Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nature ; 529(7586): 413-417, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26735014

RESUMEN

Triple-negative breast cancer (TNBC) is a heterogeneous and clinically aggressive disease for which there is no targeted therapy. BET bromodomain inhibitors, which have shown efficacy in several models of cancer, have not been evaluated in TNBC. These inhibitors displace BET bromodomain proteins such as BRD4 from chromatin by competing with their acetyl-lysine recognition modules, leading to inhibition of oncogenic transcriptional programs. Here we report the preferential sensitivity of TNBCs to BET bromodomain inhibition in vitro and in vivo, establishing a rationale for clinical investigation and further motivation to understand mechanisms of resistance. In paired cell lines selected for acquired resistance to BET inhibition from previously sensitive TNBCs, we failed to identify gatekeeper mutations, new driver events or drug pump activation. BET-resistant TNBC cells remain dependent on wild-type BRD4, which supports transcription and cell proliferation in a bromodomain-independent manner. Proteomic studies of resistant TNBC identify strong association with MED1 and hyper-phosphorylation of BRD4 attributable to decreased activity of PP2A, identified here as a principal BRD4 serine phosphatase. Together, these studies provide a rationale for BET inhibition in TNBC and present mechanism-based combination strategies to anticipate clinical drug resistance.


Asunto(s)
Azepinas/farmacología , Azepinas/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Nucleares/antagonistas & inhibidores , Estructura Terciaria de Proteína/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Triazoles/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Unión Competitiva/efectos de los fármacos , Quinasa de la Caseína II/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Resistencia a Antineoplásicos/genética , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano/efectos de los fármacos , Genoma Humano/genética , Humanos , Subunidad 1 del Complejo Mediador/metabolismo , Ratones , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Proteína Fosfatasa 2/metabolismo , Proteómica , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
PLoS Genet ; 15(3): e1008002, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30893315

RESUMEN

Mammary epithelial progenitors are the normal cell-of-origin of breast cancer. We previously defined a population of p27+ quiescent hormone-responsive progenitor cells in the normal human breast whose frequency associates with breast cancer risk. Here, we describe that deletion of the Cdkn1b gene encoding the p27 cyclin-dependent kinase inhibitor in the estrogen-induced mammary tumor-susceptible ACI rat strain leads to a decrease in the relative frequencies of Cd49b+ mammary luminal epithelial progenitors and pregnancy-related differentiation. We show by comprehensive gene expression profiling of purified progenitor and differentiated mammary epithelial cell populations that p27 deletion has the most pronounced effects on luminal progenitors. Cdkn1b-/- females have decreased fertility, but rats that are able to get pregnant had normal litter size and were able to nurse their pups implying that loss of p27 in ACI rats does not completely abrogate ovarian function and lactation. Reciprocal mammary gland transplantation experiments indicate that the p27-loss-induced changes in mammary epithelial cells are not only caused by alterations in their intrinsic properties, but are likely due to altered hormonal signaling triggered by the perturbed systemic endocrine environment observed in Cdkn1b-/- females. We also observed a decrease in the frequency of mammary epithelial cells positive for progesterone receptor (Pr) and FoxA1, known direct transcriptional targets of the estrogen receptor (Erα), and an increase in phospho-Stat5 positive cells commonly induced by prolactin (Prl). Characterization of genome-wide Pr chromatin binding revealed distinct binding patterns in mammary epithelial cells of Cdkn1b+/+ and Cdkn1b-/- females and enrichment in genes with known roles in Notch, ErbB, leptin, and Erα signaling and regulation of G1-S transition. Our data support a role for p27 in regulating the pool size of hormone-responsive luminal progenitors that could impact breast cancer risk.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Animales , Animales Modificados Genéticamente/genética , Neoplasias de la Mama/genética , Diferenciación Celular , Proliferación Celular/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Endocrinas/fisiología , Células Epiteliales , Receptor alfa de Estrógeno , Estrógenos , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Integrina alfa1 , Glándulas Mamarias Animales , Glándulas Mamarias Humanas/crecimiento & desarrollo , Embarazo , Progesterona , Ratas , Ratas Endogámicas ACI , Ratas Sprague-Dawley , Receptores de Estrógenos , Receptores de Progesterona , Factores de Riesgo , Transducción de Señal , Células Madre
3.
J Am Chem Soc ; 143(12): 4714-4724, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33739832

RESUMEN

Prodrugs engineered for preferential activation in diseased versus normal tissues offer immense potential to improve the therapeutic indexes (TIs) of preclinical and clinical-stage active pharmaceutical ingredients that either cannot be developed otherwise or whose efficacy or tolerability it is highly desirable to improve. Such approaches, however, often suffer from trial-and-error design, precluding predictive synthesis and optimization. Here, using bromodomain and extra-terminal (BET) protein inhibitors (BETi)-a class of epigenetic regulators with proven anticancer potential but clinical development hindered in large part by narrow TIs-we introduce a macromolecular prodrug platform that overcomes these challenges. Through tuning of traceless linkers appended to a "bottlebrush prodrug" scaffold, we demonstrate correlation of in vitro prodrug activation kinetics with in vivo tumor pharmacokinetics, enabling the predictive design of novel BETi prodrugs with enhanced antitumor efficacies and devoid of dose-limiting toxicities in a syngeneic triple-negative breast cancer murine model. This work may have immediate clinical implications, introducing a platform for predictive prodrug design and potentially overcoming hurdles in drug development.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Profármacos/farmacología , Proteínas/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Sustancias Macromoleculares/síntesis química , Sustancias Macromoleculares/química , Sustancias Macromoleculares/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Estructura Molecular , Profármacos/síntesis química , Profármacos/química , Proteínas/metabolismo
4.
J Biol Chem ; 289(25): 17620-33, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24802759

RESUMEN

The JmjC domain-containing H3K4 histone demethylase jumonji AT-rich interactive domain 1B (JARID1B) (also known as KDM5B and PLU1) is overexpressed in breast cancer and is a potential target for breast cancer treatment. To investigate the in vivo function of JARID1B, we developed Jarid1b(-/-) mice and characterized their phenotypes in detail. Unlike previously reported Jarid1b(-/-) strains, the majority of these Jarid1b(-/-) mice were viable beyond embryonic and neonatal stages. This allowed us to further examine phenotypes associated with the loss of JARID1B in pubertal development and pregnancy. These Jarid1b(-/-) mice exhibited decreased body weight, premature mortality, decreased female fertility, and delayed mammary gland development. Related to these phenotypes, JARID1B loss decreased serum estrogen level and reduced mammary epithelial cell proliferation in early puberty. In mammary epithelial cells, JARID1B loss diminished the expression of key regulators for mammary morphogenesis and luminal lineage specification, including FOXA1 and estrogen receptor α. Mechanistically, JARID1B was required for GATA3 recruitment to the Foxa1 promoter to activate Foxa1 expression. These results indicate that JARID1B positively regulates mammary ductal development through both extrinsic and cell-autonomous mechanisms.


Asunto(s)
Linaje de la Célula/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Glándulas Mamarias Animales/embriología , Organogénesis/fisiología , Animales , Línea Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Femenino , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Glándulas Mamarias Animales/citología , Ratones , Ratones Noqueados , Embarazo
5.
Am J Pathol ; 176(6): 2948-57, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20431030

RESUMEN

Expression of macrophage inhibitory cytokine-1 (MIC-1), a member of the transforming growth factor-beta family, normally increases during inflammation or organ injury. MIC-1 is also expressed at higher levels in melanomas; however, its role in tumorigenesis is unknown. This report identifies a novel function for MIC-1 in cancer. MIC-1 was overexpressed in approximately 67% of advanced melanomas, accompanied by fivefold to six-fold higher levels of secreted protein in serum of melanoma patients compared with normal individuals. Constitutively active mutant (V600E)B-Raf in melanoma regulated downstream MIC-1 expression. Indeed, small-interfering RNA-mediated targeting of MIC-1 or (V600E)B-Raf reduced expression and secretion by three-fold to fivefold. This decrease in MIC-1 levels reduced melanoma tumorigenesis by approximately threefold, but did not alter cultured cell growth, suggesting a unique function other than growth control. Instead, inhibition of MIC-1 was found to mechanistically retard melanoma tumor vascular development, subsequently affecting tumor cell proliferation and apoptosis. This role in melanoma angiogenesis was confirmed by comparing MIC-1 and vascular endothelial growth factor (VEGF) function in chick chorioallantoic membrane and matrigel plug assays. Similar to VEGF in melanomas, MIC-1 stimulated directional vessel development, acting as a potent angiogenic factor. Thus, MIC-1 is secreted from melanoma cells together with VEGF to promote vascular development mediated by (V600E)B-Raf signaling.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/metabolismo , Melanoma/irrigación sanguínea , Melanoma/patología , Neovascularización Patológica , Animales , Línea Celular Tumoral , Embrión de Pollo , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Melanoma/metabolismo , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Clin Cancer Res ; 15(5): 1674-85, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19208796

RESUMEN

PURPOSE: Melanoma is the most invasive and deadly form of skin cancer. Few agents are available for treating advanced disease to enable long-term patient survival, which is driving the search for new compounds inhibiting deregulated pathways causing melanoma. Akt3 is an important target in melanomas because its activity is increased in approximately 70% of tumors, decreasing apoptosis in order to promote tumorigenesis. EXPERIMENTAL DESIGN: Because naturally occurring products can be effective anticancer agents, a library was screened to identify Akt3 pathway inhibitors. Isothiocyanates were identified as candidates, but low potency requiring high concentrations for therapeutic efficacy made them unsuitable. Therefore, more potent analogs called isoselenocyanates were created using the isothiocyanate backbone but increasing the alkyl chain length and replacing sulfur with selenium. Efficacy was measured on cultured cells and tumors by quantifying proliferation, apoptosis, toxicity, and Akt3 pathway inhibition. RESULTS: Isoselenocyanates significantly decreased Akt3 signaling in cultured melanoma cells and tumors. Compounds having 4 to 6 carbon alkyl side chains with selenium substituted for sulfur, called ISC-4 and ISC-6, respectively, decreased tumor development by approximately 60% compared with the corresponding isothiocyanates, which had no effect. No changes in animal body weight or in blood parameters indicative of liver-, kidney-, or cardiac-related toxicity were observed with isoselenocyanates. Mechanistically, isoselenocyanates ISC-4 and ISC-6 decreased melanoma tumorigenesis by causing an approximately 3-fold increase in apoptosis. CONCLUSIONS: Synthetic isoselenocyanates are therapeutically effective for inhibiting melanoma tumor development by targeting Akt3 signaling to increase apoptosis in melanoma cells with negligible associated systemic toxicity.


Asunto(s)
Isocianatos/química , Melanoma Experimental/tratamiento farmacológico , Compuestos de Organoselenio/química , Compuestos de Organoselenio/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Peso Corporal/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Factores de Transcripción Forkhead/fisiología , Humanos , Isocianatos/síntesis química , Isocianatos/farmacología , Melanoma Experimental/metabolismo , Melanoma Experimental/secundario , Ratones , Ratones Desnudos , Compuestos de Organoselenio/síntesis química , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/farmacología
7.
Mol Cancer Ther ; 7(5): 1297-308, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18483317

RESUMEN

Malignant melanoma is the most deadly form of skin cancer due to its highly metastatic nature. Untargeted therapies are ineffective for treating metastatic disease, leading to the development of agents specifically inhibiting proteins or pathways deregulated in melanoma. The deregulation of inducible nitric oxide synthase (iNOS) is one such event occurring in melanoma, and is correlated with poor survival. Current iNOS inhibitors, such as PBIT [S,S'-1,4-phenylenebis(1,2-ethanediyl)bis-isothiourea], require high concentrations for clinical efficacy causing systemic toxicity. To develop more potent agents effective at significantly lower concentrations, a novel isosteric analogue of PBIT was synthesized, called PBISe [S,S'-1,4-phenylenebis(1,2-ethanediyl)bis-isoselenourea], in which sulfur was replaced with selenium. PBISe kills melanoma cells >10-fold more effectively than PBIT, and cultured cancer cells are 2- to 5-fold more sensitive than normal cells. Like PBIT, PBISe targets iNOS but also has new inhibitory properties acting as an Akt3 pathway inhibitor and mitogen-activated protein kinase (MAPK) cascade activator, which causes decreased cancer cell proliferation and increased apoptosis. Inhibition of cellular proliferation mediated by PBISe induced a G2-M phase cell cycle block linked to excessively high MAPK activity causing decreased cyclin D1 and increased p21 as well as p27 levels. PBISe promotes apoptosis by inhibiting Akt3 signaling, elevating cleaved caspase-3 and PARP levels. Compared with PBIT, PBISe reduced tumor development by 30% to 50% in mice inducing a 2-fold increase in apoptosis with negligible associated systemic toxicity. Collectively, these results suggest that PBISe is a potent chemotherapeutic agent with novel properties enabling the targeting of iNOS, Akt3, and MAPK signaling, thereby promoting melanoma cell apoptosis and inhibition of proliferation.


Asunto(s)
Antineoplásicos/uso terapéutico , Melanoma/tratamiento farmacológico , Compuestos de Organoselenio/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Urea/análogos & derivados , Animales , Apoptosis , Células CACO-2 , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/metabolismo , Ratones , Ratones Desnudos , Compuestos de Organoselenio/síntesis química , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias Cutáneas/metabolismo , Tiourea/análogos & derivados , Tiourea/química , Tiourea/farmacología , Factores de Tiempo , Urea/síntesis química , Urea/uso terapéutico
8.
Nat Commun ; 10(1): 4182, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31519911

RESUMEN

Myoepithelial cells play key roles in normal mammary gland development and in limiting pre-invasive to invasive breast tumor progression, yet their differentiation and perturbation in ductal carcinoma in situ (DCIS) are poorly understood. Here, we investigated myoepithelial cells in normal breast tissues of BRCA1 and BRCA2 germline mutation carriers and in non-carrier controls, and in sporadic DCIS. We found that in the normal breast of non-carriers, myoepithelial cells frequently co-express the p63 and TCF7 transcription factors and that p63 and TCF7 show overlapping chromatin peaks associated with differentiated myoepithelium-specific genes. In contrast, in normal breast tissues of BRCA1 mutation carriers the frequency of p63+TCF7+ myoepithelial cells is significantly decreased and p63 and TCF7 chromatin peaks do not overlap. These myoepithelial perturbations in normal breast tissues of BRCA1 germline mutation carriers may play a role in their higher risk of breast cancer. The fraction of p63+TCF7+ myoepithelial cells is also significantly decreased in DCIS, which may be associated with invasive progression.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Carcinoma Ductal de Mama/metabolismo , Mutación/genética , Animales , Proteína BRCA1/genética , Proteína BRCA2/genética , Carcinoma Ductal de Mama/genética , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Mutación de Línea Germinal/genética , Humanos , Inmunohistoquímica , Ratones , Factor 1 de Transcripción de Linfocitos T/genética , Factor 1 de Transcripción de Linfocitos T/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
9.
Nat Biomed Eng ; 2(11): 822-830, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30918745

RESUMEN

At present there are no drugs for the treatment of chronic liver fibrosis that have been approved by the Food and Drug administration of the United States. Telmisartan, a small-molecule antihypertensive drug, displays antifibrotic activity, but its clinical use is limited because it causes systemic hypotension. Here, we report the scalable and convergent synthesis of macromolecular telmisartan prodrugs optimized for preferential release in diseased liver tissue. We optimized the release of active telmisartan in fibrotic liver to be depot-like (that is, a constant therapeutic concentration) through the molecular design of telmisartan brush-arm star polymers, and show that these lead to improved efficacy and to the avoidance of dose-limiting hypotension in both metabolically and chemically induced mouse models of hepatic fibrosis, as determined by histopathology, enzyme levels in the liver, intact-tissue protein markers, hepatocyte necrosis protection, and gene-expression analyses. In rats and dogs, the prodrugs are retained long-term in liver tissue and have a well-tolerated safety profile. Our findings support the further development of telmisartan prodrugs that enable infrequent dosing in the treatment of liver fibrosis.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Diseño de Fármacos , Cirrosis Hepática/tratamiento farmacológico , Profármacos/uso terapéutico , Telmisartán/uso terapéutico , Bloqueadores del Receptor Tipo 1 de Angiotensina II/química , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Animales , Tetracloruro de Carbono/toxicidad , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Semivida , Hígado/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Polímeros/química , Profármacos/química , Profármacos/farmacocinética , Ratas , Ratas Sprague-Dawley , Telmisartán/química
10.
Nat Biomed Eng ; 2(9): 707, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31015683

RESUMEN

In the version of this Article originally published, the author Peter Blume-Jensen was not denoted as a corresponding author; this has now been amended and the author's email address has been added. The 'Correspondence and requests for materials' statement was similarly affected and has now been updated with the author's initials 'P.B-J.'

11.
Cancer Discov ; 7(10): 1098-1115, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28652380

RESUMEN

To investigate immune escape during breast tumor progression, we analyzed the composition of leukocytes in normal breast tissues, ductal carcinoma in situ (DCIS), and invasive ductal carcinomas (IDC). We found significant tissue and tumor subtype-specific differences in multiple cell types including T cells and neutrophils. Gene expression profiling of CD45+CD3+ T cells demonstrated a decrease in CD8+ signatures in IDCs. Immunofluorescence analysis showed fewer activated GZMB+CD8+ T cells in IDC than in DCIS, including in matched DCIS and recurrent IDC. T-cell receptor clonotype diversity was significantly higher in DCIS than in IDCs. Immune checkpoint protein TIGIT-expressing T cells were more frequent in DCIS, whereas high PD-L1 expression and amplification of CD274 (encoding PD-L1) was only detected in triple-negative IDCs. Coamplification of a 17q12 chemokine cluster with ERBB2 subdivided HER2+ breast tumors into immunologically and clinically distinct subtypes. Our results show coevolution of cancer cells and the immune microenvironment during tumor progression.Significance: The design of effective cancer immunotherapies requires the understanding of mechanisms underlying immune escape during tumor progression. Here we demonstrate a switch to a less active tumor immune environment during the in situ to invasive breast carcinoma transition, and identify immune regulators and genomic alterations that shape tumor evolution. Cancer Discov; 7(10); 1098-115. ©2017 AACR.See related commentary by Speiser and Verdeil, p. 1062This article is highlighted in the In This Issue feature, p. 1047.


Asunto(s)
Neoplasias de la Mama/inmunología , Carcinoma Ductal de Mama/inmunología , Carcinoma Intraductal no Infiltrante/inmunología , Perfilación de la Expresión Génica/métodos , Linfocitos T/inmunología , Antígeno B7-H1/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Complejo CD3/genética , Carcinoma Ductal de Mama/genética , Carcinoma Intraductal no Infiltrante/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Antígenos Comunes de Leucocito/genética , Receptor ErbB-2/genética , Microambiente Tumoral
12.
Cancer Res ; 76(7): 1926-34, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-26941287

RESUMEN

The frequency and proliferative activity of tissue-specific stem and progenitor cells are suggested to correlate with cancer risk. In this study, we investigated the association between breast cancer risk and the frequency of mammary epithelial cells expressing p27, estrogen receptor (ER), and Ki67 in normal breast tissue. We performed a nested case-control study of 302 women (69 breast cancer cases, 233 controls) who had been initially diagnosed with benign breast disease according to the Nurses' Health Studies. Immunofluorescence for p27, ER, and Ki67 was performed on tissue microarrays constructed from benign biopsies containing normal mammary epithelium and scored by computational image analysis. We found that the frequency of Ki67(+) cells was positively associated with breast cancer risk among premenopausal women [OR = 10.1, 95% confidence interval (CI) = 2.12-48.0]. Conversely, the frequency of ER(+) or p27(+) cells was inversely, but not significantly, associated with subsequent breast cancer risk (ER(+): OR = 0.70, 95% CI, 0.33-1.50; p27(+): OR = 0.89, 95% CI, 0.45-1.75). Notably, high Ki67(+)/low p27(+) and high Ki67(+)/low ER(+) cell frequencies were significantly associated with a 5-fold higher risk of breast cancer compared with low Ki67(+)/low p27(+) and low Ki67(+)/low ER(+) cell frequencies, respectively, among premenopausal women (Ki67(hi)/p27(lo): OR = 5.08, 95% CI, 1.43-18.1; Ki67(hi)/ER(lo): OR = 4.68, 95% CI, 1.63-13.5). Taken together, our data suggest that the fraction of actively cycling cells in normal breast tissue may represent a marker for breast cancer risk assessment, which may therefore impact the frequency of screening procedures in at-risk women. Cancer Res; 76(7); 1926-34. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/etiología , Células Epiteliales/patología , Glándulas Mamarias Humanas/patología , Adulto , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Femenino , Humanos , Premenopausia
13.
Stem Cell Reports ; 4(2): 297-311, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25619437

RESUMEN

Postnatal mammary gland development and differentiation occur during puberty and pregnancy. To explore the role of DNA methylation in these processes, we determined the genome-wide DNA methylation and gene expression profiles of CD24(+)CD61(+)CD29(hi), CD24(+)CD61(+)CD29(lo), and CD24(+)CD61(-)CD29(lo) cell populations that were previously associated with distinct biological properties at different ages and reproductive stages. We found that pregnancy had the most significant effects on CD24(+)CD61(+)CD29(hi) and CD24(+)CD61(+)CD29(lo) cells, inducing distinct epigenetic states that were maintained through life. Integrated analysis of gene expression, DNA methylation, and histone modification profiles revealed cell-type- and reproductive-stage-specific changes. We identified p27 and TGFß signaling as key regulators of CD24(+)CD61(+)CD29(lo) cell proliferation, based on their expression patterns and results from mammary gland explant cultures. Our results suggest that relatively minor changes in DNA methylation occur during luminal differentiation compared with the effects of pregnancy on CD24(+)CD61(+)CD29(hi) and CD24(+)CD61(+)CD29(lo) cells.


Asunto(s)
Metilación de ADN , Células Epiteliales/citología , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/citología , Factores de Edad , Animales , Antígenos de Superficie/metabolismo , Diferenciación Celular/genética , Análisis por Conglomerados , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Elementos de Facilitación Genéticos , Activación Enzimática , Epigénesis Genética , Células Epiteliales/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histonas/metabolismo , Inmunofenotipificación , Ratones , Ratones Noqueados , Especificidad de Órganos/genética , Fenotipo , Embarazo , Regiones Promotoras Genéticas , Maduración Sexual/genética , Transducción de Señal
14.
Immunol Lett ; 90(2-3): 93-6, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14687710

RESUMEN

Chronic rheumatoid arthritis (RA) is characterized by the hyperplasia of synovial tissue, which results from dysregulation of proliferative and antiapoptotic signals transduced in the synovial cells by unknown mechanisms. To identify candidate factors involved in the regulation of synovial hyperplasia, the expression profile of 205 apoptosis-related genes was compared between tissues from patients with RA and osteoarthritis (OA) using a cDNA microarray. Upregulated genes in the RA synovium included TNFR2, FLICE2, and signaling molecules involved in a MAP kinase pathway (GRB2, MAPK p38). In contrast, genes encoding SARP1 and various cell cycle regulators were down-regulated in the RA synovium relative to OA. Importantly, the expression levels of GRB2 and FLICE2 genes were remarkably enhanced in RA synoviocytes but not in OA synoviocytes in response to tumor necrosis factor (TNF)-alpha treatment. Thus, these results suggest that over-expression of GRB2 and FLICE2 in RA synovium is caused by TNF-alpha inducibility differentially regulated in RA synoviocytes and provide potential pathogenic roles of these genes in the hyperplasia of the RA synovium.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Artritis Reumatoide/genética , Caspasas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas/genética , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Caspasa 10 , Células Cultivadas , Proteína Adaptadora GRB2 , Perfilación de la Expresión Génica , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Membrana Sinovial/patología
15.
Cancer Prev Res (Phila) ; 4(2): 248-58, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21097713

RESUMEN

Melanoma incidence and mortality rates continue to increase despite the use of sunscreen as well as screening programs for early surgical excision of premalignant lesions. The steady increase in melanoma incidence suggests that additional preventive approaches are needed to augment these existing strategies. One unexplored area involves targeting genes whose deregulation promotes disease development to prevent melanoma. The Akt3 signaling pathway is one key signaling cascade that plays a central role by deregulating apoptosis to promote development of approximately 70% of melanomas. Isoselenocyanate-4 (ISC-4), derived from isothiocyanates by increasing the alkyl chain length and replacing sulfur with selenium, has been developed to target this important signaling pathway in melanomas; however, its chemopreventive potential is unknown. In this study, the chemopreventive efficacy of topical ISC-4 was evaluated in a laboratory-generated human skin melanoma model containing early melanocytic lesion or advanced stage melanoma cell lines and in animals containing invasive xenografted human melanoma. Repeated topical application of ISC-4 reduced tumor cell expansion in the skin model by 80% to 90% and decreased tumor development in animals by approximately 80%. Histologic examination of ISC-4-treated skin showed no obvious damage to skin cells or skin morphology, and treated animals did not exhibit markers indicative of major organ-related toxicity. Mechanistically, ISC-4 prevented melanoma by decreasing Akt3 signaling that lead to a 3-fold increase in apoptosis rates. Thus, topical ISC-4 can delay or slow down melanocytic lesion or melanoma development in preclinical models and could impact melanoma incidence rates if similar results are observed in humans.


Asunto(s)
Apoptosis/efectos de los fármacos , Cianatos/farmacología , Melanoma/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Compuestos de Selenio/farmacología , Neoplasias Cutáneas/prevención & control , Ensayos Antitumor por Modelo de Xenoinjerto , Administración Tópica , Animales , Western Blotting , Células Cultivadas , Cianatos/química , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Factores de Transcripción Forkhead/fisiología , Técnicas para Inmunoenzimas , Melanocitos/citología , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , ARN Interferente Pequeño/genética , Compuestos de Selenio/química , Piel/citología , Piel/efectos de los fármacos , Piel/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
16.
J Clin Invest ; 121(7): 2723-35, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21633165

RESUMEN

Intratumor heterogeneity is a major clinical problem because tumor cell subtypes display variable sensitivity to therapeutics and may play different roles in progression. We previously characterized 2 cell populations in human breast tumors with distinct properties: CD44+CD24- cells that have stem cell-like characteristics, and CD44-CD24+ cells that resemble more differentiated breast cancer cells. Here we identified 15 genes required for cell growth or proliferation in CD44+CD24- human breast cancer cells in a large-scale loss-of-function screen and found that inhibition of several of these (IL6, PTGIS, HAS1, CXCL3, and PFKFB3) reduced Stat3 activation. We found that the IL-6/JAK2/Stat3 pathway was preferentially active in CD44+CD24- breast cancer cells compared with other tumor cell types, and inhibition of JAK2 decreased their number and blocked growth of xenografts. Our results highlight the differences between distinct breast cancer cell types and identify targets such as JAK2 and Stat3 that may lead to more specific and effective breast cancer therapies.


Asunto(s)
Neoplasias de la Mama/patología , Antígeno CD24/metabolismo , Receptores de Hialuranos/metabolismo , Janus Quinasa 2/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Células Madre/fisiología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Antígeno CD24/genética , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Humanos , Receptores de Hialuranos/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Ratones , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT3/genética , Células Madre/citología , Trasplante Heterólogo
17.
Cancer Res ; 70(14): 6071-82, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20610626

RESUMEN

It is unknown why only a minority of circulating tumor cells trapped in lung capillaries form metastases and involvement of immune cells remains uncertain. A novel model has been developed in this study showing that neutrophils regulate lung metastasis development through physical interaction and anchoring of circulating tumor cells to endothelium. Human melanoma cells were i.v. injected into nude mice leading to the entrapment of many cancer cells; however, 24 hours later, very few remained in the lungs. In contrast, injection of human neutrophils an hour after tumor cell injection increased cancer cell retention by approximately 3-fold. Entrapped melanoma cells produced and secreted high levels of a cytokine called interleukin-8 (IL-8), attracting neutrophils and increasing tethering beta(2) integrin expression by 75% to 100%. Intercellular adhesion molecule-1 on melanoma cells and beta(2) integrin on neutrophils interacted, promoting anchoring to vascular endothelium. Decreasing IL-8 secretion from melanoma cells lowered extracellular levels by 20% to 50%, decreased beta(2) integrin on neutrophils by approximately 50%, and reduced neutrophil-mediated extravasation by 25% to 60%, resulting in approximately 50% fewer melanoma cells being tethered to endothelium and retained in lungs. Thus, transendothelial migration and lung metastasis development decreased by approximately 50%, showing that targeting IL-8 in melanoma cells has the potential to decrease metastasis development by disrupting interaction with neutrophils.


Asunto(s)
Neoplasias Pulmonares/secundario , Melanoma/secundario , Células Neoplásicas Circulantes/patología , Neutrófilos/patología , Neoplasias Cutáneas/patología , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Humanos , Interleucina-8/biosíntesis , Interleucina-8/genética , Interleucina-8/metabolismo , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/inmunología , Melanoma/sangre , Melanoma/inmunología , Melanoma/patología , Ratones , Ratones Desnudos , Células Neoplásicas Circulantes/inmunología , Neutrófilos/inmunología , ARN Interferente Pequeño/genética , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/inmunología
18.
J Natl Cancer Inst ; 102(15): 1131-47, 2010 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-20660366

RESUMEN

BACKGROUND: A putative tumor suppressor gene at chromosome 10p15, which contains KLF6 and other genes, is predicted to be lost during melanoma development, and its identity is unknown. In this study, we investigated the biological roles and identity of this tumor suppressor gene. METHODS: The human UACC 903 melanoma cell line containing introduced DNA fragments from the 10p15 region with (10E6/3, 10E6/11, and 10E6/18) and without (10ER4S.2/1) the tumor suppressor gene was used. Xenograft tumors were generated in a total of 40 mice with melanoma cell lines, and tumor size was measured. Cells were cultured on plastic or a gel of type I collagen. Viability, proliferation, and apoptosis were assessed. Expression of KLF6 protein was assessed by immunohistochemistry and immunoblot analysis. Expression of phosphorylated Erk1/2 and cyclin D1 was assessed by immunoblot analysis. Protein expression of KLF6 was inhibited with small interfering RNA (siRNA). KLF6 protein expression was assessed in 17 human nevi and human melanoma specimens from 29 patients. Statistical analyses were adjusted for multiple comparisons by use of Dunnett method. All statistical tests were two-sided. RESULTS: Melanoma cells containing KLF6 generated smaller subcutaneous xenograft tumors with fewer proliferating cells than control cells. When grown on collagen 1, viability of cells with ectopic KLF6 expression (72%) was lower than that of control cells (100%) (group difference = -28%, 95% confidence interval = -31.3% to -25.2%, P < .001). Viability of melanoma cells with or without the KLF6 tumor suppressor gene on plastic dishes was similar. When KLF6 expression was inhibited with KLF6 siRNA, viability of cells with the tumor suppressor gene on collagen I gel increased compared with that of control cells carrying scrambled siRNA. KLF6 protein was detected in all nevi examined but not in human metastatic melanoma tissue examined. Ectopic expression of KLF6 protein in melanoma cells grown on collagen I decreased levels of phosphorylated Erk1/2 and cyclin D1 in the mitogen-activated protein kinase signaling pathway. CONCLUSIONS: In melanoma cells, the tumor suppressor gene at 10p15 appears to be KLF6. Signaling from the collagen I-rich extracellular matrix appears to be involved in the tumor suppressive activity of KLF6 protein.


Asunto(s)
Cromosomas Humanos Par 10 , Colágeno Tipo I/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Melanoma/genética , Proteínas Proto-Oncogénicas/genética , Neoplasias Cutáneas/genética , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Animales , Apoptosis/genética , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Ciclina D1/metabolismo , Fragmentación del ADN , ADN Complementario/biosíntesis , Femenino , Gelatina , Regulación Neoplásica de la Expresión Génica , Humanos , Immunoblotting , Inmunohistoquímica , Factor 6 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Metaloproteinasas de la Matriz , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Nevo/genética , Fosforilación , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Trasplante Heterólogo , Proteínas Supresoras de Tumor/metabolismo
19.
J Med Chem ; 51(24): 7820-6, 2008 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19053750

RESUMEN

Synthesis and identification of novel phenylalkyl isoselenocyanates (ISCs), isosteric selenium analogues of naturally occurring phenylalkyl isothiocyanates (ITCs), as effective cytotoxic and antitumor agents are described. The structure-activity relationship comparison of ISCs with ITCs and effect of the increasing alkyl chain length in inhibiting cancer cell growth were evaluated on melanoma, prostate, breast, glioblastoma, sarcoma, and colon cancer cell lines. IC(50) values for ISC compounds were generally lower than their corresponding ITC analogues. Similarly, in UACC 903 human melanoma cells, the inhibition of cell proliferation and induction of apoptosis were more pronounced with ISCs compared to ITCs. Further, ISCs and ITCs effectively inhibited melanoma tumor growth in mice following intraperitoneal xenograft. A similar reduction in tumor size was observed at 3 times lower doses of ISCs compared to corresponding ITCs.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Isotiocianatos/química , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Química Farmacéutica/métodos , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Ratones , Modelos Químicos , Trasplante de Neoplasias , Relación Estructura-Actividad
20.
J Rheumatol ; 30(6): 1203-7, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12784390

RESUMEN

OBJECTIVE: To assess the therapeutic potential of rosmarinic acid (RosA) in an inflammatory autoimmune arthritis model. METHODS: Collagen induced arthritis is established in male DBA/1 mice. Mice were administered daily with 50 mg/kg/day of RosA for 15 days from Day 21 post-immunization and inspected daily to determine the progression of arthritis. After termination of injection, affected hindpaws were subjected to histopathological analyses and immunohistochemical assays for cyclooxygenase-2 (COX-2) expression. RESULTS: Repeated administration of RosA dramatically reduced the arthritic index and number of affected paws. Histopathologic observations closely paralleled clinical data, showing that RosA treated mice retained nearly normal architecture of synovial tissues, whereas control mice exhibited severe synovitis. Synovial tissues from RosA treated mice exhibited remarkably reduced frequency of COX-2-expressing cells, compared to those from untreated mice. CONCLUSION: RosA suppressed synovitis in a murine collagen induced arthritis model; this effect may be beneficial for treatment of rheumatoid arthritis in clinical settings.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Artritis Experimental/tratamiento farmacológico , Cinamatos/farmacología , Animales , Artritis Experimental/patología , Ciclooxigenasa 2 , Depsidos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Isoenzimas/metabolismo , Articulaciones/enzimología , Articulaciones/patología , Masculino , Ratones , Ratones Endogámicos DBA , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ácido Rosmarínico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA