Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 9(2)2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38060314

RESUMEN

Patients with cholangiocarcinoma have poor clinical outcomes due to late diagnoses, poor prognoses, and limited treatment strategies. To identify drug combinations for this disease, we have conducted a genome-wide CRISPR screen anchored on the bromodomain and extraterminal domain (BET) PROTAC degrader ARV825, from which we identified anticancer synergy when combined with genetic ablation of members of the mTOR pathway. This combination effect was validated using multiple pharmacological BET and mTOR inhibitors, accompanied by increased levels of apoptosis and cell cycle arrest. In a xenograft model, combined BET degradation and mTOR inhibition induced tumor regression. Mechanistically, the 2 inhibitor classes converged on H3K27ac-marked epigenetic suppression of the serine glycine one carbon (SGOC) metabolism pathway, including the key enzymes PHGDH and PSAT1. Knockdown of PSAT1 was sufficient to replicate synergy with single-agent inhibition of either BET or mTOR. Our results tie together epigenetic regulation, metabolism, and apoptosis induction as key therapeutic targets for further exploration in this underserved disease.


Asunto(s)
Colangiocarcinoma , Inhibidores mTOR , Humanos , Epigénesis Genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Línea Celular Tumoral , Serina-Treonina Quinasas TOR , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/genética
2.
J Invest Dermatol ; 143(9): 1779-1787.e1, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36871660

RESUMEN

Loss of protein expression of the tumor suppressor PTEN is associated with increased cancer aggressiveness, decreased tumor immune infiltration, and resistance to immune and targeted therapies in melanoma. We assessed a unique cohort of eight melanoma samples with focal loss of PTEN protein expression to understand the features and mechanisms of PTEN loss in this disease. We compared the PTEN-negative (PTEN[-]) areas to their adjacent PTEN-positive (PTEN[+]) areas using DNA sequencing, DNA methylation, RNA expression, digital spatial profiling, and immunohistochemical platforms. Variations or homozygous deletions of PTEN were identified in PTEN(-) areas that were not detected in the adjacent PTEN(+) areas in three cases (37.5%), but no clear genomic or DNA methylation basis for loss was identified in the remaining PTEN(-) samples. RNA expression data from two independent platforms identified a consistent increase in chromosome segregation gene expression in PTEN(-) versus adjacent PTEN(+) areas. Proteomic analysis showed a relative paucity of tumor-infiltrating lymphocytes in PTEN(-) versus adjacent PTEN(+) areas. The findings add to our understanding of potential molecular intratumoral heterogeneity in melanoma and the features associated with the loss of PTEN protein in this disease.


Asunto(s)
Melanoma , Fosfohidrolasa PTEN , Humanos , Fosfohidrolasa PTEN/genética , Proteómica , Melanoma/genética , Melanoma/patología , Genes Supresores de Tumor , ARN
3.
JCO Precis Oncol ; 6: e2100197, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35171660

RESUMEN

PURPOSE: IDH mutations occur in about 30% of patients with cholangiocarcinoma. Analysis of mutations in circulating tumor DNA (ctDNA) can be performed by droplet digital polymerase chain reaction (ddPCR). The analysis of ctDNA is a feasible approach to detect IDH mutations. METHODS: We isolated ctDNA from the blood of patients with IDH-mutated advanced cholangiocarcinoma collected at baseline, on therapy, and at progression to isocitrate dehydrogenase (IDH) inhibitors. RESULTS: Of 31 patients with IDH1R132 (n = 26) or IDH2R172 mutations (n = 5) in the tumor, IDH mutations were detected in 84% of ctDNA samples analyzed by ddPCR and in 83% of ctDNA samples analyzed by next-generation sequencing (NGS). Patients with a low variant allele frequency of ctDNA detected by NGS at baseline had a longer median time to treatment failure compared to patients with high variant allele frequency of ctDNA (3.6 v 1.5 months; P = .008). Patients with a decrease in IDH-mutated ctDNA on therapy by ddPCR compared with no change/increase had a trend to a longer median survival (P = .07). Most frequent emergent alterations in ctDNA by NGS at progression were ARID1A (n = 3) and TP53 mutations (n = 3). CONCLUSION: Detection of IDH mutations in ctDNA in patients with advanced cholangiocarcinoma is feasible, and dynamic changes in ctDNA can correspond with the clinical course and clonal evolution.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , ADN Tumoral Circulante , Inhibidores Enzimáticos , Isocitrato Deshidrogenasa , Neoplasias de los Conductos Biliares/sangre , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/enzimología , Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos , Colangiocarcinoma/sangre , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/enzimología , Colangiocarcinoma/genética , ADN Tumoral Circulante/sangre , ADN Tumoral Circulante/genética , Evolución Clonal , Inhibidores Enzimáticos/farmacología , Humanos , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Pronóstico
4.
Hepatology ; 75(2): 297-308, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34510503

RESUMEN

BACKGROUND AND AIMS: Cholangiocarcinoma (CCA) is a deadly and highly therapy-refractory cancer of the bile ducts, with early results from immune checkpoint blockade trials showing limited responses. Whereas recent molecular assessments have made bulk characterizations of immune profiles and their genomic correlates, spatial assessments may reveal actionable insights. APPROACH AND RESULTS: Here, we have integrated immune checkpoint-directed immunohistochemistry with next-generation sequencing of resected intrahepatic CCA samples from 96 patients. We found that both T-cell and immune checkpoint markers are enriched at the tumor margins compared to the tumor center. Using two approaches, we identify high programmed cell death protein 1 or lymphocyte-activation gene 3 and low CD3/CD4/inducible T-cell costimulator specifically in the tumor center as associated with poor survival. Moreover, loss-of-function BRCA1-associated protein-1 mutations are associated with and cause elevated expression of the immunosuppressive checkpoint marker, B7 homolog 4. CONCLUSIONS: This study provides a foundation on which to rationally improve and tailor immunotherapy approaches for this difficult-to-treat disease.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/genética , Antígenos B7/genética , Neoplasias de los Conductos Biliares/inmunología , Conductos Biliares Intrahepáticos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Linfocitos T CD4-Positivos , Línea Celular Tumoral , Colangiocarcinoma/inmunología , Femenino , Expresión Génica , Genes Supresores de Tumor , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunohistoquímica , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Mutación con Pérdida de Función , Masculino , Persona de Mediana Edad , Oncogenes/genética , Receptor de Muerte Celular Programada 1/genética , Tasa de Supervivencia , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Inhibidor 1 de la Activación de Células T con Dominio V-Set/genética , Adulto Joven , Proteína del Gen 3 de Activación de Linfocitos
5.
Cancer Discov ; 11(5): 1014-1015, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33947718

RESUMEN

Primary sclerosing cholangitis and colitis are known predisposing factors for bile duct cancer, but their exact pro-oncogenic mechanisms are unclear. In this issue of Cancer Discovery, Zhang and colleagues identify intestinal barrier impairment as a key mechanism, resulting in gut microbes spilling into the portal vein, in turn recruiting immunosuppressive myeloid-derived suppressor cells and promoting cholangiocarcinoma.See related article by Zhang et al. p. 1248.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Microbioma Gastrointestinal , Conductos Biliares Intrahepáticos , Humanos , Células Mieloides , Factores de Riesgo
6.
Mol Carcinog ; 58(9): 1680-1690, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31211467

RESUMEN

Treatment with vemurafenib, a potent and selective inhibitor of mitogen-activated protein kinase signaling downstream of the BRAFV600E oncogene, elicits dramatic clinical responses in patients with metastatic melanoma. Unfortunately, the clinical utility of this drug is limited by a high incidence of drug resistance. Thus, there is an unmet need for alternative therapeutic strategies to treat vemurafenib-resistant metastatic melanomas. We have conducted high-throughput screening of two bioactive compound libraries (Siga and Spectrum libraries) against a metastatic melanoma cell line (A2058) and identified two structurally analogous compounds, deguelin and rotenone, from a cell viability assay. Vemurafenib-resistant melanoma cell lines, A2058R and A375R (containing the BRAFV600E mutation), also showed reduced proliferation when treated with these two compounds. Deguelin, a mitochondrial complex I inhibitor, was noted to significantly inhibit oxygen consumption in cellular metabolism assays. Mechanistically, deguelin treatment rapidly activates AMPK signaling, which results in inhibition of mTORC1 signaling and differential phosphorylation of mTORC1's downstream effectors, 4E-BP1 and p70S6 kinase. Deguelin also significantly inhibited ERK activation and Ki67 expression without altering Akt activation in the same timeframe in the vemurafenib-resistant melanoma cells. These data posit that treatment with metabolic regulators, such as deguelin, can lead to energy starvation, thereby modulating the intracellular metabolic environment and reducing survival of drug-resistant melanomas harboring BRAF V600E mutations.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Rotenona/análogos & derivados , Vemurafenib/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Melanoma/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación/efectos de los fármacos , Rotenona/farmacología , Transducción de Señal/efectos de los fármacos
7.
Oncogene ; 37(19): 2481-2489, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29463860

RESUMEN

The study and comprehension of the molecular mechanisms underlying cancer biology strongly rely on mouse modeling. An ideal mouse model should have molecular, histopathological, and etiological characteristics as close as possible to those of the corresponding human tumors. Among solid tumors, colorectal cancer (CRC) is one of the malignancies that best suits reproduction in an animal model: it evolves through a progressive set of molecular events and is generally associated with a precise histopathology and a neat cataloging of stages and grades. The development of refined CRC mouse models over several decades has seen them recently evolve toward sophisticated systems that ever more closely approximate the human pathology, with different models addressing different human CRC subtypes. In particular, a metastatic CRC model has been seen as a "holy grail" in this field, and we describe in this review the path taken to achieve metastatic models and discuss the path forward.


Asunto(s)
Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Metástasis de la Neoplasia/patología , Animales , Carcinógenos/toxicidad , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Humanos , Ratones , Mutación , Metástasis de la Neoplasia/genética
8.
BMC Cancer ; 17(1): 736, 2017 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-29121869

RESUMEN

BACKGROUND: Understanding the underlying molecular mechanisms involved in the formation of cutaneous malignant melanoma is critical for improved diagnosis and treatment. Keratinocytic nuclear receptor Retinoid X Receptor α (RXRα) has a protective role against melanomagenesis and is involved in the regulation of keratinocyte and melanocyte homeostasis subsequent acute ultraviolet (UV) irradiation. METHODS: We generated a trigenic mouse model system (RXRα ep-/- | Tyr-NRAS Q61K | CDK4 R24C/R24C ) harboring an epidermal knockout of Retinoid X Receptor α (RXRα ep-/- ), combined with oncogenic NRAS Q61K (constitutively active RAS) and activated CDK4 R24C/R24C (constitutively active CDK4). Those mice were subjected to a single neonatal dose of UVB treatment and the role of RXR α was evaluated by characterizing the molecular and cellular changes that took place in the untreated and UVB treated trigenic RXRα ep-/- mice compared to the control mice with functional RXRα. RESULTS: Here we report that the trigenic mice develops spontaneous melanoma and exposure to a single neonatal UVB treatment reduces the tumor latency in those mice compared to control mice with functional RXRα. Melanomas from the trigenic RXRα ep-/- mice are substantial in size, show increased proliferation, exhibit increased expression of malignant melanoma markers and exhibit enhanced vascularization. Altered expression of several biomarkers including increased expression of activated AKT, p21 and cyclin D1 and reduced expression of pro-apoptotic marker BAX was observed in the tumor adjacent normal (TAN) skin of acute ultraviolet B treated trigenic RXRα ep-/- mice. Interestingly, we observed a significant increase in p21 and Cyclin D1 in the TAN skin of un-irradiated trigenic RXRα ep-/- mice, suggesting that those changes might be consequences of loss of functional RXRα in the melanoma microenvironment. Loss of RXRα in the epidermal keratinocytes in combination with oncogenic NRAS Q61K and CDK4 R24C/R24C mutations in trigenic mice led to significant melanoma invasion into the draining lymph nodes as compared to controls with functional RXRα. CONCLUSIONS: Our study demonstrates the protective role of keratinocytic RxRα in (1) suppressing spontaneous and acute UVB-induced melanoma, and (2) preventing progression of the melanoma to malignancy in the presence of driver mutations like activated CDK4 R24C/R24C and oncogenic NRAS Q61K .


Asunto(s)
Técnicas de Ablación/métodos , Quinasa 4 Dependiente de la Ciclina/metabolismo , GTP Fosfohidrolasas/metabolismo , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Receptor alfa X Retinoide/deficiencia , Neoplasias Cutáneas/metabolismo , Rayos Ultravioleta/efectos adversos , Enfermedad Aguda , Animales , Animales Recién Nacidos , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Masculino , Melanoma/etiología , Melanoma/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Melanoma Cutáneo Maligno
9.
Cell Rep ; 19(7): 1304-1312, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28514651

RESUMEN

Tumor evolution is an iterative process of selection for pro-oncogenic aberrations. This process can be accelerated by genomic instability, but how it interacts with different selection bottlenecks to shape the evolving genomic landscape remains understudied. Here, we assessed tumor initiation and therapy resistance bottlenecks in mouse models of melanoma, with or without genomic instability. At the initiation bottleneck, whole-exome sequencing revealed that drug-naive tumors were genomically silent, and this was surprisingly unaffected when genomic instability was introduced via telomerase inactivation. We hypothesize that the strong engineered alleles created low selection pressure. At the therapy resistance bottleneck, strong selective pressure was applied using a BRAF inhibitor. In the absence of genomic instability, tumors acquired a non-genomic drug resistance mechanism. By contrast, telomerase-deficient, drug-resistant melanomas acquired highly recurrent copy number gains. These proof-of-principle experiments demonstrate how different selection pressures can interact with genomic instability to impact tumor evolution.


Asunto(s)
Inestabilidad Genómica , Melanoma/genética , Animales , Variaciones en el Número de Copia de ADN/genética , Modelos Animales de Enfermedad , Ingeniería Genética , Ratones , Telomerasa/metabolismo
11.
J Control Release ; 220(Pt A): 503-514, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26578440

RESUMEN

Metastatic melanoma has a high mortality rate due to lymphatic progression of the disease. Current treatment is surgery followed by radiation and intravenous chemotherapy. However, drawbacks for current chemotherapeutics lie in the fact that they develop resistance and do not achieve therapeutic concentrations in the lymphatic system. We hypothesize that a three-drug nanoscale drug delivery system, tailored for lymphatic uptake, administered subcutaneously, will have decreased drug resistance and therefore offer better therapeutic outcomes. We prepared and characterized nanoparticles (NPs) with docetaxel, everolimus, and LY294002 in polyethyleneglycol-block-poly(ε-caprolactone) (PEG-PCL) polymer with different charge distributions by modifying the ratio of anionic and neutral end groups on the PEG block. These NPs are similarly sized (~48 nm), with neutral, partially charged, or fully charged surface. The NPs are able to load ~2mg/mL of each drug and are stable for 24h. The NPs are assessed for safety and efficacy in two transgenic metastatic melanoma mouse models. All the NPs were safe in both models based on general appearance, weight changes, death, and blood biochemical analyses. The partially charged NPs are most effective in decreasing the number of melanocytes at both the proximal (sentinel) lymph node (LN) and the distal LN from the injection site. The neutral NPs are efficacious at the proximal LN, while the fully charged NPs have no effect on either LNs. Thus, our data indicates that the NP surface charge and lymphatic efficacy are closely tied to each other and the partially charged NPs have the highest potential in treating metastatic melanoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cromonas/administración & dosificación , Portadores de Fármacos , Everolimus/administración & dosificación , Lactonas/química , Ganglios Linfáticos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Morfolinas/administración & dosificación , Nanopartículas , Polietilenglicoles/química , Neoplasias Cutáneas/tratamiento farmacológico , Taxoides/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromonas/química , Cromonas/metabolismo , Docetaxel , Composición de Medicamentos , Resistencia a Antineoplásicos , Everolimus/química , Everolimus/metabolismo , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Melanocitos/efectos de los fármacos , Melanocitos/patología , Melanoma/genética , Melanoma/metabolismo , Melanoma/secundario , Ratones Transgénicos , Morfolinas/química , Morfolinas/metabolismo , Nanotecnología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Solubilidad , Propiedades de Superficie , Taxoides/química , Taxoides/metabolismo , Factores de Tiempo , Carga Tumoral/efectos de los fármacos
12.
Mol Cancer Res ; 13(1): 186-96, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25189354

RESUMEN

UNLABELLED: Understanding the molecular mechanisms behind formation of melanoma, the deadliest form of skin cancer, is crucial for improved diagnosis and treatment. One key is to better understand the cross-talk between epidermal keratinocytes and pigment-producing melanocytes. Here, using a bigenic mouse model system combining mutant oncogenic NRAS(Q61K) (constitutively active RAS) or mutant activated CDK4(R24C/R24C) (prevents binding of CDK4 by kinase inhibitor p16(INK4A)) with an epidermis-specific knockout of the nuclear retinoid X receptor alpha (RXRα(ep-/-)) results in increased melanoma formation after chronic ultraviolet-B (UVB) irradiation compared with control mice with functional RXRα. Melanomas from both groups of bigenic RXRα(ep-/-) mice are larger in size with higher proliferative capacity, and exhibit enhanced angiogenic properties and increased expression of malignant melanoma markers. Analysis of tumor adjacent normal skin from these mice revealed altered expression of several biomarkers indicative of enhanced melanoma susceptibility, including reduced expression of tumor suppressor p53 and loss of PTEN, with concomitant increase in activated AKT. Loss of epidermal RXRα in combination with UVB significantly enhances invasion of melanocytic cells to draining lymph nodes in bigenic mice expressing oncogenic NRAS(Q61K) compared with controls with functional RXRα. These results suggest a crucial role of keratinocytic RXRα to suppress formation of UVB-induced melanomas and their progression to malignant cancers in the context of driver mutations such as activated CDK4(R24C/R24C) or oncogenic NRAS(Q61K). IMPLICATIONS: These findings suggest that RXRα may serve as a clinical diagnostic marker and therapeutic target in melanoma progression and metastasis.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , GTP Fosfohidrolasas/genética , Melanoma/genética , Proteínas de la Membrana/genética , Fosfohidrolasa PTEN/biosíntesis , Receptor alfa X Retinoide/genética , Proteína p53 Supresora de Tumor/biosíntesis , Animales , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Melanoma/patología , Ratones , Mutación , Microambiente Tumoral/genética , Microambiente Tumoral/efectos de la radiación , Rayos Ultravioleta
13.
PLoS Genet ; 10(5): e1004321, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24810760

RESUMEN

Understanding the molecular mechanisms of ultraviolet (UV) induced melanoma formation is becoming crucial with more reported cases each year. Expression of type II nuclear receptor Retinoid-X-Receptor α (RXRα) is lost during melanoma progression in humans. Here, we observed that in mice with melanocyte-specific ablation of RXRα and RXRß, melanocytes attract fewer IFN-γ secreting immune cells than in wild-type mice following acute UVR exposure, via altered expression of several chemoattractive and chemorepulsive chemokines/cytokines. Reduced IFN-γ in the microenvironment alters UVR-induced apoptosis, and due to this, the survival of surrounding dermal fibroblasts is significantly decreased in mice lacking RXRα/ß. Interestingly, post-UVR survival of the melanocytes themselves is enhanced in the absence of RXRα/ß. Loss of RXRs α/ß specifically in the melanocytes results in an endogenous shift in homeostasis of pro- and anti-apoptotic genes in these cells and enhances their survival compared to the wild type melanocytes. Therefore, RXRs modulate post-UVR survival of dermal fibroblasts in a "non-cell autonomous" manner, underscoring their role in immune surveillance, while independently mediating post-UVR melanocyte survival in a "cell autonomous" manner. Our results emphasize a novel immunomodulatory role of melanocytes in controlling survival of neighboring cell types besides controlling their own, and identifies RXRs as potential targets for therapy against UV induced melanoma.


Asunto(s)
Ciclo Celular/efectos de la radiación , Inmunidad Innata/fisiología , Melanocitos/fisiología , Receptor alfa X Retinoide/fisiología , Receptor beta X Retinoide/fisiología , Rayos Ultravioleta , Animales , Melanocitos/efectos de la radiación , Ratones , Ratones Transgénicos , Receptor alfa X Retinoide/genética , Receptor beta X Retinoide/genética
14.
Br J Nutr ; 107(4): 473-84, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21849094

RESUMEN

It is appreciated far and wide that increased and regular consumption of fruits and vegetables is linked with noteworthy anticancer benefits. Extensively consumed as a spice in foods and beverages worldwide, ginger (Zingiber officinale Roscoe) is an excellent source of several bioactive phenolics, including non-volatile pungent compounds such as gingerols, paradols, shogaols and gingerones. Ginger has been known to display anti-inflammatory, antioxidant and antiproliferative activities, indicating its promising role as a chemopreventive agent. Here, we show that whole ginger extract (GE) exerts significant growth-inhibitory and death-inductory effects in a spectrum of prostate cancer cells. Comprehensive studies have confirmed that GE perturbed cell-cycle progression, impaired reproductive capacity, modulated cell-cycle and apoptosis regulatory molecules and induced a caspase-driven, mitochondrially mediated apoptosis in human prostate cancer cells. Remarkably, daily oral feeding of 100 mg/kg body weight of GE inhibited growth and progression of PC-3 xenografts by approximately 56 % in nude mice, as shown by measurements of tumour volume. Tumour tissue from GE-treated mice showed reduced proliferation index and widespread apoptosis compared with controls, as determined by immunoblotting and immunohistochemical methods. Most importantly, GE did not exert any detectable toxicity in normal, rapidly dividing tissues such as gut and bone marrow. To the best of our knowledge, this is the first report to demonstrate the in vitro and in vivo anticancer activity of whole GE for the management of prostate cancer.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Suplementos Dietéticos , Extractos Vegetales/uso terapéutico , Neoplasias de la Próstata/dietoterapia , Rizoma/química , Zingiber officinale/química , Animales , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/metabolismo , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Suplementos Dietéticos/efectos adversos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Extractos Vegetales/efectos adversos , Extractos Vegetales/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/prevención & control , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...