Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Nano ; 16(12): 20021-20033, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36264003

RESUMEN

To address the key challenges in the development of next-generation drug delivery systems (DDS) with desired physicochemical properties to overcome limitations regarding safety, in vivo efficacy, and solid tumor penetration, an ultrasmall folate receptor alpha (FRα) targeted silica nanoparticle (C'Dot) drug conjugate (CDC; or folic acid CDC) was developed. A broad array of methods was employed to screen a panel of CDCs and identify a lead folic acid CDC for clinical development. These included comparing the performance against antibody-drug conjugates (ADCs) in three-dimensional tumor spheroid penetration ability, assessing in vitro/ex vivo cytotoxic efficacy, as well as in vivo therapeutic outcome in multiple cell-line-derived and patient-derived xenograft models. An ultrasmall folic acid CDC, EC112002, was identified as the lead candidate out of >500 folic acid CDC formulations evaluated. Systematic studies demonstrated that the lead formulation, EC112002, exhibited highly specific FRα targeting, multivalent binding properties that would mediate the ability to outcompete endogenous folate in vivo, enzymatic responsive payload cleavage, stability in human plasma, rapid in vivo clearance, and minimal normal organ retention organ distribution in non-tumor-bearing mice. When compared with an anti-FRα-DM4 ADC, EC112002 demonstrated deeper penetration into 3D cell-line-derived tumor spheroids and superior specific cytotoxicity in a panel of 3D patient-derived tumor spheroids, as well as enhanced efficacy in cell-line-derived and patient-derived in vivo tumor xenograft models expressing a range of low to high levels of FRα. With the growing interest in developing clinically translatable, safe, and efficacious DDSs, EC112002 has the potential to address some of the critical limitations of the current systemic drug delivery for cancer management.


Asunto(s)
Receptor 1 de Folato , Sistema de Administración de Fármacos con Nanopartículas , Neoplasias , Animales , Humanos , Ratones , Línea Celular Tumoral , Modelos Animales de Enfermedad , Receptor 1 de Folato/metabolismo , Receptor 1 de Folato/uso terapéutico , Ácido Fólico/química , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Dióxido de Silicio/uso terapéutico
2.
Gastroenterology ; 161(1): 196-210, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33745946

RESUMEN

BACKGROUND & AIMS: Understanding the mechanisms by which tumors adapt to therapy is critical for developing effective combination therapeutic approaches to improve clinical outcomes for patients with cancer. METHODS: To identify promising and clinically actionable targets for managing colorectal cancer (CRC), we conducted a patient-centered functional genomics platform that includes approximately 200 genes and paired this with a high-throughput drug screen that includes 262 compounds in four patient-derived xenografts (PDXs) from patients with CRC. RESULTS: Both screening methods identified exportin 1 (XPO1) inhibitors as drivers of DNA damage-induced lethality in CRC. Molecular characterization of the cellular response to XPO1 inhibition uncovered an adaptive mechanism that limited the duration of response in TP53-mutated, but not in TP53-wild-type CRC models. Comprehensive proteomic and transcriptomic characterization revealed that the ATM/ATR-CHK1/2 axes were selectively engaged in TP53-mutant CRC cells upon XPO1 inhibitor treatment and that this response was required for adapting to therapy and escaping cell death. Administration of KPT-8602, an XPO1 inhibitor, followed by AZD-6738, an ATR inhibitor, resulted in dramatic antitumor effects and prolonged survival in TP53-mutant models of CRC. CONCLUSIONS: Our findings anticipate tremendous therapeutic benefit and support the further evaluation of XPO1 inhibitors, especially in combination with DNA damage checkpoint inhibitors, to elicit an enduring clinical response in patients with CRC harboring TP53 mutations.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/tratamiento farmacológico , Carioferinas/antagonistas & inhibidores , Mutación , Inhibidores de Proteínas Quinasas/administración & dosificación , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Bases de Datos Genéticas , Células HCT116 , Células HT29 , Humanos , Indoles/administración & dosificación , Carioferinas/metabolismo , Ratones , Morfolinas/administración & dosificación , Piperazinas/administración & dosificación , Piridinas/administración & dosificación , Pirimidinas/administración & dosificación , Receptores Citoplasmáticos y Nucleares/metabolismo , Sulfonamidas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
3.
Nat Med ; 24(10): 1627, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30104769

RESUMEN

In the version of this article originally published, information regarding several funding sources was omitted from the Acknowledgements section. The following sentences should have been included: "This work was supported by the generous philanthropic contributions to The University of Texas MD Anderson Lung Cancer Moon Shots Program, the UT Lung SPORE 5 P50 CA07090, and the MD Anderson Cancer Center Support Grant P30CA01667. V.P is supported by R01CA155196-01A1 from the National Cancer Institute." Also, reference 18 was incorrect. The original reference was: Kim, E. S. et al. The BATTLE trial: personalizing therapy for lung cancer. Cancer Discov. 1, 44-53 (2011). It should have been: Papadimitrakopoulou, V. et al. The BATTLE-2 study: a biomarker-integrated targeted therapy study in previously treated patients with advanced non-small-cell lung cancer. J Clin. Oncol. 34, 3638-3647 (2016). The errors have been corrected in the HTML and PDF versions of this article.

4.
Nat Med ; 24(7): 1047-1057, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29892061

RESUMEN

Lung cancer is a devastating disease that remains a top cause of cancer mortality. Despite improvements with targeted and immunotherapies, the majority of patients with lung cancer lack effective therapies, underscoring the need for additional treatment approaches. Genomic studies have identified frequent alterations in components of the SWI/SNF chromatin remodeling complex including SMARCA4 and ARID1A. To understand the mechanisms of tumorigenesis driven by mutations in this complex, we developed a genetically engineered mouse model of lung adenocarcinoma by ablating Smarca4 in the lung epithelium. We demonstrate that Smarca4 acts as a bona fide tumor suppressor and cooperates with p53 loss and Kras activation. Gene expression analyses revealed the signature of enhanced oxidative phosphorylation (OXPHOS) in SMARCA4 mutant tumors. We further show that SMARCA4 mutant cells have enhanced oxygen consumption and increased respiratory capacity. Importantly, SMARCA4 mutant lung cancer cell lines and xenograft tumors have marked sensitivity to inhibition of OXPHOS by a novel small molecule, IACS-010759, that is under clinical development. Mechanistically, we show that SMARCA4-deficient cells have a blunted transcriptional response to energy stress creating a therapeutically exploitable synthetic lethal interaction. These findings provide the mechanistic basis for further development of OXPHOS inhibitors as therapeutics against SWI/SNF mutant tumors.


Asunto(s)
ADN Helicasas/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación/genética , Proteínas Nucleares/genética , Fosforilación Oxidativa , Factores de Transcripción/genética , Animales , Vías Biosintéticas , Línea Celular Tumoral , Respiración de la Célula , ADN Helicasas/deficiencia , Metabolismo Energético , Femenino , Ingeniería Genética , Humanos , Ratones Desnudos , Mitocondrias/metabolismo , Proteínas Nucleares/deficiencia , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Estrés Fisiológico/genética , Factores de Transcripción/deficiencia , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nat Med ; 24(7): 1036-1046, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29892070

RESUMEN

Metabolic reprograming is an emerging hallmark of tumor biology and an actively pursued opportunity in discovery of oncology drugs. Extensive efforts have focused on therapeutic targeting of glycolysis, whereas drugging mitochondrial oxidative phosphorylation (OXPHOS) has remained largely unexplored, partly owing to an incomplete understanding of tumor contexts in which OXPHOS is essential. Here, we report the discovery of IACS-010759, a clinical-grade small-molecule inhibitor of complex I of the mitochondrial electron transport chain. Treatment with IACS-010759 robustly inhibited proliferation and induced apoptosis in models of brain cancer and acute myeloid leukemia (AML) reliant on OXPHOS, likely owing to a combination of energy depletion and reduced aspartate production that leads to impaired nucleotide biosynthesis. In models of brain cancer and AML, tumor growth was potently inhibited in vivo following IACS-010759 treatment at well-tolerated doses. IACS-010759 is currently being evaluated in phase 1 clinical trials in relapsed/refractory AML and solid tumors.


Asunto(s)
Neoplasias/patología , Fosforilación Oxidativa , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Metabolismo Energético , Glucólisis/efectos de los fármacos , Células HEK293 , Humanos , Ácido Láctico/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Mitocondrias/metabolismo , Nucleótidos/biosíntesis , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cell Biosci ; 8: 3, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29344347

RESUMEN

PURPOSE: We investigated the genomic DNA methylation profile of prostate cancer in transgenic adenocarcinoma of the mouse prostate (TRAMP) cancer model and to analyze the crosstalk among targeted genes and the related functional pathways. METHODS: Prostate DNA samples from 24-week-old TRAMP and C57BL/6 male mice were isolated. The DNA methylation profiles were analyzed by methylated DNA immunoprecipitation (MeDIP) followed by next-generation sequencing (MeDIP-seq). Canonical pathways, diseases and function and network analyses of the different samples were then performed using the Ingenuity® Pathway Analysis (IPA) software. Some target genes with significant difference in methylation were selected for validation using methylation specific primers (MSP) and qPCR. RESULTS: TRAMP mice undergo extensive aberrant CpG hyper- and hypo-methylation. There were 2147 genes with a significant (log2-change ≥ 2) change in CpG methylation between the two groups, as mapped by the IPA software. Among these genes, the methylation of 1105 and 1042 genes was significantly decreased and increased, respectively, in TRAMP prostate tumors. The top associated disease identified by IPA was adenocarcinoma; however, the cAMP response element-binding protein (CREB)-, histone deacetylase 2 (HDAC2)-, glutathione S-transferase pi (GSTP1)- and polyubiquitin-C (UBC)-related pathways showed significantly altered methylation profiles based on the canonical pathway and network analyses. MSP and qPCR results of genes of interests corroborated with MeDIP-seq findings. CONCLUSIONS: This is the first MeDIP-seq with IPA analysis of the TRAMP model to provide novel insight into the genome-wide methylation profile of prostate cancer. Studies on epigenetics, such as DNA methylation, will potentially provide novel avenues and strategies for further development of biomarkers targeted for treatment and prevention approaches for prostate cancer.

7.
Cancer Res ; 77(21): e62-e66, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29092942

RESUMEN

Patient-derived tumor xenograft (PDX) mouse models have emerged as an important oncology research platform to study tumor evolution, mechanisms of drug response and resistance, and tailoring chemotherapeutic approaches for individual patients. The lack of robust standards for reporting on PDX models has hampered the ability of researchers to find relevant PDX models and associated data. Here we present the PDX models minimal information standard (PDX-MI) for reporting on the generation, quality assurance, and use of PDX models. PDX-MI defines the minimal information for describing the clinical attributes of a patient's tumor, the processes of implantation and passaging of tumors in a host mouse strain, quality assurance methods, and the use of PDX models in cancer research. Adherence to PDX-MI standards will facilitate accurate search results for oncology models and their associated data across distributed repository databases and promote reproducibility in research studies using these models. Cancer Res; 77(21); e62-66. ©2017 AACR.


Asunto(s)
Neoplasias , Ensayos Antitumor por Modelo de Xenoinjerto/estadística & datos numéricos , Animales , Bases de Datos como Asunto , Modelos Animales de Enfermedad , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Pacientes
8.
AAPS J ; 19(6): 1703-1714, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28828752

RESUMEN

Oxidative stress is an important pathogenic factor in various hepatic diseases. Nuclear factor-erythroid 2-related factor-2 (Nrf2), which coordinates the expression of an array of antioxidant and detoxifying genes, has been proposed as a potential target for prevention and treatment of liver disease. Dibenzoylmethane (DBM) is a minor ingredient in licorice that activates Nrf2 and prevents various cancers and oxidative damage. In the present study, the mechanisms by which DBM activates Nrf2 signaling were delineated, and its protective effect against carbon tetrachloride (CCl4)-induced liver injury was examined. DBM potently induced the expression of HO-1 in cells and in the livers of mice, but this induction was diminished in Nrf2-deficient mice and cells. Overexpression of Nrf2 enhanced DBM-induced HO-1 expression, while overexpression of a dominant-negative fragment of Nrf2 inhibited this induction. DBM treatment resulted in dissociation from Keap1 and nuclear translocation of Nrf2. Moreover, DBM activated Akt/protein kinase B, mitogen-activated protein kinases, and AMP-activated protein kinase and increased intracellular calcium levels. Inhibition of JNK, AMPK, or intracellular calcium signaling significantly suppressed the induction of HO-1 expression by DBM. Finally, DBM treatment significantly inhibited CCl4-induced acute liver injury in wild-type but not in Nrf2-deficient mice. Taken together, our results revealed the mechanisms by which DBM activates Nrf2 and induces HO-1 expression, and provide molecular basis for the design and development of DBM and its derivatives for prevention or treatment of liver diseases by targeting Nrf2.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Señalización del Calcio/fisiología , Chalconas/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Factor 2 Relacionado con NF-E2/fisiología , Animales , Tetracloruro de Carbono/toxicidad , Línea Celular Tumoral , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/fisiología , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones
9.
Br J Cancer ; 116(4): 515-523, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-28118322

RESUMEN

BACKGROUND: Screening of patients for cancer-driving mutations is now used for cancer prognosis, remission scoring and treatment selection. Although recently emerged targeted next-generation sequencing-based approaches offer promising diagnostic capabilities, there are still limitations. There is a pressing clinical need for a well-validated, rapid, cost-effective mutation profiling system in patient specimens. Given their speed and cost-effectiveness, quantitative PCR mutation detection techniques are well suited for the clinical environment. The qBiomarker mutation PCR array has high sensitivity and shorter turnaround times compared with other methods. However, a direct comparison with existing viable alternatives are required to assess its true potential and limitations. METHODS: In this study, we evaluated a panel of 117 patient-derived tumour xenografts by the qBiomarker array and compared with other methods for mutation detection, including Ion AmpliSeq sequencing, whole-exome sequencing and droplet digital PCR. RESULTS: Our broad analysis demonstrates that the qBiomarker's performance is on par with that of other labour-intensive and expensive methods of cancer mutation detection of frequently altered cancer-associated genes, and provides a foundation for supporting its consideration as an option for molecular diagnostics. CONCLUSIONS: This large-scale direct comparison and validation of currently available mutation detection approaches is extremely relevant for the current scenario of precision medicine and will lead to informed choice of screening methodologies, especially in lower budget conditions or time frame limitations.


Asunto(s)
Análisis Mutacional de ADN/métodos , Xenoinjertos , Neoplasias/genética , Animales , Xenoinjertos/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias/patología , Reacción en Cadena de la Polimerasa/métodos , Reproducibilidad de los Resultados , Células Tumorales Cultivadas
10.
Mol Nutr Food Res ; 60(6): 1427-36, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26820911

RESUMEN

SCOPE: Tumor metastasis greatly contributes to the mortality of prostate cancer. The glucosinolate-derived phenethyl isothiocyanate (PEITC) has been widely documented to reduce the risk of prostate cancer by modulating multiple biologically relevant processes. Emerging evidence suggests that PEITC may exert its anti-cancer effects through epigenetic mechanisms including microRNAs. Altered levels of miRNA have been linked to tumor malignancy due to their capacity to regulate functional gene expression in carcinogenesis. Here, we assessed the effects of PEITC on miRNA expression which is related to PCa cell invasiveness. METHODS AND RESULTS: Utilizing oligonucleotide microarray first identified the most affected miRNAs in LNCaP cells after PEITC treatment. Several top altered miRNAs were further validated using quantitative PCR. Interestingly, overexpression of miR-194 suppressed PC3 cell invasion in matrigel-coated Transwell chambers. Bone morphogenetic protein 1 (BMP1) was shown to be a direct target of miR-194. Downregulation of BMP1 by miR-194 or PEITC led to decreased expression of key oncogenic matrix metalloproteinases, MMP2 and MMP9. This in turn resulted in the suppression of tumor invasion. CONCLUSION: Our results indicate that miR-194 downregulates the expression of oncogenic MMP2 and MMP9 by targeting BMP1, which suggests a potential new mechanistic target by which PEITC suppresses prostate cancer cell invasiveness.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Isotiocianatos/farmacología , MicroARNs/metabolismo , Invasividad Neoplásica/genética , Neoplasias de la Próstata/genética , Proteína Morfogenética Ósea 1/genética , Proteína Morfogenética Ósea 1/metabolismo , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , MicroARNs/genética , Neoplasias de la Próstata/tratamiento farmacológico
11.
J Pharmacokinet Pharmacodyn ; 42(4): 401-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26138223

RESUMEN

3,3'-Diindolylmethane (DIM) has been investigated as a potential anti-cancer chemopreventive agent in many preclinical and clinical studies. In this study, we sought to characterize the pharmacokinetics of DIM and to build a pharmacokinetic (PK) and pharmacodynamic (PD) model of the DIM-induced gene expression of phase II drug metabolizing enzymes (DME), which potentially links DIM's molecular effects to its in vivo chemopreventive efficacy. DIM (10 mg/kg) was administered intravenously (i.v.) to male Sprague-Dawley rats and blood samples were collected at selected time points for 48 h. The plasma concentration of DIM was determined using a validated HPLC method. The mRNA expression of NQO1, GSTP1 and UGT1A1 in blood lymphocytes was measured using quantitative PCR. An indirect response model was employed to relate the concentration of DIM to the expression of the genes NQO1, GSTP1 and UGT1A1, which were chosen as PD markers for DIM. After i.v. administration, the plasma concentration of DIM declined quickly, and the expression of target genes increased significantly, peaking at 1-2 h and then returning to basal levels after 24 h. The parameters in the PK-PD model were estimated. The PK-PD model aptly described the time delay and magnitude of gene expression induced by DIM. Our results indicate that DIM is effective at inducing various phase II DME, which are capable of detoxify carcinogens. This PK-PD modeling approach provides a framework for evaluating the acute effects of DIM or other similar drugs in clinical trials.


Asunto(s)
Anticarcinógenos/farmacocinética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucuronosiltransferasa/genética , Gutatión-S-Transferasa pi/genética , Indoles/farmacocinética , Modelos Biológicos , NAD(P)H Deshidrogenasa (Quinona)/genética , Animales , Anticarcinógenos/sangre , Anticarcinógenos/farmacología , Indoles/sangre , Indoles/farmacología , Inyecciones Intravenosas , Masculino , Fase II de la Desintoxicación Metabólica , Ratas Sprague-Dawley
12.
Cancer Prev Res (Phila) ; 7(12): 1186-97, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25266896

RESUMEN

Epigenetic control of NRF2, a master regulator of many critical antioxidative stress defense genes in human prostate cancer (CaP), is unknown. Our previous animal study found decreased Nrf2 expression through promoter CpG methylation/histone modifications during prostate cancer progression in TRAMP mice. In this study, we evaluated CpG methylation of human NRF2 promoter in 27 clinical prostate cancer samples and in LNCaP cells using MAQMA analysis and bisulfite genomic DNA sequencing. Prostate cancer tissue microarray (TMA) containing normal and prostate cancer tissues was studied by immunohistochemistry. Luciferase reporter assay using specific human NRF2 DNA promoter segments and chromatin immunoprecipitation (ChIP) assay against histone modifying proteins were performed in LNCaP cells. Three specific CpG sites in the NRF2 promoter were found to be hypermethylated in clinical prostate cancer samples (BPH

Asunto(s)
Metilación de ADN , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica , Factor 2 Relacionado con NF-E2/genética , Neoplasias de la Próstata/genética , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Islas de CpG/genética , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
13.
Cell Biosci ; 4: 39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25228981

RESUMEN

Nrf2 plays a critical role in defending against oxidative stress and inflammation. We previously reported that Nrf2 confers protection against ultraviolet-B (UVB)-induced inflammation, sunburn reaction, and is involved in sulforaphane-mediated photo-protective effects in the skin. In this study, we aimed to demonstrate the protective role of Nrf2 against inflammation-mediated extracellular matrix (ECM) damage induced by UVB irradiation. Ear biopsy weights were significantly increased in both Nrf2 wild-type (Nrf2 WT) and knockout (Nrf2 KO) mice one week after UVB irradiation. However, these weights increased more significantly in KO mice compared to WT mice, suggesting a greater inflammatory response in KO mice. In addition, we analyzed the protein expression of numerous markers, including macrophage inflammatory protein-2 (MIP-2), pro-matrix metalloproteinase-9 (MMP-9), and p53. p53, a regulator of DNA repair, was overexpressed in Nrf2 KO mice, indicating that the absence of Nrf2 led to more sustained DNA damage. There was also more substantial ECM degradation and increased inflammation in UVB-irradiated Nrf2 KO mice compared to UVB-irradiated WT mice. Furthermore, the protective effects of Nrf2 in response to UVB irradiation were mediated by increased HO-1 protein expression. Collectively, our results show that Nrf2 plays a key role in protecting against UVB irradiation and that the photo-protective effect of Nrf2 is closely related to the inhibition of ECM degradation and inflammation.

14.
Cancer Prev Res (Phila) ; 7(3): 319-29, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24441674

RESUMEN

Nrf2 is a transcription factor that plays critical roles in regulating the expression of cellular defensive antioxidants and detoxification enzymes. However, the role of Nrf2 and Nrf2's epigenetics reprogramming in skin tumor transformation is unknown. In this study, we investigated the inhibitory role and epigenetics of Nrf2 on tumor transformation induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) in mouse skin epidermal JB6 (JB6 P+) cells and the anticancer effect of sulforaphane (SFN), an isothiocyanate found in cruciferous vegetables. After five days of treatment, SFN significantly inhibited TPA-induced JB6 cellular transformation and SFN enhanced the nuclear translocation of Nrf2 and increased the mRNA and protein levels of the Nrf2 target genes HO-1, NQO1 and UGT1A1. Knockdown of Nrf2 attenuated the induction of Nrf2, HO-1 and NQO1 by SFN, enhanced TPA-induced colony formation and dampened the inhibitory effect of SFN on TPA-induced JB6 transformation. Epigenetics investigation using bisulfite genomic sequencing showed that SFN decreased the methylation ratio of the first 15 CpGs of the Nrf2 gene promoter, which was corroborated by increased Nrf2 mRNA expression. Furthermore, SFN strongly reduced the protein expression of DNA methyltransferases (DNMT1, DNMT3a and DNMT3b). SFN also inhibited the total histone deacetylase (HDAC) activity and decreased the protein expression of HDAC1, HDAC2, HDAC3 and HDAC4. Collectively, these results suggest that the anti-cancer effect of SFN against TPA-induced neoplastic transformation of mouse skin could involve the epigenetic reprogramming of anti-cancer genes such as Nrf2, leading to the epigenetic reactivation of Nrf2 and the subsequent induction of downstream target genes involved in cellular protection.


Asunto(s)
Anticarcinógenos/uso terapéutico , Transformación Celular Neoplásica/efectos de los fármacos , Epigénesis Genética/fisiología , Isotiocianatos/uso terapéutico , Factor 2 Relacionado con NF-E2/genética , Neoplasias Cutáneas/prevención & control , Animales , Carcinógenos , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Células Cultivadas , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor/efectos de los fármacos , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Sulfóxidos , Acetato de Tetradecanoilforbol
15.
Mol Carcinog ; 53(1): 77-84, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22911891

RESUMEN

Mutations in adenomatous polyposis coli (APC) gene are found in more than 80% of colorectal cancer (CRC) patients. The nuclear transcription factor Nrf2 plays a central role in the regulation of oxidative stress and inflammation. Previously, we have shown that chronic inflammation in Nrf2(-/-) (Nrf2 knockout; KO) mice resulted in higher expression of inflammatory markers and cytokines, coupled with higher inflammatory damage to the colonic crypt cells, as compared to the Nrf2(+/+) (wild type; WT) mice. Induction of mutation in the colon by administration of carcinogen, AOM prior to DSS-induced inflammation resulted in higher tumor incidence and numbers in Nrf2KO mice. These results indicate that Nrf2-dependent inhibition of inflammation appears to be critical in inhibiting mutation-initiated colorectal carcinogenesis. In this study, we aim to investigate if loss of Nrf2 would dose-dependently promote intestinal tumorigenesis in Apc(min/+) mice. To demonstrate the in vivo mechanisms, we constructed both Apc mutated and Nrf2 deficient strain Apc(min/+) mice with C57BL/6 Nrf2KO mice to obtain F1, Apc(min/+) ;Nrf2(+/-) and F2, Apc(min/+) ;Nrf2(-/-) mice. Nrf2KO decreased the protein expression of antioxidant enzyme NQO1 in Apc(min/+) . In contrast, Nrf2KO enhanced the expression of inflammatory markers such as COX-2, cPLA, LTB4 in Apc(min/+) . Finally, Nrf2KO resulted in higher level of PCNA and c-Myc expression in intestinal tissue, indicating the deficiency of Nrf2 promotes proliferation of intestinal crypt cells in Apc(min/+) . Taken together, our results suggest that Nrf2KO attenuates anti-oxidative stress pathway, induces inflammation, and increases proliferative potential in the intestinal crypts leading to enhanced intestinal carcinogenesis and adenomas in Apc(min/+) .


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Mucosa Intestinal/metabolismo , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/genética , Transducción de Señal , Animales , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Técnicas de Inactivación de Genes , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Pólipos Intestinales/genética , Pólipos Intestinales/patología , Intestinos/patología , Leucotrieno B4/metabolismo , Masculino , Ratones , Ratones Noqueados , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo
16.
Curr Drug Metab ; 14(6): 688-94, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23869812

RESUMEN

Cancer chemopreventive activities of various phytochemicals have been attributed to the modulation of xenobiotic disposition, which includes absorption, distribution, metabolism, and excretion. The interaction between xenobiotics and xenobiotic-metabolizing enzymes (XMEs) is bidirectional. XMEs are responsible for the biotransformation of xenobiotics such as bioactivation and detoxification. Conversely, xenobiotics affect XMEs through transcriptional regulation (induction or suppression) and post-translational interactions (inhibition or activation). Similar relationships also exist between xenobiotics and their transporters. Studies conducted over the past decade have demonstrated that the transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2), plays a critical role in the regulation of detoxifying enzymes and transporters through a signaling system that senses and responds to redox imbalance. The role of Nrf2 in the interaction between chemopreventive phytochemicals and detoxifying enzymes/transporters has become an important topic in cancer chemoprevention. In this review, the genetic and epigenetic factors that contribute to Nrf2-mediated regulation of detoxifying XMEs and transporters are discussed in the context of cancer chemoprevention. Phytochemicals may modulate the genome as well as epigenome, altering the regulation of XMEs and transporters, which may be critical for both cancer chemoprevention and the prevention of other oxidative stress- and inflammatory-related diseases, including cardiovascular, metabolic and neurological pathologies. The pharmacogenomic expression of XMEs and transporters, with an emphasis on both genomics and epigenetics, will also be discussed.


Asunto(s)
Anticarcinógenos/farmacología , Proteínas de Transporte de Membrana/genética , Factor 2 Relacionado con NF-E2/metabolismo , Oxidorreductasas/genética , Fitoquímicos/farmacología , Transferasas/genética , Animales , Epigenómica , Humanos , Proteínas de Transporte de Membrana/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/prevención & control , Oxidorreductasas/metabolismo , Polimorfismo Genético , Transferasas/metabolismo , Xenobióticos/metabolismo
17.
Biopharm Drug Dispos ; 34(6): 303-11, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23640758

RESUMEN

The root of Angelica sinensis (Oliv.) Diels (abbreviated as AS) (Danggui) has a long history in Asian herbal medicine. Recently, it was demonstrated that AS possesses anti-cancer and anti-oxidant activities. Because the transcription factor Nrf2 mediates the expression of many cellular anti-oxidative stress genes, including genes that are involved in phase II drug metabolism and anti-oxidative stress, this study sought to investigate whether pure compounds from AS or an AS extract could activate antioxidant response element (ARE)-mediated gene expression and induce anti-inflammatory activities. Z-Ligustilide (Ligu), 3-butylidenephthalide (Buty) and CO2 supercritical fluid-extracted lipophilic AS extract (SFE) were tested in HepG2-C8 cells stabilized with ARE luciferase reporter gene. Ligu and Buty caused significant toxicity only at 100 µm. All three samples induced ARE-luciferase activity; however, SFE at 8.5 µg/ml induced ARE-luciferase activity 2-3 fold more potently than did either of the pure compounds. SFE also significantly increased the endogenous mRNA of Nrf2 and the Nrf2 target anti-oxidative gene NAD(P)H dehydrogenase, quinone 1 (NQO1). The protein expression of NQO1 was also significantly induced by SFE. In RAW 264.7 cells, SFE suppressed lipopolysaccharide (LPS)-induced IL-1ß and TNF-α expression about 2 fold stronger than sulforaphane, whereas both pure compounds and SFE suppressed inflammatory nitric oxide (NO) production. In summary, this study demonstrates that AS has anti-inflammatory effects and activates the Nrf2 pathway, which protects against oxidative stress.


Asunto(s)
4-Butirolactona/análogos & derivados , Angelica sinensis , Antiinflamatorios/farmacología , Factor 2 Relacionado con NF-E2/genética , Anhídridos Ftálicos/farmacología , Extractos Vegetales/farmacología , 4-Butirolactona/farmacología , Animales , Línea Celular Tumoral , Expresión Génica , Células Hep G2 , Humanos , Interleucina-1beta/metabolismo , Ratones , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico/metabolismo , Fitoquímicos/farmacología , ARN Mensajero/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Factor de Necrosis Tumoral alfa/genética
18.
AAPS J ; 15(3): 864-74, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23658110

RESUMEN

3,3'-diindolylmethane (DIM) is currently being investigated in many clinical trials including prostate, breast, and cervical cancers and has been shown to possess anticancer effects in several in vivo and in vitro models. Previously, DIM has been reported to possess cancer chemopreventive effects in prostate carcinogenesis in TRAMP mice; however, the in vivo mechanism is unclear. The present study aims to investigate the in vitro and in vivo epigenetics modulation of DIM in TRAMP-C1 cells and in TRAMP mouse model. In vitro study utilizing TRAMP-C1 cells showed that DIM suppressed DNMT expression and reversed CpG methylation status of Nrf2 resulting in enhanced expression of Nrf2 and Nrf2-target gene NQO1. In vivo study, TRAMP mice fed with DIM-supplemented diet showed much lower incidence of tumorigenesis and metastasis than the untreated control group similar to what was reported previously. DIM increased apoptosis, decreased cell proliferation and enhanced Nrf2 and Nrf2-target gene NQO1 expression in prostate tissues. Importantly, immunohistochemical analysis showed that DIM reduced the global CpG 5-methylcytosine methylation. Focusing on one of the early cancer chemopreventive target gene Nrf2, bisulfite genomic sequencing showed that DIM decreased the methylation status of the first five CpGs of the Nrf2 promoter region, corroborating with the results of in vitro TRAMP-C1 cells. In summary, our current study shows that DIM is a potent cancer chemopreventive agent for prostate cancer and epigenetic modifications of the CpG including Nrf2 could be a potential mechanism by which DIM exerts its chemopreventive effects.


Asunto(s)
Anticarcinógenos/uso terapéutico , Epigénesis Genética/fisiología , Indoles/uso terapéutico , Factor 2 Relacionado con NF-E2/fisiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Animales , Anticarcinógenos/farmacología , Línea Celular Tumoral , Femenino , Indoles/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
19.
Biochem Pharmacol ; 85(9): 1398-404, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23416117

RESUMEN

Growing evidence suggests epigenetic alteration is involved during the development and progression of prostate cancer. Previously, we found Nrf2, a key regulator of cellular antioxidant defense systems, was silenced through epigenetic mechanism during tumorigenesis in vivo TRAMP mice and in vitro TRAMP C1 cells. Sulforaphane (SFN) in cruciferous vegetable has been demonstrated to be a potent cancer prevention agent for years. The aim of this study is to investigate the potential of SFN to restore Nrf2 expression in TRAMP C1 cells through epigenetic modifications. Bisulfite genomic sequencing results indicated that SFN treatment led to demethylation of the first 5 CpGs in the promoter region of the Nrf2 gene in TRAMP C1 cells. Using methylation DNA immunoprecipitation (MeDIP) assay, SFN significantly reduced the ratio of anti-mecyt antibody binding to the Nrf2 promoter containing the first 5 CpGs. SFN increased mRNA and protein expressions of Nrf2 and Nrf2 downstream target gene NQO-1. In addition, SFN decreased the protein levels of DNMT1 and DNMT3a. SFN treatment also attenuated the protein expression levels of HDACs 1, 4, 5, and 7 while increased the level of active chromatin marker acetyl-Histone 3 (Ac-H3). SFN treatments also increased chromatin-immunoprecipitated DNA of Nrf2 gene promoter using anti-Ac-H3 antibody. Taken together, our current study shows that SFN regulates Nrf2's CpGs demethylation and reactivation in TRAMP C1 cells, suggesting SFN may exert its chemopreventive effect in part via epigenetic modifications of Nrf2 gene with subsequent induction of its downstream anti-oxidative stress pathway.


Asunto(s)
Anticarcinógenos/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias de la Próstata/metabolismo , Tiocianatos/farmacología , Animales , Línea Celular Tumoral , Islas de CpG , Epigénesis Genética , Isotiocianatos , Masculino , Metilación , Ratones , Neoplasias de la Próstata/genética , Sulfóxidos
20.
Chem Res Toxicol ; 26(3): 477-85, 2013 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-23441843

RESUMEN

Cancer development has been linked to epigenetic modifications of cancer oncogenes and tumor suppressor genes; in advanced metastatic cancers, severe epigenetic modifications are present. We previously demonstrated that the progression of prostate tumors in TRAMP mice is associated with methylation silencing of the Nrf2 promoter and a reduced level of transcription of Nrf2 and Nrf2 target genes. Radix Angelicae Sinensis (RAS; Danggui) is a medicinal herb and health food supplement that has been widely used in Asia for centuries. Z-Ligustilide (Lig) is one of the bioactive components of RAS. We investigated the potential of Lig and RAS to restore Nrf2 gene expression through epigenetic modification in TRAMP C1 cells. Lig and RAS induced the mRNA and protein expression of endogenous Nrf2 and Nrf2 downstream target genes, such as HO-1, NQO1, and UGT1A1. Bisulfite genomic sequencing revealed that Lig and RAS treatment decreased the level of methylation of the first five CpGs of the Nrf2 promoter. A methylation DNA immunoprecipitation assay demonstrated that Lig and RAS significantly decreased the relative amount of methylated DNA in the Nrf2 gene promoter region. Lig and RAS also inhibited DNA methyltransferase activity in vitro. Collectively, these results suggest that Lig and RAS are able to demethylate the Nrf2 promoter CpGs, resulting in the re-expression of Nrf2 and Nrf2 target genes. Epigenetic modifications of genes, including Nrf2, may therefore contribute to the overall health benefits of RAS, including the anticancer effect of RAS and its bioactive component, Lig.


Asunto(s)
4-Butirolactona/análogos & derivados , Angelica sinensis/química , Antineoplásicos Fitogénicos/farmacología , Medicamentos Herbarios Chinos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Neoplasias de la Próstata/tratamiento farmacológico , 4-Butirolactona/química , 4-Butirolactona/farmacología , Animales , Antineoplásicos Fitogénicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Islas de CpG/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Medicamentos Herbarios Chinos/química , Epigénesis Genética/efectos de los fármacos , Masculino , Ratones , Regiones Promotoras Genéticas/efectos de los fármacos , Próstata/efectos de los fármacos , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...