Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 10(1): 3589, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31399573

RESUMEN

Overcoming cellular growth restriction, including the evasion of cellular senescence, is a hallmark of cancer. We report that PAK4 is overexpressed in all human breast cancer subtypes and associated with poor patient outcome. In mice, MMTV-PAK4 overexpression promotes spontaneous mammary cancer, while PAK4 gene depletion delays MMTV-PyMT driven tumors. Importantly, PAK4 prevents senescence-like growth arrest in breast cancer cells in vitro, in vivo and ex vivo, but is not needed in non-immortalized cells, while PAK4 overexpression in untransformed human mammary epithelial cells abrogates H-RAS-V12-induced senescence. Mechanistically, a PAK4 - RELB - C/EBPß axis controls the senescence-like growth arrest and a PAK4 phosphorylation residue (RELB-Ser151) is critical for RELB-DNA interaction, transcriptional activity and expression of the senescence regulator C/EBPß. These findings establish PAK4 as a promoter of breast cancer that can overcome oncogene-induced senescence and reveal a selective vulnerability of cancer to PAK4 inhibition.


Asunto(s)
Neoplasias de la Mama/patología , Factor de Transcripción ReIB/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Mama/citología , Mama/patología , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Senescencia Celular/genética , Células Epiteliales , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Cultivo Primario de Células , Pronóstico , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas p21 Activadas/genética
2.
J Natl Cancer Inst ; 111(9): 983-995, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30816935

RESUMEN

BACKGROUND: A better definition of biomarkers and biological processes related to local recurrence and disease progression is highly warranted for ductal breast carcinoma in situ (DCIS). Stromal-epithelial interactions are likely of major importance for the biological, clinical, and pathological distinctions between high- and low-risk DCIS cases. METHODS: Stromal platelet derived growth factor receptor (PDGFR) was immunohistochemically assessed in two DCIS patient cohorts (n = 458 and n = 80). Cox proportional hazards models were used to calculate the hazard ratios of recurrence. The molecular mechanisms regulating stromal PDGFR expression were investigated in experimental in vitro co-culture systems of DCIS cells and fibroblasts and analyzed using immunoblot and quantitative real-time PCR. Knock-out of JAG1 in DCIS cells and NOTCH2 in fibroblasts was obtained through CRISPR/Cas9. Experimental data were validated by mammary fat pad injection of DCIS and DCIS-JAG1 knock-out cells (10 mice per group). All statistical tests were two-sided. RESULTS: PDGFRα(low)/PDGFRß(high) fibroblasts were associated with increased risk for recurrence in DCIS (univariate hazard ratio = 1.59, 95% confidence interval [CI] = 1.02 to 2.46; P = .04 Wald test; multivariable hazard ratio = 1.78, 95% CI = 1.07 to 2.97; P = .03). Tissue culture and mouse model studies indicated that this fibroblast phenotype is induced by DCIS cells in a cell contact-dependent manner. Epithelial Jagged1 and fibroblast Notch2 were identified through loss-of-function studies as key juxtacrine signaling components driving the formation of the poor prognosis-associated fibroblast phenotype. CONCLUSIONS: A PDGFRα(low)/PDGFRß(high) fibroblast subset was identified as a marker for high-risk DCIS. The Jagged-1/Notch2/PDGFR stroma-epithelial pathway was described as a novel signaling mechanism regulating this poor prognosis-associated fibroblast subset. In general terms, the study highlights epithelial-stromal crosstalk in DCIS and contributes to ongoing efforts to define clinically relevant fibroblast subsets and their etiology.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Intraductal no Infiltrante/metabolismo , Comunicación Celular , Células Epiteliales/metabolismo , Células del Estroma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores de Tumor , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Carcinoma Intraductal no Infiltrante/mortalidad , Carcinoma Intraductal no Infiltrante/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Biología Computacional/métodos , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Ratones , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo
3.
Nat Med ; 24(4): 463-473, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29529015

RESUMEN

Breast tumors of the basal-like, hormone receptor-negative subtype remain an unmet clinical challenge, as there is high rate of recurrence and poor survival in patients following treatment. Coevolution of the malignant mammary epithelium and its underlying stroma instigates cancer-associated fibroblasts (CAFs) to support most, if not all, hallmarks of cancer progression. Here we delineate a previously unappreciated role for CAFs as determinants of the molecular subtype of breast cancer. We identified paracrine crosstalk between cancer cells expressing platelet-derived growth factor (PDGF)-CC and CAFs expressing the cognate receptors in human basal-like mammary carcinomas. Genetic or pharmacological intervention of PDGF-CC activity in mouse models of cancer resulted in conversion of basal-like breast cancers into a hormone receptor-positive state that enhanced sensitivity to endocrine therapy in previously resistant tumors. We conclude that specification of breast cancer to the basal-like subtype is under microenvironmental control and is therapeutically actionable.


Asunto(s)
Neoplasias de la Mama/patología , Linfocinas/metabolismo , Comunicación Paracrina , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Microambiente Tumoral , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/irrigación sanguínea , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células Epiteliales/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Fibrosis , Humanos , Linfocinas/deficiencia , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neovascularización Patológica/patología , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Pronóstico , Modelos de Riesgos Proporcionales , Transducción de Señal , Células del Estroma/patología , Análisis de Supervivencia , Resultado del Tratamiento
4.
J Natl Cancer Inst ; 107(10)2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26296362

RESUMEN

BACKGROUND: Pericytes are members of the tumor stroma; however, little is known about their origin, function, or interaction with other tumor components. Emerging evidence suggest that pericytes may regulate leukocyte transmigration. Myeloid-derived suppressor cells (MDSC) are immature myeloid cells with powerful inhibitory effects on T-cell-mediated antitumor reactivity. METHODS: We generated subcutaneous tumors in a genetic mouse model of pericyte deficiency (the pdgfb (ret/ret) mouse) and littermate control mice (n = 6-25). Gene expression profiles from 253 breast cancer patients (stage I-III) were evaluated for clinic-pathological parameters and survival using Cox proportional hazard ratios (HRs) and 95% confidence intervals (CIs) based on a two-sided Wald test. RESULTS: We report that pericyte deficiency leads to increased transmigration of Gr1(+)/CD11b(+) cells in experimentally induced tumors. Pericyte deficiency produced defective tumor vasculature, resulting in a more hypoxic microenvironment promoting IL-6 upregulation in the malignant cells. Silencing IL-6 expression in tumor cells attenuated the observed differences in MDSC transmigration. Restoring the pericyte coverage in tumors abrogated the increased MDSC trafficking to pericyte-deficient tumors. MDSC accumulation in tumors led to increases in tumor growth and in circulating malignant cells. Finally, gene expression analysis from human breast cancer patients revealed increased expression of the human MDSC markers CD33 and S100A9 with concomitant decreased expression of pericyte genes and was associated with poor prognosis (HR = 1.88, 95% CI = 1.08 to 3.25, P = .03). CONCLUSIONS: Our data uncovers a novel paracrine interaction between tumor pericytes and inflammatory cells and delineates the cellular events resulting in the recruitment of MDSC to tumors. Furthermore, we propose for the first time a role for tumor pericytes in modulating the expression of immune mediators in malignant cells by promoting a hypoxic microenvironment.


Asunto(s)
Neoplasias de la Mama/patología , Antígeno CD11b/metabolismo , Movimiento Celular , Células Mieloides , Neoplasias Experimentales/patología , Pericitos , Receptores de Quimiocina/metabolismo , Animales , Antígenos de Superficie/metabolismo , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula , Femenino , Citometría de Flujo , Silenciador del Gen , Humanos , Interleucina-6/genética , Ratones , Neoplasias Experimentales/metabolismo , Tejido Subcutáneo , Suecia , Transcriptoma , Microambiente Tumoral
5.
Cancer Res ; 75(12): 2445-56, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-26077471

RESUMEN

Exploration of new strategies for the prevention of breast cancer metastasis is justifiably at the center of clinical attention. In this study, we combined a computational biology approach with mechanism-based preclinical trials to identify inhibitors of activin-like receptor kinase (ALK) 1 as effective agents for blocking angiogenesis and metastasis in breast cancer. Pharmacologic targeting of ALK1 provided long-term therapeutic benefit in mouse models of mammary carcinoma, accompanied by strikingly reduced metastatic colonization as a monotherapy or part of combinations with chemotherapy. Gene-expression analysis of breast cancer specimens from a population-based nested case-control study encompassing 768 subjects defined endothelial expression of ALK1 as an independent and highly specific prognostic factor for metastatic manifestation, a finding that was corroborated in an independent clinical cohort. Overall, our results suggest that pharmacologic inhibition of endothelial ALK1 constitutes a tractable strategy for interfering with metastatic dissemination of breast cancer.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Activinas Tipo II/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Endotelio/enzimología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Metástasis de la Neoplasia
6.
Oncotarget ; 5(23): 12418-27, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25360764

RESUMEN

Therapeutic vaccination targeting self-molecules is an attractive alternative to monoclonal antibody-based therapies for cancer and various inflammatory diseases. However, development of cancer vaccines targeting self-molecules has proven difficult. One complicating factor is that tumor cells have developed strategies to escape recognition by the immune system. Antigens specifically expressed by the tumor vasculature can therefore provide alternative targets. The alternatively spliced extra domain-A and B (ED-A and ED-B) of fibronectin are expressed during vasculogenesis in the embryo, but essentially undetectable under normal conditions in the adult. However, these domains are re-expressed during tumor angiogenesis and matrix remodeling, which renders them highly interesting for targeted cancer therapies. Using the MMTV-PyMT transgenic model of metastatic mammary carcinoma, we show that tumor burden can be significantly decreased by immunization against ED-A in a therapeutic setting. Furthermore, we found that in mice carrying anti-ED-A antibodies the number of metastases was reduced. ED-A immunization increased infiltration of macrophages and compromised tumor blood vessel function. These findings implicate an attack of the tumor vasculature by the immune system, through a polyclonal antibody response. We conclude that tumor vascular antigens are promising candidates for development of therapeutic vaccines targeting growth of primary tumors as well as disseminated disease.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Fibronectinas/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Animales , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Transgénicos , Embarazo
7.
Semin Cancer Biol ; 25: 3-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24412106

RESUMEN

In the field of tumor biology, increasing attention is now focused on the complex interactions between various constituent cell types within the tumor microenvironment as being functionally important for the etiology of the disease. The detailed description of tumor-promoting properties of cancer-associated fibroblasts, endothelial cells, pericytes, and immune cells, introduces novel potential drug targets for improved cancer treatments, as well as a rationale for exploring the tumor stroma as a previously unchartered source for prognostic or predictive biomarkers. However, recent work highlights the fact that cellular identity is perhaps too broadly defined and that subdivision of each cell type may reveal functionally distinct subsets of cells. Here, we will review our current understanding of the diversity of different subsets of mesenchymal cells, i.e., cancer-associated fibroblasts and pericytes, residing within the tumor parenchyma.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Neoplasias/patología , Microambiente Tumoral , Animales , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio/metabolismo , Endopeptidasas , Fibroblastos/fisiología , Gelatinasas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Pericitos/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína de Unión al Calcio S100A4 , Serina Endopeptidasas/metabolismo
8.
Dev Cell ; 23(3): 587-99, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22975327

RESUMEN

Angiogenesis, the process by which new blood vessels arise from preexisting ones, is critical for embryonic development and is an integral part of many disease processes. Recent studies have provided detailed information on how angiogenic sprouts initiate, elongate, and branch, but less is known about how these processes cease. Here, we show that S1PR1, a receptor for the blood-borne bioactive lipid sphingosine-1-phosphate (S1P), is critical for inhibition of angiogenesis and acquisition of vascular stability. Loss of S1PR1 leads to increased endothelial cell sprouting and the formation of ectopic vessel branches. Conversely, S1PR1 signaling inhibits angiogenic sprouting and enhances cell-to-cell adhesion. This correlates with inhibition of vascular endothelial growth factor-A (VEGF-A)-induced signaling and stabilization of vascular endothelial (VE)-cadherin localization at endothelial junctions. Our data suggest that S1PR1 signaling acts as a vascular-intrinsic stabilization mechanism, protecting developing blood vessels against aberrant angiogenic responses.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Neovascularización Fisiológica , Receptores de Lisoesfingolípidos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores de Lisoesfingolípidos/deficiencia , Receptores de Esfingosina-1-Fosfato , Pez Cebra
9.
BMC Cancer ; 12: 378, 2012 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-22931209

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) and their role during tumor development have been studied in great detail during the last decade, albeit their expression pattern and regulation during normal development are however not so well established. Previous studies have shown that miRNAs are differentially expressed in solid human tumors. Platelet-derived growth factor (PDGF) signaling is known to be involved in normal development of the brain as well as in malignant primary brain tumors, gliomas, but the complete mechanism is still lacking. We decided to investigate the expression of the oncogenic miR-21 during normal mouse development and glioma, focusing on PDGF signaling as a potential regulator of miR-21. METHODS: We generated mouse glioma using the RCAS/tv-a system for driving PDGF-BB expression in a cell-specific manner. Expression of miR-21 in mouse cell cultures and mouse brain were assessed using Northern blot analysis and in situ hybridization. Immunohistochemistry and Western blot analysis were used to investigate SOX2 expression. LNA-modified siRNA was used for irreversible depletion of miR-21. For inhibition of PDGF signaling Gleevec (imatinib mesylate), Rapamycin and U0126, as well as siRNA were used. Statistical significance was calculated using double-sided unpaired Student's t-test. RESULTS: We identified miR-21 to be highly expressed during embryonic and newborn brain development followed by a gradual decrease until undetectable at postnatal day 7 (P7), this pattern correlated with SOX2 expression. Furthermore, miR-21 and SOX2 showed up-regulation and overlapping expression pattern in RCAS/tv-a generated mouse brain tumor specimens. Upon irreversible depletion of miR-21 the expression of SOX2 was strongly diminished in both mouse primary glioma cultures and human glioma cell lines. Interestingly, in normal fibroblasts the expression of miR-21 was induced by PDGF-BB, and inhibition of PDGF signaling in mouse glioma primary cultures resulted in suppression of miR-21 suggesting that miR-21 is indeed regulated by PDGF signaling. CONCLUSIONS: Our data show that miR-21 and SOX2 are tightly regulated already during embryogenesis and define a distinct population with putative tumor cell of origin characteristics. Furthermore, we believe that miR-21 is a mediator of PDGF-driven brain tumors, which suggests miR-21 as a promising target for treatment of glioma.


Asunto(s)
Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glioma/genética , MicroARNs/genética , Factores de Transcripción SOXB1/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/genética , Becaplermina , Northern Blotting , Western Blotting , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Línea Celular Transformada , Línea Celular Tumoral , Pollos , Glioma/metabolismo , Glioma/patología , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Interferencia de ARN , Transducción de Señal/genética , Trasplante Heterólogo
10.
Ups J Med Sci ; 117(2): 196-201, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22432595

RESUMEN

INTRODUCTION: The initial clinical experience from targeted therapy for breast cancer has been mixed. While important progress has been made in the care of a subset of patients characterized by amplification of HER2 through the use of trastuzumab, other targeted therapies have failed to improve the outcome for large, unselected groups of patients. Thus, efforts to find prognostic or predictive biomarkers to enable tailored therapy are highly warranted. Genetically engineered mouse models of human cancer provide a convenient setting in which to perform explorative studies. However, there is a paucity of comparative studies between mouse and human tumours in order to validate the use of mouse models as discovery tools. MATERIALS AND METHODS: Here, we have compared the localization of markers for cancer-associated fibroblasts in the MMTV-PyMT mouse model of mammary carcinoma with that of human breast cancer. The expression of α-smooth muscle actin, platelet-derived growth factor receptor-α, and fibroblast-specific protein-1 was assessed by immunostaining of sections from tumours of MMTV-PyMT mice. Information about the distribution of the same markers in human breast cancer was derived from the publicly available database the Human Protein Atlas. RESULTS: Both mouse and human mammary carcinomas were infused by a rich fibrotic stroma. While no marker was capable of identifying all stromal fibroblasts, the expression pattern of each marker was remarkably similar in mouse and human. DISCUSSION: We conclude that the MMTV-PyMT mouse model of breast cancer will have utility as a discovery tool for biomarkers of cancer-associated fibroblasts during malignant conversion.


Asunto(s)
Fibroblastos/patología , Neoplasias Mamarias Experimentales/patología , Animales , Humanos , Inmunohistoquímica , Neoplasias Mamarias Experimentales/virología , Virus del Tumor Mamario del Ratón/aislamiento & purificación , Ratones , Ratones Transgénicos
11.
Neuro Oncol ; 13(12): 1277-87, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21926087

RESUMEN

Faithful replication and DNA repair are vital for maintenance of genome integrity. RAD51 is a central protein in homologous recombination repair and during replication, when it protects and restarts stalled replication forks. Aberrant RAD51 expression occurs in glioma, and high expression has been shown to correlate with prolonged survival. Furthermore, genes involved in DNA damage response (DDR) are mutated or deleted in human glioblastomas, corroborating the importance of proper DNA repair to suppress gliomagenesis. We have analyzed DDR and genomic instability in PDGF-B-induced gliomas and investigated the role of RAD51 in glioma development. We show that PDGF-B-induced gliomas display genomic instability and that co-expression of RAD51 can suppress PDGF-B-induced tumorigenesis and prolong survival. Expression of RAD51 inhibited proliferation and genomic instability of tumor cells independent of Arf status. Our results demonstrate that the RAD51 pathway can prevent glioma initiation and maintain genome integrity of induced tumors, suggesting reactivation of the RAD51 pathway as a potential therapeutic avenue.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Inestabilidad Genómica , Glioma/prevención & control , Hiperplasia/prevención & control , Proteínas Proto-Oncogénicas c-sis/metabolismo , Recombinasa Rad51/metabolismo , Factor 1 de Ribosilacion-ADP/fisiología , Animales , Western Blotting , Encéfalo/citología , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Pollos , Daño del ADN , Reparación del ADN , Replicación del ADN , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Glioma/genética , Glioma/patología , Proteínas Fluorescentes Verdes/genética , Recombinación Homóloga , Humanos , Hiperplasia/genética , Hiperplasia/patología , Técnicas para Inmunoenzimas , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-sis/genética , Recombinasa Rad51/genética , Células Madre/metabolismo
12.
Blood ; 118(10): 2906-17, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21778339

RESUMEN

Endothelial cells (ECs) in blood vessels under formation are stabilized by the recruitment of pericytes, both in normal tissues and during angiogenesis in pathologic situations, including neoplasia. In the tumor vasculature, besides supporting the functionality of blood flow, pericytes protect ECs from antiangiogenic therapies, and have thus been implicated in clinical resistance to vascular targeting drugs. However, the molecular nature of the crosstalk between pericytes and ECs is largely unchartered. Herein, we identified pericyte-induced survival signals in ECs by isolation of vascular fragments derived from tumors that had been genetically or pharmacologically engineered to be either pericyte-rich or pericyte-poor. Pericytes induced the antiapoptotic protein Bcl-w in tumor ECs both in vivo and in vitro, thereby conveying protection from cytotoxic damage. The pericyte-dependent survival signaling in ECs was consequential to enforcement of an autocrine loop involving VEGF-A expression in ECs. Through molecular and functional studies, we delineated a signal transduction pathway in ECs downstream of integrin α(v) involving activation of NF-κB as the initiating event of the protective crosstalk from pericytes. Our elucidation of pericyte-derived pro-survival signaling in tumor ECs has potentially important implications for clinical development of antiangiogenic drugs, and suggests new therapeutic targets for rational multitargeting of cancer.


Asunto(s)
Apoptosis , Comunicación Autocrina , Endotelio Vascular/patología , Melanoma Experimental/patología , Neovascularización Patológica , Pericitos/patología , Proteínas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Proteínas Reguladoras de la Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Adhesión Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Proteínas Activadoras de GTPasa/fisiología , Perfilación de la Expresión Génica , Hibridación in Situ , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Pericitos/metabolismo , Proteínas/genética , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Vinblastina/farmacología
13.
BMC Mol Biol ; 12: 28, 2011 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-21733196

RESUMEN

BACKGROUND: CGGBP1 is a CGG-triplet repeat binding protein, which affects transcription from CGG-triplet-rich promoters such as the FMR1 gene and the ribosomal RNA gene clusters. Earlier, we reported some previously unknown functions of CGGBP1 in gene expression during heat shock stress response. Recently we had found CGGBP1 to be a cell cycle regulatory midbody protein required for normal cytokinetic abscission in normal human fibroblasts, which have all the cell cycle regulatory mechanisms intact. RESULTS: In this study we explored the role of CGGBP1 in the cell cycle in various cancer cell lines. CGGBP1 depletion by RNA interference in tumor-derived cells caused an increase in the cell population at G0/G1 phase and reduced the number of cells in the S phase. CGGBP1 depletion also increased the expression of cell cycle regulatory genes CDKN1A and GAS1, associated with reductions in histone H3 lysine 9 trimethylation in their promoters. By combining RNA interference and genetic mutations, we found that the role of CGGBP1 in cell cycle involves multiple mechanisms, as single deficiencies of CDKN1A, GAS1 as well as TP53, INK4A or ARF failed to rescue the G0/G1 arrest caused by CGGBP1 depletion. CONCLUSIONS: Our results show that CGGBP1 expression is important for cell cycle progression through multiple parallel mechanisms including the regulation of CDKN1A and GAS1 levels.


Asunto(s)
Ciclo Celular , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Neoplasias/patología , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ADN/genética , Fase G1 , Proteínas Ligadas a GPI/genética , Humanos , Ratones , Neoplasias/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/genética , Fase de Descanso del Ciclo Celular , Fase S , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
14.
Thyroid ; 16(2): 143-50, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16676399

RESUMEN

Anaplastic thyroid carcinoma (ATC) is one of the most malignant tumors in humans, and currently there is no effective treatment. In the present study we investigated the effect of an endogenous estrogen metabolite, 2-methoxyestradiol (2-ME), on the growth of human ATC cells. 2-ME treatment had a strong growth inhibitory effect on five human ATC cell lines (HTh7, HTh 74, HTh83, C643, and SW1736), but showed no effect on one cell line (KAT-4). Cell cycle analysis of the growth-inhibited cells showed that 2-ME induced a G2/M-arrest, followed by an increased fraction of cells in sub-G1. Analysis of internucleosomal DNA laddering as well as DNA fragmentation in a terminal deoxynucleotide transferase-mediated dUTP nick-end labeling (TUNEL) assay demonstrated a high number of cells undergoing apoptosis after 2-ME treatment. An increased activation of caspase-3 and caspase-8 by 2-ME was observed, and inhibition of caspase-3 decreased the apoptotic effect. Addition of 2-ME increased activity of p38 mitogen-activated protein kinase (MAPK) in the sensitive HTh7 as well as the refractory KAT-4 cells, however, activation of stress-activated protein kinase/c-jun aminoterminal kinase (SAPK/JNK) was seen only in the HTh7 cells. Inhibitors of p38 MAPK and SAPK/JNK significantly attenuated the 2-ME effect. Taken together, our data demonstrate an antiproliferative and apoptotic effect of 2-ME on ATC cells involving activation of MAPKs.


Asunto(s)
Apoptosis , Carcinoma/tratamiento farmacológico , Carcinoma/metabolismo , Estradiol/análogos & derivados , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/metabolismo , 2-Metoxiestradiol , Antineoplásicos/farmacología , Western Blotting , Carcinoma/patología , Caspasa 3 , Caspasa 8 , Caspasas/metabolismo , Línea Celular Tumoral , Fragmentación del ADN , Estradiol/farmacología , Citometría de Flujo , Fase G1 , Humanos , Etiquetado Corte-Fin in Situ , Sistema de Señalización de MAP Quinasas , Modelos Estadísticos , Ósmosis , ARN Mensajero/metabolismo , Ribonucleasas/metabolismo , Neoplasias de la Tiroides/patología , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Lab Invest ; 85(4): 512-21, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15711566

RESUMEN

A pathologically elevated interstitial fluid pressure (IFP) is a characteristic of both clinical and experimental carcinoma. The soluble TGF-beta receptor type II-murine Fc:IgG2A chimeric protein (Fc:TbetaRII) lowers IFP in the KAT-4 experimental model for anaplastic thyroid carcinoma. Analyses of messenger RNA (mRNA) expressions by Affymetrix microarrays and RNase protection assays, as well as of protein expressions identified tumor macrophages as targets for Fc:TbetaRII. Treatment with Fc:TbetaRII reduced albumin extravasation, increased coverage of alpha-smooth muscle actin-positive cells and reduced expression of NG2, a marker of activated pericytes, in KAT-4 carcinoma blood vessels. Specific inhibition of interleukin-1 (IL-1), a major cytokine produced by activated macrophages, lowered carcinoma IFP to a similar degree as Fc:TbetaRII but had no significant effect on the parameters of blood vessel maturation. Neither Fc:TbetaRII nor inhibition of IL-1 changed blood vessel density. Finally, pretreatment of KAT-4 carcinomas with Fc:TbetaRII increased the antitumor efficacy of doxorubicin. Our data emphasize a potential role of tumor macrophages in carcinoma physiology and identify these cells as potential stromal targets for treatment aimed to improve efficacy of chemotherapy.


Asunto(s)
Regulación hacia Abajo/fisiología , Líquido Extracelular , Macrófagos/citología , Neoplasias de la Tiroides/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Doxorrubicina/farmacología , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos
16.
Biochem Biophys Res Commun ; 305(4): 1017-23, 2003 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-12767932

RESUMEN

Mechanism(s) for generation of the high tumor interstitial fluid pressure (TIFP) that is characteristic of carcinoma is not known. We investigated the role of hyaluronan, the major water-binding polysaccharide of the extracellular matrix, for the generation of a high TIFP. A human anaplastic thyroid carcinoma (KAT-4) xenografted to athymic mice and a syngeneic rat colon carcinoma (PROb) were used. Neither KAT-4 nor PROb cells produced hyaluronan (HA) in culture, however, both cell lines produced factors that stimulated HA-synthesis by cultured fibroblasts. Modulating hyaluronan levels by transfection of PROb carcinoma cells with hyaluronan synthase-2 revealed no correlation between hyaluronan content and TIFP. Furthermore, lowering of TIFP by treating KAT-4 tumors with a specific inhibitor of TGF-beta 1 and -beta 3 did not change the concentration of hyaluronan in the tumors. In summary, our results suggest that a modulation of hyaluronan content is not a major pathogenetic mechanism for the generation of the characteristically high TIFP in malignant carcinomas.


Asunto(s)
Carcinoma/química , Ácido Hialurónico/análisis , Animales , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular , Neoplasias Colorrectales/química , Neoplasias Colorrectales/metabolismo , Espacio Extracelular , Femenino , Fibroblastos/metabolismo , Humanos , Ácido Hialurónico/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Presión , Ratas , Neoplasias de la Tiroides/química , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta3 , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int J Cancer ; 102(5): 453-62, 2002 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-12432546

RESUMEN

A high tumor interstitial fluid pressure (TIFP) is a pathologic characteristic distinguishing the stroma of carcinomas from normal interstitial loose connective tissues. The role of TGF-beta1 and -beta3 in generating a high TIFP was investigated in xenografted experimental anaplastic thyroid carcinoma (ATC) derived from the human ATC cell line KAT-4. A single intravenous injection of a soluble recombinant TGF-beta receptor type II-murine Fc:IgG(2A) chimeric protein that specifically inhibits TGF-beta1 and -beta3, significantly lowered TIFP in a time and concentration dependent manner but did not change total tissue water content in the tumors. Tumor growth rate was higher in tumors treated with the TGF-beta1 and -beta3 inhibitor compared to control tumors during the first 10 days after administration of the inhibitor. The apoptotic index of carcinoma cells, and expression of the cell cycle inhibitor p27(Kip1), were, however, increased in TGF-beta1 and -beta3 inhibitor-treated tumors. Prolonged treatment periods and administration of a second dose of the inhibitor decreased tumor growth rate. The TGF-beta1 and -beta3 inhibitor did not affect proliferation or expression of phosphorylated Smad2 protein in KAT-4 cells cultured in vitro. Our results indicate that members of the TGF-beta family are potential targets for novel anti-cancer treatment directed to the stroma. First by controlling TIFP and by that potentially the uptake of anticancer drugs into tumors and second by their suggested role in maintaining a supportive tumor stroma.


Asunto(s)
Carcinoma/metabolismo , Espacio Extracelular/fisiología , Neoplasias de la Tiroides/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Apoptosis , Carcinoma/patología , Proteínas de Ciclo Celular/metabolismo , División Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Humanos , Ratones , Trasplante de Neoplasias , Presión , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta , Proteínas Recombinantes de Fusión , Células del Estroma/metabolismo , Neoplasias de la Tiroides/patología , Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta3 , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...