Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Plant Cell Environ ; 46(8): 2507-2522, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37212208

RESUMEN

Field-grown rice (Oryza sativa L.), when exposed to various environmental stresses, produces high amounts of reactive oxygen species, such as H2 O2 . MicroRNAs (miRNAs) play crucial roles in plant stress responses. This study characterized the functions of H2 O2 -regulated miRNAs in rice. Small RNA deep sequencing revealed that miR156 levels decreased following H2 O2 treatment. Searches of the rice transcriptome and degradome databases indicated that OsSPL2 and OsTIFY11b are miR156-target genes. Interactions between miR156 and OsSPL2 and OsTIFY11b were confirmed using transient expression assays through agroinfiltration. In addition, the levels of OsSPL2 and OsTIFY11b transcripts were lower in transgenic rice plants overexpressing miR156 than in wild-type plants. The OsSPL2-GFP and OsTIFY11b-GFP proteins were localized to the nucleus. Yeast two-hybrid and bimolecular fluorescence complementation assays indicated interactions between OsSPL2 and OsTIFY11b. Furthermore, OsTIFY11b interacted with OsMYC2 to regulate the expression of OsRBBI3-3, which encodes a proteinase inhibitor. The results suggested that H2 O2 accumulation in rice suppresses the expression of miR156, and induces the expression of its target genes, OsSPL2 and OsTIFY11b, whose proteins interact in the nucleus to regulate the expression of OsRBBI3-3, which is involved in plant defense.


Asunto(s)
MicroARNs , Oryza , Oryza/genética , Oryza/metabolismo , Peróxido de Hidrógeno/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Secuencia de Bases , Estrés Fisiológico/genética , Plantas Modificadas Genéticamente/metabolismo , Regulación de la Expresión Génica de las Plantas , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo
2.
Bioresour Technol ; 370: 128566, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36592866

RESUMEN

The disposal of waste activated sludge (WAS) accounts for approximately 60 % of wastewater treatment plant operating costs. In this study, according to the reaction time and water quality parameters, ultrasonic hydrolysis of WAS is divided into three stages, including floc-disintegration (0-25.2 kJ/g TS), cell-disruption (25.2-36 kJ/g TS), and cell-degradation (over 36 kJ/g TS). The results show that more than 70 % carbon distributes inside the cell, which also contains 63.8 % protein enhancing denitrification capacity. Moreover, cell-degradation hydrolysate has a higher proportion of readily biodegradable COD, indicating that intracellular organic matter is more capable of denitrification than extracellular. Therefore, the optimal ultrasonic operating range is Es = 36-72 kJ/g TS as carbon source, and obtain the hydrolysate with high ratio of soluble chemical oxygen demand to total nitrogen for denitrification. Furthermore, this study supports the comprehensive interpretation of ultrasonic hydrolyzed WAS and the characteristics of hydrolysate as carbon source for enhancing denitrification.


Asunto(s)
Aguas del Alcantarillado , Eliminación de Residuos Líquidos , Eliminación de Residuos Líquidos/métodos , Desnitrificación , Ultrasonido , Nitrógeno/metabolismo , Carbono/metabolismo , Reactores Biológicos
3.
Cancer Lett ; 526: 66-75, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34808284

RESUMEN

Autologous cancer vaccines (ACVs) are a desirable approach for personalized medicine, but the efficiency of ACVs remains unsatisfactory due to their low immunogenicity. This study developed a platform that can enhance the immunogenicity of ACVs by transplanting the tumors into immunodeficient mice. The CT26 cell line was inoculated into severe combined immunodeficient mice (SCID) for vaccine preparation where escalates tumor development, subsequently diversifying the tumor antigenic topology. CT26/SCID cancer vaccines significantly inhibited tumor growth, increased the amount of tumor infiltrating lymphocytes, and triggered Th-1 predominant immune responses. Tumor antigenic profiles of CT26/SCID cells were further analyzed by liquid chromatography-tandem mass spectrometry. Compared to CT26 parental cells, a total of 428 differentially expressed proteins (DEPs) were detected. These DEPs revealed that CT26/SCID cells overexpressed several novel therapeutic targets, including KNG1, apoA-I and, ß2-GPI, which can trigger cytotoxic T cells towards Th-1 predominant immune responses and directly suppress proliferation in tumors. CT26/SCID cancer vaccines can be easily manufactured, while traits of triggering stronger antigen-specific Th-1 immune activity against tumors, are retained. Results of this study provide an effective proof-of-concept of an ACV for personalized cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Inmunoterapia/métodos , Animales , Vacunas contra el Cáncer/farmacología , Femenino , Humanos , Ratones
4.
Oncogene ; 39(37): 5950-5963, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32778768

RESUMEN

TMPRSS2 is an important membrane-anchored serine protease involved in human prostate cancer progression and metastasis. A serine protease physiologically often comes together with a cognate inhibitor for execution of proteolytically biologic function; however, TMPRSS2's cognate inhibitor is still elusive. To identify the cognate inhibitor of TMPRSS2, in this study, we applied co-immunoprecipitation and LC/MS/MS analysis and isolated hepatocyte growth factor activator inhibitors (HAIs) to be potential inhibitor candidates for TMPRSS2. Moreover, the recombinant HAI-2 proteins exhibited a better inhibitory effect on TMPRSS2 proteolytic activity than HAI-1, and recombinant HAI-2 proteins had a high affinity to form a complex with TMPRSS2. The immunofluorescence images further showed that TMPRSS2 was co-localized to HAI-2. Both KD1 and KD2 domain of HAI-2 showed comparable inhibitory effects on TMPRSS2 proteolytic activity. In addition, HAI-2 overexpression could suppress the induction effect of TMPRSS2 on pro-HGF activation, extracellular matrix degradation and prostate cancer cell invasion. We further determined that the expression levels of TMPRSS2 were inversely correlated with HAI-2 levels during prostate cancer progression. In orthotopic xenograft animal model, TMPRSS2 overexpression promoted prostate cancer metastasis, and HAI-2 overexpression efficiently blocked TMPRSS2-induced metastasis. In summary, the results together indicate that HAI-2 can function as a cognate inhibitor for TMPRSS2 in human prostate cancer cells and may serve as a potential factor to suppress TMPRSS2-mediated malignancy.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Serina Endopeptidasas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Masculino , Glicoproteínas de Membrana/química , Invasividad Neoplásica , Neoplasias de la Próstata/etiología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Proteínas Inhibidoras de Proteinasas Secretoras/metabolismo , Proteolisis
5.
BMC Cancer ; 20(1): 603, 2020 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-32600429

RESUMEN

BACKGROUND: Modulated electro-hyperthermia (mEHT) is a form of hyperthermia used in cancer treatment. mEHT has demonstrated the ability to activate host immunity by inducing the release of heat shock proteins, triggering apoptosis, and destroying the integrity of cell membranes to enhance cellular uptake of chemo-drugs in tumor cells. Both curcumin and resveratrol are phytochemicals that function as effective antioxidants, immune activators, and potential inhibitors of tumor development. However, poor bioavailability is a major obstacle for use in clinical cancer treatment. METHODS: This purpose of this study was to investigate whether mEHT can increase anti-cancer efficacy of nanosized curcumin and resveratrol in in vitro and in vivo models. The in vitro study included cell proliferation assay, cell cycle, and apoptosis analysis. Serum concentration was analyzed for the absorption of curcumin and resveratrol in SD rat model. The in vivo CT26/BALB/c animal tumor model was used for validating the safety, tumor growth curve, and immune cell infiltration within tumor tissues after combined mEHT/curcumin/resveratrol treatment. RESULTS: The results indicate co-treatment of mEHT with nano-curcumin and resveratrol significantly induced cell cycle arrest and apoptosis of CT26 cells. The serum concentrations of curcumin and resveratrol were significantly elevated when mEHT was applied. The combination also inhibited the growth of CT26 colon cancer by inducing apoptosis and HSP70 expression of tumor cells while recruiting CD3+ T-cells and F4/80+ macrophages. CONCLUSIONS: The results of this study have suggested that this natural, non-toxic compound can be an effective anti-tumor strategy for clinical cancer therapy. mEHT can enable cellular uptake of potential anti-tumor materials and create a favorable tumor microenvironment for an immunological chain reaction that improves the success of combined treatments of curcumin and resveratrol.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Neoplasias Colorrectales/terapia , Curcumina/administración & dosificación , Terapia por Estimulación Eléctrica/métodos , Hipertermia Inducida/métodos , Resveratrol/administración & dosificación , Animales , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/farmacocinética , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Disponibilidad Biológica , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Línea Celular Tumoral/trasplante , Neoplasias Colorrectales/patología , Terapia Combinada/métodos , Curcumina/efectos adversos , Curcumina/farmacocinética , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Nanopartículas/administración & dosificación , Ratas , Resveratrol/efectos adversos , Resveratrol/farmacocinética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
6.
Sci Rep ; 10(1): 8936, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488092

RESUMEN

Modulated electro-hyperthermia (mEHT) is a form of mild hyperthermia (HT) used for cancer treatment. The principle utility of HT is the ability not only to increase cell temperature, but also to increase blood flow and associated pO2 to the microenvironment. While investigational evidence has shown the unique ability of mEHT to elicit apoptosis in cancer cells, in vivo and in vitro, the same trait has not been observed with conventional HT. There is dissension as to what allows mEHT to elicit apoptosis despite heating to only mild temperatures, with the predominant opinion in favor of increased temperature at a cellular level as the driving force. For this study, we hypothesized that in addition to temperature, the amount of electrical energy delivered is a major factor in induction of apoptosis by mEHT. To evaluate the impact of electrical energy on apoptosis, we divided generally practiced mEHT treatment into 3 phases: Phase I (treatment start to 10 min. mark): escalation from 25 °C to 37 °C Phase II (10 min. mark to 15 min. mark): escalation from 37 °C to 42 °C Phase III (15 min. mark to 45 min. mark): maintenance at 42 °C Combinations of mEHT at 18 W power, mEHT at 7.5 W power, water bath, and incubator were applied to each of the three phases. Power output was recorded per second and calculated as average power per second. Total number of corresponding Joules emitted per each experiment was also recorded. The biological effect of apoptotic cell death was assayed by annexin-V assay. In group where mEHT was applied for all three phases, apoptosis rate was measured at 31.18 ± 1.47%. In group where mEHT was only applied in Phases II and III, apoptosis rate dropped to 20.2 ± 2.1%. Where mEHT was only applied in Phase III, apoptosis was 6.4 ± 1.7%. Interestingly, when mEHT was applied in Phases I and II, whether Phase III was conducted in either water bath at 42 °C or incubator at 37 °C, resulted in nearly identical apoptosis rates, 26 ± 4.4% and 25.9 ± 3.1%, respectively. These results showed that accumulation of mEHT at high-powered setting (18 W/sec) during temperature escalation (Phase I and Phase II), significantly increased apoptosis of tested cancer cells. The data also showed that whereas apoptosis rate was significantly increased during temperature escalation by higher power (18 W/sec), apoptosis was limited during temperature maintenance with lower power (7.5 W/sec). This presents that neither maintenance of 42 °C nor accumulation of Joules by mEHT has immediate correlating effect on apoptosis rate. These findings may offer a basis for direction of clinical application of mEHT treatment.


Asunto(s)
Hipertermia Inducida/métodos , Neoplasias/terapia , Microambiente Tumoral/fisiología , Células A549 , Apoptosis , Línea Celular Tumoral , Humanos , Oxígeno/sangre , Flujo Sanguíneo Regional/fisiología
7.
J Neurooncol ; 146(3): 417-426, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32020472

RESUMEN

INTRODUCTION: The failure of immune checkpoint inhibitor (ICPi) on glioblastoma (GBM) treatment underscores the need for improving therapeutic strategy. We aimed to change tumor associated macrophage (TAM) from M2 type (anti-inflammatory) to M1 (pro-inflammatory) type to increase the therapeutic response of ICPi. We proposed that combined rapamycin (R) and hydroxychloroquine (Q) preferentially induce M2 cells death, as fatty acid oxidation was their major source of energy. METHODS: Macrophage polarization was characterized on mice and human macrophage cell lines by specific cytokines stimulation with or without RQ treatment under single culture or co-culture with GBM cell lines. Tumor sizes were evaluated on subcutaneous and intracranial GL261 mice models with or without RQ, anti-PD1 mAb treatment. Tumor volumes assessed by MRI scan and proportions of tumor infiltrating immune cells analyzed by flow cytometry were compared. RESULTS: In vitro RQ treatment decreased the macrophages polarization of M2, increased the phagocytic ability, and increased the lipid droplets accumulation. RQ treatment decreased the expression levels of CD47 and SIRPα on tumor cells and macrophage cells in co-culture experiments. The combination of RQ and anti-PD1 treatment was synergistic in action. Enhanced the intra-tumoral M1/M2 ratio, the CD8/CD4 ratio in the intracranial GL261 tumor model after RQ treatment were evident. CONCLUSION: We provide a rationale for manipulating the macrophage phenotype and increased the therapeutic effect of ICPi. To re-educate and re-empower the TAM/microglia opens an interesting avenue for GBM treatment.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Hidroxicloroquina/administración & dosificación , Macrófagos/efectos de los fármacos , Sirolimus/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Encefálicas/metabolismo , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Femenino , Glioblastoma/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores
8.
Int J Nanomedicine ; 14: 1269-1279, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30863059

RESUMEN

PURPOSE: Modulated electro-hyperthermia (mEHT) stands to be a significant technological advancement in the hyperthermia field, utilizing autofocusing electromagnetic power on the cell membrane to create massive apoptosis. Since mEHT possesses the unique ability to excite cell membranes, we hypothesized that mEHT could enhance the uptake of liposomal drugs by enhancing phagocytic activity. MATERIALS AND METHODS: Water bath control and mEHT were used to compare the enhancement of liposome-encapsulated doxorubicin (Lipodox®) uptake by cancer cells. Cancer cells were made visible by doxorubicin fluorescence to investigate drug uptake. Viable cell yield was determined via the Trypan Blue exclusion method. Various substrates were used to investigate the mechanism of drug-uptake enhancement. The murine colon carcinoma model, CT26, was used to confirm the tissue infiltration of Lipodox® and its therapeutic effect. RESULTS: mEHT treatment showed a significant enhancement of Lipodox® uptake of doxorubicin fluorescence compared with 37°C or 42°C water bath treatment. Tumor tissue sections also confirmed that mEHT treatment achieved the highest doxorubicin concentration in vivo (1.44±0.32 µg/g in mEHT group and 0.79±0.32 µg/g in 42°C water bath). Wortmannin was used to inhibit the macropinocytosis effect and 70 kDa dextran-FITC served as uptake substance. The uptake of dextran-FITC by cancer cells significantly increased after mEHT treatment whereas such enhancement was significantly inhibited by wortmannin. CONCLUSION: The result showed mEHT-induced particle-uptake through macropinocytosis. mEHT-enhanced uptake of Lipodox® may amplify the therapeutic effect of liposomal drugs. This novel finding warrants further clinical investigation.


Asunto(s)
Doxorrubicina/análogos & derivados , Hipertermia Inducida , Neoplasias/tratamiento farmacológico , Células A549 , Animales , Antibióticos Antineoplásicos , Apoptosis , Membrana Celular , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Células Hep G2 , Humanos , Ratones Endogámicos BALB C , Neoplasias/patología , Tamaño de la Partícula , Pinocitosis , Polietilenglicoles/farmacología , Polietilenglicoles/uso terapéutico , Transducción de Señal
9.
Sci Rep ; 9(1): 20403, 2019 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-31892709

RESUMEN

The p53 gene is an important tumour suppressor gene. Mutant p53 genes account for about half of all lung cancer cases. There is increasing evidence for the anti-tumour effects of statins via inhibition of the mevalonate pathway. We retrospectively investigated the correlation between statin use and lung cancer prognosis using the Taiwanese National Health Insurance Research Database, mainly focusing on early-stage lung cancer. This study reports the protective effects of statin use in early-stage lung cancer patients regardless of chemotherapy. Statin treatments reduced the 5-year mortality (odds ratio, 0.43; P < 0.001) in this population-based study. Significantly higher levels of cellular apoptosis, inhibited cell growth, and regulated lipid raft content were observed in mutant p53 lung cancer cells treated with simvastatin. Further, simvastatin increased the caspase-dependent apoptotic pathway, promotes mutant p53 protein degradation, and decreased motile activity in lung cancer cells with p53 missense mutations. These data suggest that statin use in selected lung cancer patients may have clinical benefits.


Asunto(s)
Adenocarcinoma del Pulmón/mortalidad , Apoptosis/efectos de los fármacos , Enfermedades Cardiovasculares/prevención & control , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neoplasias Pulmonares/mortalidad , Proteína p53 Supresora de Tumor/genética , Adenocarcinoma del Pulmón/genética , Adulto , Anciano , Anciano de 80 o más Años , Ciclo Celular/efectos de los fármacos , Bases de Datos Factuales , Femenino , Humanos , Neoplasias Pulmonares/genética , Masculino , Microdominios de Membrana/efectos de los fármacos , Persona de Mediana Edad , Pronóstico , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología , Tasa de Supervivencia
10.
Oncotarget ; 9(6): 6883-6896, 2018 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-29467937

RESUMEN

Glioblastoma (GBM) cells are characterized by high phagocytosis, lipogenesis, exocytosis activities, low autophagy capacity and high lysosomal demand are necessary for survival and invasion. The lysosome stands at the cross roads of lipid biosynthesis, transporting, sorting between exogenous and endogenous cholesterol. We hypothesized that three already approved drugs, the autophagy inducer, sirolimus (rapamycin, Rapa), the autophagy inhibitor, chloroquine (CQ), and DNA alkylating chemotherapy, temozolomide (TMZ) could synergize against GBM. This repurposed triple therapy combination induced GBM apoptosis in vitro and inhibited GBM xenograft growth in vivo. Cytotoxicity is caused by induction of lysosomal membrane permeabilization and release of hydrolases, and may be rescued by cholesterol supplementation. Triple treatment inhibits lysosomal function, prevents cholesterol extraction from low density lipoprotein (LDL), and causes clumping of lysosome associated membrane protein-1 (LAMP-1) and lipid droplets (LD) accumulation. Co-treatment of the cell lines with inhibitor of caspases and cathepsin B only partially reverse of cytotoxicities, while N-acetyl cysteine (NAC) can be more effective. A combination of reactive oxygen species (ROS) generation from cholesterol depletion are the early event of underling mechanism. Cholesterol repletion abolished the ROS production and reversed the cytotoxicity from QRT treatment. The shortage of free cholesterol destabilizes lysosomal membranes converting aborted autophagy to apoptosis through either direct mitochondria damage or cathepsin B release. This promising anti-GBM triple therapy combination severely decreases mitochondrial function, induces lysosome-dependent apoptotic cell death, and is now poised for further clinical testing and validation.

11.
Oncotarget ; 7(51): 84082-84092, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27556507

RESUMEN

Radiofrequency-induced hyperthermia (HT) treatments for cancer include conventional capacitive coupling hyperthermia (cCHT) and modulated electro-hyperthermia (mEHT). In this study, we directly compared these methods with regard to in vitro cytotoxicity and mechanisms of action under isothermal conditions. Hepatoma (HepG2) cells were exposed to HT treatment (42°C for 30 min) using mEHT, cCHT or a water bath. mEHT produced a much higher apoptosis rate (43.1% ± 5.8%) than cCHT (10.0% ± 0.6%), the water bath (8.4% ± 1.7%) or a 37°C control (6.6% ± 1.1%). The apoptosis-inducing effect of mEHT at 42°C was similar to that achieved with a water bath at 46°C. mEHT also increased expression of caspase-3, 8 and 9. All three hyperthermia methods increased intracellular heat shock protein 70 (Hsp70) levels, but only mEHT greatly increased the release of Hsp70 from cells. Calreticulin and E-cadherin levels in the cell membrane also increased after mEHT treatment, but not after cCHT or water bath. These results suggest that mEHT selectively deposits energy on the cell membrane and may be a useful treatment modality that targets cancer cell membranes.


Asunto(s)
Apoptosis , Calor , Hipertermia Inducida/métodos , Cadherinas/metabolismo , Calreticulina/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Células Hep G2 , Humanos , Células MCF-7 , Neoplasias/metabolismo , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , beta Catenina/metabolismo
12.
BMC Cancer ; 15: 708, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26472466

RESUMEN

BACKGROUND: The treatment of intratumoral dentritic cells (DCs) commonly fails because it cannot evoke immunity in a poor tumor microenvironment (TME). Modulated electro-hyperthermia (mEHT, trade-name: oncothermia) represents a significant technological advancement in the hyperthermia field, allowing the autofocusing of electromagnetic power on a cell membrane to generate massive apoptosis. This approach turns local immunogenic cancer cell death (apoptosis) into a systemic anti-tumor immune response and may be implemented by treatment with intratumoral DCs. METHODS: The CT26 murine colorectal cancer model was used in this investigation. The inhibition of growth of the tumor and the systemic anti-tumor immune response were measured. The tumor was heated to a core temperature of 42 °C for 30 min. The matured synergetic DCs were intratumorally injected 24 h following mEHT was applied. RESULTS: mEHT induced significant apoptosis and enhanced the release of heat shock protein70 (Hsp70) in CT26 tumors. Treatment with mEHT-DCs significantly inhibited CT26 tumor growth, relative to DCs alone or mEHT alone. The secondary tumor protection effect upon rechallenging was observed in mice that were treated with mEHT-DCs. Immunohistochemical staining of CD45 and F4/80 revealed that mEHT-DC treatment increased the number of leukocytes and macrophages. Most interestingly, mEHT also induced infiltrations of eosinophil, which has recently been reported to be an orchestrator of a specific T cell response. Cytotoxic T cell assay and ELISpot assay revealed a tumor-specific T cell activity. CONCLUSIONS: This study demonstrated that mEHT induces tumor cell apoptosis and enhances the release of Hsp70 from heated tumor cells, unlike conventional hyperthermia. mEHT can create a favorable tumor microenvironment for an immunological chain reaction that improves the success rate of intratumoral DC immunotherapy.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Neoplasias Colorrectales/terapia , Células Dendríticas/inmunología , Inmunoterapia , Microambiente Tumoral/inmunología , Animales , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Células Dendríticas/trasplante , Humanos , Hipertermia Inducida , Ratones , Linfocitos T Citotóxicos/inmunología
13.
Cancer Res ; 75(14): 2949-60, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26018085

RESUMEN

Dysregulation of androgen signaling and pericellular proteolysis is necessary for prostate cancer progression, but the links between them are still obscure. In this study, we show how the membrane-anchored serine protease TMPRSS2 stimulates a proteolytic cascade that mediates androgen-induced prostate cancer cell invasion, tumor growth, and metastasis. We found that matriptase serves as a substrate for TMPRSS2 in mediating this proinvasive action of androgens in prostate cancer. Further, we determined that higher levels of TMPRSS2 expression correlate with higher levels of matriptase activation in prostate cancer tissues. Lastly, we found that the ability of TMPRSS2 to promote prostate cancer tumor growth and metastasis was associated with increased matriptase activation and enhanced degradation of extracellular matrix nidogen-1 and laminin ß1 in tumor xenografts. In summary, our results establish that TMPRSS2 promotes the growth, invasion, and metastasis of prostate cancer cells via matriptase activation and extracellular matrix disruption, with implications to target these two proteases as a strategy to treat prostate cancer.


Asunto(s)
Andrógenos/farmacología , Matriz Extracelular/metabolismo , Neoplasias de la Próstata/patología , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/fisiología , Animales , Células CHO , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cricetinae , Cricetulus , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Matriz Extracelular/patología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Serina Endopeptidasas/genética , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Cancer Res ; 74(18): 5229-43, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25060522

RESUMEN

The transmembrane cell adhesion protein ADAM9 has been implicated in cancer cell migration and lung cancer metastasis to the brain, but the underpinning mechanisms are unclear and clinical support has been lacking. Here, we demonstrate that ADAM9 enhances the ability of tissue plasminogen activator (tPA) to cleave and stimulate the function of the promigratory protein CDCP1 to promote lung metastasis. Blocking this mechanism of cancer cell migration prolonged survival in tumor-bearing mice and cooperated with dexamethasone and dasatinib (a dual Src/Abl kinase inhibitor) treatment to enhance cytotoxic treatment. In clinical specimens, high levels of ADAM9 and CDCP1 correlated with poor prognosis and high risk of mortality in patients with lung cancer. Moreover, ADAM9 levels in brain metastases derived from lung tumors were relatively higher than the levels observed in primary lung tumors. Our results show how ADAM9 regulates lung cancer metastasis to the brain by facilitating the tPA-mediated cleavage of CDCP1, with potential implications to target this network as a strategy to prevent or treat brain metastatic disease.


Asunto(s)
Proteínas ADAM/metabolismo , Adenocarcinoma/patología , Neoplasias Encefálicas/secundario , Neoplasias Pulmonares/patología , Proteínas de la Membrana/metabolismo , Activadores Plasminogénicos/metabolismo , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma del Pulmón , Animales , Antígenos CD/metabolismo , Antígenos de Neoplasias , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Dasatinib , Dexametasona/administración & dosificación , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones SCID , Metástasis de la Neoplasia , Proteínas de Neoplasias/metabolismo , Activadores Plasminogénicos/genética , Pirimidinas/administración & dosificación , Tiazoles/administración & dosificación
15.
PLoS One ; 9(4): e94065, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24705471

RESUMEN

Lung cancer is the leading cause of cancer death worldwide, and brain metastasis is a major cause of morbidity and mortality in lung cancer. CDH2 (N-cadherin, a mesenchymal marker of the epithelial-mesenchymal transition) and ADAM9 (a type I transmembrane protein) are related to lung cancer brain metastasis; however, it is unclear how they interact to mediate this metastasis. Because microRNAs regulate many biological functions and disease processes (e.g., cancer) by down-regulating their target genes, microRNA microarrays were used to identify ADAM9-regulated miRNAs that target CDH2 in aggressive lung cancer cells. Luciferase assays and western blot analysis showed that CDH2 is a target gene of miR-218. MiR-218 was generated from pri-mir-218-1, which is located in SLIT2, in non-invasive lung adenocarcinoma cells, whereas its expression was inhibited in aggressive lung adenocarcinoma. The down-regulation of ADAM9 up-regulated SLIT2 and miR-218, thus down-regulating CDH2 expression. This study revealed that ADAM9 activates CDH2 through the release of miR-218 inhibition on CDH2 in lung adenocarcinoma.


Asunto(s)
Proteínas ADAM/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Cadherinas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/genética , Regiones no Traducidas 3' , Proteínas ADAM/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Antígenos CD/química , Antígenos CD/genética , Secuencia de Bases , Sitios de Unión , Cadherinas/química , Línea Celular Tumoral , Movimiento Celular/genética , Análisis por Conglomerados , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Proteínas de la Membrana/genética , MicroARNs/química , Interferencia de ARN , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...