Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Methods Mol Biol ; 2386: 129-145, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34766269

RESUMEN

Mass cytometry, or cytometry by time-of-flight (the basis for Fluidigm® CyTOF® technology), is a system for single-cell detection using antibodies tagged with metal probes. Without the need for compensation, the highly parametric Helios™ mass cytometer has a detection range of 135 distinct mass channels (75-209 Da). Optimized for mass cytometry, the Maxpar® Direct™ Immune Profiling Assay™ is a dry, metal-tagged antibody cocktail for immunophenotyping 37 immune cell populations found in human peripheral blood in a single tube. The Maxpar Direct Assay utilizes 31 mass channels for marker detection and live/dead viability staining, with at least 14 additional marker channels available from the Fluidigm catalog for flexible custom panel design. Here, we describe a workflow combining the assay with additional surface and intracellular cytokine antibodies for peripheral blood mononuclear cell (PBMC) staining using lanthanide-, bismuth-, and cadmium-tagged antibodies.


Asunto(s)
Citocinas/análisis , Anticuerpos , Biomarcadores , Citometría de Flujo , Humanos , Inmunofenotipificación , Espacio Intracelular , Leucocitos Mononucleares/inmunología , Coloración y Etiquetado , Flujo de Trabajo
2.
Gastroenterology ; 160(4): 1359-1372.e13, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33307028

RESUMEN

BACKGROUND & AIMS: Pancreatic ductal adenocarcinomas (PDACs) are characterized by fibrosis and an abundance of cancer-associated fibroblasts (CAFs). We investigated strategies to disrupt interactions among CAFs, the immune system, and cancer cells, focusing on adhesion molecule CDH11, which has been associated with other fibrotic disorders and is expressed by activated fibroblasts. METHODS: We compared levels of CDH11 messenger RNA in human pancreatitis and pancreatic cancer tissues and cells with normal pancreas, and measured levels of CDH11 protein in human and mouse pancreatic lesions and normal tissues. We crossed p48-Cre;LSL-KrasG12D/+;LSL-Trp53R172H/+ (KPC) mice with CDH11-knockout mice and measured survival times of offspring. Pancreata were collected and analyzed by histology, immunohistochemistry, and (single-cell) RNA sequencing; RNA and proteins were identified by imaging mass cytometry. Some mice were given injections of PD1 antibody or gemcitabine and survival was monitored. Pancreatic cancer cells from KPC mice were subcutaneously injected into Cdh11+/+ and Cdh11-/- mice and tumor growth was monitored. Pancreatic cancer cells (mT3) from KPC mice (C57BL/6), were subcutaneously injected into Cdh11+/+ (C57BL/6J) mice and mice were given injections of antibody against CDH11, gemcitabine, or small molecule inhibitor of CDH11 (SD133) and tumor growth was monitored. RESULTS: Levels of CDH11 messenger RNA and protein were significantly higher in CAFs than in pancreatic cancer epithelial cells, human or mouse pancreatic cancer cell lines, or immune cells. KPC/Cdh11+/- and KPC/Cdh11-/- mice survived significantly longer than KPC/Cdh11+/+ mice. Markers of stromal activation entirely surrounded pancreatic intraepithelial neoplasias in KPC/Cdh11+/+ mice and incompletely in KPC/Cdh11+/- and KPC/Cdh11-/- mice, whose lesions also contained fewer FOXP3+ cells in the tumor center. Compared with pancreatic tumors in KPC/Cdh11+/+ mice, tumors of KPC/Cdh11+/- mice had increased markers of antigen processing and presentation; more lymphocytes and associated cytokines; decreased extracellular matrix components; and reductions in markers and cytokines associated with immunosuppression. Administration of the PD1 antibody did not prolong survival of KPC mice with 0, 1, or 2 alleles of Cdh11. Gemcitabine extended survival of KPC/Cdh11+/- and KPC/Cdh11-/- mice only or reduced subcutaneous tumor growth in mT3 engrafted Cdh11+/+ mice when given in combination with the CDH11 antibody. A small molecule inhibitor of CDH11 reduced growth of pre-established mT3 subcutaneous tumors only if T and B cells were present in mice. CONCLUSIONS: Knockout or inhibition of CDH11, which is expressed by CAFs in the pancreatic tumor stroma, reduces growth of pancreatic tumors, increases their response to gemcitabine, and significantly extends survival of mice. CDH11 promotes immunosuppression and extracellular matrix deposition, and might be developed as a therapeutic target for pancreatic cancer.


Asunto(s)
Cadherinas/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Ductal Pancreático/inmunología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/inmunología , Animales , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Fibroblastos Asociados al Cáncer/inmunología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/cirugía , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Matriz Extracelular/inmunología , Matriz Extracelular/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metalotioneína 3 , Ratones , Ratones Noqueados , Páncreas/citología , Páncreas/inmunología , Páncreas/patología , Páncreas/cirugía , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/cirugía , Pancreaticoduodenectomía , Escape del Tumor/efectos de los fármacos , Escape del Tumor/genética , Escape del Tumor/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Gemcitabina
3.
Cancer Res ; 74(2): 508-19, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24282282

RESUMEN

Improved clinical management of prostate cancer has been impeded by an inadequate understanding of molecular genetic elements governing tumor progression. Gene signatures have provided improved prognostic indicators of human prostate cancer. The TGF-ß/BMP-SMAD4 signaling pathway, which induces epithelial-mesenchymal transition (EMT), is known to constrain prostate cancer progression induced by Pten deletion. Herein, cyclin D1 inactivation reduced cellular proliferation in the murine prostate in vivo and in isogenic oncogene-transformed prostate cancer cell lines. The in vivo cyclin D1-mediated molecular signature predicted poor outcome of recurrence-free survival for patients with prostate cancer (K-means HR, 3.75, P = 0.02) and demonstrated that endogenous cyclin D1 restrains TGF-ß, Snail, Twist, and Goosecoid signaling. Endogenous cyclin D1 enhanced Wnt and ES cell gene expression and expanded a prostate stem cell population. In chromatin immunoprecipitation sequencing, cyclin D1 occupied genes governing stem cell expansion and induced their transcription. The coordination of EMT restraining and stem cell expanding gene expression by cyclin D1 in the prostate may contribute to its strong prognostic value for poor outcome in biochemical-free recurrence in human prostate cancer.


Asunto(s)
Ciclina D1/fisiología , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfohidrolasa PTEN/metabolismo , Pronóstico , Recurrencia , Transducción de Señal , Resultado del Tratamiento
4.
Oncotarget ; 4(6): 923-35, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23798621

RESUMEN

Breast cancer is a leading form of cancer in the world. The Drosophila Dac gene was cloned as an inhibitor of the hyperactive epidermal growth factor (EGFR), ellipse. Herein, endogenous DACH1 co-localized with p53 in a nuclear, extranucleolar compartment and bound to p53 in human breast cancer cell lines, p53 and DACH1 bound common genes in Chip-Seq. Full inhibition of breast cancer contact-independent growth by DACH1 required p53. The p53 breast cancer mutants R248Q and R273H, evaded DACH1 binding. DACH1 phosphorylation at serine residue (S439) inhibited p53 binding and phosphorylation at p53 amino-terminal sites (S15, S20) enhanced DACH1 binding. DACH1 binding to p53 was inhibited by NAD-dependent deacetylation via DACH1 K628. DACH1 repressed p21CIP1 and induced RAD51, an association found in basal breast cancer. DACH1 inhibits breast cancer cellular growth in an NAD and p53-dependent manner through direct protein-protein association.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas del Ojo/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Secuencia de Aminoácidos , Apoptosis/fisiología , Sitios de Unión , Neoplasias de la Mama/genética , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Daño del ADN , Proteínas del Ojo/genética , Femenino , Expresión Génica , Células HEK293 , Humanos , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Homología de Secuencia de Aminoácido , Transducción de Señal , Factores de Transcripción/genética , Transfección , Proteína p53 Supresora de Tumor/genética
5.
J Proteome Res ; 11(4): 2236-46, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22356716

RESUMEN

Progression of invasive carcinoma involves the deregulation of molecular signaling pathways that results in the acquisition of oncogenic phenotypes. Functional enrichment analysis allows for the identification of deregulated pathways from omics scale expression data. Given the importance of post-transcriptional regulatory mechanisms on protein expression and function, identification of deregulated pathways on the basis of protein expression data is likely to provide new insights. In this study, we have developed methods for label-based mass spectrometry in a large number of samples and applied these methods toward identification and quantification of protein expression in samples of infiltrating ductal carcinoma, benign breast growths, and normal adjacent tissue. We identified 265 proteins with differential expression patterns in infiltrating ductal carcinoma relative to benign growths or normal breast tissue. Analysis of the differentially expressed proteins indicated the deregulation of signaling pathways related to proliferation, invasion and metastasis, and immune response. Our approach provides complementary information to gene expression microarray data and identifies a number of deregulated molecular signaling pathways indicative of breast cancer progression that may enable more accurate, biologically relevant diagnoses and provide a stepping stone to personalized treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Proteoma/análisis , Proteómica/métodos , Microambiente Tumoral , Anciano , Neoplasias de la Mama/química , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/química , Carcinoma Ductal de Mama/patología , Cromatografía Liquida , Análisis por Conglomerados , Electroforesis en Gel Bidimensional , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Proteoma/metabolismo , Transducción de Señal , Espectrometría de Masas en Tándem
6.
Adv Bioinformatics ; 2011: 608295, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22190923

RESUMEN

Identification of regulatory molecules in signaling pathways is critical for understanding cellular behavior. Given the complexity of the transcriptional gene network, the relationship between molecular expression and phenotype is difficult to determine using reductionist experimental methods. Computational models provide the means to characterize regulatory mechanisms and predict phenotype in the context of gene networks. Integrating gene expression data with phenotypic data in transcriptional network models enables systematic identification of critical molecules in a biological network. We developed an approach based on fuzzy logic to model cell budding in Saccharomyces cerevisiae using time series expression microarray data of the cell cycle. Cell budding is a phenotype of viable cells undergoing division. Predicted interactions between gene expression and phenotype reflected known biological relationships. Dynamic simulation analysis reproduced the behavior of the yeast cell cycle and accurately identified genes and interactions which are essential for cell viability.

7.
J Proteomics ; 75(2): 366-74, 2011 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21851866

RESUMEN

Molecular biomarkers of early stage breast cancer may improve the sensitivity and specificity of diagnosis. Plasma biomarkers have additional value in that they can be monitored with minimal invasiveness. Plasma biomarker discovery by genome-wide proteomic methods is impeded by the wide dynamic range of protein abundance and the heterogeneity of protein expression in healthy and disease populations which requires the analysis of a large number of samples. We addressed these issues through the development of a novel protocol that couples a combinatorial peptide ligand library protein enrichment strategy with isobaric label-based 2D LC-MS/MS for the identification of candidate biomarkers in high throughput. Plasma was collected from patients with stage I breast cancer or benign breast lesions. Low abundance proteins were enriched using a bead-based combinatorial library of hexapeptides. This resulted in the identification of 397 proteins, 22% of which are novel plasma proteins. Twenty-three differentially expressed plasma proteins were identified, demonstrating the effectiveness of the described protocol and defining a set of candidate biomarkers to be validated in independent samples. This work can be used as the basis for the design of properly powered investigations of plasma protein expression for biomarker discovery in larger cohorts of patients with complex disease.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias de la Mama/química , Neoplasias de la Mama/sangre , Neoplasias de la Mama/patología , Cromatografía Liquida/métodos , Técnicas Químicas Combinatorias/métodos , Diagnóstico Precoz , Femenino , Humanos , Persona de Mediana Edad , Biblioteca de Péptidos , Espectrometría de Masas en Tándem/métodos
8.
Cancer Res ; 70(24): 10464-73, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21159656

RESUMEN

The role of mammary epithelial cell (MEC) NF-κB in tumor progression in vivo is unknown, as murine NF-κB components and kinases either are required for murine survival or interfere with normal mammary gland development. As NF-κB inhibitors block both tumor-associated macrophages (TAM) and MEC NF-κB, the importance of MEC NF-κB to tumor progression in vivo remained to be determined. Herein, an MEC-targeted inducible transgenic inhibitor of NF-κB (IκBαSR) was developed in ErbB2 mammary oncomice. Inducible suppression of NF-κB in the adult mammary epithelium delayed the onset and number of new tumors. Within similar sized breast tumors, TAM and tumor neoangiogenesis was reduced. Coculture experiments demonstrated MEC NF-κB enhanced TAM recruitment. Genome-wide expression and proteomic analysis showed that IκBαSR inhibited tumor stem cell pathways. IκBαSR inhibited breast tumor stem cell markers in transgenic tumors, reduced stem cell expansion in vitro, and repressed expression of Nanog and Sox2 in vivo and in vitro. MEC NF-κB contributes to mammary tumorigenesis. As we show that NF-κB contributes to expansion of breast tumor stem cells and heterotypic signals that enhance TAM and vasculogenesis, these processes may contribute to NF-κB-dependent mammary tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/patología , Neoplasias Mamarias Experimentales/patología , FN-kappa B/metabolismo , Células Madre Neoplásicas/patología , Animales , Procesos de Crecimiento Celular/fisiología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Femenino , Proteínas I-kappa B/biosíntesis , Proteínas I-kappa B/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Transgénicos , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , Células Madre Neoplásicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor ErbB-2/biosíntesis , Transfección
9.
Proc Natl Acad Sci U S A ; 107(18): 8231-6, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20406904

RESUMEN

microRNAs are thought to regulate tumor progression and invasion via direct interaction with target genes within cells. Here the microRNA17/20 cluster is shown to govern cellular migration and invasion of nearby cells via heterotypic secreted signals. microRNA17/20 abundance is reduced in highly invasive breast cancer cell lines and node-positive breast cancer specimens. Cell-conditioned medium from microRNA17/20-overexpressing noninvasive breast cancer cell MCF7 was sufficient to inhibit MDA-MB-231 cell migration and invasion through inhibiting secretion of a subset of cytokines, and suppressing plasminogen activation via inhibition of the secreted plasminogen activators (cytokeratin 8 and alpha-enolase). microRNA17/20 directly repressed IL-8 by targeting its 3' UTR, and inhibited cytokeratin 8 via the cell cycle control protein cyclin D1. At variance with prior studies, these results demonstrated a unique mechanism of how the altered microRNA17/20 expression regulates cellular secretion and tumor microenvironment to control migration and invasion of neighboring cells in breast cancer. These findings not only reveal an antiinvasive function of miR-17/20 in breast cancer, but also identify a heterotypic secreted signal that mediates the microRNA regulation of tumor metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , MicroARNs/genética , Transducción de Señal , Regiones no Traducidas 3' , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Plasminógeno/metabolismo , Unión Proteica
10.
Cancer Res ; 70(5): 2105-14, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20179208

RESUMEN

Cyclin D1 belongs to a family of proteins that regulate progression through the G1-S phase of the cell cycle by binding to cyclin-dependent kinase (cdk)-4 to phosphorylate the retinoblastoma protein and release E2F transcription factors for progression through cell cycle. Several cancers, including breast, colon, and prostate, overexpress the cyclin D1 gene. However, the correlation of cyclin D1 overexpression with E2F target gene regulation or of cdk-dependent cyclin D1 activity with tumor development has not been identified. This suggests that the role of cyclin D1 in oncogenesis may be independent of its function as a cell cycle regulator. One such function is the role of cyclin D1 in cell adhesion and motility. Filamin A (FLNa), a member of the actin-binding filamin protein family, regulates signaling events involved in cell motility and invasion. FLNa has also been associated with a variety of cancers including lung cancer, prostate cancer, melanoma, human bladder cancer, and neuroblastoma. We hypothesized that elevated cyclin D1 facilitates motility in the invasive MDA-MB-231 breast cancer cell line. We show that MDA-MB-231 motility is affected by disturbing cyclin D1 levels or cyclin D1-cdk4/6 kinase activity. Using mass spectrometry, we find that cyclin D1 and FLNa coimmunoprecipitate and that lower levels of cyclin D1 are associated with decreased phosphorylation of FLNa at Ser2152 and Ser1459. We also identify many proteins related to cytoskeletal function, biomolecular synthesis, organelle biogenesis, and calcium regulation whose levels of expression change concomitant with decreased cell motility induced by decreased cyclin D1 and cyclin D1-cdk4/6 activities.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular/fisiología , Proteínas Contráctiles/metabolismo , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Proteínas de Microfilamentos/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Quinasa 6 Dependiente de la Ciclina/metabolismo , Filaminas , Humanos , Datos de Secuencia Molecular , Invasividad Neoplásica , Fosfoproteínas/metabolismo
11.
J Biol Chem ; 285(13): 9792-9802, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20093358

RESUMEN

Activation of the inflammasome generates the pro-inflammatory cytokines interleukin-1 beta and -18, which are important mediators of inflammation. Abnormal activation of the inflammasome leads to many inflammatory diseases, including gout, silicosis, neurodegeneration, and genetically inherited periodic fever syndromes. Therefore, identification of small molecule inhibitors that target the inflammasome is an important step toward developing effective therapeutics for the treatment of inflammation. Here, we show that the herbal NF-kappaB inhibitory compound parthenolide inhibits the activity of multiple inflammasomes in macrophages by directly inhibiting the protease activity of caspase-1. Additional investigations of other NF-kappaB inhibitors revealed that the synthetic I kappaB kinase-beta inhibitor Bay 11-7082 and structurally related vinyl sulfone compounds selectively inhibit NLRP3 inflammasome activity in macrophages independent of their inhibitory effect on NF-kappaB activity. In vitro assays of the effect of parthenolide and Bay 11-7082 on the ATPase activity of NLRP3 demonstrated that both compounds inhibit the ATPase activity of NLRP3, suggesting that the inhibitory effect of these compounds on inflammasome activity could be mediated in part through their effect on the ATPase activity of NLRP3. Our results thus elucidate the molecular mechanism for the therapeutic anti-inflammatory activity of parthenolide and identify vinyl sulfones as a new class of potential therapeutics that target the NLRP3 inflammasome.


Asunto(s)
Antiinflamatorios/farmacología , Inflamación/tratamiento farmacológico , Nitrilos/farmacología , Sesquiterpenos/farmacología , Sulfonas/farmacología , Animales , Células de la Médula Ósea/metabolismo , Caspasa 1/metabolismo , Muerte Celular , Humanos , Immunoblotting , L-Lactato Deshidrogenasa/metabolismo , Macrófagos/metabolismo , Ratones , FN-kappa B/metabolismo , Sulfonas/química
12.
Am J Pathol ; 174(3): 746-61, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19234134

RESUMEN

Recently, we reported that human breast cancer-associated fibroblasts show functional inactivation of the retinoblastoma (RB) tumor suppressor and down-regulation of caveolin-1 (Cav-1) protein expression. However, it remains unknown whether loss of Cav-1 is sufficient to confer functional RB inactivation in mammary fibroblasts. To establish a direct cause-and-effect relationship, mammary stromal fibroblasts (MSFs) were prepared from Cav-1(-/-) null mice and subjected to phenotypic analysis. Here, we provide evidence that Cav-1(-/-) MSFs share many characteristics with human cancer-associated fibroblasts. The Cav-1(-/-) MSF transcriptome significantly overlaps with human cancer-associated fibroblasts; both show a nearly identical profile of RB/E2F-regulated genes that are up-regulated, which is consistent with RB inactivation. This Cav-1(-/-) MSF gene signature is predictive of poor clinical outcome in breast cancer patients treated with tamoxifen. Consistent with these findings, Cav-1(-/-) MSFs show RB hyperphosphorylation and the up-regulation of estrogen receptor co-activator genes. We also evaluated the paracrine effects of "conditioned media" prepared from Cav-1(-/-) MSFs on wild-type mammary epithelia. Our results indicate that Cav-1(-/-) MSF "conditioned media" is sufficient to induce an epithelial-mesenchymal transition, indicative of an invasive phenotype. Proteomic analysis of this "conditioned media" reveals increased levels of proliferative/angiogenic growth factors. Consistent with these findings, Cav-1(-/-) MSFs are able to undergo endothelial-like transdifferentiation. Thus, these results have important implications for understanding the role of cancer-associated fibroblasts and RB inactivation in promoting tumor angiogenesis.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Caveolina 1/deficiencia , Caveolina 1/genética , Fibroblastos/patología , Células del Estroma/patología , Western Blotting , Mama/citología , Mama/fisiología , Neoplasias de la Mama/mortalidad , Técnicas de Cultivo de Célula , División Celular , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Células Epiteliales/citología , Células Epiteliales/patología , Células Epiteliales/fisiología , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Neoplásico/genética , Células del Estroma/citología , Células del Estroma/fisiología , Análisis de Supervivencia
13.
Am J Pathol ; 174(2): 613-29, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19164602

RESUMEN

Here, we show that functional loss of a single gene is sufficient to confer constitutive milk protein production and protection against mammary tumor formation. Caveolin-3 (Cav-3), a muscle-specific caveolin-related gene, is highly expressed in muscle cells. We demonstrate that Cav-3 is also expressed in myoepithelial cells within the mammary gland. To determine whether genetic ablation of Cav-3 expression affects adult mammary gland development, we studied the phenotype(s) of Cav-3(-/-)-null mice. Interestingly, Cav-3(-/-) virgin mammary glands developed lobulo-alveolar hyperplasia, akin to the changes normally observed during pregnancy and lactation. Genome-wide expression profiling revealed up-regulation of gene transcripts associated with pregnancy/lactation, mammary stem cells, and human breast cancers, consistent with a constitutive lactogenic phenotype. Expression levels of three key transcriptional regulators of lactation, namely Elf5, Stat5a, and c-Myc, were also significantly elevated. Experiments with pregnant mice directly showed that Cav-3(-/-) mice underwent precocious lactation. Finally, using orthotopic tumor cell implantation, we demonstrated that virgin Cav-3(-/-) mice were dramatically protected against mammary tumor formation. Thus, Cav-3(-/-) mice are a novel preclinical model to study the protective effects of a lactogenic microenvironment on mammary tumor onset and progression. Our current studies have broad implications for using the lactogenic microenvironment as a paradigm to discover new therapies for the prevention and/or treatment of human breast cancers.


Asunto(s)
Caveolina 3/genética , Caveolina 3/metabolismo , Expresión Génica , Lactancia/fisiología , Neoplasias Mamarias Experimentales/genética , Animales , Movimiento Celular/fisiología , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Mutantes , Leche Humana/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Reacción en Cadena de la Polimerasa , Embarazo
14.
Cancer Res ; 66(17): 8529-41, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16951165

RESUMEN

Expression of the cyclin-dependent kinase (Cdk) inhibitor (p27(Kip1)) is frequently reduced in human tumors, often correlating with poor prognosis. p27(Kip1) functions as a haploinsufficient tumor suppressor; however, the mechanism by which one allele of p27(Kip1) regulates oncogenic signaling in vivo is not well understood. We therefore investigated the mechanisms by which p27(Kip1) inhibits mammary tumor onset. Using the common background strain of FVB, p27(Kip1) heterozygosity (p27(+/-)) accelerated ErbB2-induced mammary tumorigenesis. We conducted microarray analyses of mammary tumors developing in mice with genetic haploinsufficiency for p27(Kip1) expressing a mammary-targeted ErbB2 oncogene. Global gene expression profiling and Western blot analysis of ErbB2/p27(+/-) tumors showed that the loss of p27(Kip1) induced genes promoting lymphangiogenesis, cellular proliferation, and collaborative oncogenic signaling (Wnt/beta-catenin/Tcf, Cdc25a, Smad7, and Skp2). Skp2 expression was induced by ErbB2 and repressed by p27(Kip1). Degradation of p27(Kip1) involves an SCF-type E3 ubiquitin ligase, including Skp2. The Skp2 component of the SCF(SKP2) complex that degrades p27(Kip1) was increased in ErbB2 tumors correlating with earlier tumor onset. In both murine and human ErbB2-overexpressing breast cancers, p27(Kip1) levels correlated inversely with Skp2. p27(Kip1) haploinsufficiency activated Wnt/beta-catenin/hedgehog signaling. Reintroduction of p27(Kip1) inhibited beta-catenin induction of Tcf-responsive genes (Siamosis, c-Myc, and Smad7). p27(Kip1) is haploinsufficient for ErbB2 mammary tumor suppression in vivo and functions to repress collaborative oncogenic signals including Skp2 and Wnt/beta-catenin signaling.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Neoplasias Mamarias Experimentales/patología , beta Catenina/fisiología , Animales , Núcleo Celular/fisiología , Núcleo Celular/ultraestructura , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Cartilla de ADN , Femenino , Eliminación de Gen , Glándulas Mamarias Animales/fisiología , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
15.
Mol Cell Biol ; 26(21): 8122-35, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16923962

RESUMEN

The NAD-dependent histone deacetylase Sir2 plays a key role in connecting cellular metabolism with gene silencing and aging. The androgen receptor (AR) is a ligand-regulated modular nuclear receptor governing prostate cancer cellular proliferation, differentiation, and apoptosis in response to androgens, including dihydrotestosterone (DHT). Here, SIRT1 antagonists induce endogenous AR expression and enhance DHT-mediated AR expression. SIRT1 binds and deacetylates the AR at a conserved lysine motif. Human SIRT1 (hSIRT1) repression of DHT-induced AR signaling requires the NAD-dependent catalytic function of hSIRT1 and the AR lysine residues deacetylated by SIRT1. SIRT1 inhibited coactivator-induced interactions between the AR amino and carboxyl termini. DHT-induced prostate cancer cellular contact-independent growth is also blocked by SIRT1, providing a direct functional link between the AR, which is a critical determinant of progression of human prostate cancer, and the sirtuins.


Asunto(s)
Dihidrotestosterona/metabolismo , Regulación de la Expresión Génica , Neoplasias de la Próstata , Receptores Androgénicos/metabolismo , Sirtuinas/metabolismo , Acetilación , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proliferación Celular , Genes Reporteros , Histona Acetiltransferasas/metabolismo , Humanos , Lisina/metabolismo , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/genética , Péptidos/metabolismo , Receptores Androgénicos/genética , Transducción de Señal/fisiología , Sirtuina 1 , Sirtuinas/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Factores de Transcripción p300-CBP
16.
Mol Cell Biol ; 26(14): 5449-69, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16809779

RESUMEN

The cyclin D1 gene encodes a regulatory subunit of the holoenzyme that phosphorylates and inactivates the pRb tumor suppressor to promote nuclear DNA synthesis. cyclin D1 is overexpressed in human breast cancers and is sufficient for the development of murine mammary tumors. Herein, cyclin D1 is shown to perform a novel function, inhibiting mitochondrial function and size. Mitochondrial activity was enhanced by genetic deletion or antisense or small interfering RNA to cyclin D1. Global gene expression profiling and functional analysis of mammary epithelial cell-targeted cyclin D1 antisense transgenics demonstrated that cyclin D1 inhibits mitochondrial activity and aerobic glycolysis in vivo. Reciprocal regulation of these genes was observed in cyclin D1-induced mammary tumors. Cyclin D1 thus integrates nuclear DNA synthesis and mitochondrial function.


Asunto(s)
Ciclina D1/metabolismo , Mitocondrias/metabolismo , Animales , Secuencia de Bases , Ciclina D1/deficiencia , Ciclina D1/genética , ADN/genética , Femenino , Perfilación de la Expresión Génica , Glucólisis , Hexoquinasa/genética , Hexoquinasa/metabolismo , Humanos , Lipogénesis/genética , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/genética , Modelos Biológicos , Familia de Multigenes , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética
17.
Mol Cell Biol ; 26(11): 4240-56, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16705174

RESUMEN

Cyclin D1 is overexpressed in human tumors, correlating with cellular metastasis, and is induced by activating Rho GTPases. Herein, cyclin D1-deficient mouse embryo fibroblasts (MEFs) exhibited increased adhesion and decreased motility compared with wild-type MEFs. Retroviral transduction of cyclin D1 reversed these phenotypes. Mutational analysis of cyclin D1 demonstrated that its effects on cellular adhesion and migration were independent of the pRb and p160 coactivator binding domains. Genomewide expression arrays identified a subset of genes regulated by cyclin D1, including Rho-activated kinase II (ROCKII) and thrombospondin 1 (TSP-1). cyclin D1(-/-) cells showed increased Rho GTP and ROCKII activity and signaling, with increased phosphorylation of LIM kinase, cofilin (Ser3), and myosin light chain 2 (Thr18/Ser19). Cyclin D1 repressed ROCKII and TSP-1 expression, and the migratory defect of cyclin D1(-/-) cells was reversed by ROCK inhibition or TSP-1 immunoneutralizing antibodies. cyclin E knockin to the cyclin D1(-/-) MEFs rescued the DNA synthesis defect of cyclin D1(-/-) MEFs but did not rescue either the migration defect or the abundance of ROCKII. Cyclin D1 promotes cellular motility through inhibiting ROCK signaling and repressing the metastasis suppressor TSP-1.


Asunto(s)
Movimiento Celular , Ciclina D1/metabolismo , Regulación de la Expresión Génica , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Trombospondina 1/antagonistas & inhibidores , Animales , Adhesión Celular , Células Cultivadas , Ciclina D1/química , Ciclina D1/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , ADN/biosíntesis , Fibroblastos/citología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Mutación/genética , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fibras de Estrés/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo , Quinasas Asociadas a rho
18.
J Cell Biol ; 173(4): 533-44, 2006 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-16717128

RESUMEN

The activity of the p53 gene product is regulated by a plethora of posttranslational modifications. An open question is whether such posttranslational changes act redundantly or dependently upon one another. We show that a functional interference between specific acetylated and phosphorylated residues of p53 influences cell fate. Acetylation of lysine 320 (K320) prevents phosphorylation of crucial serines in the NH(2)-terminal region of p53; only allows activation of genes containing high-affinity p53 binding sites, such as p21/WAF; and promotes cell survival after DNA damage. In contrast, acetylation of K373 leads to hyperphosphorylation of p53 NH(2)-terminal residues and enhances the interaction with promoters for which p53 possesses low DNA binding affinity, such as those contained in proapoptotic genes, leading to cell death. Further, acetylation of each of these two lysine clusters differentially regulates the interaction of p53 with coactivators and corepressors and produces distinct gene-expression profiles. By analogy with the "histone code" hypothesis, we propose that the multiple biological activities of p53 are orchestrated and deciphered by different "p53 cassettes," each containing combination patterns of posttranslational modifications and protein-protein interactions.


Asunto(s)
Ciclo Celular/genética , Regulación de la Expresión Génica/genética , Procesamiento Proteico-Postraduccional/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Secuencia de Aminoácidos/fisiología , Apoptosis/genética , Sitios de Unión/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Genes cdc/fisiología , Humanos , Lisina/metabolismo , Fosforilación , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Estructura Terciaria de Proteína/fisiología , Elementos Reguladores de la Transcripción/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/química
19.
J Biol Chem ; 280(11): 10264-76, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15632193

RESUMEN

The SIR2 family of nicotinamide adenosine dinucleotide (NAD)-dependent deacetylases modulates diverse biological functions in different species, including longevity, apoptosis, cell cycle exit, and cellular differentiation. SIRT1, the closest mammalian ortholog of the yeast SIR2 (silent information regulator 2) gene, represses several transcription factors, including p53, NFkappaB and forkhead proteins. The p300 protein serves as a rate-limiting transcriptional cointegrator of diverse transcription factors either to activate or to repress transcription through modular subdomains. Herein, SIRT1 physically interacted with and repressed p300 transactivation, requiring the NAD-dependent deacetylase activity of SIRT1. SIRT1 repression involved the CRD1 transcriptional repression domain of p300. Two residues within the CRD1 domain (Lys-1020 and Lys-1024) were required for SIRT1 repression and served as substrates for SIRT1 deacetylation. These residues also serve as acceptor lysines for modification by the ubiquitin-like SUMO protein. The SUMO-specific protease SSP3 relieved SIRT1 repression of p300. SSP3 antagonism of SIRT1 required the SUMO-deconjugating function of SSP3. Thus, p300 serves as a deacetylase substrate for SIRT1 through a conserved SUMO consensus motif. Because p300 is a limiting transcriptional cofactor, deacetylation and repression of p300 by SIRT1 may serve an important integration point during metabolism and cellular differentiation.


Asunto(s)
Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Lisina/química , Proteínas Nucleares/metabolismo , Sirtuinas/química , Sirtuinas/metabolismo , Transactivadores/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Western Blotting , Diferenciación Celular , Línea Celular , Células Cultivadas , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Proteína p300 Asociada a E1A , Fibroblastos/citología , Genes Reporteros , Vectores Genéticos , Humanos , Inmunoprecipitación , Luciferasas/metabolismo , Espectrometría de Masas , Ratones , Datos de Secuencia Molecular , NAD/metabolismo , FN-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/química , Péptidos/química , Unión Proteica , Estructura Terciaria de Proteína , ARN/química , Retroviridae/metabolismo , Serina Endopeptidasas/metabolismo , Sirtuina 1 , Factores de Tiempo , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo
20.
Chem Biodivers ; 2(11): 1495-502, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17191949

RESUMEN

Time-Of-Flight Mass Spectrometry (TOF-SIMS) was used to determine elemental and biomolecular ions from isolated protein samples. We identified a set of 23 mass-to-charge ratio (m/z) peaks that represent signatures for distinguishing biological samples. The 23 peaks were identified by Singular Value Decomposition (SVD) and Canonical Analysis (CA) to find the underlying structure in the complex mass-spectra data sets. From this modified data, SVD was used to identify sets of m/z peaks, and we used these patterns from the TOF-SIMS data to predict the biological source from which individual mass spectra were generated. The signatures were validated using an additional data set different from the initial training set used to identify the signatures. We present a simple method to identify multiple variables required for sample classification based on mass spectra that avoids overfit. This is important in a variety of studies using mass spectrometry, including the ability to identify proteins in complex mixtures and for the identification of new biomarkers.


Asunto(s)
Productos Biológicos/análisis , Espectrometría de Masa de Ion Secundario/métodos , Proteínas/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA