Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Manag Res ; 13: 6377-6387, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34408497

RESUMEN

OBJECTIVE: Prostate cancer (PCa) ranks as the second common malignancy in males worldwide. Although conspicuous progressions in diagnosis and treatment have been achieved in the past decades, the prognosis expectation of PCa remains unsatisfied yet. To improve the prognosis prediction of PCa, more specific biomarkers are needed. In this retrospective research, we focused on ßKlotho and ETS-like transcription factor 4 (ELK4), aiming to identify potential prognostic biomarkers for PCa. METHODS: Western blotting was used to determine the expression of ßKlotho, ELK4, and PARP in C4-2B and PC3 PCa cell lines. CCK-8 assay and colony formation assay were applied to examine the roles of ßKlotho and ELK4 in the proliferation of PCa cells. The expression of ßKlotho and ELK4 in PCa tissue samples was determined by immunochemistry. Pearson's χ2 test and Fisher's exact test were performed to investigate the associations among ßKlotho, ELK4 and various clinical factors. Kaplan-Meier curves and Cox regression model were established to reveal the correlation among ßKlotho, ELK4 expression and the prognosis of patients. RESULTS: ßKlotho overexpression down-regulated the ELK4 expression, induced apoptosis and inhibited cell proliferation in both C4-2B and PC3 cells, which were reversed by ELK4 overexpression. ßKlotho expression in PCa tissue samples had negative correlation with the ELK4 expression, and higher ßKlotho expression was associated with lower Gleason score, absent distant metastasis and lower prostate-specific antigen (PSA) level. On the contrast, higher ELK4 expression was correlated with distant metastasis and higher PSA level. Moreover, ßKlotho and ELK4 were both recognized as independent factors for the prognosis of patients with PCa. CONCLUSION: ßKlotho inhibits proliferation of prostate cancer cells by downregulating ELK4. Both ßKlotho and ELK4 expressions correlate with the prognosis of PCa, which may serve as potential biomarkers for follow-up surveillance and prognostic assessments.

2.
Exp Cell Res ; 384(1): 111558, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31472118

RESUMEN

Hydroxyacyl-CoA dehydrogenase alpha subunit (HADHA) is a key lipid metabolic enzyme with a novel role in carcinogenesis. We previously reported that HADHA, a prognostic marker, was downregulated in clear cell renal cell carcinoma (ccRCC). Herein, the tumor inhibitory role of HADHA overexpression in ccRCC was investigated further. The quantitative proteomic analysis displayed that a total of 1293 and 1293 proteins were identified in HADHA overexpressed 786-O-hadha and vector-transfected control 786-O-vc cells, respectively, and 206 proteins were found to be up- or downregulated. PANTHER, OmicsNet, STRING, and DAVID tools were utilized on the dysregulated proteins in order to elucidate multiple metabolic pathways (especial lipid metabolism) and lipid metabolism-related proteins (e.g. ACAT1, ACLY). The dysregulation of the lipid metabolic enzymes, ACAT1, ACLY, CYB5R3 and FASN, were confirmed by Western blotting. Further assays demonstrated that HADHA overexpression significantly inhibited cell growth, induced cell apoptosis, and decreased the formation of cytoplasmic lipid droplets (LDs); moreover, it also inhibited tumor growth and lessened the formation of LDs in xenografted mouse. Collectively, these data revealed that HADHA overexpression disrupted lipid metabolism and inhibited tumor growth, which shed light on HADHA as a potential therapeutic target for clinical intervention of ccRCC.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Renales/genética , Metabolismo de los Lípidos/genética , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Animales , Apoptosis/genética , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/genética , Regulación hacia Abajo/genética , Femenino , Humanos , Neoplasias Renales/patología , Gotas Lipídicas/patología , Ratones , Ratones Endogámicos BALB C , Proteómica/métodos
3.
Biomed Pharmacother ; 91: 202-207, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28458158

RESUMEN

Previous studies showed that HCRP1 is decreased in tumor cells compared with normal tissue, and functions as a tumor suppressor. However, its expression pattern and function in human prostate cancer remain unclear. In this study we examined HCRP1 expression in prostate cancer cell lines via western blotting. Thereafter, we performed CCK-8 assay and matrigel invasion assay after cells were transfected with HCRP1 overexpression plasmid or siRNA. We further investigated the possible mechanism involved in HCRP1's regulation to prostate cancer cell proliferation and invasion. We found that HCRP1 negatively regulates EGFR activity and expression of its downstream proteins. Moreover, we found that HCRP1 is negatively correlated with multi-drug resistant related proteins after cells were treated with paclitaxel, cisplatin or gefitinib, indicating its inhibiting effect of chemotherapy resistance. In summary, our results provided evidence that HCRP1 is a negative regulator in prostate cancer progression, metastasis and multi-drug resistance.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Receptores ErbB/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Línea Celular Tumoral , Proliferación Celular , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Humanos , Masculino , Modelos Biológicos , Invasividad Neoplásica
4.
Urol Int ; 98(1): 120-124, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-26159694

RESUMEN

Renal angiomyolipoma (AML) is a benign tumor. However, rare cases of renal AML demonstrate aggressive behaviors such as tumor thrombus extension into the inferior vena cava (IVC). We successfully treated a case of epithelioid AML in the right kidney involving the IVC. We also reviewed and analyzed 45 case reports of the common type of AML. Radiologists and clinicians should know that epithelioid AML can be an aggressive tumor.


Asunto(s)
Angiomiolipoma/patología , Neoplasias Renales/patología , Células Neoplásicas Circulantes , Vena Cava Inferior , Adulto , Humanos , Masculino , Invasividad Neoplásica
5.
Oncol Lett ; 12(5): 3403-3410, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27900012

RESUMEN

Renal cell carcinoma (RCC) is resistant to standard radiotherapy. Ubenimex, an aminopeptidase N inhibitor, is widely used as an adjunct therapy after surgery to enhance the function of immunocompetent cells and confer antitumor effects. Our previous study demonstrated that ubenimex induces autophagic cell death in RCC cells. Recently, the molecular mechanism of autophagy induction has been associated with radiosensitivity in RCC cells. In the present study, the ability of ubenimex to enhance RCC cell sensitivity to radiation via the induction of autophagic cell death was determined, and the mechanism of action of this effect was investigated. The 786-O and OS-RC-2 human RCC cell lines were treated with 0.5 mg/ml ubenimex and different doses of irradiation (IR). The cell viability was measured using a colony-formation assay and flow cytometry. Acridine orange (AO)-ethidium bromide (EB) staining was assessed by fluorescence microscopy as an indicator of autophagic cell death. Protein expression was assessed by western blotting. Autophagosomes were evaluated using transmission electron microscopy. RCC cells were used to evaluate the sensitivity to radiation using clonogenic survival and lactate dehydrogenase assays. Furthermore, these parameters were also tested at physiological oxygen levels. The AO-EB staining and flow cytometry of the OS-RC-2 cells indicated that the combined treatment significantly enhanced autophagic cell death compared with ubenimex or IR alone. Therefore, treatment with ubenimex did not significantly alter cell cycle progression but increased cell death when combined with radiation. An Akt agonist could significantly weaken this effect, indicating that ubenimex may act as an Akt inhibitor. Furthermore, the western blot analysis indicated that the combined treatment inhibited the Akt signaling pathway compared with ubenimex treatment or IR alone. Ubenimex may enhance RCC cell sensitivity to radiation by inducing cell autophagy. This induction changes the role of autophagy from protective to lethal in vitro, and this switch is associated with the inhibition of the Akt signaling pathway.

6.
Tumour Biol ; 37(11): 15133-15143, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27677286

RESUMEN

Renal cell carcinoma (RCC) accounts for 3 % of all adult malignancies and is the most lethal urological cancer. Livin is a member of the inhibitor of apoptosis protein (IAP) family, which is associated with tumor resistance to radiotherapy and chemotherapy. Clinical data also showed that patients with high tumor grades and stages have higher expression levels of Livin in RCC cells. Autophagy is a survival mechanism activated in response to nutrient deprivation. A possible role of Livin in the autophagy of RCC cells has not been investigated; therefore, this pioneer study was carried out. Livin was silenced in RCC cells (slow virus infection [SVI]-shLivin cells) by lentiviral transfection. Then, mRNA and protein expression levels in the transfected cells were assessed by quantitative fluorescence PCR and Western blotting, respectively. In addition, acridine orange staining and electron microscopy were used to assess autophagy in SVI-shLivin cells. The cisplatin IC50 values for RCC cells were measured by the CCK8 assay. Potent antitumor activities were observed in xenograft mouse models generated with Livin-silenced RCC cells in terms of delayed tumor onset and suppressed tumor growth. These results suggested that Livin silencing could increase the chemotherapeutic sensitivity of RCC cells to cisplatin and induce autophagic cell death. A possible mechanism of Bcl-2 and Akt pathway involvement was discussed specifically in this study. Overall, Livin silencing induces apoptotic and autophagic cell death and increases chemotherapeutic sensitivity of RCC cells to cisplatin.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinoma de Células Renales/patología , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antineoplásicos/farmacología , Biomarcadores de Tumor , Western Blotting , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Técnicas para Inmunoenzimas , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/genética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Clasificación del Tumor , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Tumour Biol ; 37(9): 12731-12742, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27448305

RESUMEN

Although GC (gemcitabine and cisplatin) chemotherapy remains an effective method for treating bladder cancer (BCa), chemoresistance is a major obstacle in chemotherapy. In this study, we determined whether gemcitabine resistance correlates with migratory/invasive potential in BCa and whether this relationship is regulated by the cylindromatosis (CYLD)-Livin module. First, we independently investigated the correlation of CYLD/Livin and gemcitabine resistance with the potential for tumor migration and invasiveness. Second, we found that co-transfected CYLD and Livin dramatically improved sensitivity to gemcitabine chemotherapy and decreased migration/invasion potential. Next, we determined that CYLD may regulate Livin by the NF-κB-dependent pathway. We also found that CYLD overexpression and Livin knockdown might improve gemcitabine chemosensitivity by decreasing autophagy and increasing apoptosis in BCa cells. Finally, the effects of CYLD-Livin on tumor growth in vivo were evaluated. Our study demonstrates that CYLD-Livin might represent a potential therapeutic for chemoresistant BCa.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Movimiento Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas de Neoplasias/genética , Proteínas Supresoras de Tumor/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Western Blotting , Línea Celular Tumoral , Movimiento Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Desoxicitidina/farmacología , Enzima Desubiquitinante CYLD , Regulación hacia Abajo , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/metabolismo , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
8.
Biomed Pharmacother ; 81: 25-30, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27261573

RESUMEN

Prostate cancer is the leading cause of cancer deaths among men in the worldwide, it's important to find new prognostic factors and therapeutic targets. microRNAs play critical roles in the development and progression of prostate cancer. Here we revealed miR-1301 promoted prostate cancer progression. miR-1301 was upregulated in prostate cancer tissues and cells, overexpression of miR-1301 promoted anchorage-dependent and -independent growth using MTT analysis, colony formation analysis and soft agar growth analysis, whereas knockdown of miR-1301 suppressed anchorage-dependent and -independent growth. We also found overexpression of miR-1301 inhibited p27 expression and promoted Cyclin D1 expression, whereas knockdown of miR-1301 reduced this effect, suggesting miR-1301 promoted the G1/S transition. These results suggested miR-1301 promoted cell proliferation of prostate cancer. microRNAs can inhibit target mRNA translation or/and induce mRNA degradation, we found tumor suppresser PPP2R2C was the target of miR-1301, simultaneous downregualtion of PPP2R2C and miR-1301 promoted anchorage-dependent and -independent growth. These findings suggested miR-1301 promoted prostate cancer proliferation by inhibiting PPP2R2C, and might a therapeutic target for prostate cancer.


Asunto(s)
MicroARNs/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteína Fosfatasa 2/metabolismo , Secuencia de Bases , Adhesión Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , MicroARNs/genética , Proteína Fosfatasa 2/genética , Regulación hacia Arriba/genética
9.
Oncotarget ; 7(11): 12053-62, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26919249

RESUMEN

BACKGROUND: Benign prostatic hyperplasia (BPH) is one of the most common diseases in middle-age or older men. Increasing evidence has shown that BPH is associated with hypoxia microenvironment. METHODS: We retrospectively collected patient data and tissue samples from fetal prostates(FP), normal prostates(NP), intra-acinar of BPH, peri-acinar of BPH, prostate cancers and sarcomas of prostate. The expression of HIF-1α, as well as VEGF was visualized by immunohistochemistry and statistically analyzed with clinical parameters. RESULTS: Expression of HIF-lα was observed in intra-acinar of BPH (69.5%), prostate cancer (85.7%) and all FPs, while NP and peri-acinar of BPH tissues were all stained negative. HIF-lα levels in FPs and the malignant tumors were higher than BPH tissues(p < 0.05), and the expression of HIF-lα in intra-acinar of BPH was higher than NP and peri-acinar of BPH (p < 0.05). The expression of HIF-lα was correlated with the weight of intra-acinar of prostate (p < 0.05). And patients with prostate weight larger that 72.45g were prone to have HIF-lα moderate-positive expression, according to the ROC curve (AUC = 0.734, 95%CI = 0.630-0.838). Moreover, the risk of acute urine retention (AUR) for HIF-lα moderate-positive patients increased significantly (OR=5.517, 95%CI = 2.434-12.504). CONCLUSIONS: HIF-lα expression is increased in highly proliferative prostate tissues and correlated with the weight of intra-acinar prostate. Moreover, HIF-lα is also an independent risk factor for AUR occurrence in BPH patients.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Próstata/crecimiento & desarrollo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Adulto , Humanos , Inmunohistoquímica , Masculino , Próstata/patología , Hiperplasia Prostática/patología , Estudios Retrospectivos , Adulto Joven
10.
Oncol Rep ; 35(4): 2121-30, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26846372

RESUMEN

Prostate cancer is the second most frequently diagnosed cancer in males worldwide and is commonly associated with metastasis. Moreover, in prostate cancer, aminopeptidase N (APN) expression is closely correlated with metastasis. Ubenimex, an APN inhibitor, is widely used as an adjunct therapy for cancer, enhancing the function of immunocompetent cells and conferring antitumor effects. However, due to the low expression of APN, it is rarely used to treat prostate cancer. Recently, the induction of autophagy as a molecular mechanism has been strongly connected with tumor cell death. Thus, we investigated whether ubenimex could inhibit cell proliferation, migration and invasion by downregulating APN expression to induce autophagic cell death in prostate cancer cells. The LNCaP and PC-3 cell lines were treated with different doses of ubenimex. Cell viability was measured using growth curve analysis and WST-8 proliferation assay. Autophagic cell death was assessed using fluorescence microscopy and acridine orange/ethidium bromide (AO/EB) staining. Protein expression was assessed by immunofluorescence and western blot analyses. Autophagosomes were evaluated using transmission electron microscopy. Wound-healing migration assays were performed to determine the migratory ability of the PC-3 cells. In addition, nude mice were used in the present study to examine PC-3 cell proliferation in vivo. The results revealed that APN expression differed between the metastatic and non-metastatic prostate cancer cells. In addition, ubenimex inhibited APN expression in the prostate cancer cells. Ubenimex increased prostate cancer cell death, as determined using the lactate dehydrogenase (LDH) cytotoxicity assay. This effect was accompanied by increased levels of LC3B. Furthermore, ubenimex inhibited PC-3 cell proliferation in vivo and in vitro. Ubenimex inhibited the cell migration and invasion in prostate cancer cells by downregulating APN expression. Finally, ubenimex induced autophagic cell death in both metastatic and non-metastatic prostate cancer cells. Based on these results, ubenimex appears to be an excellent adjunctive therapy for the treatment of prostate cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Autofagia , Antígenos CD13/antagonistas & inhibidores , Leucina/análogos & derivados , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucina/administración & dosificación , Leucina/farmacología , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neoplasias de la Próstata/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
PLoS One ; 10(12): e0144484, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26658059

RESUMEN

Development of resistance to gemcitabine is a major concern in bladder cancer therapy, and the mechanism remains unclear. Eg5 has been recently identified as an attractive target in cancer chemotherapy, so novel targeted chemotherapy with Eg5 inhibitor is expected to improve the anticancer effect in gemcitabine-resistant bladder cancer. In this research, RT112-Gr cells were 350-fold less sensitive to gemcitabine than the parental cell lines, while KU7-Gr cells were 15-fold less sensitive to gemcitabine than the parental cell lines. Human OneArray Microarray analysis was performed to obtain broad spectrum information about the genes differentially expressed in RT112 and RT112-Gr cells. The anti-proliferative activity of S(MeO)TLC, an Eg5 inhibitor, was analyzed in RT112-Gr cell lines using a cell viability assay. Furthermore, the inhibitory effect was evaluated in vivo using subcutaneous xenograft tumor model. According to the result of Human OneArray GeneChip, RRM1 and RRM2 were up-regulated, while there was no significant change in Eg5. Trypan blue staining confirmed that in S(MeO)TLC and Gemcitabine combining S(MeO)TLC group cell viability were significantly decreased in RT112-Gr cells as compared with other groups. S(MeO)TLC and S(MeO)TLC+gemcitabine groups prominently suppressed tumor growth in comparison with other groups' in vivo. There were no significant differences in S(MeO)TLC and gemcitabine+S(MeO)TLC group in the effect of inhibition of bladder cancer in vivo and in vitro. Our data collectively demonstrated that S(MeO)TLC represents a novel strategy for the treatment of gemcitabine resistant bladder cancer.


Asunto(s)
Cisteína/análogos & derivados , Cinesinas/antagonistas & inhibidores , Compuestos de Tritilo/farmacología , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Análisis por Conglomerados , Cisteína/farmacología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Cinesinas/genética , Cinesinas/metabolismo , Ratones Endogámicos BALB C , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleósido Difosfato Reductasa/efectos adversos , Ribonucleósido Difosfato Reductasa/genética , Ribonucleósido Difosfato Reductasa/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
12.
Med Sci Monit ; 21: 2421-7, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26286430

RESUMEN

BACKGROUND: Cnidium lactone is a natural coumarin compound that can inhibit a variety of cancer cell proliferation and induce cancer cell apoptosis. This experiment investigated the effect of cnidium lactone on molecular marker expression in prostate cancer nude mice to study its effect in inducing apoptosis. MATERIAL AND METHODS: We randomly and equally divided 30 male BALB/C nude mice inoculated with human prostate cancer cells PC-3 into a negative control group, a cyclophosphamide group (500 mg/Kg), and cnidium lactone groups at 3 doses (280 mg/Kg, 140 mg/Kg, and 70 mg/Kg). The mice were weighed at 2 weeks after administration. Tunnel assay was applied to test the nude mice tumor cell apoptosis. ELISA was performed to detect serum AMACR, CD147, mutant P53, BCL-2, AND BAX expression levels. Tumor tissue was separated and weighed. RESULTS: Mice weight did not change significantly in the groups receiving 3 different doses of cnidium lactone(p>0.05), while it decreased obviously in the cyclophosphamide group (p<0.05). Tumor weight, CD147, mutant P53, and BCL-2 levels were significantly lower in the groups receiving 3 different doses of cnidium lactone than in the negative control group (p<0.05). Among them, the abovementioned indexes decreased markedly in the 280 mg/Kg and 140 mg/Kg dose groups than in the cyclophosphamide group (p<0.05). AMACR and BAX levels showed no significant difference in the cnidium lactone group or the cyclophosphamide group (p>0.05). CONCLUSIONS: Cnidium lactone may induce prostate cancer cell apoptosis and inhibit its proliferation through regulating CD147, mutant P53, and BCL-2 expression in nude mice.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Lactonas/farmacología , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/sangre , Proteína p53 Supresora de Tumor/sangre , Proteína X Asociada a bcl-2/sangre , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Apoptosis/efectos de los fármacos , Basigina/sangre , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Cnidium/química , Cumarinas/administración & dosificación , Cumarinas/farmacología , Ciclofosfamida/farmacología , Humanos , Lactonas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Mutantes/sangre , Proteínas Mutantes/genética , Neoplasias de la Próstata/genética , Racemasas y Epimerasas/sangre , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Int J Clin Exp Pathol ; 8(2): 1302-11, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25973014

RESUMEN

Renal cell carcinoma (RCC) carries a high risk of malignancy and metastasis. The inducible isoform of prostaglandin synthase, cyclooxygenase (COX)-2, and ICAM-1 may be involved in tumor metastasis. CCN3, also called nephroblastoma overexpressed gene (NOV), has been found to regulate the proliferation and differentiation of cancer cells. The effects of NOV on RCC cell migration and expression of COX-2 and ICAM-1 have not described yet in detail. But here, NOV was found to promote the migration and expression of COX-2 and ICAM-1 in human RCC cells. Akt inhibitor was found to interfere with this NOV-induced migration and up-regulation of COX-2 and ICAM-1 in RCC cells. NOV stimulation was here found to promote the phosphorylation of Akt. RCC tissue chips were subjected to IHC staining, which showed COX-2 expression in RCC tissues to be a significantly closely correlated with NOV expression, with significance determined using Pearson correlation testing (P < 0.05). The results of the current work indicate that NOV activates COX-2 and ICAM-1 through Akt, promoting the migration of RCC cells.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Movimiento Celular/fisiología , Ciclooxigenasa 2/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Neoplasias Renales/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Ciclooxigenasa 2/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/genética , Neoplasias Renales/genética , Neoplasias Renales/patología , Proteína Hiperexpresada del Nefroblastoma/genética , Fosforilación , Transducción de Señal/fisiología , Regulación hacia Arriba
14.
Comput Biol Chem ; 56: 71-83, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25889321

RESUMEN

OBJECTIVES: To explore molecular mechanisms of bladder cancer (BC), network strategy was used to find biomarkers for early detection and diagnosis. METHODS: The differentially expressed genes (DEGs) between bladder carcinoma patients and normal subjects were screened using empirical Bayes method of the linear models for microarray data package. Co-expression networks were constructed by differentially co-expressed genes and links. Regulatory impact factors (RIF) metric was used to identify critical transcription factors (TFs). The protein-protein interaction (PPI) networks were constructed by the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and clusters were obtained through molecular complex detection (MCODE) algorithm. Centralities analyses for complex networks were performed based on degree, stress and betweenness. Enrichment analyses were performed based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. RESULTS: Co-expression networks and TFs (based on expression data of global DEGs and DEGs in different stages and grades) were identified. Hub genes of complex networks, such as UBE2C, ACTA2, FABP4, CKS2, FN1 and TOP2A, were also obtained according to analysis of degree. In gene enrichment analyses of global DEGs, cell adhesion, proteinaceous extracellular matrix and extracellular matrix structural constituent were top three GO terms. ECM-receptor interaction, focal adhesion, and cell cycle were significant pathways. CONCLUSIONS: Our results provide some potential underlying biomarkers of BC. However, further validation is required and deep studies are needed to elucidate the pathogenesis of BC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Neoplasias de la Vejiga Urinaria/genética , Teorema de Bayes , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Mapas de Interacción de Proteínas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/metabolismo
15.
Tumour Biol ; 35(8): 7659-68, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24801905

RESUMEN

Eg5 is critical for mitosis and overexpressed in various malignant tumors, which has now been identified as a promising target in cancer therapy. However, the anti-cancer activity of Eg5 inhibitor in renal cell carcinoma (RCC) remains an open issue. In this paper, we evaluated, for the first time, the therapeutic benefit of blocking Eg5 by S-(methoxytrityl)-L-cysteine (S(MeO)TLC) in RCC both in vitro and vivo. The expression of Eg5 was examined in clinical tissue samples and various kidney cell lines, including 293T, 786-0, and OS-RC-2. The anti-proliferative activity of Eg5 inhibitors, (S)-trityl-L-cysteine (STLC) and S(MeO)TLC, was evaluated by a cell viability assay. An apoptosis assay with Hoechst nuclear staining and flow cytometry was applied to investigate the efficacy of the S(MeO)TLC, which is more potent than STLC. Immunofluorescence was used to research the possible mechanism. Furthermore, in vivo studies were performed by using subcutaneous xenograft models, which were used to confirm its role as a potential anti-neoplastic drug. The Eg5 expression was detected in kidney cell lines and RCC tissues, which was low in normal kidney samples. STLC and S(MeO)TLC exhibited their optimal anti-proliferative activity in 72 h, and cells treated with S(MeO)TLC presented characteristic monoastral spindle phenotype in 24 h and apoptotic cells in 48 h. In vivo, S(MeO)TLC effectively suppressed tumor growth in subcutaneous xenograft models. Inhibition of Eg5 represses the proliferation of RCC in vitro and in vivo. All these findings collectively demonstrate that S(MeO)TLC, a potent Eg5 inhibitor, is a promising anti-cancer agent for the treatment of RCC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Cisteína/análogos & derivados , Neoplasias Renales/tratamiento farmacológico , Cinesinas/antagonistas & inhibidores , Terapia Molecular Dirigida , Compuestos de Tritilo/farmacología , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Cisteína/farmacología , Femenino , Humanos , Neoplasias Renales/patología , Cinesinas/análisis , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Med Oncol ; 30(1): 476, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23371254

RESUMEN

Eg5 is a member of the kinesin family of proteins, which associates with bipolar spindle formation in dividing tumor cells during mitosis. The aim of our study is to investigate the prognostic role of Eg5 expression in patients with renal cell carcinoma (RCC). RCC tissue specimens from 164 consecutively treated patients who underwent surgery between 2005 and 2011 were evaluated. The Eg5 expression was determined by immunohistochemistry, and correlated with clinicopathological parameters. The prognostic significance of Eg5 expression was explored using the univariate and multivariate survival analysis of 164 patients who were followed; one hundred and sixty-four tissue specimens "of patients" who were regularly followed with the mean 35.8 months (from 5 to 80 months). The expression of Eg5 was significantly associated with tumor nuclear grade (P = 0.019) and stage (P = 0.007), as well as tumor size (P = 0.033). In univariate analysis, Eg5 overexpression showed unfavorable influence on recurrence-free survival with statistical significance (P = 0.003). Clinical stage, nuclear grade and tumor size also showed strong statistical relation with adverse recurrence-free survival (P < 0.001). Multivariate analysis revealed that tumor stage, nuclear grade and Eg5 reactivity (P < 0.001, P = 0.002, P = 0.032) were identified as independent prognostic factors for recurrence-free survival in patients with RCC. In our opinion, the result of this study proved the relationship between Eg5 expression and worse clinical outcome in RCC. This finding suggested that Eg5 served as a prognostic factor, which could be useful to predict cancer evolution and provide appropriate treatments for RCC patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Cinesinas/biosíntesis , Adulto , Anciano , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Cinesinas/análisis , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , Adulto Joven
18.
Oncol Lett ; 3(5): 1099-1104, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22783399

RESUMEN

The CCN3/nephroblastoma overexpressed gene belongs to the CCN family of genes that encode secreted proteins involved in a variety of processes including tumorigenesis. Altered expression of CCN3 has been observed in human nephroblastoma and renal cell carcinoma (RCC), suggesting that CCN3 plays a role in kidney tumorigenesis. The aim of the present study was to examine the role of CCN3 in clear cell RCC biology. In particular, we studied the expression of CCN3 in 32 pairs of RCC tissues and corresponding normal kidney tissues using immunohistochemistry. The CCN3 gene was transfected into the 786-O cell line and the behaviors of stably transfected clones were analyzed. Results showed the expression of CCN3 was lower in RCC tissues compared to corresponding normal kidney tissues and the expression of CCN3 was inversely correlated with the Ki67 index. CCN3-expressing clones exhibited significantly inhibited cell proliferation. Furthermore, CCN3-transfected 786-O cells exhibited increased adhesion to extracellular matrix proteins, migration and invasion in Matrigel. Our data indicated that CCN3 plays an anti-proliferative role in clear cell RCC cells and promotes the adhesion, migration and invasion of clear cell RCC cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...