Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 986
Filtrar
1.
Cell Cycle ; 19(23): 3260-3276, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33222607

RESUMEN

Non-small cell lung cancer (NSCLC) is a leading cause of cancer death in both men and women. microRNAs (miRs) can exert important functions in cancer development. However, the role of miR-877 in NSCLC as it relates to tartrate resistant acid phosphatase 5 (ACP5) is unknown. For this study, the gain-and-loss-of-function experiments were performed to explore the effects of miR-877 and ACP5 on NSCLC. miR-877 expression in LC and paracancerous tissues, lung epithelial cell line and NSCLC cell lines was detected, and the association between miR-877 expression and clinical features of LC patients was analyzed. The levels of ACP5, epithelial-mesenchymal transition (EMT) markers and apoptosis-related proteins were measured. In vivo experiments were conducted for further validation. Consequently, we found that miR-877 expression was lowered in LC tissues and cell lines, and correlated with clinical stage, differentiation, lymph node metastasis and prognosis of NSCLC patients. Additionally, miR-877 was determined to inhibit ACP5 activity, and miR-877 downregulated the PI3K/AKT pathway by silencing ACP5. Furthermore, overexpression of miR-877 inhibited the viability, migration, invasion and EMT of NSCLC cells, but promoted cell apoptosis. In conclusion, miR-877 overexpression inhibited malignant biological behaviors of NSCLC cells by downregulating ACP5 and inactivating the PI3K/AKT pathway.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , MicroARNs/biosíntesis , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatasa Ácida Tartratorresistente/metabolismo , Células A549 , Anciano , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Fosfatasa Ácida Tartratorresistente/antagonistas & inhibidores , Fosfatasa Ácida Tartratorresistente/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
2.
J Immunol Res ; 2020: 7263602, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33062724

RESUMEN

Serine/threonine protein kinase-3 (STK3) is a critical molecule of the Hippo pathway but little is known about its biological functions in the ovarian cancer development. We demonstrated the roles of STK3 in ovarian cancer. Existing databases were used to study the expression profile of STK3. STK3 was significantly downregulated in OC patients, and the low STK3 expression was correlated with a poor prognosis. In vitro cell proliferation, apoptosis, and migration assays, and in vivo subcutaneous xenograft tumor models were used to determine the roles of STK3. The overexpression of STK3 significantly inhibited cell proliferation, apoptosis, and migration of ovarian cancer cells in vitro and tumor growth in vivo. Bisulfite sequencing PCR analysis was performed to validate the methylation of STK3 in ovarian cancer. RNA sequencing and gene set enrichment analysis (GSEA) were used to compare the transcriptome changes in the STK3 overexpression ovarian cancer and control cells. The signaling pathway was analyzed by western blotting. STK3 promoted the migration of CD8+ T-cells by activating nuclear transcription factor κB (NF-κB) signaling. STK3 is a potential predictor of OC. It plays an important role in suppressing tumor growth of ovarian cancer and in chemotaxis of CD8+ T-cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inhibidores de Crecimiento/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Carcinogénesis , Línea Celular Tumoral , Quimiotaxis , Citotoxicidad Inmunológica , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Inhibidores de Crecimiento/genética , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Neoplasias Ováricas/mortalidad , Proteínas Serina-Treonina Quinasas/genética , Serina-Treonina Quinasa 3 , Transducción de Señal , Análisis de Supervivencia
3.
J Immunol Res ; 2020: 7671502, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33062725

RESUMEN

Glioblastoma (GBM) is an aggressive brain tumor with shorter median overall survival time. It is urgent to find novel methods to enhance the therapeutic efficiency clinically. miR-373 is related to the biological development process of cancers, but there are no reports whether modulation on miR-373 could affect GBM development or modify the efficiency of chemo- or radiotherapy yet. Our current study found that the higher level of miR-373 was observed in U-251 cells. Inhibition on miR-373 could reduce the U-251 cell number by 65% and PCNA expression obviously. In addition, inhibition on miR-373 sensitized U-251 cells to chemo- or radiotherapy. The cell cycle of U-251 cells could be modulated by miR-373 knockdown, which could enhance the p21 expression and reduce the cdc2 level. Anti-miR-373 could increase the Bax/Bcl-2 ratio of U-251 cells and induce cell apoptosis significantly. These above effects of miR-373 could be reversed by Limk1 overexpression. Thus, our experimental data confirmed the fact that miR-373 could be a new therapeutic target to enhance the efficiency of chemo- or radiotherapy for clinical GBM patients.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Inhibidores de Crecimiento/genética , Quinasas Lim/metabolismo , MicroARNs/genética , Apoptosis , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Quinasas Lim/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Fármacos Sensibilizantes a Radiaciones , Proteína X Asociada a bcl-2/metabolismo
4.
Metab Eng ; 54: 54-68, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30851381

RESUMEN

Chinese hamster ovary (CHO) cells in fed-batch cultures are known to consume large amounts of nutrients and divert significant portion of them towards the formation of byproducts, some of which, including lactate and ammonia, are known to be growth inhibitory in nature. A major fraction of these inhibitory metabolites are byproducts or intermediates of amino acid catabolism. Limiting the supply of amino acids has been shown to curtail the production of corresponding inhibitory byproducts resulting in enhanced growth and productivities in CHO cell fed-batch cultures (Mulukutla et al., 2017). In the current study, metabolic engineering of CHO cells was undertaken in order to reduce the biosynthesis of these novel growth inhibitors. Phenylalanine-tyrosine (Phe-Tyr) and branched chain amino acid (BCAA) catabolic pathways were engineered as part of this effort. Four genes that encode enzymes in the Phe-Tyr pathway, which were observed to be minimally expressed in CHO cells, were in turn overexpressed. Metabolically engineered cells were prototrophic to tyrosine and had reduced production of the inhibitory byproducts from Phe-Tyr pathway including 3-phenyllactate and 4-hydroxyphenyllactate. In case of BCAA catabolic pathway, branched chain aminotransferase 1 (BCAT1) gene, which encodes the enzyme that catalyzes the first step in the catabolism of BCAAs, was knocked out in CHO cells. Knockout (KO) of BCAT1 function completely eliminated production of inhibitory byproducts from BCAA catabolic pathway, including isovalerate, isobutyrate and 2-methylbutyrate, resulting in significantly enhanced cell growth and productivities in fed-batch cultures. This study is first of its kind to demonstrate that metabolic engineering of essential amino acid metabolism of CHO cells can significantly improve cell culture process performance.


Asunto(s)
Técnicas de Cultivo Celular por Lotes , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Ingeniería Metabólica , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus
5.
Cytokine ; 118: 80-92, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30017387

RESUMEN

Many tumor cells escape from cancer immunosurveillance and resist treatment with interferons (IFNs). Although the mechanism underlying IFN resistance is mostly attributed to a deficiency of components of the IFN-signaling pathway, some types of tumor cells resist IFN-mediated cell growth arrest despite the presence of an intact JAK/STAT signaling pathway. However, the molecular mechanisms underlying the unresponsiveness to IFNs independent of the defective JAK/STAT pathway remain to be clarified. To elucidate the mechanisms underlying IFNγ resistance, we examined the anti-proliferative effect of IFNγ on mouse tumor cell lines. Mouse squamous cell carcinoma (SCCVII) cells were resistant to IFNγ-mediated cell growth arrest despite the presence of the IFNγ-induced STAT1-dependent signaling pathway, whereas IFNγ inhibited cell growth of B16/F1 cells, a well-known IFNγ-sensitive mouse melanoma cell line, at the G1 phase of the cell cycle. Treatment of SCCVII cells with IFNγ neither downregulated the expression of cyclin D1, cyclin A2, and cyclin E1 nor induced a hypo-phosphorylated, active form of retinoblastoma protein (pRb). Interestingly, the hyper-phosphorylated, inactive form of pRb was exclusively localized in the cytoplasm in SCCVII cells. The IFN-inducible 204 gene (Ifi204), whose gene product, p204, binds to pRb and exerts an anti-proliferative effect, was repressed in SCCVII cells. p204 overexpression in SCCVII significantly inhibited cell growth, and mutation of a pRb-binding LXCXE motif decreased the anti-proliferative effect. These results suggest that silencing of Ifi204/p204 induces resistance to IFNγ-mediated cell growth arrest in SCCVII cells.


Asunto(s)
Proliferación Celular/genética , Inhibidores de Crecimiento/genética , Interferón gamma/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo/genética , Fase G1/genética , Melanoma/genética , Melanoma/patología , Ratones , Fosforilación/genética , Unión Proteica/genética , Proteína de Retinoblastoma/genética , Transducción de Señal/genética
7.
Cell Mol Neurobiol ; 35(8): 1165-73, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26012781

RESUMEN

In recent years, microRNA has become a hotspot in research on diseases, especially in the initiation and progression of different types of cancer. In this study, we found that miR-218 could inhibit growth and metabolism in gliomas by directly targeting E2F2. First, we obtained data from the Chinese Glioma Genome Atlas (CGGA) database to analyze miR-218 expression in different grades of gliomas. The effects of miR-218 on cell cycle progression and cell proliferation in U87 and U251 cell lines were investigated by flow cytometry, specifically CCK8 assay and tablet cloning, respectively. Glucose consumption and lactate production of glioma cell lines were measured by correlative test kits. Furthermore, we used Western blot analysis and luciferase reporter assay to identify the direct and functional target of miR-218. Data from the CGGA database and real-time quantitative reverse transcription-PCR demonstrated that miR-218 was obviously reduced in human glioblastoma tissues, as well as in the cell lines. When miR-218 level was elevated in vitro, cell cycle progression was arrested in the G1 phase, and cell proliferation was dramatically inhibited. Both glucose consumption and lactate production of glioma cells were significantly reduced. Western blot analysis and luciferase reporter assay revealed that E2F2 was a direct target of miR-218 in glioma cells. This investigation demonstrated that elevated E2F2 expression could partly weaken the effect of miR-218 in vitro. This study also showed that miR-218 may be a repressor in glioma by directly targeting E2F2, as well as a potential therapeutic target in gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Factor de Transcripción E2F2/biosíntesis , Glioblastoma/metabolismo , Inhibidores de Crecimiento/biosíntesis , MicroARNs/biosíntesis , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/prevención & control , Línea Celular Tumoral , Factor de Transcripción E2F2/genética , Marcación de Gen/métodos , Glioblastoma/genética , Glioblastoma/prevención & control , Inhibidores de Crecimiento/genética , Humanos , MicroARNs/genética
8.
PLoS One ; 10(4): e0123407, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25853862

RESUMEN

The dormancy of tumor cells is a major problem in chemotherapy, since it limits the therapeutic efficacy of anti-tumor drugs that only target dividing cells. One potential way to overcome chemo-resistance is to "wake up" these dormant cells. Here we show that the opioid antagonist methylnaltrexone (MNTX) enhances the effect of docetaxel (Doc) by blocking a cell growth-suppressive pathway. We found that PENK, which encodes opioid growth factor (OGF) and suppresses cell growth, is predominantly expressed in diffuse-type gastric cancers (GCs). The blockade of OGF signaling by MNTX releases cells from their arrest and boosts the effect of Doc. In comparison with the use of Doc alone, the combined use of Doc and MNTX significantly prolongs survival, alleviates abdominal pain, and diminishes Doc-resistant spheroids on the peritoneal membrane in model mice. These results suggest that blockade of the pathways that suppress cell growth may enhance the effects of anti-tumor drugs.


Asunto(s)
Naltrexona/análogos & derivados , Receptores Opioides/biosíntesis , Neoplasias Gástricas/tratamiento farmacológico , Taxoides/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Docetaxel , Sinergismo Farmacológico , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Humanos , Ratones , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Compuestos de Amonio Cuaternario/administración & dosificación , Receptores Opioides/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
9.
Int Immunopharmacol ; 25(1): 169-73, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25639227

RESUMEN

Mothers against decapentaplegic homologue 4 (Smad4) is associated with several human cancers. However, the exact mechanism of Smad4 in human glioma is still unknown. In this study, we constructed a recombinant adenovirus encoding Smad4 and transduced it into glioma cells. The results demonstrated that the overexpression of Smad4 not only suppressed glioma cell proliferation but also increased cell apoptosis by promoting cell cycle arrest at G1 phase. Furthermore, an adenovirus encoding Smad4 suppressed tumor formation in nude mice. These findings clearly demonstrate that Smad4 plays an important role in human glioma development by regulating cell proliferation. Moreover, Smad4 may represent a potential therapeutic target in glioma.


Asunto(s)
Proliferación Celular , Glioma/metabolismo , Proteína Smad4/metabolismo , Adenoviridae/genética , Animales , Apoptosis/genética , Carcinogénesis/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Vectores Genéticos/genética , Glioma/genética , Inhibidores de Crecimiento/genética , Humanos , Ratones , Ratones Desnudos , Proteína Smad4/genética , Transgenes/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Prostate ; 74(12): 1189-98, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24975685

RESUMEN

BACKGROUND: Regucalcin (RGN) is a calcium (Ca(2+) )-binding protein underexpressed in prostate adenocarcinoma comparatively to non-neoplastic prostate or benign prostate hyperplasia cases. Moreover, RGN expression is negatively associated with the cellular differentiation of prostate adenocarcinoma, suggesting that loss of RGN may be associated with tumor onset and progression. However, the RGN actions over the control of prostate cell growth have not been investigated. METHODS: Androgens are implicated in the promotion of prostate cell proliferation, thus we studied the in vivo effect of androgens on RGN expression in rat prostate. The role of RGN modulating cell proliferation and apoptotic pathways in rat prostate was investigated using transgenic animals (Tg-RGN) overexpressing the protein. RESULTS: In vivo stimulation with 5α-dihydrotestosterone (DHT) down-regulated RGN expression in rat prostate. Cell proliferation index and prostate weight were reduced in Tg-RGN, which was concomitant with altered expression of cell-cycle regulators. Tg-RGN presented diminished expression of the oncogene H-ras and increased expression of cell-cycle inhibitor p21. Levels of anti-apoptotic Bcl-2, as well as the Bcl-2/Bax protein ratio were increased in prostates overexpressing RGN. Both caspase-3 expression and enzyme activity were decreased in the prostates of Tg-RGN. CONCLUSIONS: Overexpression of RGN resulted in inhibition of cell proliferation and apoptotic pathways, which demonstrated its role maintaining prostate growth balance. Thus, deregulation of RGN expression may be an important event favoring the development of prostate cancer. Moreover, the DHT effect down-regulating RGN expression in rat prostate highlighted for the importance of this protein in prostatic physiology.


Asunto(s)
Andrógenos/genética , Apoptosis/genética , Proteínas de Unión al Calcio/genética , Ciclo Celular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Próstata/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Hidrolasas de Éster Carboxílico , Regulación hacia Abajo/genética , Inhibidores de Crecimiento/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Próstata/citología , Próstata/patología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Ratas Wistar , Transducción de Señal/genética
11.
Braz. j. med. biol. res ; 47(4): 273-278, 8/4/2014. graf
Artículo en Inglés | LILACS | ID: lil-705769

RESUMEN

Overexpression of cytokine-induced apoptosis inhibitor 1 (CIAPIN1) contributes to multidrug resistance (MDR) in breast cancer. This study aimed to evaluate the potential of CIAPIN1 gene silencing by RNA interference (RNAi) as a treatment for drug-resistant breast cancer and to investigate the effect of CIAPIN1 on the drug resistance of breast cancer in vivo. We used lentivirus-vector-based RNAi to knock down CIAPIN1 in nude mice bearing MDR breast cancer tumors and found that lentivirus-vector-mediated silencing of CIAPIN1 could efficiently and significantly inhibit tumor growth when combined with chemotherapy in vivo. Furthermore, Western blot analysis showed that both CIAPIN1 and P-glycoprotein expression were efficiently downregulated, and P53 was upregulated, after RNAi. Therefore, we concluded that lentivirus-vector-mediated RNAi targeting of CIAPIN1 is a potential approach to reverse MDR of breast cancer. In addition, CIAPIN1 may participate in MDR of breast cancer by regulating P-glycoprotein and P53 expression.


Asunto(s)
Animales , Femenino , Humanos , Antibióticos Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/genética , Silenciador del Gen , Péptidos y Proteínas de Señalización Intracelular/genética , Western Blotting , Neoplasias de la Mama/genética , Carcinoma/tratamiento farmacológico , Carcinoma/genética , Modelos Animales de Enfermedad , Genes MDR , Vectores Genéticos/genética , Inhibidores de Crecimiento/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lentivirus/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/genética , /efectos de los fármacos
12.
Braz J Med Biol Res ; 47(4): 273-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24676475

RESUMEN

Overexpression of cytokine-induced apoptosis inhibitor 1 (CIAPIN1) contributes to multidrug resistance (MDR) in breast cancer. This study aimed to evaluate the potential of CIAPIN1 gene silencing by RNA interference (RNAi) as a treatment for drug-resistant breast cancer and to investigate the effect of CIAPIN1 on the drug resistance of breast cancer in vivo. We used lentivirus-vector-based RNAi to knock down CIAPIN1 in nude mice bearing MDR breast cancer tumors and found that lentivirus-vector-mediated silencing of CIAPIN1 could efficiently and significantly inhibit tumor growth when combined with chemotherapy in vivo. Furthermore, Western blot analysis showed that both CIAPIN1 and P-glycoprotein expression were efficiently downregulated, and P53 was upregulated, after RNAi. Therefore, we concluded that lentivirus-vector-mediated RNAi targeting of CIAPIN1 is a potential approach to reverse MDR of breast cancer. In addition, CIAPIN1 may participate in MDR of breast cancer by regulating P-glycoprotein and P53 expression.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/genética , Silenciador del Gen , Péptidos y Proteínas de Señalización Intracelular/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Animales , Western Blotting , Neoplasias de la Mama/genética , Carcinoma/tratamiento farmacológico , Carcinoma/genética , Modelos Animales de Enfermedad , Femenino , Genes MDR , Vectores Genéticos/genética , Inhibidores de Crecimiento/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lentivirus/genética , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteína p53 Supresora de Tumor/efectos de los fármacos
13.
PLoS One ; 8(10): e76895, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204697

RESUMEN

G-protein-coupled receptors (GPCR) are the largest family of cell surface molecules that play important role/s in a number of biological and pathological processes including cancers. Earlier studies have highlighted the importance of Wnt7a signaling via its cognate receptor Frizzled9, a GPCR, in inhibition of cell proliferation, anchorage-independent growth, and reversal of transformed phenotype in non small cell lung cancer primarily through activation of the tumor suppressor, PPARγ. However, the G-protein effectors that couple to this important tumor suppressor pathway have not been identified, and are of potential therapeutic interest. In this study, by using two independent Wnt7a/Frizzled9-specific read-outs, we identify Gα16 as a novel downstream effector of Wnt7a/Frizzled9 signaling. Interestingly, Gα16 expression is severely down-regulated, both at the messenger RNA levels and protein levels, in many non small cell lung cancer cell lines. Additionally, through gene-specific knock-downs and expression of GTPase-deficient forms (Q212L) of Gα16, we also establish Gα16 as a novel regulator of non small cell lung cancer cell proliferation and anchorage-independent cell growth. Taken together, our data not only establish the importance of Gα16 as a critical downstream effector of the non-canonical Wnt signaling pathway but also as a potential therapeutic target for the treatment of non small cell lung cancer.


Asunto(s)
Proliferación Celular , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Inhibidores de Crecimiento/metabolismo , Vía de Señalización Wnt , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular , Línea Celular Tumoral , Activación Enzimática , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Inhibidores de Crecimiento/genética , Humanos , Immunoblotting , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Mutación , PPAR gamma/metabolismo , Interferencia de ARN , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
14.
J Microbiol Biotechnol ; 23(11): 1560-8, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23966021

RESUMEN

Salmonella, a main cause of foodborne diseases, encounters a variety of environmental stresses and overcomes the stresses by multiple resistance strategies. One of the general responses to hyperosmotic stress is to import or produce compatible solutes so that cells maintain fluid balance and protect proteins and lipids from denaturation. The ProP and ProU systems are the main transport systems for compatible solutes. The OsmU system, recently identified as a third osmoprotectant transport system, debilitates excessive growth as well by reducing production of trehalose. We studied a fourth putative osmoprotectant transport system, YehZYXW, with high sequence similarity with the OsmU system. A Salmonella strain lacking YehZ, a predicted substrate-binding protein, did not suffer from hyperosmolarity but rather grew more rapidly than the wild type regardless of glycine betaine, an osmoprotectant, suggesting that the YehZYXW system controls bacterial growth irrespective of transporting glycine betaine. However, the growth advantage of ΔyehZ was not attributable to an increase in OtsBA-mediated trehalose production, which is responsible for the outcompetition of the ΔosmU strain. Overexpressed YehZ in trans was capable of deaccelerating bacterial growth vice versa, supporting a role of YehZ in dampening growth. The expression of yehZ was increased in response to nutrient starvation, acidic pH, and the presence of glycine betaine under hyperosmotic stress. Identifying substrates for YehZ will help decipher the role of the YehZYXW system in regulating bacterial growth in response to environmental cues.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Inhibidores de Crecimiento/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Presión Osmótica , Salmonella typhimurium/enzimología , Salmonella typhimurium/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Portadoras/genética , Eliminación de Gen , Expresión Génica , Inhibidores de Crecimiento/genética , Proteínas de Transporte de Membrana/genética , Salmonella typhimurium/efectos de los fármacos
15.
Brain ; 136(Pt 5): 1488-507, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23587805

RESUMEN

Tripeptidyl peptidase 1 (TPP1) deficiency causes CLN2 disease, late infantile (or classic late infantile neuronal ceroid lipofuscinosis), a paediatric neurodegenerative disease of autosomal recessive inheritance. Patients suffer from blindness, ataxia, epilepsy and cognitive defects, with MRI indicating widespread brain atrophy, and profound neuron loss is evident within the retina and brain. Currently there are no effective therapies for this disease, which causes premature death in adolescence. Zebrafish have been successfully used to model a range of neurological and behavioural abnormalities. The aim of this study was to characterize the pathological and functional consequences of Tpp1 deficiency in zebrafish and to correlate these with human CLN2 disease, thereby providing a platform for drug discovery. Our data show that homozygous tpp1(sa0011) mutant (tpp1(sa0011)(-/-)) zebrafish display a severe, progressive, early onset neurodegenerative phenotype, characterized by a significantly small retina, a small head and curved body. The mutant zebrafish have significantly reduced median survival with death occurring 5 days post-fertilization. As in human patients with CLN2 disease, mutant zebrafish display storage of subunit c of mitochondrial ATP-synthase, hypertrophic lysosomes as well as localized apoptotic cell death in the retina, optic tectum and cerebellum. Further neuropathological phenotypes of these mutants provide novel insights into mechanisms of pathogenesis in CLN2 disease. Secondary neurogenesis in the retina, optic tectum and cerebellum is impaired and axon tracts within the spinal cord, optic nerve and the posterior commissure are disorganized, with the optic nerve failing to reach its target. This severe neurodegenerative phenotype eventually results in functional motor impairment, but this is preceded by a phase of hyperactivity that is consistent with seizures. Importantly, both of these locomotion phenotypes can be assayed in an automated manner suitable for high-throughput studies. Our study provides proof-of-principle that tpp1(sa0011)(-/-) mutants can utilize the advantages of zebrafish for understanding pathogenesis and drug discovery in CLN2 disease and other epilepsies.


Asunto(s)
Aminopeptidasas/deficiencia , Proliferación Celular , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/deficiencia , Progresión de la Enfermedad , Lipofuscinosis Ceroideas Neuronales/enzimología , Lipofuscinosis Ceroideas Neuronales/patología , Serina Proteasas/deficiencia , Aminopeptidasas/genética , Aminopeptidasas/fisiología , Animales , Animales Modificados Genéticamente , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/fisiología , Modelos Animales de Enfermedad , Inhibidores de Crecimiento/deficiencia , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/fisiología , Humanos , Actividad Motora/fisiología , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Lipofuscinosis Ceroideas Neuronales/genética , Serina Proteasas/genética , Serina Proteasas/fisiología , Tripeptidil Peptidasa 1 , Pez Cebra
16.
Parasitol Res ; 112(6): 2207-13, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23532543

RESUMEN

Haemaphysalis longicornis is known as one of the most important ticks transmitting Babesia parasites in East Asian countries, including Babesia ovata and Babesia gibsoni, as well as Theileria parasites. H. longicornis is not the natural vector of Babesia bovis and Babesia bigemina. Vector ticks and transmitted parasites are thought to have established unique host-parasite interaction for their survival, meaning that vector ticks may have defensive molecules for the growth control of parasites in their bodies. However, the precise adaptation mechanism of tick-Babesia parasites is still unknown. Recently, cyclophilin A (CyPA) was reported to be important for the development of Babesia parasites in ticks. To reveal a part of their adaptation mechanism, the current study was conducted. An injection of B. bovis-infected RBCs into adult female H. longicornis ticks was found to upregulate the expression profiles of the gene and protein of CyPA in H. longicornis (HlCyPA). In addition, recombinant HlCyPA (rHlCyPA) purified from Escherichia coli exhibited significant inhibitory growth effects on B. bovis and B. bigemina cultivated in vitro, without any hemolytic effect on bovine RBCs at all concentrations used. In conclusion, our results suggest that HlCyPA might play an important role in the growth regulation of Babesia parasites in H. longicornis ticks, during natural acquisition from an infected host. Furthermore, rHlCyPA may be a potential alternative chemotherapeutic agent against babesiosis.


Asunto(s)
Proteínas de Artrópodos/metabolismo , Babesia/efectos de los fármacos , Babesia/crecimiento & desarrollo , Ciclofilina A/metabolismo , Inhibidores de Crecimiento/metabolismo , Ixodidae/parasitología , Animales , Proteínas de Artrópodos/genética , Ciclofilina A/genética , Escherichia coli/genética , Perfilación de la Expresión Génica , Inhibidores de Crecimiento/genética , Ixodidae/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
17.
Tumour Biol ; 34(3): 1801-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23494180

RESUMEN

Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. However, the molecular development of UM is not fully understood and current therapeutic modalities result in poor outcomes. Increasingly, data have shown that human papillomaviruses (HPVs) contribute to the development of cervical cancer and other malignancies, and the key viral oncoprotein E6/E7 has become the target of gene therapy in HPV-related cancers. In this study, we identified HPV 18 infection in the UM cell line, VUP, for the first time and silenced HPV 18 E6/E7 expression using siRNA. Our results demonstrated that down regulation of HPV 18E6/E7 led to growth inhibition and cell cycle block in VUP cells by activation of the p53 and Rb pathways. We propose that HPV is possibly involved in the development of UM, and provide a novel target for the development of therapeutic strategies for UM.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Inhibidores de Crecimiento/genética , Papillomavirus Humano 18/fisiología , Melanoma/genética , Proteínas Oncogénicas Virales/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Neoplasias de la Úvea/genética , Apoptosis , Western Blotting , Ciclo Celular , Proteínas de Unión al ADN/genética , Regulación Viral de la Expresión Génica , Humanos , Melanoma/patología , Melanoma/virología , Proteínas Oncogénicas Virales/genética , Proteína de Retinoblastoma/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias de la Úvea/patología , Neoplasias de la Úvea/virología
18.
J Immunol ; 190(6): 2485-9, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23420891

RESUMEN

During early stages of B-lineage differentiation in bone marrow, signals emanating from IL-7R and pre-BCR are thought to synergistically induce proliferative expansion of progenitor cells. Paradoxically, loss of pre-BCR-signaling components is associated with leukemia in both mice and humans. Exactly how progenitor B cells perform the task of balancing proliferative burst dependent on IL-7 with the termination of IL-7 signals and the initiation of L chain gene rearrangement remains to be elucidated. In this article, we provide genetic and functional evidence that the cessation of the IL-7 response of pre-B cells is controlled via a cell-autonomous mechanism that operates at a discrete developmental transition inside Fraction C' (large pre-BII) marked by transient expression of c-Myc. Our data indicate that pre-BCR cooperates with IL-7R in expanding the pre-B cell pool, but it is also critical to control the differentiation program shutting off the c-Myc gene in large pre-B cells.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Regulación hacia Abajo/inmunología , Interleucina-7/fisiología , Proteínas Proto-Oncogénicas c-myc/genética , Células Madre/inmunología , Células Madre/metabolismo , Animales , Subgrupos de Linfocitos B/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Regulación hacia Abajo/genética , Técnicas de Sustitución del Gen , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Interleucina-7/antagonistas & inhibidores , Interleucina-7/metabolismo , Ratones , Ratones Noqueados , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Receptores de Interleucina-7/antagonistas & inhibidores , Receptores de Interleucina-7/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Madre/patología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Células del Estroma/patología
19.
J Immunol ; 190(4): 1447-56, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23296704

RESUMEN

Nucleic acid-reactive B cells frequently arise in the bone marrow but are tolerized by mechanisms including receptor editing, functional anergy, and/or deletion. TLR9, a sensor of endosomal dsDNA, both promotes and regulates systemic autoimmunity in vivo, but the precise nature of its apparently contradictory roles in autoimmunity remained unclear. In this study, using the 3H9 anti-DNA BCR transgene in the autoimmune-prone MRL.Fas(lpr) mouse model of systemic lupus erythematosus, we identify the stages at which TLR9 contributes to establishing and breaking B cell tolerance. Although TLR9 is dispensable for L chain editing during B cell development in the bone marrow, TLR9 limits anti-DNA B cell life span in the periphery and is thus tolerogenic. In the absence of TLR9, anti-DNA B cells have much longer life spans and accumulate in the follicle, neither activated nor deleted. These cells retain some characteristics of anergic cells, in that they have elevated basal BCR signaling but impaired induced responses and downregulate their cell-surface BCR expression. In contrast, whereas TLR9-intact anergic B cells accumulate near the T/B border, TLR9-deficient anti-DNA B cells are somewhat more dispersed throughout the follicle. Nonetheless, in older autoimmune-prone animals, TLR9 expression specifically within the B cell compartment is required for spontaneous peripheral activation of anti-DNA B cells and their differentiation into Ab-forming cells via an extrafollicular pathway. Thus, TLR9 has paradoxical roles in regulating anti-DNA B cells: it helps purge the peripheral repertoire of autoreactive cells, yet is also required for their activation.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Anergia Clonal/inmunología , ADN/inmunología , Inhibidores de Crecimiento/fisiología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Receptor Toll-Like 9/fisiología , Animales , Subgrupos de Linfocitos B/citología , Anergia Clonal/genética , Inhibidores de Crecimiento/genética , Humanos , Ratones , Ratones Endogámicos MRL lpr , Ratones Noqueados , Ratones Transgénicos , Quimera por Radiación/inmunología , Receptor Toll-Like 9/genética
20.
J Immunol ; 190(1): 159-67, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23225884

RESUMEN

MEK1 phosphorylates ERK1/2 and regulates T cell generation, differentiation, and function. MEK1 has recently been shown to translocate to the nucleus. Its nuclear function is largely unknown. By studying human CD4 T cells, we demonstrate that a low level of MEK1 is present in the nucleus of CD4 T cells under basal conditions. T cell activation further increases the nuclear translocation of MEK1. MEK1 interacts with the nuclear receptor corepressor silencing mediator of retinoid and thyroid hormone receptor (SMRT). MEK1 reduces the nuclear level of SMRT in an activation-dependent manner. MEK1 is recruited to the promoter of c-Fos upon TCR stimulation. Conversely, SMRT is bound to the c-Fos promoter under basal conditions and is removed upon TCR stimulation. We examined the role of SMRT in regulation of T cell function. Small interfering RNA-mediated knockdown of SMRT results in a biphasic effect on cytokine production. The production of the cytokines IL-2, IL-4, IL-10, and IFN-γ increases in the early phase (8 h) and then decreases in the late phase (48 h). The late-phase decrease is associated with inhibition of T cell proliferation. The late-phase inhibition of T cell activation is, in part, mediated by IL-10 that is produced in the early phase and, in part, by ß-catenin signaling. Thus, we have identified a novel nuclear function of MEK1. MEK1 triggers a complex pattern of early T cell activation, followed by a late inhibition through its interaction with SMRT. This biphasic dual effect most likely reflects a homeostatic regulation of T cell function by MEK1.


Asunto(s)
Transporte Activo de Núcleo Celular/inmunología , Linfocitos T CD4-Positivos/inmunología , MAP Quinasa Quinasa 1/fisiología , Co-Represor 1 de Receptor Nuclear/fisiología , Co-Represor 2 de Receptor Nuclear/antagonistas & inhibidores , Co-Represor 2 de Receptor Nuclear/fisiología , Transporte Activo de Núcleo Celular/genética , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/metabolismo , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Silenciador del Gen/inmunología , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/metabolismo , Inhibidores de Crecimiento/fisiología , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Co-Represor 2 de Receptor Nuclear/genética , Regiones Promotoras Genéticas/inmunología , Unión Proteica/genética , Unión Proteica/inmunología , Proteínas Proto-Oncogénicas c-fos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...