Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int Immunopharmacol ; 124(Pt A): 110838, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37633235

RESUMO

Colorectal cancer (CRC) is a growing concern due to its high morbidity and mortality, and the search for effective and less toxic active substances against inflammatory bowel diseases has been a hot topic in the research and development of drugs against CRC. It is reported that monotropein isolated from the roots of Morinda officinalis, can improve Dextran Sodium Sulfate (DSS)-induced ulcerative colitis in mice, but its therapeutic effects and mechanisms for CRC treatment are still to be investigated. In the present study, we first used molecular docking, BLI, CESTA, and DARTS methods to detest whether monotropein targets VDR proteins. In addition, we used tumor cell conditioned co-culture and four models of macrophage polarisation to investigate the regulation of four macrophage polarisations by monotropein using RT-PCR, IF and western blot. Furthermore, we further validated the target of action of monotropein for the treatment of Azoxymethane (AOM)/DSS induced colitis associated cancer (CAC) using knockout animals. Meanwhile, we further explored the mechanism of action of monotropein in regulating polarisation by detecting JAK/STAT1-related genes and proteins. Molecular docking and biofilm interference techniques showed that monotropein bound to the VDR, and additional results from CESTA and DARTS suggested that VDR proteins are targets of monotropein. Furthermore, in tumor cell conditioned co-cultures or LPS + IFN-γ induced RAW264.7 cells, VDR translocation to the nucleus was reduced, JAK1/STAT1 signaling pathway proteins were up-regulated, and macrophages were polarised towards the M1-type after monotropein intervention. Animal models in which normal VDR or myeloid VDR was knocked out confirmed that JAK1 levels in intestinal tissues were increased after monotropein intervention, macrophages were polarised towards the M1 type, and CAC paracarcinomas were ameliorated. Taken together, the present study concluded that monotropein inhibited colitis-associated cancers through macrophage polarisation regulated by VDR/JAK1/STAT1.

2.
J Agric Food Chem ; 71(1): 546-556, 2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-36538589

RESUMO

It is extremely important to promote angiogenesis-dependent osteogenesis and ameliorate bone loss for the prevention and treatment of osteoporosis (OP) development. Vitexin, as one of the major active components in pigeonpea leave, promoted the proliferation of osteoblast and HUVECs in hypoxia. The present study aimed to investigate the effect of vitexin on alleviating osteoporosis in ovariectomized (OVX) rats and further explore its underlying mechanisms. Herein, the OVX rat model was established and treated with vitexin (10 mg kg-1) for 3 months. After being sacrificed, we performed hematoxylin-eosin (H&E) staining and micro-computed tomography (micro-CT) to assess bone mass, which found that trabecular bone was damaged in the OVX rat model. Vitexin could repair bone injury and promote osteoblast biochemical indicators and angiogenesis indicators. Furthermore, EAhy926 cells were used to further explore the effect of vitexin on improving hypoxia-induced endothelial injury in vitro. Vitexin had a protective effect on hypoxia-treated EAhy926 cells and up-regulated vitamin D receptor (VDR) signaling and promoted phosphorylation of phosphatidylinositol-3-kinase (PI3K), protein kinase B (AKT), and endothelial NO synthase (eNOS), which enhanced endothelial cell migration and tube formation. VDR small-interfering RNA (siRNA) transfection significantly decreased both VDR and p-eNOS proteins, and VDR siRNA transfection + vitexin could not further increase VDR and downstream proteins. Overall, this study presented that vitexin regulates angiogenesis and osteogenesis in ovariectomy-induced osteoporosis of rats via the VDR/eNOS signaling pathway.


Assuntos
Osteoporose , Fosfatidilinositol 3-Quinase , Feminino , Ratos , Animais , Humanos , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Osteogênese , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Microtomografia por Raio-X , Osteoporose/etiologia , Osteoporose/genética , Transdução de Sinais , RNA Interferente Pequeno , Ovariectomia/efeitos adversos , Receptores de Calcitriol/genética
3.
J Leukoc Biol ; 112(1): 143-155, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35224772

RESUMO

In hyperglycemia-induced complications, macrophages play important roles in disease progression, and altered digestion is a key feature that dictates macrophage function. Recent evidence indicates that kakonein (Ka) possesses anti-inflammatory activities for hyperglycemia-induced complication. In this study, we established a mouse model of Nlrp3+/+ and Nlrp3-/- hyperglycemia and administering Ka, primary culture macrophages were tested by engulfing and digesting microbes. The role of macrophages in the cathepsin B-NLRP3 pathway involved in the mechanism of Ka in restoring macrophage digestion function was investigated using biochemical analyses, molecular biotechnology, and microbiology. Ka restored the function of macrophage digestion, which were same characterized by Nlrp3-/- mice. Meanwhile, kakonein could decrease NLRP3 inflammasome products expression and NLRP3/ASC or NLRP3/Casp1 colocalization in macrophage. Interestingly, Ka suppressed inflammasome response not by reducing NLRP3 and ASC expression but by reducing cathepsin B release and activation. And Ka restored macrophage digestion and inhibited NLRP3 inflammasome activation consistent with cathepsin B inhibitor. It is concluded that Ka reduced the release of lysosomal cathepsin B and consequently inhibited NLRP3 inflammasome activation to prevent macrophage digestion. Hence, Ka may contribute to new targets for treatment of hyperglycemia-associated dysfunction of macrophage digestion and development of innovative drugs.


Assuntos
Anti-Inflamatórios , Hiperglicemia , Isoflavonas , Macrófagos , Fagocitose , Animais , Anti-Inflamatórios/farmacologia , Catepsina B/metabolismo , Modelos Animais de Doenças , Hiperglicemia/metabolismo , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Isoflavonas/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fagocitose/efeitos dos fármacos
4.
Front Pharmacol ; 9: 1347, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524287

RESUMO

Patchouli alcohol (PA), a natural tricyclic sesquiterpene extracted from Pogostemon cablin (Blanco) Benth. (Labiatae), has been found to exhibit anti-Helicobacter pylori and anti-inflammatory properties. In this study, we investigated the protective effect of PA against H. pylori-induced gastritis in vitro and in vivo, and determined the underlying mechanism. In the in vivo experiment, a C57BL/6 mouse model of gastritis was established using H. pylori SS1, and treatments with standard triple therapy or 5, 10, and 20 mg/kg PA were performed for 2 weeks. Results indicated that PA effectively attenuated oxidative stress by decreasing contents of intracellular reactive oxygen species (ROS) and malonyldialdehyde (MDA), and increasing levels of non-protein sulfhydryl (NP-SH), catalase and glutathione (GSH)/glutathione disulphide (GSSG). Additionally, treatment with PA significantly attenuated the secretions of interleukin 1 beta (IL-1ß), keratinocyte chemoattractant and interleukin 6 (IL-6). PA (20 mg/kg) significantly protected the gastric mucosa from H. pylori-induced damage. In the in vitro experiment, GES-1 cells were cocultured with H. pylori NCTC11637 at MOI = 100:1 and treated with different doses of PA (5, 10, and 20 µg/ml). Results indicated that PA not only significantly increased the cell viability and decreased cellular lactate dehydrogenase (LDH) leakage, but also markedly elevated the mitochondrial membrane potential and remarkably attenuated GES-1 cellular apoptosis, thereby protecting gastric epithelial cells against injuries caused by H. pylori. PA also inhibited the secretions of pro-inflammatory factors, such as monocyte chemotactic protein 1 (MCP-1), tumor necrosis factor-α (TNF-α) and IL-6. Furthermore, after PA treatment, the combination of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) and cysteine-aspartic proteases 1 (CASPASE-1), the expression levels of NLRP3 inflammasome-related proteins, such as thioredoxin-interacting protein (TXNIP), pro-CASPASE-1, cle-CASPASE-1, and NLRP3 and genes (NLRP3 and CASPASE1) were significantly decreased as compared to the model group. In conclusion, treatment with PA for 2 weeks exhibited highly efficient protective effect against H. pylori-induced gastritis and related damages. The underlying mechanism might involve antioxidant activity, inhibition of pro-inflammatory factor and regulation of NLRP3 inflammasome function. PA exerted anti-H. pylori and anti-gastritis effects and thus had the potential to be a promising candidate for treatment of H. pylori-related diseases.

5.
Cell Physiol Biochem ; 50(4): 1585-1600, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30359991

RESUMO

BACKGROUND/AIMS: Angiotensin II (Ang II) is an octapeptide hormone that plays a significant role in mediating hypertension. Although hypertension is considered a chronic inflammatory disease, the molecular basis of the sterile inflammatory response involved in hypertension remains unclear. METHODS: We investigated the role of macrophage NLRP3 inflammasomes in engulfing and digesting microbes, a key macrophage function, and in early onset of hypertension-associated macrophage injury using biochemical analyses, gene silencing, molecular biotechnology, immunofluorescence, and microbiology. RESULTS: Ang II stimulation decreased nitric oxide (NO) release and macrophage digestion in cultured THP-1 cells and markedly increased NLRP3 inflammasome formation and activation. NO release and macrophage digestion were restored by NLRP3 inflammasome inhibition with isoliquiritigenin and gene silencing. This Ang II-induced upregulation of NLRP3 inflammasomes in macrophages was attributed to lysosomal damage and release of cathepsin B. Mechanistically, losartan, a nonpeptide Ang II receptor antagonist, decreased Ang II-induced NLRP3 inflammasome activation, lysosomal membrane permeability, lysosomal cathepsin B release, and macrophage digestion dysfunction. Similarly, Ang II-induced macrophage microbe digestion and NO production, which were blocked by ATI gene silencing. In addition, in vivo experiments showed that the bacteria scavenging function was clearly decreased in macrophages from Ang II-induced hypertensive mice. CONCLUSION: Angiotensin II enhances lysosomal membrane permeabilization and the consequent release of lysosomal cathepsin B, resulting in activation of the macrophage NLRP3 inflammasome. This may contribute to NO mediation of dysfunction in digesting microbes.


Assuntos
Angiotensina II/farmacologia , Catepsina B/metabolismo , Inflamassomos/metabolismo , Macrófagos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Catepsina B/antagonistas & inibidores , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Chalconas/farmacologia , Escherichia coli/fisiologia , Hipertensão/metabolismo , Hipertensão/patologia , Losartan/farmacologia , Lisossomos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Óxido Nítrico/metabolismo , Fagocitose/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo
6.
Zhongguo Zhong Yao Za Zhi ; 43(15): 3171-3175, 2018 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-30200714

RESUMO

The aim of this paper is to investigate the effect of patchouli alcohol in enhancing Helicobater pylori's action in eradicating macrophages and its mechanism. H. pylori was co-cultured with macrophages at a ratio of MOI=100 in different concentrations of patchouli alcohol. The effect of patchouli alcohol in eradicating macrophages was detected by agar dilution method. The effect of patchouli alcohol on NO and myeloperoxidase (MPO) levels in macrophages were measured by H. pylori by biochemical methods. Patchouli alcohol effect on H. pylori-induced pro-inflammatory gene expression and protein secretion in macrophages were detected by RT-qPCR and ELISA method. The eradication of H. pylori has significantly enhanced, and the destabilization of lysosomes has been reversed. Meanwhile, patchouli alcohol has an effect in inhibiting pro-inflammation and oxidation. The mechanism of patchouli alcohol in eradicating H. pylori and resisting oxidative stress may be associated to the blocking of bacteria escape lysosome combination procedures.


Assuntos
Antibacterianos/farmacologia , Helicobacter pylori/efeitos dos fármacos , Lisossomos/imunologia , Macrófagos/imunologia , Sesquiterpenos/farmacologia , Células Cultivadas , Humanos , Macrófagos/efeitos dos fármacos , Estresse Oxidativo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA