia/tratamento farmacológico , Proteína HMGB1/antagonistas & inibidores , Fitoterapia , Extratos Vegetais/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/isolamento & purificação , Modelos Animais de Doenças , Endotoxemia/induzido quimicamente , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipopolissacarídeos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos
16.
Naunyn Schmiedebergs Arch Pharmacol ; 390(5): 483-492, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28124089

RESUMO

Celecoxib, a selective cyclooxygenase-2 inhibitor, produces thrombotic events in patients predisposed to cardiovascular risk factors. One theory reported an increase in endothelial expression of tissue factor (TF) as a predisposing factor. This work explored the effect of evening primrose oil (EPO), a source of prostaglandin E1, and forskolin (a cyclic adenosine monophosphate stimulator) against the prothrombotic effect of celecoxib in mice. Lipopolysaccharide mouse model of endotoxemia was used to induce an upregulation of TF activity. Male mice received celecoxib (25 mg/kg), celecoxib plus EPO, or celecoxib plus forskolin for 4 weeks and then subjected to a prothrombotic challenge in the form of an intraperitoneal injection of lipopolysaccharide. Results showed an increase in plasma TF activity, endothelial TF expression, and thrombin-antithrombin (TAT) but lower antithrombin III (ATIII) level in mice that received celecoxib in comparison to those that received the vehicle. Adding EPO or forskolin to celecoxib regimen significantly decreased the prothrombotic effect of celecoxib. A positive correlation (r = 0.8501) was found between TF activity and TAT. Co-administration of EPO or forskolin decreased the activity of TF and mitigated the prothrombotic effect of celecoxib. Therefore, these combinations may have the utility to abrogate the prothrombotic adverse effect of celecoxib in clinical setting.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Celecoxib , Colforsina/farmacologia , Inibidores de Ciclo-Oxigenase 2 , Endotoxemia/induzido quimicamente , Fibrinolíticos/farmacologia , Ácidos Linoleicos/farmacologia , Lipopolissacarídeos , Óleos de Plantas/farmacologia , Tromboplastina/metabolismo , Trombose/prevenção & controle , Ácido gama-Linolênico/farmacologia , Animais , Antitrombina III/metabolismo , Modelos Animais de Doenças , Endotoxemia/sangue , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Oenothera biennis , Peptídeo Hidrolases/sangue , Trombose/sangue , Trombose/induzido quimicamente , Regulação para Cima
17.
PLoS One ; 12(1): e0169614, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28125622

RESUMO

Calgranulin genes (S100A8, S100A9 and S100A12) play key immune response roles in inflammatory disorders, including cardiovascular disease. Long-chain omega-3 polyunsaturated fatty acids (LC n-3 PUFA) may have systemic and adipose tissue-specific anti-inflammatory and cardio-protective action. Interactions between calgranulins and the unsaturated fatty acid arachidonic acid (AA) have been reported, yet little is known about the relationship between calgranulins and the LC n-3 PUFA eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). We explored tissue-specific action of calgranulins in the setting of evoked endotoxemia and n-3 PUFA supplementation. Expression of calgranulins in adipose tissue in vivo was assessed by RNA sequencing (RNASeq) before and after n-3 PUFA supplementation and evoked endotoxemia in the fenofibrate and omega-3 fatty acid modulation of endotoxemia (FFAME) Study. Subjects received n-3 PUFA (n = 8; 3600mg/day EPA/DHA) or matched placebo (n = 6) for 6-8 weeks, before completing an endotoxin challenge (LPS 0.6 ng/kg). Calgranulin genes were up-regulated post-LPS, with greater increase in n-3 PUFA (S100A8 15-fold, p = 0.003; S100A9 7-fold, p = 0.003; S100A12 28-fold, p = 0.01) compared to placebo (S100A8 2-fold, p = 0.01; S100A9 1.4-fold, p = 0.4; S100A12 5-fold, p = 0.06). In an independent evoked endotoxemia study, calgranulin gene expression correlated with the systemic inflammatory response. Through in vivo and in vitro interrogation we highlight differential responses in adipocytes and mononuclear cells during inflammation, with n-3 PUFA leading to increased calgranulin expression in adipose, but decreased expression in circulating cells. In conclusion, we present a novel relationship between n-3 PUFA anti-inflammatory action in vivo and cell-specific modulation of calgranulin expression during innate immune activation.


Assuntos
Antioxidantes/administração & dosagem , Calgranulina A/genética , Calgranulina B/genética , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácido Eicosapentaenoico/administração & dosagem , Endotoxemia/prevenção & controle , Proteína S100A12/genética , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Adulto , Nádegas , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Estudos de Casos e Controles , Endotoxemia/induzido quimicamente , Endotoxemia/genética , Endotoxemia/patologia , Feminino , Regulação da Expressão Gênica , Voluntários Saudáveis , Humanos , Lipopolissacarídeos/farmacologia , Masculino , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/patologia , Proteína S100A12/metabolismo , Transdução de Sinais
18.
Br J Pharmacol ; 174(12): 1620-1632, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27435156

RESUMO

BACKGROUND AND PURPOSE: Excessive inflammation in sepsis causes microvascular thrombosis and thrombocytopenia associated with organ dysfunction and high mortality. The present studies aimed to investigate whether inhibition of dipeptidyl peptidase-4 (DPP-4) and supplementation with glucagon-like peptide-1 (GLP-1) receptor agonists improved endotoxaemia-associated microvascular thrombosis via immunomodulatory effects. EXPERIMENTAL APPROACH: Endotoxaemia was induced in C57BL/6J mice by a single injection of LPS (17.5 mg kg-1 for survival and 10 mg kg-1 for all other studies). For survival studies, treatment was started 6 h after LPS injection. For all other studies, drugs were injected 48 h before LPS treatment. KEY RESULTS: Mice treated with LPS alone showed severe thrombocytopenia, microvascular thrombosis in the pulmonary circulation (fluorescence imaging), increased LDH activity, endothelial dysfunction and increased markers of inflammation in aorta and whole blood (leukocyte-dependent oxidative burst, nitrosyl-iron haemoglobin, a marker of nitrosative stress, and expression of inducible NOS). Treatment with the DPP-4 inhibitor linagliptin or the GLP-1 receptor agonist liraglutide, as well as genetic deletion of DPP-4 (DPP4-/- mice) improved all these parameters. In GLP-1 receptor-deficient mice, both linagliptin and liraglutide lost their beneficial effects and improvement of prognosis. Incubation of platelets and cultured monocytes (containing GLP-1 receptor protein) with GLP-1 receptor agonists inhibited the monocytic oxidative burst and platelet activation, with a GLP-1 receptor-dependent elevation of cAMP levels and PKA activation. CONCLUSIONS AND IMPLICATIONS: GLP-1 receptor activation in platelets by linagliptin and liraglutide strongly attenuated endotoxaemia-induced microvascular thrombosis and mortality by a cAMP/PKA-dependent mechanism, preventing systemic inflammation, vascular dysfunction and end organ damage. LINKED ARTICLES: This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.


Assuntos
Endotoxemia/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Microvasos/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais , Trombose Venosa/metabolismo , Animais , Dipeptidil Peptidase 4/deficiência , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Endotoxemia/induzido quimicamente , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/deficiência , Lipopolissacarídeos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microvasos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Trombose Venosa/induzido quimicamente
19.
Transfusion ; 57(1): 53-59, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27696454

RESUMO

BACKGROUND: Transfusion of a single unit of stored red blood cells (RBCs) has been hypothesized to induce supra-physiological levels of non-transferrin bound iron (NTBI), which may enhance inflammation and act as a nutrient for bacteria. We investigated the relation between RBC storage time and iron levels in a clinically relevant "two-hit" human transfusion model. STUDY DESIGN AND METHODS: Eighteen healthy male volunteers (ages 18-35 years) were infused with 2 ng lipopolysaccharide (LPS)/kg to induce systemic inflammatory response syndrome. Two hours later, each participant received either 1 unit of 2-day stored (2D) autologous RBCs, 35-day stored (35D) autologous RBCs, or an equal volume of saline. Every 2 hours up to 8 hours after LPS infusion, hemoglobin, hemolysis parameters, and iron parameters, including NTBI, were measured. RESULTS: Transfusion of both 2D and 35D RBCs caused increases in hemoglobin, plasma iron, and transferrin saturation; whereas levels remained stable in the saline group. Transfusion of 35D RBCs did not result in hemolysis nor did it lead to increased levels of NTBI compared with 2D RBCs or saline. LPS induced increases in ferritin, haptoglobin, bilirubin, and lactate dehydrogenase that were similar in all three groups. CONCLUSION: We conclude that 35D autologous RBCs do not cause hemolysis or increased levels of NTBI during human endotoxemia.


Assuntos
Preservação de Sangue , Transfusão de Sangue Autóloga , Endotoxemia/sangue , Endotoxemia/terapia , Transfusão de Eritrócitos , Ferro/sangue , Adolescente , Adulto , Bilirrubina/sangue , Endotoxemia/induzido quimicamente , Ferritinas/sangue , Haptoglobinas/metabolismo , Hemoglobinas/metabolismo , Humanos , L-Lactato Desidrogenase/sangue , Lipopolissacarídeos/toxicidade , Masculino , Fatores de Tempo
20.
Biomed Pharmacother ; 86: 48-56, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27939519

RESUMO

High mobility group box 1 (HMGB1), a highly conserved non-histone DNA-binding protein, plays an important role in the pathogenesis of sepsis. Previously, the authors reported 13-ethylberberine (13-EBR) has anti-inflammatory and antibacterial effects. However, the effect of 13-EBR on HMGB1 release was not investigated. In the present study, it was hypothesized 13-EBR might reduce HMGB1 release by activating AMPK under septic conditions. The results obtained showed 13-EBR significantly reduced HMGB1 release from LPS-activated RAW264.7 cells, and that this reduction was reversed by silencing p38, or AMPK, or by co-treating cells with p38 MAPKinase inhibitor. 13-EBR increased the phosphorylations of p38 and AMPK, and the phosphorylation of p38 by 13-EBR was inhibited by AMPK-siRNA, indicating AMPK acted upstream of p38. In the lung tissues of LPS-treated mice, 13-EBR administration significantly increased p-AMPK but reduced inducible nitric oxide synthase (iNOS) protein levels. Hematoxylin and eosin staining revealed 13-EBR significantly reduced LPS-induced lung and liver damage. In addition, 13-EBR inhibited NF-kB in LPS-activated RAW264.7 cells, and in LPS-treated mice, 13-EBR administration significantly increased survival. Furthermore, co-administration of 13-EBR plus LPS prevented LPS-induced aortic rings hypocontractile response to phenylephrine in vitro. Taken together, these results indicate 13-EBR might offer a means of treating sepsis through AMPK activation.


Assuntos
Berberina/uso terapêutico , Endotoxemia/metabolismo , Endotoxemia/prevenção & controle , Proteína HMGB1/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Proteínas Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Berberina/farmacologia , Endotoxemia/induzido quimicamente , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Proteína HMGB1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Insuficiência de Múltiplos Órgãos/induzido quimicamente , Insuficiência de Múltiplos Órgãos/metabolismo , Insuficiência de Múltiplos Órgãos/prevenção & controle , Células RAW 264.7 , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA