Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Gen Subj ; 1868(8): 130651, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38825256

RESUMEN

Cannabidiol (CBD) has antioxidant and anti-inflammatory activities. However, the anti-tumor effect of CBD on hepatocellular carcinoma (HCC) remains unclear. Here, we investigated whether CBD displays anti-tumorigenic effects in HCC cells and whether it could reduce tumorigenesis and metastases in vivo. First, this study treated HCC cells with different concentrations of CBD, followed by analyzing the changes in the proliferative, apoptotic, migratory and invasive abilities. The effects of CBD on the growth and metastasis of HCC cells in vivo were verified by tumorigenesis and metastasis assays. Subsequently, the target genes of CBD were predicted through the SwissTarget website and the genes differentially expressed in cells after CBD treatment were analyzed by microarray for intersection. The enrichment of the pathways after CBD treatment was analyzed by KEGG enrichment analysis, followed by western blot validation. Finally, rescue assays were used to validate the functions of genes as well as pathways in the growth and metastasis of HCC cells. A significant weakening of the ability of HCC cells to grow and metastasize in vitro and in vivo was observed upon CBD treatment. Mechanistically, CBD reduced GRP55 expression in HCC cells, along with increased TP53 expression and blocked MAPK signaling activation. In CBD-treated cells, the anti-tumor of HCC cells was restored after overexpression of GRP55 or deletion of TP53. CBD inhibits the MAPK signaling activation and increases the TP53 expression by downregulating GRP55 in HCC cells, thereby suppressing the growth and metastasis of HCC cells.


Asunto(s)
Cannabidiol , Carcinoma Hepatocelular , Neoplasias Hepáticas , Receptores de Cannabinoides , Proteína p53 Supresora de Tumor , Cannabidiol/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Humanos , Proteína p53 Supresora de Tumor/metabolismo , Receptores de Cannabinoides/metabolismo , Receptores de Cannabinoides/genética , Animales , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Ratones , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Fenotipo , Ratones Desnudos
2.
Phytother Res ; 37(7): 2965-2978, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36879546

RESUMEN

Acute lung injury (ALI) caused by acute bacterial infection remains a common life-threatening lung disease. An increased inflammatory response is the basis for the occurrence and development of ALI. Most antibiotics can only reduce the bacterial load but do not protect from lung damage because of an excessive immune response. Chrysophanol (chrysophanic acid, Chr), as a natural anthraquinone extracted from Rheum palmatum L., has various biological functions, including anti-inflammatory, anti-cancer activities, and ameliorative effects on cardiovascular diseases. Considering these properties, we investigated the effect of Chr in Klebsiella pneumoniae (KP)-induced ALI mice and its potential mechanism. Our results showed that Chr had protective effects against KP-infected mice, including increased survival rate, decreased bacterial burden, reduced recruitment of immune cells, and reduced reactive oxygen species level of lung macrophages. Chr reduced the expression of inflammatory cytokines by inhibiting the toll-like receptor 4/nuclear factor kappa-B (TLR4/NF-κB) signaling pathway and inflammasome activation and strengthening autophagy. Overactivation of the TLR4/NF-κB signaling pathway by the activator Neoseptin 3 led to Chr losing control of inflammatory cytokines in cells, resulting in increased cell death. Similarly, overactivation of the c-Jun N-terminal kinase signaling pathway using the activator anisomycin resulted in Chr losing its inhibitory effect on NOD-like receptor thermal protein domain associated protein 3 (NFRP3) inflammasome activation, and cell viability was reduced. In addition, autophagy was blocked by siBeclin1, so Chr could not reduce inflammatory factors, and cell viability was markedly inhibited. Collectively, this work unravels the molecular mechanism underpinning Chr-alleviated ALI via inhibiting pro-inflammatory cytokines. Thus, Chr is a potential therapeutic agent for KP-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda , FN-kappa B , Ratones , Animales , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Klebsiella pneumoniae/metabolismo , Inflamasomas , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inducido químicamente , Antraquinonas/farmacología , Antraquinonas/uso terapéutico , Pulmón , Citocinas/metabolismo , Lipopolisacáridos/farmacología
3.
Phytomedicine ; 93: 153742, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34624808

RESUMEN

BACKGROUND: Lung cancer is the leading cause of cancer death worldwide, yet no effective medication for this disease is available. Cochlioquinone B derivative (CoB1), purified from Salvia miltiorrhiza endophytic Bipolaris sorokiniana, affects the defense against pulmonary pathogens by regulating inflammatory responses. However, the effect of CoB1 on lung cancer and the underlying molecular mechanisms remain unknown. In the present study, we investigate the protective effects of CoB1 on lung cancer and explore its underlying mechanism. METHOD: We examined the inhibitory effect of CoB1 on lung cancer cells (A549 cells) by MTT and colony formation assay. The effect of CoB1 on cytostatic autophagy in lung cancer cells was verified by Western blot, transmission electron microscopy, and confocal microscopy. The differentially expressed miRNAs were identified using quantitative RT-PCR. Luciferase assay and Northern blot were performed to verify the correlation between miRNA-125b and Foxp3. Protein expression in autophagy-related pathways was detected by Western blot. Xenograft tumor models were constructed to explore the inhibitory effect of CoB1 and the role of miRNA-125b as a suppressor in lung cancer in vivo. RESULT: CoB1 inhibited lung cancer cell proliferation by inducing cytostatic autophagy both in vitro and in vivo. CoB1-induced autophagy was related to blocking of the PI3K/Akt1/mTOR signaling pathway. In addition, CoB1 induced miR-125b expression via activating the TAK1/MKK4/JNK/Smad axis, thereby reducing Foxp3 expression and further inducing autophagy. CONCLUSION: This study is the first to report the specific inhibitory function of CoB1 purified from Salvia miltiorrhiza endophytic Bipolaris sorokiniana in lung cancer, which may be due to the induction of autophagy. This study provides evidence and novel insights into the anticancer efficacy of CoB1.


Asunto(s)
Citostáticos , Neoplasias Pulmonares , MicroARNs , Autofagia , Línea Celular Tumoral , Proliferación Celular , Factores de Transcripción Forkhead , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
4.
Mol Ther Oncolytics ; 20: 82-93, 2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33575473

RESUMEN

Lung cancer is the most frequent and fatal malignancy in humans worldwide, yet novel successful drugs for control of this disease are still lacking. Ipomoea batatas polysaccharides (IBPs) have been implicated in inhibiting diverse cancer types, but their functions in mitigating lung cancer are largely unknown. In this study, we identify a role of IBP in inhibiting lung cancer proliferation. We found that IBP significantly impedes the proliferation of lung cancer cells by inducing cytostatic macroautophagy both in vitro and in vivo. Mechanistically, IBP specifically promotes ubiquitination-mediated degradation of PAK1 (p21-activated kinase 1) and blocks its downstream Akt1/mTOR signaling pathway, leading to increased autophagic flux. In lung cancer xenografts in mice, IBP-induced cytostatic autophagy suppresses tumor development. Through site-directed mutational analysis, the underlying signaling augments ubiquitination via PAK1-ubiquitin interaction. Collectively, this work unravels the molecular mechanism underpinning IBP-induced cytostatic autophagy in lung cancer and characterizes IBP as a potential therapeutic agent for lung cancer treatment.

5.
Sci Rep ; 10(1): 20013, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33203903

RESUMEN

The study aimed to investigate the antibacterial effect and potential mechanisms of chlorogenic acid (CA) in Klebsiella pneumonia (KPN) induced infection in vitro and in vivo. 62 KPN strains were collected from the First People's Hospital of Yunnan Province. CA and CA combined Levofloxacin (LFX) were detected for KPN biofilm (BF) formation in vitro. The lung infection mice model were established by KPN. The effect of CA (500 mg/kg), LFX (50 mg/kg) and CA combined LFX (250 mg/kg + 25 mg/kg) were evaluated through the survival of mice, the changes of inflammation factors of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1ß and IL-6 in serum, the histopathological analysis of lung and the protein expression of NLRP3 signaling pathway in vivo. A total of 62 KPNs were isolated and identified, of which 13 (21%) strains were BF positive. 8 (13%) strains were extended spectrum ß-lactamase strains (ESBLs), and 20 (32%) strains are ESBLs biofilm positive. In vitro study, CA and LFX showed a synergistic effect on KPN biofilm formation. In vivo mice experiment, CA, especially CA + LFX treated group significantly decreased the serum levels of TNF-α, IL-1ß and IL-6, improved the survival ratio and lung pathology changes, and also reduced the protein expression of ASC, caspase 1 p20, IL-1ß and phosphor NF-κB p65. CA could effectively alleviate lung infection of KPN infected mice, and the antibacterial effection is strengthened by combined with LFX. The study provide a theroy basis for making rational and scientific antibacterial therapy strategy in clinic.


Asunto(s)
Antibacterianos/farmacología , Ácido Clorogénico/farmacología , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae/efectos de los fármacos , Levofloxacino/farmacología , Animales , Antibacterianos/uso terapéutico , Biopelículas/efectos de los fármacos , Ácido Clorogénico/uso terapéutico , Farmacorresistencia Bacteriana , Sinergismo Farmacológico , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/mortalidad , Levofloxacino/uso terapéutico , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/metabolismo
6.
Front Cell Dev Biol ; 8: 552020, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240872

RESUMEN

BACKGROUND: Salmonella typhimurium (ST) causes several intestinal diseases. Polyphenols including chlorogenic acid (CGA) inhibit pathogenesis. OBJECTIVE: This study aimed to investigate the mechanisms of CGA in ST infection. METHODS: The intestinal pathological changes and survival rate of ST-infected mice were measured to verify the protection of CGA on ST infection. The antibacterial effects of CGA in vitro on the invasion to intestinal epithelial cells and autophagy was evaluated. The relationships among GAS5, miR-23a, and PTEN were verified. Expression of inflammation- and autophagy-related proteins was detected. RESULTS: CGA treatment alleviated pathological damage, improved the secretion disturbance of intestinal cytokines caused by ST infection, and reduced the mortality of mice. Intestinal GAS5 was upregulated after CGA treatment. LncRNA GAS5 competitively bound to miR-23a to upregulate PTEN and inhibit the p38 MAPK pathway. CGA regulated the p38 MAPK pathway through lncRNA GAS5/miR-23a/PTEN axis to promote autophagy in ST infection. The functional rescue experiments of miR-23a and PTEN further identified these effects. CONCLUSION: CGA promotes autophagy and inhibits ST infection through the GAS5/miR-23a/PTEN axis and the p38 MAPK pathway.

8.
J Immunol ; 205(5): 1293-1305, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32747503

RESUMEN

Owing to multiple antibiotic resistance, Pseudomonas aeruginosa causes the most intractable infections to human beings worldwide, thus exploring novel drugs to defend against this bacterium remains of great importance. In this study, we purified a novel cochlioquinone B derivative (CoB1) from Salvia miltiorrhiza endophytic Bipolaris sorokiniana and reveal its role in host defense against P. aeruginosa infection by activating cytoprotective autophagy in alveolar macrophages (AMs) both in vivo and in vitro. Using a P. aeruginosa infection model, we observed that CoB1-treated mice manifest weakened lung injury, reduced bacterial systemic dissemination, decreased mortality, and dampened inflammatory responses, compared with the wild type littermates. We demonstrate that CoB1-induced autophagy in mouse AMs is associated with decreased PAK1 expression via the ubiquitination-mediated degradation pathway. The inhibition of PAK1 decreases the phosphorylation level of Akt, blocks the Akt/mTOR signaling pathway, and promotes the release of ULK1/2-Atg13-FIP200 complex from mTOR to initiate autophagosome formation, resulting in increased bacterial clearance capacity. Together, our results provide a molecular basis for the use of CoB1 to regulate host immune responses against P. aeruginosa infection and indicate that CoB1 is a potential option for the treatment of infection diseases.


Asunto(s)
Autofagia/efectos de los fármacos , Benzoquinonas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Células Cultivadas , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
9.
Colloids Surf B Biointerfaces ; 185: 110616, 2020 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-31740323

RESUMEN

Multidrug-resistant (MDR) bacterial strains have led to notable heathy threats to human beings. The demand for the development of effective antibacterial materials is increasing. Silver nanoparticles (AgNPs) and graphene-based nanomaterials are two major types of nanomaterials that are studied to inhibit and/or kill bacteria. In this study, by combining the excellent photothermal effect of graphene and antibacterial activity of AgNPs, we have applied reduced graphene oxide/silver (RGO/Ag) nanocomposite to destroy the MDR bacteria. The antibacterial activity of the RGO/Ag nanocomposite was systematically investigated using a regular bacterium of Escherichia coli (E. coli) and an MDR bacterium of Klebsiella pneumoniae (Kp). Compared with AgNPs, graphene oxide (GO) and RGO, the RGO/Ag nanocomposite showed significant higher antibacterial efficiency for both regular bacteria and MDR bacteria. Under a near-infrared (NIR) irradiation (0.30 W/cm2 for 10 min), the RGO/Ag nanocomposite demonstrated an enhanced synergetic antibacterial activity through the photothermal effect. Nearly 100 % of E. coli and Kp were killed by the treatment of 15 µg/mL and 30 µg/mL of RGO/Ag nanocomposite, respectively. Moreover, a membrane integrity assay and a reactive oxygen species (ROS) assay demonstrated that the RGO/Ag nanocomposite under NIR irradiation induced the cell membrane disruption and generation of ROS, providing possible mechanisms for their high antibacterial activity besides the photothermal effect. Finally, the cytotoxicity of the RGO/Ag nanocomposites toward different mammalian cells was studied, the cell viabilities retained above 60 % at higher concentrations of RGO/Ag, indicating that the RGO/Ag nanocomposites may be a low cytotoxic, efficient antibacterial agent with the irradiation.


Asunto(s)
Antibacterianos/farmacología , Grafito/farmacología , Hipertermia Inducida , Nanocompuestos/química , Fototerapia , Plata/farmacología , Bacterias/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
10.
mBio ; 10(4)2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31337721

RESUMEN

Pseudomonas aeruginosa, one of the most common pathogens in hospital-acquired infections, is tightly controlled by a multilayered regulatory network, including the quorum sensing system (QS), the type VI secretion system (T6SS), and resistance to host immunity. We found that the P. aeruginosa 3880 (PA3880) gene, which encodes an unknown protein, acts as a regulator of anaerobic metabolism in response to oxidative stress and virulence in P. aeruginosa More than 30 PA3880 homologs were found in other bacterial genomes, indicating that PA3880 is widely distributed in the Bacteria kingdom as a highly conserved gene. Deletion of the PA3880 gene changed the expression levels of more than 700 genes, including a group of virulence genes, under both aerobic and anaerobic conditions. To further study the mechanisms of PA3880-mediated regulation in virulence, we utilized a bacterial two-hybrid assay and found that the PA3880 protein interacted directly with QS regulator MvfR and anaerobic regulator Anr. Loss of the PA3880 protein significantly blunted the pathogenicity of P. aeruginosa, resulting in increased host survival, decreased bacterial burdens, reduced inflammatory responses, and fewer lung injuries in challenged mice hosts. Mechanistically, we found that Cys44 was a critical site for the full function of PA3880 in influencing alveolar macrophage phagocytosis and bacterial clearance. We also found that AnvM directly interacted with host receptors Toll-like receptor 2 (TLR2) and TLR5, which might lead to activation of the host immune response. Hence, we gave the name AnvM (anaerobic and virulence modulator) to the PA3880 protein. This characterization of AnvM could help to uncover new targets and strategies to treat P. aeruginosa infections.IMPORTANCE Infections by Pseudomonas aeruginosa, one of the most frequently isolated human pathogens, can create huge financial burdens. However, knowledge of the molecular mechanisms involved in the pathogenesis of P. aeruginosa remains elusive. We identified AnvM as a novel regulator of virulence in P. aeruginosa Deletion of anvM altered the expression levels of more than 700 genes under aerobic and anaerobic conditions, including quorum sensing system genes and oxidative stress resistance genes. AnvM directly interacted with MvfR and Anr, thus regulating their downstream genes. More importantly, AnvM directly bound to TLR2 and TLR5, which turn on the host immune response. These findings provide insights into the significance of AnvM homologs in pathogenic bacteria and suggest a potential drug target against bacterial infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Factores de Virulencia/metabolismo , Anaerobiosis , Animales , Proteínas Bacterianas/genética , Femenino , Regulación Bacteriana de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Percepción de Quorum , Receptor Toll-Like 5/metabolismo , Receptores Toll-Like/metabolismo , Virulencia , Factores de Virulencia/genética
11.
Breast Cancer ; 26(6): 766-775, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31172425

RESUMEN

BACKGROUND: Icariin is a major component isolated from Epimedium brevicornum Maxim and has been reported to exhibit anti-tumor activity. However, whether icariin could reverse the acquired drug resistance in breast cancer remains largely unclear. Therefore, this study was designed to explore the antitumor effects of icariin and its underlying mechanisms in a tamoxifen-resistant breast cancer cell line MCF-7/TAM. METHODS: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and Lactate dehydrogenase (LDH) assay were performed to determine the effects of icariin on cell viability and cell death. Cell cycle progression and apoptosis were detected by flow cytometry analysis. Transmission electron microscopy (TEM) assay was utilized to observe cell autophagy. The downstream protein levels were measured using western blotting. RESULTS: Here, we observed that icariin treatment not only inhibited the growth of MCF-7 but also has a potential function to overcome tamoxifen resistance in MCF-7/TAM. Moreover, icariin significantly induced cell cycle G0/G1 phase arrest and apoptosis, as well as suppressed autophagy. At molecular levels, icariin treatment remarkably down-regulated the expression levels of CDK2, CDK4, Cyclin D1, Bcl-2, LC3-1, LC3-II, AGT5, Beclin-1, but upregulated the expression levels of caspase-3, PARP and p62. Most importantly, we found inhibition of autophagy via 3-MA treatment could significantly enhance the effects of icariin on cell viability and apoptosis. Enhanced autophagy via autophagy related 5 (ATG5) overexpression could partially reverse the effects of icariin on cell viability and apoptosis. CONCLUSION: These results revealed that icariin might potentially be useful as an adjuvant agent in cancer chemotherapy to enhance the effect of tamoxifen through suppression of autophagy in vitro and provide insight into the therapeutic potential of icariin for the treatment of chemo-resistant breast cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Flavonoides/farmacología , Extractos Vegetales/farmacología , Tamoxifeno/efectos adversos , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Caspasa 3/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Epimedium/química , Femenino , Humanos , Células MCF-7 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/efectos adversos , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Transducción de Señal/efectos de los fármacos , Tamoxifeno/uso terapéutico , Transfección
12.
Mol Med Rep ; 19(3): 2057-2064, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30664158

RESUMEN

The present study aimed to investigate the anti­arthritic effects of curculigoside isolated from the rhizome of Curculigo orchioides Gaertn in vivo and in vitro, as well as to determine the potential underlying mechanisms. A rat model of arthritis was induced with type II collagen. Arthritic rats were treated with curculigoside (50 mg/kg) and blood samples were collected to determine serum levels of tumor necrosis factor (TNF)­α, interleukin (IL)­1ß, IL­6, IL­10, IL­12 and IL­17A. Furthermore, indices of the thymus and spleen were determined. The anti­proliferative effects of curculigoside were detected with Cell Counting kit­8 assays in rheumatoid arthritis­derived fibroblast­like synoviocyte MH7A cells. In addition, expression levels of Janus kinase (JAK)1, JAK3, signal transducer and activator of transcription (STAT)3, nuclear factor (NF)­κB p65 and its inhibitor (IκB) were determined by western blotting. The results revealed that curculigoside inhibited paw swelling and arthritis scores in type II collagen­induced arthritic (CIA) rats. Additionally, curculigoside decreased serum levels of TNF­α, IL­1ß, IL­6, IL­10, IL­12 and IL­17A in CIA rats. Curculigoside also significantly inhibited MH7A cell proliferation in a time and concentration­dependent manner. Furthermore, treatment downregulated the expression of JAK1, JAK3 and STAT3, and upregulated cytosolic nuclear factor (NF)­κB p65 and IκB. In conclusion, the results of the present study indicated that curculigoside exhibited significant anti­arthritic effects in vivo and in vitro, and the molecular mechanism may be associated with the JAK/STAT/NF­κB signaling pathway.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Benzoatos/administración & dosificación , Glucósidos/administración & dosificación , Janus Quinasa 1/genética , Janus Quinasa 3/genética , Factor de Transcripción STAT3/genética , Animales , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Proliferación Celular/efectos de los fármacos , Colágeno Tipo II/toxicidad , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Humanos , Proteínas I-kappa B/genética , Interleucinas/genética , Ratas , Transducción de Señal , Sinoviocitos/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Necrosis Tumoral alfa/genética
13.
Int J Clin Exp Pathol ; 12(6): 2075-2083, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31934029

RESUMEN

This study aimed to investigate the therapeutic effects and mechanism of action of curcumin against a MRL/lpr lupus model. Eight-week-old female MRL/lpr mice were used to establish the lupus nephritis model. Histologic and immunohistochemical analysis was conducted for lupus nephritis. Anti-dsDNA IgG and BAFF level were detected by ELISA. Cells directly isolated from the spleen were used to detect macrophage subsets and activation status by FACS. Curcumin reduced the total IgG and anti-dsDNA IgG levels in blood and reduced the activation of B cells in MRL/lpr mice. Moreover, curcumin prevented activation of macrophages in MRL/lpr mice. Levels of BAFF in serum, spleens and kidneys were also reduced in curcumin-treated MRL/lpr mice. In vitro experiments showed that curcumin reduced the activation of macrophage and leaded to the decrease of BAFF from them upon toll like receptor (TLR) 4 stimulation. Curcumin attenuates lupus nephritis in MRL/lpr mice by inhibiting macrophages activation and their secreting BAFF, which may be a potential therapeutic candidate for the treatment of SLE.

14.
Int J Clin Exp Pathol ; 11(1): 76-87, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-31938089

RESUMEN

BACKGROUND: Lung epithelial cell dysfunction induced by hyperoxia-associated oxidative stress is a prominent feature involved in the development of acute lung injury (ALI). How the underlying molecular mechanisms contributed to this process are poorly defined. In the present study, we sought to identify the role of miR-124 in hyperoxia-induced cell apoptosis and excessive inflammatory response in pulmonary epithelial cell. METHODS: The miR-124 levels in pulmonary epithelial cell were assayed by qRT-PCR. MiR-124 mimics and inhibitors were transfected to gain or loss of miR-124 function. Cell proliferation was analyzed by CCK8 assay. Cell apoptosis was analyzed by flow cytometry. The targeted genes were predicted by a bioinformatics algorithm and confirmed by a dual luciferase reporter assay. The protein levels were assayed by western blotting. RESULTS: The results showed that miR-124 was significantly down-regulated in Beas2B cells and primary LECs upon hyperoxia exposure conditions. However, overexpression of miR-124 dramatically attenuated hyperoxia-provoked TLR4, NF-κB and pro-inflammatory cytokines production. In vitro, the cell viability and apoptosis was significantly reversed following transfection with miR-124 mimics in the presence of hyperoxia. Furthermore, the 3'-untranslated region (3'-UTR) of CCL2 was bound by miR-124. CONCLUSION: It was concluded that miR-124 inhibited hyperoxia-induced apoptosis and excessive inflammatory response in Beas2B cells and primary LECs, at least partially, through the inhibition of TLR4/NF-κB/CCL2 signaling cascades.

17.
Oncol Rep ; 38(2): 611-624, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28627697

RESUMEN

Nanomaterials are increasingly used as drug carriers for cancer therapy. Nanomaterials also appeal to researchers in the areas of cancer diagnosis and biomarker discovery. Several antitumor nanodrugs are currently being tested in preclinical and clinical trials and show promise in therapeutic and other settings. We review the development of nanomaterial drug carriers, including liposomes, polymer nanoparticles, dendritic polymers, and nanomicelles, for the diagnosis and treatment of various cancers. The prospects of nanomaterials as drug carriers for future clinical applications are also discussed.


Asunto(s)
Sistemas de Liberación de Medicamentos/tendencias , Nanotecnología/tendencias , Neoplasias/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Dendrímeros/uso terapéutico , Humanos , Liposomas/uso terapéutico , Nanopartículas/uso terapéutico
18.
Colloids Surf B Biointerfaces ; 157: 1-9, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28554055

RESUMEN

Graphene is a novel two-dimensional nanomaterial with a growing number of practical applications across numerous fields. In this work, we explored potential biomedical applications of graphene oxide (GO) by systematically studying antibacterial capacity of GO in both macrophages and animal models. Three types of bacteria, including Klebsiella pneumoniae (Kp), Escherichia coli (E. coli) and P. aeruginosa (Pa) were used for in vitro study. Kp was also selected as a representative multidrug resistant (MDR) bacterium for in vivo study. In in vitro study, GO effectively eradicated Kp in agar dishes and thus protected alveolar macrophages (AM) from Kp infection in the culture. In the in vivo evaluation, GO were introduced intranasally into mouse lungs followed by testing organ tissue damage including lung, liver, spleen, and kidneys, polymorphonuclear neutrophil (PMN) penetration, bacterial dissemination, and mortality in Kp-infected mice. We found that GO can prohibit the growth and spread of Kp both in vitro and in vivo, resulting in significantly increased cell survival rate, less tissue injury, subdued inflammatory response, and prolonged mice survival. These findings indicate that GO could be a promising biomaterial for effectively controlling MDR pathogens.


Asunto(s)
Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Grafito/farmacología , Grafito/uso terapéutico , Nanoestructuras/química , Animales , Antibacterianos/química , Farmacorresistencia Bacteriana Múltiple , Escherichia coli/efectos de los fármacos , Femenino , Grafito/química , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/patogenicidad , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa/efectos de los fármacos
19.
J Immunol ; 198(8): 3205-3213, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28258192

RESUMEN

Sepsis is a severe and complicated syndrome that is characterized by dysregulation of host inflammatory responses and organ failure, with high morbidity and mortality. The literature implies that autophagy is a crucial regulator of inflammation in sepsis. In this article, we report that autophagy-related protein 7 (Atg7) is involved in inflammasome activation in Pseudomonas aeruginosa abdominal infection. Following i.p. challenge with P. aeruginosa, atg7fl/fl mice showed impaired pathogen clearance, decreased survival, and widespread dissemination of bacteria into the blood and lung tissue compared with wild-type mice. The septic atg7fl/fl mice also exhibited elevated neutrophil infiltration and severe lung injury. Loss of Atg7 resulted in increased production of IL-1ß and pyroptosis, consistent with enhanced inflammasome activation. Furthermore, we demonstrated that P. aeruginosa flagellin is a chief trigger of inflammasome activation in the sepsis model. Collectively, our results provide insight into innate immunity and inflammasome activation in sepsis.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/inmunología , Inflamasomas/inmunología , Infecciones por Pseudomonas/inmunología , Piroptosis/inmunología , Sepsis/inmunología , Animales , Proteína 7 Relacionada con la Autofagia/deficiencia , Modelos Animales de Enfermedad , Inmunidad Innata/inmunología , Immunoblotting , Inflamasomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/inmunología , Sepsis/metabolismo
20.
J Immunol ; 198(7): 2844-2853, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28202616

RESUMEN

Oxygen is supplied as a supportive treatment for patients suffering from acute respiratory distress syndrome. Unfortunately, high oxygen concentration increases reactive oxygen species generation, which causes DNA damage and ultimately cell death in the lung. Although 8-oxoguanine-DNA glycosylase (OGG-1) is involved in repairing hyperoxia-mediated DNA damage, the underlying molecular mechanism remains elusive. In this study, we report that ogg-1-deficient mice exhibited a significant increase of proinflammatory cytokines (TNF-α, IL-6, and IFN-γ) in the lung after being exposed to 95% oxygen. In addition, we found that ogg-1 deficiency downregulated (macro)autophagy when exposed to hyperoxia both in vitro and in vivo, which was evident by decreased conversion of LC3-I to LC3-II, reduced LC3 punctate staining, and lower Atg7 expression compared with controls. Using a chromatin immunoprecipitation assay, we found that OGG-1 associated with the promoter of Atg7, suggesting a role for OGG1 in regulation of Atg7 activity. Knocking down OGG-1 decreased the luciferase reporter activity of Atg7. Further, inflammatory cytokine levels in murine lung epithelial cell line cells were downregulated following autophagy induction by starvation and rapamycin treatment, and upregulated when autophagy was blocked using 3-methyladenine and chloroquine. atg7 knockout mice and Atg7 small interfering RNA-treated cells exhibited elevated levels of phospho-NF-κB and intensified inflammatory cytokines, suggesting that Atg7 impacts inflammatory responses to hyperoxia. These findings demonstrate that OGG-1 negatively regulates inflammatory cytokine release by coordinating molecular interaction with the autophagic pathway in hyperoxia-induced lung injury.


Asunto(s)
Lesión Pulmonar Aguda/patología , Autofagia , ADN Glicosilasas/metabolismo , Reparación del ADN , Hiperoxia/patología , Pulmón/patología , Lesión Pulmonar Aguda/metabolismo , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Ensayo Cometa , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Hiperoxia/metabolismo , Inmunoprecipitación , Inflamación/metabolismo , Inflamación/patología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...