Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Methods Mol Biol ; 2318: 337-346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019301

RESUMEN

Oncoproteins encoded by dominant oncogenes have long been considered as targets for chemotherapeutic intervention. However, oncogenic transcription factors have often been dismissed as "undruggable." Members of the Myc family of transcription factors have been identified as promising targets for cancer chemotherapy in multiple publications reporting the requirement of Myc proteins for maintenance of almost every type of tumor. Here, we describe cell-based approaches to identify c-Myc small molecule inhibitors by screening complex libraries of diverse small molecules based on Myc functionality and specificity.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Genes myc/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Línea Celular Tumoral , Genes myc/genética , Genes myc/fisiología , Humanos , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
2.
Theranostics ; 10(8): 3622-3635, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32206112

RESUMEN

The transcriptional co-regulators YAP and TAZ pair primarily with the TEAD family of transcription factors to elicit a gene expression signature that plays a prominent role in cancer development, progression and metastasis. YAP and TAZ endow cells with various oncogenic traits such that they sustain proliferation, inhibit apoptosis, maintain stemness, respond to mechanical stimuli, engineer metabolism, promote angiogenesis, suppress immune response and develop resistance to therapies. Therefore, inhibiting YAP/TAZ- TEAD is an attractive and viable option for novel cancer therapy. It is exciting to know that many drugs already in the clinic restrict YAP/TAZ activities and several novel YAP/TAZ inhibitors are currently under development. We have classified YAP/TAZ-inhibiting drugs into three groups. Group I drugs act on the upstream regulators that are stimulators of YAP/TAZ activities. Many of the Group I drugs have the potential to be repurposed as YAP/TAZ indirect inhibitors to treat various solid cancers. Group II modalities act directly on YAP/TAZ or TEADs and disrupt their interaction; targeting TEADs has emerged as a novel option to inhibit YAP/TAZ, as TEADs are major mediators of their oncogenic programs. TEADs can also be leveraged on using small molecules to activate YAP/TAZ-dependent gene expression for use in regenerative medicine. Group III drugs focus on targeting one of the oncogenic downstream YAP/TAZ transcriptional target genes. With the right strategy and impetus, it is not far-fetched to expect a repurposed group I drug or a novel group II drug to combat YAP and TAZ in cancers in the near future.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Neoplasias/terapia , Transactivadores/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/farmacología , Antineoplásicos/farmacología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Proteínas de Unión al ADN/farmacología , Diseño de Fármacos , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Transactivadores/farmacología , Factores de Transcripción/metabolismo , Factores de Transcripción/farmacología , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP
3.
Nat Rev Clin Oncol ; 16(6): 341-355, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30718843

RESUMEN

The era of personalized medicine for advanced-stage non-small-cell lung cancer (NSCLC) began when biomarker-based evidence of molecular pathway and/or oncogene addiction of the tumour became mandatory for the allocation of specific targeted therapies. More recently, the immunotherapy revolution, specifically, the development of immune-checkpoint inhibitors (ICIs), has dramatically altered the NSCLC treatment landscape. Herein, we compare and contrast the clinical development of immunotherapy and oncogene-directed therapy for NSCLC, focusing on the role of predictive biomarkers. Immunotherapy biomarkers are fundamentally different from oncogene biomarkers in that they are continuous rather than categorical (binary), spatially and temporally variable and reliant on multiple complex interactions rather than a single, dominant determinant. The performance of predictive biomarkers for ICIs might be improved by combining different markers to reduce the assumptive risks associated with each one. Novel combinations with chemotherapy and ICIs complicate biomarker discovery but do not decrease the value of the markers identified. Perfectly predictive biomarkers of benefit from immunotherapy are unlikely to be identified, although exclusionary biomarkers of minimal benefit or an unacceptable risk of toxicity might be feasible. The clinical adoption and applicability of such biomarkers might vary depending on line of treatment, the available therapeutic alternatives and health economic considerations.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/inmunología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia , Neoplasias Pulmonares/inmunología , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/genética , Medicina de Precisión
4.
Int J Mol Sci ; 18(7)2017 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-28714919

RESUMEN

Immunotherapy is being tested intensively in clinical trials for prostate cancer; it includes immune checkpoint inhibition, prostate specific antigen (PSA) vaccines and dendritic cell-based strategies. Despite increasing evidence for clinical responses, the consensus of multiple trials is that prostate cancers are poorly responsive to immunotherapy. Prostate cancer has a high degree of pathological and genetic heterogeneity compared to other cancer types, which may account for immunotherapeutic resistance. This hypothesis also implies that select types of prostate tumors may be differentially responsive to immune-based strategies and that the clinical stage, pathological grade and underlying genetic landscape may be important criteria in identifying tumors that respond to immune therapies. One strategy is to target oncogenic driver pathways in combination with immunotherapies with the goal of overcoming tumor immunity and broadening the number of patients achieving a clinical response. In this analysis, we address the hypothesis that driver oncogenic signaling pathways regulate cancer progression, tumor immunity and resistance to current immune therapeutics in prostate cancer. We propose that increased responsiveness may be achieved through the combined use of immunotherapies and inhibitors targeting tumor cell autonomous pathways that contribute towards anti-tumor immunity in patients with prostate cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas Oncogénicas/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Inmunoterapia/métodos , Masculino , Terapia Molecular Dirigida/métodos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
5.
Nat Med ; 22(7): 735-43, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27294876

RESUMEN

Immunomodulatory drugs (IMiDs), such as thalidomide and its derivatives lenalidomide and pomalidomide, are key treatment modalities for hematologic malignancies, particularly multiple myeloma (MM) and del(5q) myelodysplastic syndrome (MDS). Cereblon (CRBN), a substrate receptor of the CRL4 ubiquitin ligase complex, is the primary target by which IMiDs mediate anticancer and teratogenic effects. Here we identify a ubiquitin-independent physiological chaperone-like function of CRBN that promotes maturation of the basigin (BSG; also known as CD147) and solute carrier family 16 member 1 (SLC16A1; also known as MCT1) proteins. This process allows for the formation and activation of the CD147-MCT1 transmembrane complex, which promotes various biological functions, including angiogenesis, proliferation, invasion and lactate export. We found that IMiDs outcompete CRBN for binding to CD147 and MCT1, leading to destabilization of the CD147-MCT1 complex. Accordingly, IMiD-sensitive MM cells lose CD147 and MCT1 expression after being exposed to IMiDs, whereas IMiD-resistant cells retain their expression. Furthermore, del(5q) MDS cells have elevated CD147 expression, which is attenuated after IMiD treatment. Finally, we show that BSG (CD147) knockdown phenocopies the teratogenic effects of thalidomide exposure in zebrafish. These findings provide a common mechanistic framework to explain both the teratogenic and pleiotropic antitumor effects of IMiDs.


Asunto(s)
Basigina/efectos de los fármacos , Proteínas de Ciclo Celular/efectos de los fármacos , Factores Inmunológicos/farmacología , Inmunosupresores/farmacología , Proteínas Oncogénicas/efectos de los fármacos , Péptido Hidrolasas/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Teratogénesis/efectos de los fármacos , Talidomida/farmacología , Proteínas Adaptadoras Transductoras de Señales , Basigina/genética , Basigina/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Humanos , Lenalidomida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Teratogénesis/genética , Talidomida/análogos & derivados , Ubiquitina-Proteína Ligasas
6.
Blood ; 125(22): 3455-65, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-25795919

RESUMEN

Nucleophosmin (NPM1) mutations represent an attractive therapeutic target in acute myeloid leukemia (AML) because they are common (∼30% AML), stable, and behave as a founder genetic lesion. Oncoprotein targeting can be a successful strategy to treat AML, as proved in acute promyelocytic leukemia by treatment with all-trans retinoic acid (ATRA) plus arsenic trioxide (ATO), which degrade the promyelocytic leukemia (PML)-retinoic acid receptor fusion protein. Adjunct of ATRA to chemotherapy was reported to be beneficial for NPM1-mutated AML patients. Leukemic cells with NPM1 mutation also showed sensibility to ATO in vitro. Here, we explore the mechanisms underlying these observations and show that ATO/ATRA induce proteasome-dependent degradation of NPM1 leukemic protein and apoptosis in NPM1-mutated AML cell lines and primary patients' cells. We also show that PML intracellular distribution is altered in NPM1-mutated AML cells and reverted by arsenic through oxidative stress induction. Interestingly, similarly to what was described for PML, oxidative stress also mediates ATO-induced degradation of the NPM1 mutant oncoprotein. Strikingly, NPM1 mutant downregulation by ATO/ATRA was shown to potentiate response to the anthracyclin daunorubicin. These findings provide experimental evidence for further exploring ATO/ATRA in preclinical NPM1-mutated AML in vivo models and a rationale for exploiting these compounds in chemotherapeutic regimens in clinics.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Leucemia Mieloide Aguda/metabolismo , Proteínas Nucleares/metabolismo , Óxidos/farmacología , Tretinoina/farmacología , Animales , Apoptosis/genética , Trióxido de Arsénico , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones SCID , Proteínas Mutantes/efectos de los fármacos , Proteínas Mutantes/metabolismo , Mutación , Proteínas Nucleares/efectos de los fármacos , Nucleofosmina , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Células Tumorales Cultivadas , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Exp Mol Pathol ; 98(3): 375-81, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25445497

RESUMEN

Gene transcription analysis in clinical tumor samples can help with diagnosis, prognosis, and treatment of cancers. We aimed to identify the optimal reference genes for reliable expression analysis in various tumor samples by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). Using a one-step TaqMan-based qRT-PCR, 5 commonly used reference genes (ACTB, GAPDH, RPLPO, GUSB, and TFRC) and 10 anticancer drug-related genes (TYMS, RRM1, TUBB3, STMN1, TOP2A, EGFR, VEGFR2, HER2, ERCC1, and BRCA1) were analyzed in 327 tissue samples from lung, rectal, colon, gastric, esophageal, and breast tumors. According to the expression stability assessments obtained by using three programs (geNorm, NormFinder, and BestKeeper) and a comprehensive ranking method, the optimal reference genes for lung, gastric, esophageal, and breast tumors were RPLPO, GAPDH, ACTB, and ACTB, respectively. For rectal tumors, a combination of the 3 most stable genes (GUSB, ACTB, and RPLPO) was suitable for qRT-PCR, whereas for colon tumors, a combination of the 4 most stable genes (GAPDH, ACTB, GUSB, and RPLPO) was optimal for qRT-PCR. Based on the expression data of target genes normalized against selected reference genes, the principal component analysis revealed 4 expression patterns in 6 different tissues. One pattern was observed in gastric, rectal, and colon tumor tissues, which are gastrointestinal tumors. Expressions in the breast, lung, and esophageal tissues were separately represented as one pattern. Our results could facilitate the practice of personalized cancer medicine based on the gene expression profile of the patients.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Gastrointestinales/genética , Perfilación de la Expresión Génica/normas , Neoplasias Pulmonares/genética , Proteínas Oncogénicas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Proteínas Supresoras de Tumor/genética , Antineoplásicos/farmacología , Células HeLa , Humanos , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Especificidad de Órganos , Estándares de Referencia , Proteínas Supresoras de Tumor/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo
8.
J Med Food ; 18(2): 173-81, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25162762

RESUMEN

Lutein is a carotenoid pigment present in fruits and vegetables that has anti-inflammatory and antitumor properties. In this study, we examined the effect of lutein on proliferation and survival-associated genes in prostate cancer (PC-3) cells. We found that in vitro culture of PC-3 cells with lutein induced mild decrease in proliferation that improved in combination treatment with peroxisome proliferator-activated receptor gamma (PPARγ) agonists and other chemotherapeutic agents. Flow cytometry analyses showed that lutein improved drug-induced cell cycle arrest and apoptosis in prostate cancer. Gene array and quantitative reverse transcription-polymerase chain reaction analyses showed that lutein altered the expression of growth and apoptosis-associated biomarker genes in PC-3 cells. These findings highlight that lutein modulates the expression of growth and survival-associated genes in prostate cancer cells.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proliferación Celular/efectos de los fármacos , Luteína/farmacología , Proteínas Oncogénicas/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Carotenoides/farmacología , Bovinos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo , Frutas/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Luteína/administración & dosificación , Masculino , PPAR gamma/administración & dosificación , PPAR gamma/agonistas , Neoplasias de la Próstata/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Verduras/química
9.
J Periodontal Res ; 49(6): 810-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24823913

RESUMEN

BACKGROUND AND OBJECTIVE: Sonic hedgehog protein (SHH) is a mitogen that stimulates cell proliferation. Cyclosporine A enhances the proliferation of gingival cells; however, the relationships of SHH to cyclosporine A or to cyclosporine A-enhanced gingival cell proliferation have not been described. MATERIAL AND METHODS: Here, we investigated SHH expression in gingiva in vitro and in vivo after cyclosporine A treatment and tested the effect of SHH inhibition on cyclosporine A-enhanced gingival fibroblast proliferation in vitro. RESULTS: In human gingival fibroblasts, cyclosporine A treatment increased the expression of SHH transcripts and SHH protein, and stimulated cell proliferation; the addition of cyclopamine, an SHH signaling inhibitor, suppressed cyclosporine A-enhanced cell proliferation. Up-regulated expression of SHH and up-regulation of proliferating cell nuclear antigen transcripts and protein were observed in the edentulous gingiva of cyclosporine A-treated rats. CONCLUSION: Cyclosporine A up-regulates gingival SHH expression in vitro and in vivo, and the inhibition of the SHH pathway counteracts the stimulatory effect of cyclosporine A on gingival fibroblast proliferation. Therefore, we suggest that SHH mediates a novel molecular mechanism for cyclosporine A-induced gingival complications.


Asunto(s)
Ciclosporina/farmacología , Encía/efectos de los fármacos , Proteínas Hedgehog/efectos de los fármacos , Mitógenos/farmacología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Encía/citología , Proteínas Hedgehog/análisis , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Arcada Edéntula/patología , Masculino , Modelos Animales , Proteínas Oncogénicas/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/análisis , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transactivadores/efectos de los fármacos , Regulación hacia Arriba , Alcaloides de Veratrum/farmacología , Proteína con Dedos de Zinc GLI1 , Dedos de Zinc/efectos de los fármacos
10.
Am J Respir Crit Care Med ; 188(7): 770-5, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23841470

RESUMEN

The remarkable success of epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors in patients with EGFR mutations and ALK rearrangements, respectively, introduced the era of targeted therapy in advanced non-small cell lung cancer (NSCLC), shifting treatment from platinum-based combination chemotherapy to molecularly tailored therapy. Recent genomic studies in lung adenocarcinoma identified other potential therapeutic targets, including ROS1 rearrangements, RET fusions, MET amplification, and activating mutations in BRAF, HER2, and KRAS in frequencies exceeding 1%. Lung cancers that harbor these genomic changes can potentially be targeted with agents approved for other indications or under clinical development. The need to generate increasing amounts of genomic information should prompt health-care providers to be mindful of the amounts of tissue needed for these assays when planning diagnostic procedures. In this review, we summarize oncogenic drivers in NSCLC that can be currently detected, highlight their potential therapeutic implications, and discuss practical considerations for successful application of tumor genotyping in clinical decision making.


Asunto(s)
Adenocarcinoma/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Proteínas Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Tirosina Quinasas Receptoras/genética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma del Pulmón , Quinasa de Linfoma Anaplásico , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/uso terapéutico , Genómica , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida/métodos , Mutación/efectos de los fármacos , Mutación/genética , Proteínas Oncogénicas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-ret/efectos de los fármacos , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas p21(ras) , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/uso terapéutico , Receptor ErbB-2/efectos de los fármacos , Receptor ErbB-2/genética , Proteínas ras/efectos de los fármacos , Proteínas ras/genética
11.
Cancer Chemother Pharmacol ; 71(3): 713-25, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23292117

RESUMEN

PURPOSE: To investigate the effects of (-)-epigallocatechin-3-gallate (EGCG) on human papillomavirus (HPV)-16 oncoprotein-induced angiogenesis in non-small cell lung cancer (NSCLC) cells and the underlying mechanisms. METHODS: NSCLC cells (A549 and NCI-H460) transfected with EGFP plasmids containing HPV-16 E6 or E7 oncogene were treated with different concentrations of EGCG for 16 h. The effects of EGCG on angiogenesis in vitro and in vivo were observed. The expression of HIF-1α, p-Akt, and p-ERK1/2 proteins in NSCLC cells was analyzed by Western blot. The levels of HIF-1α mRNA in NSCLC cells were detected by real-time RT-PCR. The concentration of VEGF and IL-8 in the conditioned media was determined by ELISA. HIF-1α, VEGF, and CD31 expression in A549 xenografted tumors of nude mice was analyzed by immunohistochemistry. RESULTS: HPV-16 E6 and E7 oncoproteins HIF-1α-dependently promoted angiogenesis in vitro and in vivo, which was inhibited by EGCG. Mechanistically, EGCG inhibited HPV-16 oncoprotein-induced HIF-1α protein expression but had no effect on HIF-1α mRNA expression in NSCLC cells. Additionally, 50 and 100 µmol/L of EGCG significantly reduced the secretion of VEGF and IL-8 proteins induced by HPV-16 E7 oncoprotein in NSCLC A549 cells. Meanwhile, HPV-16 E6 and E7 oncoproteins HIF-1α-dependently enhanced Akt activation in A549 cells, which was suppressed by EGCG. Furthermore, EGCG inhibited HPV-16 oncoprotein-induced HIF-1α and HIF-1α-dependent VEGF and CD31 expression in A549 xenografted tumors. CONCLUSIONS: EGCG inhibited HPV-16 oncoprotein-induced angiogenesis conferred by NSCLC through the inhibition of HIF-1α protein expression and HIF-1α-dependent expression of VEGF, IL-8, and CD31 as well as activation of Akt, suggesting that HIF-1α may be a potential target of EGCG against HPV-related NSCLC angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Catequina/análogos & derivados , Papillomavirus Humano 16/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/prevención & control , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Proteínas Oncogénicas Virales/toxicidad , Proteínas Oncogénicas/efectos de los fármacos , Proteínas E7 de Papillomavirus/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/toxicidad , Animales , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/patología , Catequina/farmacología , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Interleucina-8/biosíntesis , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transfección , Factor A de Crecimiento Endotelial Vascular/biosíntesis
12.
Hypertension ; 59(2): 446-52, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22215708

RESUMEN

The D(2) dopamine receptor (D(2)R) is important in the pathogenesis of essential hypertension. We have already reported that systemic deletion of the D(2)R gene in mice results in reactive oxygen species (ROS)-dependent hypertension, suggesting that the D(2)R has antioxidant effects. However, the mechanism of this effect is unknown. DJ-1 is a protein that has antioxidant properties. D(2)R and DJ-1 are expressed in the mouse kidney and colocalize and coimunoprecipitate in mouse renal proximal tubule cells. We hypothesized that D(2)Rs regulate renal ROS production in the kidney through regulation of DJ-1 expression or function. Heterozygous D(2)(+/-) mice have increased blood pressure, urinary 8-isoprostanes, and renal Nox 4 expression, but decreased renal DJ-1 expression. Silencing D(2)R expression in mouse renal proximal tubule cells increases ROS production and decreases the expression of DJ-1. Conversely, treatment of these cells with a D(2)R agonist increases DJ-1 expression and decreases Nox 4 expression and NADPH oxidase activity, effects that are partially blocked by a D(2)R antagonist. Silencing DJ-1 expression in mouse renal proximal tubule cells increases ROS production and Nox 4 expression. Selective renal DJ-1 silencing by the subcapsular infusion of DJ-1 siRNA in mice increases blood pressure, renal Nox4 expression, and NADPH oxidase activity. These results suggest that the inhibitory effects of D(2)R on renal ROS production are at least, in part, mediated by a positive regulation of DJ-1 expression/function and that DJ-1 may have a role in the prevention of hypertension associated with increased ROS production.


Asunto(s)
Hipertensión/etiología , Hipertensión/metabolismo , Riñón/metabolismo , Proteínas Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Hipertensión/fisiopatología , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 4 , NADPH Oxidasas/metabolismo , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/efectos de los fármacos , Peroxirredoxinas , Proteína Desglicasa DJ-1 , ARN Interferente Pequeño/farmacología , Receptores de Dopamina D2/deficiencia , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo
13.
J Oral Pathol Med ; 41(4): 322-31, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22103929

RESUMEN

BACKGROUND: To clarify the efficacy of grape seed procyanidin (GSP) on antiproliferative effects related to p53 functional status of oral squamous cell carcinoma (OSCC) for its chemoadjuvant potential. METHODS: We used GSP to investigate SCC-25 cells with wild-type p53 gene and OEC-M1 cells with mutant p53 gene for the assessment of antiproliferative effects including cell viability, cell cycle, apoptosis, migration and invasion potential, and alterations of associated oncoproteins involved in cellular and molecular events. RESULTS: The findings suggest that GSP on OEC-M1 cells leads to cell cycle arrest by increasing the expression of p21(Cip1) /p27(Kip1) protein without functioning mitochondria-mediated apoptosis, whereas GSP on SCC-25 cells inhibits cell proliferation via both G1-phase arrest and mitochondria-mediated apoptosis in a dose-dependent manner as a result of alterations of Bcl-2. GSP also inhibits the migration and invasion of both cells, which are associated with the suppression of matrix metalloproteinases (MMPs), MMP-2 and MMP-9. CONCLUSION: Antiproliferative effectiveness of GSP is closely associated with the p53 status of OSCC cells. GSP displays chemoadjuvant potential via cell cycle blockage and apoptotic induction. Our findings clearly suggest that GSP may play a role as a novel chemopreventive or therapeutic agent for OSCC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Escamosas/patología , Extracto de Semillas de Uva/farmacología , Neoplasias de la Boca/patología , Preparaciones de Plantas/farmacología , Proantocianidinas/farmacología , Proteína p53 Supresora de Tumor/efectos de los fármacos , Vitis , Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Fase G1/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Invasividad Neoplásica , Proteínas Oncogénicas/efectos de los fármacos , Mutación Puntual/genética , Inhibidores de Proteínas Quinasas/análisis , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética
14.
J Biol Chem ; 286(17): 14941-51, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21372141

RESUMEN

Parkinson disease is caused by the death of midbrain dopamine neurons from oxidative stress, abnormal protein aggregation, and genetic predisposition. In 2003, Bonifati et al. (23) found that a single amino acid mutation in the DJ-1 protein was associated with early-onset, autosomal recessive Parkinson disease (PARK7). The mutation L166P prevents dimerization that is essential for the antioxidant and gene regulatory activity of the DJ-1 protein. Because low levels of DJ-1 cause Parkinson, we reasoned that overexpression might stop the disease. We found that overexpression of DJ-1 improved tolerance to oxidative stress by selectively up-regulating the rate-limiting step in glutathione synthesis. When we imposed a different metabolic insult, A53T mutant α-synuclein, we found that DJ-1 turned on production of the chaperone protein Hsp-70 without affecting glutathione synthesis. After screening a number of small molecules, we have found that the histone deacetylase inhibitor phenylbutyrate increases DJ-1 expression by 300% in the N27 dopamine cell line and rescues cells from oxidative stress and mutant α-synuclein toxicity. In mice, phenylbutyrate treatment leads to a 260% increase in brain DJ-1 levels and protects dopamine neurons against 1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine (MPTP) toxicity. In a transgenic mouse model of diffuse Lewy body disease, long-term administration of phenylbutyrate reduces α-synuclein aggregation in brain and prevents age-related deterioration in motor and cognitive function. We conclude that drugs that up-regulate DJ-1 gene expression may slow the progression of Parkinson disease by moderating oxidative stress and protein aggregation.


Asunto(s)
Proteínas Oncogénicas/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , Fenilbutiratos/farmacología , Animales , Células Cultivadas , Inhibidores de Histona Desacetilasas , Ratones , Neuronas , Fármacos Neuroprotectores , Proteínas Oncogénicas/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Peroxirredoxinas , Fenilbutiratos/uso terapéutico , Proteína Desglicasa DJ-1 , Proteínas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
Anticancer Drugs ; 20(4): 230-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19288605

RESUMEN

A number of polyamine analogs are currently used in various clinical trials as cancer treatment and it is important to investigate their effects not only on cancer cells but also on normal cells. Treatment with polyamine analogs depletes cells of polyamines and inhibits cell proliferation, but the analogs cannot take over the normal function of the natural polyamines in the cell. In this study, the normal-like breast epithelial cell line MCF-10A was treated with the polyamine analog N',N"-diethylnorspermine (DENSPM). The cells were then studied using a bromodeoxyuridine- DNA flow cytometry method as well as western blot. The ability of both normal-like and breast cancer cells to recover from DENSPM treatment was also studied. DENSPM treatment of MCF-10A cells resulted in a prolongation of the S and G2 +M phases, followed by a G1/S block. The p53/p21/RB1 pathway was involved in the G1/S block as shown by increased levels of p53 and p21 detected by western blot. Decreased levels of cyclin E1, cyclin A2, and cyclin B1 in DENSPM-treated cells can explain the prolongation of cell cycle phases that occurred before the G1/S block. We also show that MCF-10A cells rapidly recover from DENSPM-induced growth inhibition in contrast to four human breast cancer cell lines. The goal of cancer treatment is to cause minimal and reversible damage to normal cells, while cancer cells should be eliminated. Altogether, the data show that treatment with polyamine analogs spares normal cells, while negatively affecting the cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Espermina/análogos & derivados , Western Blotting , Neoplasias de la Mama/patología , Bromodesoxiuridina , Línea Celular , Línea Celular Tumoral , Ciclina A/efectos de los fármacos , Ciclina A/metabolismo , Ciclina A2 , Ciclina B/efectos de los fármacos , Ciclina B/metabolismo , Ciclina B1 , Ciclina E/efectos de los fármacos , Ciclina E/metabolismo , ADN , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Citometría de Flujo , Humanos , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Espermina/farmacología
16.
J Pharmacol Sci ; 109(3): 463-8, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19276614

RESUMEN

Although a loss-of-function type mutation was identified in familial Parkinson's disease PARK7, the wild-type of DJ-1 is known to act as an oxidative stress sensor in neuronal cells. Recently, we found a DJ-1 modulator UCP0054278 by in silico virtual screening. In this study, we determined the neuroprotective effects of UCP0054278 against focal ischemia-induced neurodegeneration in rats. Hydrogen peroxide-induced cell death and the production of reactive oxygen species were significantly inhibited by UCP0054278 in normal SH-SY5Y cells, but not in DJ-1-knockdown cells. These results suggest that UCP0054278 interacts with endogenous DJ-1 and then exhibits antioxidant and neuroprotective responses.


Asunto(s)
Benzamidas/farmacología , Benzodioxoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteínas Oncogénicas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/fisiopatología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Peróxido de Hidrógeno , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Degeneración Nerviosa/prevención & control , Neuroblastoma/metabolismo , Proteínas Oncogénicas/metabolismo , Enfermedad de Parkinson/genética , Proteína Desglicasa DJ-1 , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
17.
Pediatr Blood Cancer ; 50(4): 806-11, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17554788

RESUMEN

BACKGROUND: Use of retinoic acid (RA) has become the standard of care in the treatment of high risk neuroblastoma (NB). In vitro, RA induces growth arrest and differentiation, an effect that likely underlies its activity in the clinical setting. An important event in differentiation is the transcriptional downregulation of the MYCN oncogene, which is frequently activated in aggressive tumors. While it is known that Sp1/Sp3 and E2F are necessary to drive basal MYCN expression, the mechanism for its downregulation by RA remains enigmatic. Changes in E2F binding have been reported, however these occurred after the actual transcriptional response. Here, post-translational modifications of Sp proteins were examined as an alternate mechanism of RA-mediated promoter regulation. PROCEDURE: Western blot was used to evaluate steady state levels of nuclear/cytoplasmic Sp1/Sp3. Promoter binding and DNA conformation were determined by gel shift, circular permutation, and chromatin immunoprecipitation assays. Immunoprecipitation/western and (32)P-phosphoamino analyses were used to detect glycosylation, acetylation, sumoylation, and phosphorylation. RESULTS: RA did not affect the cellular level of Sp1/Sp3 proteins, their nuclear/cytoplasmic distribution, ability to bind the MYCN promoter, degree of Sp-induced DNA bending, or post-translational modifications. CONCLUSIONS: MYCN RA response is not mediated solely though the region controlling basal activity. RA may be exerting its effects via multiple non-adjacent regulatory regions, potentially including basal motifs, either within the MYCN promoter or distally, on the same or even different chromosomes. Such cooperative trans-type DNA-protein interactions could explain the inaccessibility of this mechanism to the locus-specific approaches employed up to this point.


Asunto(s)
Antineoplásicos/farmacología , Inmunoglobulinas/efectos de los fármacos , Proteínas Nucleares/efectos de los fármacos , Proteínas Oncogénicas/efectos de los fármacos , Factor de Transcripción Sp3/efectos de los fármacos , Tretinoina/farmacología , Western Blotting , Línea Celular Tumoral , Regulación hacia Abajo , Ensayo de Cambio de Movilidad Electroforética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Inmunoprecipitación , Proteína Proto-Oncogénica N-Myc , Neuroblastoma , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Factor de Transcripción Sp3/genética , Factor de Transcripción Sp3/metabolismo
19.
Chin J Dig Dis ; 7(4): 219-24, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17054584

RESUMEN

BACKGROUND: Non-steroidal anti-inflammatory drugs such as indomethacin can inhibit the growth of tumors through both the cyclooxygenase-2 (COX-2) dependant and COX-2 independent pathways, but the exact mechanism has not yet been shown. In our previous study, COX-2 independent proteins (Bfl-1, WISP-1 and proliferating cell nuclear antigens [PCNA]) in indomethacin-treated colorectal cancer cells with the use of proteomics technology had been identified. OBJECTIVES: To study and confirm the effect of indomethacin on the expression of Bfl-1, WISP-1 and PCNA in human colon cancer line HCT116 cells and the COX-2 independent tumor inhibiting pathway. METHODS: Human colon cancer cell line HCT116 cells were divided into a treatment with indomethacin (IC 50) group, and a treatment with dimethyl sulfoxide (DMSO) as a control group for 48 h. The expression of Bfl-1, WISP-1 and PCNA, mRNA and protein were determined by a real-time quantitative PCR and Western blot, respectively. RESULTS: Indomethacin down-regulated the expression of Bfl-1, WISP-1 and PCNA mRNA in vitro (9.53 +/- 0.15 vs 27.87 +/- 0.12, 7.37 +/- 0.58 vs 20.17 +/- 0.58, 5.17 +/- 0.06 vs 0.87 +/- 0.06). Indomethacin also down-regulated the expression of Bfl-1, WISP-1 and PCNA protein (40.01 +/- 1.61 vs 43.76 +/- 1.63, 22.50 +/- 1.17 vs 30.30 +/- 1.55, 17.69 +/- 1.18 vs 20.80 +/- 1.08). CONCLUSIONS: Inducing apoptosis and inhibiting proliferation contribute to the anticancer activity of indomethacin via COX-2 independent pathway of Bfl-1, WISP-1 and PCNA. This further confirms the results of our previous study.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Indometacina/farmacología , Proteínas Oncogénicas/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proteínas CCN de Señalización Intercelular , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Antígenos de Histocompatibilidad Menor , Proteínas Oncogénicas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo
20.
J Am Chem Soc ; 128(27): 8939-46, 2006 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-16819890

RESUMEN

Inteins are naturally occurring protein elements that catalyze their own excision from within a larger protein together with the ligation of the flanking "extein" sequences. Previously we reported the directed evolution of an intein-based molecular switch in which intein splicing in yeast cells was made dependent on the cell-permeable small molecule 4-hydroxytamoxifen (4-HT). Here we show that these evolved inteins are effective means of rendering protein function and biological signaling pathway activation dependent on 4-HT in mammalian cells. We have characterized the generality, speed, and dose dependence of ligand-induced protein splicing in murine NIH3T3 cells and in human HEK293 cells. Evolved inteins were used to control in mammalian cells the function of Gli1 and a truncated form of Gli3, two transcriptional mediators of the Hedgehog signaling pathway. Finally, we show that a complex biological process such as osteoblast differentiation can be made dependent on 4-HT using the evolved intein system. Our findings suggest that evolved small-molecule-dependent inteins may serve as a general means of achieving gene-specific, dose-dependent, post-translational, and small-molecule-induced control over protein activity in mammalian systems.


Asunto(s)
Inteínas , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Tamoxifeno/análogos & derivados , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Catálisis , Diferenciación Celular/efectos de los fármacos , Línea Celular , Evolución Molecular Dirigida , Relación Dosis-Respuesta a Droga , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/efectos de los fármacos , Proteínas Fluorescentes Verdes/fisiología , Humanos , Inteínas/genética , Factores de Transcripción de Tipo Kruppel/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ligandos , Ratones , Peso Molecular , Células 3T3 NIH , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Osteoblastos/química , Transducción de Señal/fisiología , Tamoxifeno/química , Tamoxifeno/farmacología , Factores de Tiempo , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/química , Proteína con Dedos de Zinc GLI1 , Proteína Gli3 con Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...