Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-36881383

RESUMEN

The NLRP3 inflammasome, which plays a central role in innate immunity, is linked to a variety of inflammatory diseases, and thus it may provide a new target for the treatment of those diseases. Biosynthesized silver nanoparticles (AgNPs), particularly those synthesized using medicinal plant extracts, have recently been shown to be a promising therapeutic option. Herein, the aqueous extract of Ageratum conyzoids was used to prepare a series of sized AgNPs (AC-AgNPs), in which the smallest mean particle size was 30 ± 1.3 nm with a polydispersity of 0.328 ± 0.009. The ζ potential value was -28.77 with a mobility of -1.95 ± 0.24 cm2/(v·s). Its main ingredient, elemental silver, accounted for about 32.71 ± 4.87% of its mass, and other ingredients included amentoflavone-7,7⁗-dimethyl ether, 1,3,5-tricaffeoylquinic acid, kaempferol 3,7,4'-triglucoside, 5,6,7,3',4',5'-hexamethoxyflavone, kaempferol, and ageconyflavone B. In LPS+ATP-stimulated RAW 264.7 and THP-1 cells, AC-AgNPs significantly inhibited the release of IL-1ß, IL-18, TNF-α, and caspase-1, indicating that AC-AgNPs can inhibit the activation of the NLRP3 inflammasome. The mechanistic study revealed that AC-AgNPs could decrease the phosphorylation levels of IκB-α and p65, resulting in decreased expression of NLRP3 inflammasome-related proteins, including pro-IL-1ß, IL-1ß, procaspase 1, caspase 1P20, NLRP3, and ASC, and also scavenge the level of intracellular ROS to prevent NLRP3 inflammasome assembly. Furthermore, AC-AgNPs attenuated the in vivo expression of inflammatory cytokines by suppressing NLRP3 inflammasome activation in a peritonitis mouse model. Our study provides evidence that the as-prepared AC-AgNPs can inhibit the inflammatory process by suppressing NLRP3 inflammasome activation and might be used to treat NLRP3 inflammasome-driven inflammatory diseases.

2.
J Ethnopharmacol ; 309: 116353, 2023 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-36907476

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Ageratum conyzoides L. (Asteraceae), a well-known and widely distributed traditional tropical medicinal herb, has been used to treat diverse diseases. Our preliminary research has shown that aqueous extracts of A. conyzoides leaf (EAC) have anti-inflammatory activity. However, the detailed underlying anti-inflammatory mechanism of EAC is still unclear. AIM OF THE STUDY: To determine the anti-inflammatory mechanism of action of EAC. MATERIALS AND METHODS: The major constituents of EAC were identified by ultra-performance liquid chromatography (UPLC) combined with quadrupole-time-of-flight mass/mass spectrometry (UPLC-Q-TOF-MS/MS). LPS and ATP were used to activate the NLRP3 inflammasome in two types of macrophages (RAW 264.7 and THP-1 cells). The cytotoxicity of EAC was measured by the CCK8 assay. The levels of inflammatory cytokines and NLRP3 inflammasome-related proteins were detected by ELISA and western blotting (WB), respectively. The oligomerization of NLRP3 and ASC and the resulting inflammasome complex formation were observed by immunofluorescence. The intracellular reactive oxygen species (ROS) level was measured by flow cytometry. Finally, an MSU-induced peritonitis model was established to evaluate the anti-inflammatory effects of EAC in vivo. RESULTS: Twenty constituents were identified in the EAC. Kaempferol 3,7-diglucoside, 1,3,5-tricaffeoylquinic acid, and kaempferol 3,7,4'-triglucoside were found to be the most potent ingredients. EAC significantly reduced the levels of IL-1ß, IL-18, TNF-α, and caspase-1 in the two types of activated macrophages, implying that EAC can inhibit the activation of the NLRP3 inflammasome. A mechanistic study revealed that EAC inhibited NLRP3 inflammasome activation by blocking NF-κB signalling pathway activation and scavenging the level of intracellular ROS to prevent NLRP3 inflammasome assembly in macrophages. Furthermore, EAC attenuated the in vivo expression of inflammatory cytokines by suppressing NLRP3 inflammasome activation in a peritonitis mouse model. CONCLUSION: Our results demonstrated that EAC inhibited inflammation by suppressing NLRP3 inflammasome activation, highlighting that this traditional herbal medicine might be used to treat NLRP3 inflammasome-driven inflammatory diseases.


Asunto(s)
Ageratum , Peritonitis , Animales , Ratones , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Quempferoles/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Masas en Tándem , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Citocinas/metabolismo , Peritonitis/inducido químicamente , Peritonitis/tratamiento farmacológico , Interleucina-1beta/metabolismo
3.
Eur J Pharm Sci ; 179: 106299, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36179970

RESUMEN

There is an urgent need to find new antibiotics to fight against the increasing drug resistance of microorganisms. A novel natural compound, Penicilazaphilone C (PAC), was isolated from a marine-derived fungus. It has displayed broad bactericidal activities against Gram-negative and Gram-positive bacteria. However, its bactericidal mechanism is still unknown. Herein, time-kill assays verified that PAC is a fast and efficient bactericidal agent. Furthermore, data from 4D label-free quantitative proteome assays revealed that PAC significantly influences over 898 proteins in Escherichia coli. Combining the results of biofilm formation, ß-galactosidase measurement, TEM observation, soft agar plate swimming, reactive oxygen species measurement, qRT-PCR, and west-blotting, the mode of PAC action against E. coli was to block respiration, inhibit assimilatory nitrate reduction and dissimilar sulfur reduction, facilitate assimilatory sulfate reduction, suppress cysteine and methionine biosynthesis, down-regulate antioxidant protein expression and induced intracellular ROS accumulation, weaken bacterial chemotaxis, destroy flagellar assembly, etc., and finally cause the bacteria's death. Our findings suggest that PAC could have a multi-target regulatory effect on E. coli and could be used as a new antibiotic in medicine.


Asunto(s)
Escherichia coli , Proteómica , Antibacterianos/farmacología , Antibacterianos/metabolismo , Bacterias , Pruebas de Sensibilidad Microbiana
4.
Front Pharmacol ; 13: 973611, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36105203

RESUMEN

Background: Ferroptosis is a newly discovered and promising non-apoptotic programmed cell death (PCD), and inducing ferroptosis in cancer cells could open up a novel avenue for drug screening and cancer therapy. S-3'-hydroxy-7', 2', 4'-trimethoxyisoxane (ShtIX), a new isoflavane compound, has been reported to possess cytotoxicity in non-small cell lung cancer (NSCLC). The aim of this research is to explore the ShtIX-induced cell death form and its underlying molecular mechanism in NSCLC cells. Methods: Cell proliferation, cell cycle arrest, and cell death tests were used to assess the ability of ShtIX to kill NSCLC cells. Iron metabolism, Fe2+ content, reactive oxygen species (ROS) production, lipid peroxide (MDA) level, glutathione (GSH) level, and glutathione peroxidase 4 (GPX4) level were used to determine ferroptosis caused by ShtIX. We employed western blot, quantitative real-time PCR, and Nrf2 interference in NSCLC cells to investigate the roles of Nrf2/HO-1 in ShtIX-induced ferroptosis. In a xenograft nude mouse model, the anticancer efficacy of ShtIX and the function of ferroptosis were studied. Results: Our research shows that ShtIX can selectively kill NSCLC cells while sparing normal cells and that ShtIX-induced cell death can be efficiently reversed by the ferroptosis inhibitors and the iron chelator, but not by other cell death inhibitors. After cells were treated with ShtIX, there was an increase in Fe2+ content and lipid peroxidation accumulation, as well as a drop in GSH and GPX4 levels, all of which are indicators of ferroptosis. ShtIX also reduced the expression of Nrf2 and HO-1, and genetic Nrf2 silencing in NSCLC enhanced the effect of ShtIX-induced ferroptosis. Additionally, ShtIX retards tumor growth and induced ferroptosis through Nrf2/HO-1 signal pathway in the A549 xenograft model, whereas Fer-1 lessens the anticancer effect. Conclusion: This work provided the evidence that ShtIX caused ferroptosis in NSCLC cells, and inhibiting the Nrf2/HO-1 pathway can considerably exacerbate the effect of ShtIX-induced ferroptosis. The study establishes ShtIX as a promising natural ferroptosis inducer for the treatment of NSCLC.

5.
Nutr Cancer ; 74(8): 2996-3006, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35067132

RESUMEN

Background: Eryngium foetidum has long been used as a food ingredient and folk medicine in tropical regions. The anticancer activity of EF extract and the mechanisms remains unclear. Herein, we prepared four solvent extracts of EF leaves, detected the cytotoxic effects, and explored the potential mechanism by which these extracts induce cell death. Methods: The anticancer activity of the EF extracts was measured by MTT, CCK-8 and BrdU assays. The cell cycle was evaluated by flow cytometry and western blot. Apoptotic events were investigated with Hoechst, Annexin V/PI assays and western blot. The mitochondrial membrane potential was monitored using JC-1 staining, and ROS production was assessed with immunofluorescence. Results: The ethanol extract of EF leaves exhibited the strongest cytotoxic effect against SGC-7901 cells. The EFE extract significantly inhibited the SGC-7901 cells viability, arrested the cell cycle, increased the numbers of apoptotic cells, caused the loss of MMP, increased the Bax/Bcl-2 ratio, and led to cytochrome c release, and triggered ROS production. Conclusion: Our findings demonstrated for the first time that EFE extract induces mitochondrial associated apoptosis via ROS generation in SGC-7901 cells. Thus, EFE extract could be identified as a potential edible phytotherapy for the treatment of human gastric cancer.


Asunto(s)
Antineoplásicos , Eryngium , Neoplasias Gástricas , Antineoplásicos/farmacología , Apoptosis , Eryngium/metabolismo , Etanol , Humanos , Potencial de la Membrana Mitocondrial , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Hojas de la Planta , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Gástricas/tratamiento farmacológico
6.
Front Oncol ; 11: 629008, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708631

RESUMEN

BACKGROUND: Stigmasterol (SS) has been proven to possess potential anticancer activities in several cancer cell lines, but its molecular mechanism is still unknown. Thus, we investigated whether SS has the capabilities of inducing autophagy and its molecular mechanisms in gastric cancer cells. METHODS: We used CCK8 assay, clone formation assay, and EdU proliferation assay to assess the effects of SS on cell proliferation in SGC-7901 and MGC-803 cells in vitro, and its inhibition on the tumor growth of gastric cancer was observed in vivo. Apoptosis induced by SS was demonstrated using Hoechst and TUNEL staining, annexin V-FITC/PI assay. Immunofluorescence staining is used to detect the formation of autophagosomes triggered by SS. Apoptosis and autophagy related proteins were analyzed by western blot. RESULTS: The results indicated that SS treatment inhibited cell proliferation in SGC-7901 and MGC-803 cells. Furthermore, SS treatment induced apoptosis and autophagy by blocking Akt/mTOR signaling pathway. The pretreatment with the Akt inhibitor MK-2206 could promote apoptosis and autophagy induced by SS, predicting that Akt/mTOR pathway is involved in SS-induced apoptosis and autophagy. In addition, blockade of autophagy with 3-MA (an inhibitor of autophagy) enhanced SS-induced apoptosis in SGC-7901 and MGC-803 cells, implying that autophagy mediated by SS plays a cytoprotective role against apoptosis. Finally, an in vivo study demonstrated that tumor growth of gastric cancer was suppressed by SS in a xenograft model. CONCLUSION: Our findings illustrate for the first time that SS simultaneously induces apoptosis and protective autophagy by inhibiting Akt/mTOR pathway in gastric cancer cells, and SS may become a potential anticancer drug in treating gastric cancer in the future.

7.
Front Oncol ; 10: 575415, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194686

RESUMEN

Biosynthetic silver nanoparticles (AgNPs), specifically formed using medicinal plant extracts, have recently exhibited a remarkable therapeutic effect due to their anticancer potential. Here, we synthesized AgNPs using an aqueous extract of Ginkgo biloba leaves and evaluated its activity against cervical cancer (CCa) and the related molecular mechanisms. The physiochemical properties of the AgNPs were measured by ultraviolet-visible spectrophotometry, nanometre particle size analyzer and transmission electron microscopy. The AgNPs effects on cell proliferation and apoptosis were investigated through MTT, MTS, and colony formation assay; Hoechst 33258 staining; and flow cytometry. The intracellular ROS and oxidative stress levels were assessed using the appropriate commercial kits. Apoptosis-related protein levels were determined by western blotting. We prepared a series of different sized ginkgo extract synthesized AgNPs (GB-AgNPs), and the smallest mean particle size was 40.2 ± 1.2 nm with low polydispersity (0.091 ± 0.011), zeta potential values showed -34.56 mV. Compared to the controls, the GB-AgNP treatment inhibited the cell proliferation and induced the apoptosis of HeLa and SiHa cells. In addition, GB-AgNP treatment led to markedly increased levels of intracellular ROS, the release of cytochrome c (Cyt C) from mitochondria into the cytosol and the cleavage of caspase -9 and -3 in both CCa cell lines. Moreover, NAC, an ROS scavenger, eliminated the effect of GB-AgNPs on the HeLa and SiHa cells. This study reveals that GB-AgNPs suppresses cancer cell proliferation and induces apoptosis by upregulating intracellular ROS generation and inducing the activation of the caspase-dependent mitochondrial apoptotic pathway in CCa cells. Thus, GB-AgNPs may be a potential alternative drug for CCa therapy.

8.
Front Oncol ; 10: 116, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117763

RESUMEN

Penicilazaphilone C (PAC) is a novel azaphilonidal derivative isolated by our group that demonstrates good anticancer activities. Considering that its molecular mechanisms remain largely unknown, here we explore the molecular mechanisms of the anticancer activities of PAC against gastric cancer. The in vitro effects of PAC on cell growth, proliferation, and apoptosis were evaluated by MTT, BrdU, MTS, colony formation assays, Hoechst 33258 staining, and flow cytometry. Related proteins were examined by western blotting. Notch receptor expression was analyzed by RT-PCR. In vivo antitumor activities of PAC were observed in a nude mouse model. We found that compared to the controls, PAC treatment suppressed cell proliferation and promoted apoptosis in MGC-803 and SGC-7901 cells, and the Notch/PTEN/AKT axis was involved in the activating PAC-induced apoptosis. PAC treatment led to decreased levels of Notch (NTM), NICD, pPTEN, and pAKT compared to controls. PAC-induced inhibition of Notch-related protein expression levels and the resulting apoptosis were reversed by overexpression of Notch1 (NTM) or/and Notch2 (NTM). Moreover, PAC treatment clearly inhibited tumor growth in mice both bearing tumors derived from both MGC-803 and SGC-7901 cells. This work reveals that PAC induces the apoptosis by suppressing activation of Notch receptor proteolytic cleavage and subsequently blocking the PTEN/AKT signaling axis in gastric cancer cells. Thus, PAC is a potential alternative agent for the treatment of gastric cancer.

9.
Arch Pharm Res ; 41(10): 986-994, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29992400

RESUMEN

Toxicarioside N (Tox N), a natural product extract from Antiaris toxicaria, has been reported to induce apoptosis in human gastric cancer cells. However, the mechanism and actual role of autophagy in Tox N-induced apoptosis of human gastric cancer cells remains poorly understood. In the current study, we demonstrated that Tox N could induce autophagy by inhibiting the Akt/mTOR signaling pathway in SGC-7901 cells. Moreover, we found that the inhibition of autophagy by 3-methyladenine, an autophagy inhibitor, enhanced Tox N-induced apoptotic cell death. However, the stimulation of autophagy by rapamycin, an autophagy activator, remarkably suppressed Tox N-induced apoptosis, suggesting that autophagy plays a protective role in Tox N-induced apoptosis. Thus, the results from this study suggested that Tox N combination with an autophagy inhibitor might be a promising strategy to enhance the anticancer activity of Tox N for the treatment of human gastric cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Glicósidos/toxicidad , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Neoplasias Gástricas , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adenina/análogos & derivados , Adenina/farmacología , Apoptosis/fisiología , Autofagia/fisiología , Línea Celular Tumoral , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Neoplasias Gástricas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
10.
Arch Pharm Res ; 41(1): 71-78, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28940036

RESUMEN

Natural plant compounds with potent proliferation inhibition and apoptosis induction properties have been screened as novel anticancer drugs. Toxicarioside N (Tox N) was isolated from the seeds of the tropical plant Antiaris toxicaria in Hainan province, China. To our knowledge, the effects that Tox N has on the apoptosis of SGC-7901 cells and its potential mechanism have never been investigated. In this study, we detected the anticancer activities of Tox N and explored the potential mechanism in the human gastrointestinal cancer cell line SGC-7901. Here, we found that Tox N inhibited SGC-7901 cell growth in a dose- and time-dependent manner and induced apoptosis in cells based on cell morphology and flow cytometry analyses. Additionally, the SGC-7901 cell treated with Tox N up-regulated the expression level of cleaved caspase-3/9 and PARP, increased the Bax/Bcl-2 ratio, and led to the release of cytochrome c into the cytoplasm. In addition, Tox N treatment led to the phosphorylation of p38MAPK. SB203580, a p38MAPK inhibitor, partially attenuated Tox N induced apoptosis by preventing the activation of caspase-3/9 and PARP. Our results indicated for the first time that Tox N can induce SGC-7901 cells apoptosis by activating the p38MAPK pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Glicósidos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glicósidos/química , Glicósidos/aislamiento & purificación , Humanos , Conformación Molecular , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
11.
Springerplus ; 5(1): 1966, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27933244

RESUMEN

In recent years, marine fungi have become an important source of active marine natural products. Former researches are limited in habitats selection of fungi with bioactive compounds. In this paper were to measure antibacterial and antitumor cell activity for secondary metabolites of marine fungi, which were isolated from different habitats in coastal regions. 195 strains of marine fungi were isolated and purified from three different habitats. They biologically active experiment results showed that fungi isolation from the mangrove habitats had stronger antibacterial activity than others, and the stains isolated from the estuarial habitats had the least antibacterial activity. However, the strains separated from beach habitats strongly inhibited tumor cell proliferation in vitro, and fungi of mangrove forest habitats had the weakest activity of inhibiting tumor. Meanwhile, 195 fungal strains belonged to 46 families, 84 genera, 142 species and also showed 137 different types of activity combinations by analyzing the inhibitory activity of the metabolites fungi for 4 strains of pathogenic bacteria and B-16 cells. The study investigated the biological activity of marine fungi isolated from different habitats in Haikou coastal regions. The results help us to understand bioactive metabolites of marine fungi from different habitats, and how to selected biological activity fungi from various marine habitats effectively.

12.
Arch Pharm Res ; 39(12): 1621-1627, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27605109

RESUMEN

Two azaphilonidal derivatives [penicilazaphilones B (1) and C (2)], have been isolated from the fermented products of marine fungus strain Penicillium sclerotiorum M-22, penicilazaphilones C was a new compound. The compound's structures were identified by the analysis of spectroscopic data including 1D and 2D NMR techniques (1H-NMR, 13C-NMR, COSY, HMQC, and HMBC). Biological evaluation revealed that penicilazaphilones B and C showed selective cytotoxicity against melanoma cells B-16 and human gastric cancer cells SGC-7901 with IC50 values of 0.291, 0.449 and 0.065, 0.720 mM, respectively, while exhibiting no significant toxicity to normal mammary epithelial cells M10 at the same concentration. Moreover, penicilazaphilones C also exhibited strong antibacterial activity against Staphylococcus aureus, Pseudomonas aeruginosa, Klebsiella pneumonia and Escherichia coli with MIC values 0.037-0.150 mM, while penicilazaphilones B's bacteriostatic action was weaker.


Asunto(s)
Antibacterianos/farmacología , Antineoplásicos/farmacología , Benzopiranos/farmacología , Penicillium , Pigmentos Biológicos/farmacología , Animales , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Benzopiranos/química , Benzopiranos/aislamiento & purificación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Hongos , Humanos , Melanoma Experimental , Ratones , Pruebas de Sensibilidad Microbiana/métodos , Pigmentos Biológicos/química , Pigmentos Biológicos/aislamiento & purificación
13.
Biomaterials ; 53: 554-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25890751

RESUMEN

The combination of several potential strategies so as to develop new tumor vaccines is an attractive field of translational medicine. Pulsing tumor lysates with dendritic cells (DCs), in-vivo attraction of DCs by macrophage inflammatory protein 3α (MIP-3α), and reversion of the tumor suppressive microenvironment have been tested as strategies to develop tumor vaccines. In this study, we generated an alginate microsphere (named PaLtTcAdMIP3α) that encapsulated tumor lysates, live tumor cells engineering with a recombinant MIP-3α adenovirus and BCG. We used PaLtTcAdMIP3α as a model vaccine to test its antitumor activities. Our results showed that PaLtTcAdMIP3α expressed and excreted MIP-3α, which effectively attracted DCs ex vivo and in vivo. Injection of PaLtTcAdMIP3α into tumor-bearing mice effectively induced both therapeutic and prophylactic antitumor immunities in CT26, Meth A, B16-F10 and H22 models, but without any ensuing increase in adverse effects. Both tumor-specific cellular and humoral immune responses, especially the CD8(+) T cell-dependent cytotoxic T immunity, were found in the mice injected with PaLtTcAdMIP3α. The anti-tumor activity was abrogated completely by depletion of CD8(+) and partially by CD4(+) T lymphocytes. In addition, the number of IFN-γ-producing CD8(+) T cells in spleen and tumor tissues was significantly increased; but the number of CD4(+)CD25(+)FOXP3(+) regulatory T cells (Treg) in tumor tissues was decreased. These data strongly suggest that a combination of multi-current-using strategies such as the novel approach of using our PaLtTcAdMIP3α microspheres could be an effective tumor model vaccine.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Quimiocina CCL20/inmunología , Composición de Medicamentos , Animales , Línea Celular Tumoral , Ratones , Microambiente Tumoral
14.
Immunology ; 143(2): 230-40, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24750112

RESUMEN

The IgE Fcε3 domain is an active immunotherapeutic target for asthma and other allergic diseases. However, previous methods for preparing IgE fusion protein vaccines are complex. Antigen 43 (Ag43) is a surface protein found in Escherichia coli that contains α and ß subunits (the α subunit contains multiple T epitopes). Here we constructed a novel Ag43 surface display system (Ag43 system) to express Ag43 chimeric proteins to disrupt immune tolerance against IgE. The Ag43 system was constructed from the E. coli strain Tan109, in which the Ag43 gene was deleted and a recombinant plasmid (pETAg43) expressing a partial Ag43 gene was introduced. The Fcε3 domain of the IgE gene was then subcloned into plasmid pETAg43, resulting in a recombinant plasmid pETAg43/Fcε3, which was used to transform Tan109 for Ag43/Fcε3 surface expression. Thereafter, Ag43/Fcε3 was investigated as an asthma vaccine in a mouse model. Ag43/Fcε3 was expressed on and could be separated from the bacterial surface by heating to 60° while retaining activity. Ag43/Fcε3, as a protein vaccine, produced neutralizing autoantibodies to murine IgE, induced significant anti-asthma effects, and regulated IgE and T helper cytokines in a murine asthma model. Data show that Ag43/Fcε3 chimeric protein is a potential model vaccine for asthma treatment, and that the Ag43 system may be an effective tool for novel vaccine preparation to break immune tolerance to other self-molecules.


Asunto(s)
Adhesinas de Escherichia coli/inmunología , Asma/prevención & control , Hiperreactividad Bronquial/prevención & control , Receptores de IgE/inmunología , Vacunas Sintéticas/inmunología , Adhesinas de Escherichia coli/biosíntesis , Adhesinas de Escherichia coli/genética , Traslado Adoptivo , Animales , Anticuerpos Neutralizantes/sangre , Asma/sangre , Asma/inmunología , Asma/fisiopatología , Autoanticuerpos/sangre , Hiperreactividad Bronquial/sangre , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/fisiopatología , Broncoconstricción , Células Cultivadas , Clonación Molecular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Histamina/metabolismo , Tolerancia Inmunológica , Inmunoglobulina E/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Ovalbúmina/inmunología , Receptores de IgE/biosíntesis , Receptores de IgE/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Tiempo , Vacunas Sintéticas/biosíntesis , Vacunas Sintéticas/genética
15.
Int J Cancer ; 134(8): 1981-90, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24150860

RESUMEN

Various angiogenesis-related self-molecules have been considered to be therapeutic targets. However, the direct use of self-molecules as vaccines is not recommended because of the inherent ability of the host to develop immune tolerance. Antigen 43 (Ag43) is a surface protein found in E. coli and contains an α and a ß subunits, which contains multiple T epitopes in α subunit. Here we construct a novel Ag43 surface display system (Ag43 system) to express Ag43 chimeric proteins to disrupt immune tolerance against self-molecules. The Ag43 system was constructed from an Escherichia coli strain Tan109, derived from JM109, in which the Ag43 gene was deleted and a recombinant plasmid (pETAg43') expressing a partial Ag43 gene was introduced. The extracellular domain of angiogenesis-related endoglin gene was then subcloned into plasmid pETAg43', resulting in a recombinant plasmid pETAg43'/END(e) which was then used to transform Tan109 for protein expression. We found that Ag43 and endoglin chimeric protein (Ag43'/END(e) ) was expressed on the bacterial surface. The chimeric protein could be separated from the bacterial surface by heating to 60°C and yet retain activity. We used Ag43'/END(e) as a protein vaccine and found that it could disrupt immune tolerance against endoglin by inducing significant antitumor activities and inhibit angiogenesis in several tumor models without significant side effects. These data suggest that Ag43'/END(e) chimeric protein is a potential model vaccine for active tumor immunotherapy, and that Ag43 system could be an effective tool for novel vaccine preparation to break immune tolerance to other angiogenesis-related self-molecules for cancer therapy.


Asunto(s)
Adhesinas de Escherichia coli/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma Pulmonar de Lewis/terapia , Tolerancia Inmunológica/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Neovascularización Patológica/terapia , Adhesinas de Escherichia coli/genética , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Endoglina , Epítopos de Linfocito T , Escherichia coli/genética , Escherichia coli/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neovascularización Patológica/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología
16.
Oncol Rep ; 30(1): 478-84, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23615686

RESUMEN

Cytochalasin D (CytD) targets actin, a ubiquitous protein in eukaryotic cells. Previous studies have focused mainly on the antitumor effects of CytD. We previously found CytD to promote lung metastasis in B16 melanoma cells, which we had not anticipated, and, therefore, in the present study we investigated the possible underlying mechanisms. B16 melanoma cells were co-cultured with CytD and other agents and used to establish a lung metastatic model. In this B16 melanoma metastatic model, significantly increased lung metastasis and lung weight were found in CytD-treated mice, which was almost completely suppressed by tissue factor (TF) RNA interference expressed via lentivirus. The results of northern and western blot, and real-time RT-PCR analysis showed that the expression of TF was significantly upregulated in B16 cells treated with CytD but was significantly inhibited by TF RNA interference. In addition, upregulation and phosphorylation of mitogen-activated protein kinase p38 were also found in the metastatic lung tissues treated with CytD and in the B16 cells co-cultured with CytD and factor VIIa (FVIIa), but not in cells cultured with CytD, dimethyl sulfoxide or FVIIa alone. These results indicate that CytD stimulates the expression of TF in B16 melanoma cells, activating both coagulation-dependent and -independent pathways via binding to FVIIa, eventually promoting lung metastasis. TF interference is a potential approach to the prevention of B16 melanoma metastasis.


Asunto(s)
Citocalasina D/farmacología , Neoplasias Pulmonares/secundario , Melanoma Experimental/metabolismo , Tromboplastina/metabolismo , Animales , Línea Celular Tumoral , Factor VIIa/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Melanoma Experimental/genética , Ratones , Ratones Endogámicos C57BL , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Tromboplastina/biosíntesis , Tromboplastina/genética , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Artículo en Chino | MEDLINE | ID: mdl-24812827

RESUMEN

OBJECTIVE: To observe the immune response induced by complex gene vaccine pcSAG1-ROP5 of Toxoplasma gondii in mice. METHODS: The recombinant eukaryotic expression plasmids pcSAG1, pcROP5 and pcSAG1-ROP5 were constructed and identified by PCR, restriction enzyme digestion, and sequencing. The three recombinant plasmids were transfected into HeLa cells to express in vitro and identified by Western blotting analysis. Seventy Kunming mice were randomly divided into 5 groups with 14 each, i.e. pcSAG1 group, pcROP5 group, pcSAG1-ROP5 group, blank plasmid group and PBS control group. The mice were immunized intramuscularly with pcSAG1, pcROP5, pcSAG1-ROP5, pcDNA3.1, and PBS, respectively, every two weeks for three times. Sera were collected before each injection and 2 weeks after the last immunization. The titer of mice serum in pcSAG1-ROP5 group combined with recombinant protein SAG1, ROP5 and SAG1-ROP5 and the level of IgG against T. gondii in 5 groups were determined by ELISA. Three weeks after the last immunization, ten mice of each group were challenged with 10(3) tachyzoites of the virulent T. gondii RH strain to observe the survival time. One week later, the rest four mice in each group were sacrificed and the supernatant of cultured splenocytes was collected for the detection of IFN-gamma and IL-4. RESULTS: Western blotting showed that the recombinant plasmids pcSAG1, pcROP5 and pcSAG1-ROP5 were expressed in HeLa cells with M(r) 31 000, 57 000, and 88 000, respectively. The serum titer in pcSAG1-ROP5 group combined with SAG1, ROP5 and SAG1-ROP5 was 1:320, 1:160, and 1:2560, respectively. The IgG level kept rising in pcSAG1, pcROP5 and pcSAG1-ROP5 groups. Two weeks after the last immunization, the IgG level in pcSAG1-ROP5 group was higher than those in other groups (P<0.05). After a lethal challenge of T. gondii RH strain, the survival time of the mice in pcSAG1-ROP5 group was (288 +/- 7) h, which was 48 h and 96h longer than the groups of pcSAG1 and pcROP5, respectively (P< 0.05). Four weeks after the last immunization, IFN-gamma in splenocyte culture of pcSAG1-ROP5 group [(908.52 +/- 6.31) pg/ml] was higher than other groups (P<0.05), with no significant difference in IL-4 (P>0.05). CONCLUSION: Compared with the single gene vaccines pcSAG1 and pcROP5, higher levels of IgG and IFN-gamma and longer survival time are observed in mice immunized with pcSAG1-ROP5.


Asunto(s)
Vacunas Antiprotozoos/inmunología , Toxoplasma/inmunología , Toxoplasmosis Animal/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/inmunología , Femenino , Células HeLa , Humanos , Inmunización , Inmunoglobulina G/sangre , Interferón gamma/inmunología , Ratones , Ratones Endogámicos , Plásmidos , Proteínas Protozoarias/inmunología , Proteínas Recombinantes/genética , Toxoplasma/genética , Toxoplasmosis Animal/prevención & control , Transfección
18.
Artículo en Inglés | MEDLINE | ID: mdl-24812879

RESUMEN

OBJECTIVE: To examine the immunoprotection effect induced by MIC8 DNA vaccine co-immunized with a plasmid encoding murine IL-12 (pcIL-12) as an adjuvant in mice against the challenge of Toxoplasma gondii. METHODS: The gene sequence encoding MIC8 of T. gondii RH strain was inserted into eukaryotic expression vector pcDNA3.1 to construct the pcMIC8 expression plasmid. The recombinant plasmid was transfected into HeLa cells to test its expression and the recombinant protein was then characterized by Western blotting. Eighty Kunming mice were randomly divided into 5 groups (16 per group): 3 control groups (PBS, pcDNA3.1, and pcIL-12), pcMIC8 group, and pcMIC8 plus pcIL-12 group. Mice in the pcMIC8 plus pcIL-12 group were co-injected intramuscularly at a dosage of 100 microl each of pcMIC8 and pcIL-12 suspended in 100 microg sterile PBS. Mice in other groups were inoculated with PBS, pcDNA3.1, pcIL-12, and pcMIC8 respectively following the same protocol. All the mice received three immunizations at 2-week intervals. Serum samples were collected on day 0, 13, 27, 41, and 55 before each inoculation for determining antibody IgG, IgG subclass IgG2a. Four weeks after the final immunization, IFN-gamma and IL-4 levels in splenocytes cultures from immunized mice were detected by ELISA. The mice were challenged with 10(3) tachyzoites of the virulent T. gondii RH strain three weeks after the last immunization to observe the survival time. RESULTS: Western blotting showed that the protein extracts in HeLa cells upon transfection with pcMIC8 were effectively expressed in cells. The levels of IgG (0.51 +/- 0.028) and IgG2a (0.261 +/- 0.04)(on day 55) in mice immunized with pcMIC8 plus pcIL-12 were higher than pcMIC8 group (497.65 +/- 98.15) and control groups (PBS 47.18 +/- 2.73, pcDNA3.1 50.08 +/- 4.62, pcIL-12 118.15 +/- 12.73) (P < 0.05). There was no significant difference in the level of IgG 1 and IL-4 among the five groups (P > 0.05). After a lethal challenge of T. gondii RH strain, the survival time in mice immunized with pcMIC8 plus pcIL-12 (15d) was prolonged in comparison to that of pcMIC8 (10d) and control groups (PBS 5 d, pcDNA3.1 6d, pcIL-12 8 d) (P < 0.05). CONCLUSION: The immune responses induced by the combined use of the recombinant plasmid encoding MIC8 of T. gondii with murine IL-2 gene adjuvant can be enhanced.


Asunto(s)
Adyuvantes Inmunológicos , Moléculas de Adhesión Celular/inmunología , Interleucina-12/inmunología , Proteínas Protozoarias/inmunología , Toxoplasmosis/prevención & control , Vacunas de ADN/inmunología , Adyuvantes Inmunológicos/genética , Animales , Moléculas de Adhesión Celular/genética , Células HeLa , Humanos , Inmunización , Interleucina-12/genética , Ratones , Ratones Endogámicos , Plásmidos , Proteínas Protozoarias/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Toxoplasma , Transfección , Vacunas de ADN/genética , Vacunas de ADN/uso terapéutico
19.
PLoS One ; 7(11): e50351, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209720

RESUMEN

Toxicarioside A is a cardenolide isolated mainly from plants and animals. Emerging evidence demonstrate that cardenolides not only have cardiac effects but also anticancer effects. In this study, we used in vivo models to investigate the antitumor activities of toxicarioside A and the potential mechanisms behind them. Murine colorectal carcinoma (CT26) and Lewis lung carcinoma (LL/2) models were established in syngeneic BALB/c and C57BL/6 mice, respectively. We found that the optimum effective dose of toxicarioside A treatment significantly suppressed tumor growth and angiogenesis in CT and LL/2 tumor models in vivo. Northern and Western blot analysis showed significant inhibition of endoglin expression in toxicarioside A-treated human umbilical vein endothelial cells (HUVECs) in vitro and tumor tissues in vivo. Toxicarioside A treatment significantly inhibited cell proliferation, migration and invasion, but did not cause significant cell apoptosis and affected other membrane protein (such as CD31 and MHC I) expression. In addition, TGF-ß expression was also significantly inhibited in CT26 and LL/2 tumor cells treated with toxicarioside A. Western blot analysis indicated that Smad1 and phosphorylated Smad1 but not Smad2/3 and phosphorylated Smad2/3 were attenuated in HUVECs treated with toxicarioside A. Smad1 and Smad2/3 signaling remained unchanged in CT26 and LL/2 tumor cells treated with toxicarioside A. Endoglin knockout by small interfering RNA against endoglin induced alternations in Smad1 and Smad2/3 signaling in HUVECs. Our results indicate that toxicarioside A suppresses tumor growth through inhibition of endoglin-related tumor angiogenesis, which involves in the endoglin/TGF-ß signal pathway.


Asunto(s)
Glicósidos Cardíacos/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neovascularización Patológica , Factor de Crecimiento Transformador beta/metabolismo , Alginatos/química , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Endoglina , Células Endoteliales/citología , Femenino , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microcirculación , Modelos Químicos , Invasividad Neoplásica , Trasplante de Neoplasias , Neoplasias/patología , Fosforilación , ARN Interferente Pequeño/metabolismo , Transducción de Señal
20.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 43(3): 340-3, 2012 May.
Artículo en Chino | MEDLINE | ID: mdl-22812233

RESUMEN

OBJECTIVE: To study the antitumor efficacy of an immunoconjugate composed of adriamycin (ADM) and downsized Fab fragment of mouse anti-Endoglin monoclonal antibody. METHODS: The Fab fragment of mouse anti-Endoglin monoclonal antibody was downsized and conjugated with ADM by m-Maleimidobenzoyl-N-hydroxysuccinimide ester (MBS). The antitumor effect of the conjugate was tested in mice bearing subcutaneous injection of H22 tumor in vivo. RESULTS: The molecular ratio of Fab:ADM in conjugate was approximately 1:2. The Fab-ADM conjugate inhibited the growth of H22 by 91.94% on day 14 after injection at the dose of 0.4 mg/ kg, much higher the inhibition rate of 25.00% by the equivalent dose of free ADM. The median survival time of the mice treated with the conjugate was longer than those treated with free ADM. The Fab-ADM conjugate was significantly more effective than free ADM in tumor suppression and life span prolongation. CONCLUSION: Fab -ADM displayed more significant antitumor efficacy than free ADM in vivo and might be a novel candidate for cancer treatment.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Doxorrubicina/farmacología , Inmunotoxinas/farmacología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Animales , Antibióticos Antineoplásicos/farmacología , Endoglina , Masculino , Ratones , Neoplasias Experimentales/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...