Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Br J Cancer ; 117(1): 65-77, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28524160

RESUMEN

BACKGROUND: The HMGA2 protein has experimentally been linked to EMT and cancer stemness. Recent studies imply that tumour-stroma interactions regulate these features and thereby contribute to tumour aggressiveness. METHODS: We analysed 253 cases of pancreatic ductal adenocarcinoma (PDAC) and 155 cases of ampullary adenocarcinoma (AAC) for HMGA2 expression by IHC. The data were correlated with stroma abundance and supplemented by experimental studies. RESULTS: HMGA2 acts as an independent prognostic marker associated with a significantly shorter overall survival in both tumour types. Overall, HMGA2-positivity was more frequent in patients with PDAC than with AAC. The HMGA2 status in tumour cells significantly correlated with the abundance of PDGFRß-defined stroma cells. In vivo co-injection of Panc-1 cancer cells with pancreatic stellate cells increased tumour growth in a manner associated with increased HMGA2 expression. Furthermore, in vitro treatment of Panc-1 with conditioned media from PDGF-BB-activated stellate cells increased their ability to form tumour spheroids. CONCLUSIONS: This study identifies HMGA2 expression in tumour cells as an independent prognostic marker in PDAC and AAC. Correlative data analysis gives novel tissue-based evidence for a heterotypic cross-talk with stroma cells as a possible mechanism for HMGA2 induction, which is further supported by experimental models.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias del Conducto Colédoco/metabolismo , Proteína HMGA2/metabolismo , Neoplasias Pancreáticas/metabolismo , Adenocarcinoma/patología , Anciano , Ampolla Hepatopancreática , Animales , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Neoplasias del Conducto Colédoco/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Análisis Multivariante , Estadificación de Neoplasias , Trasplante de Neoplasias , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/metabolismo , Pronóstico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/metabolismo , Tasa de Supervivencia
2.
Breast Cancer Res Treat ; 158(2): 253-61, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27349429

RESUMEN

Following female sex and age, mammographic density is considered one of the strongest risk factors for breast cancer. Despite the association between mammographic density and breast cancer risk, little is known about the underlying histology and biological basis of breast density. To better understand the mechanisms behind mammographic density we assessed morphology, proliferation and hormone receptor status in relation to mammographic density in breast tissues from healthy women. Tissues were obtained from 2012-2013 by ultrasound-guided core needle biopsy from 160 women as part of the Karma (Karolinska mammography project for risk prediction for breast cancer) project. Mammograms were collected through routine mammography screening and mammographic density was calculated using STRATUS. The histological composition, epithelial and stromal proliferation status and hormone receptor status were assessed through immunohistochemical staining. Higher mammographic density was significantly associated with a greater proportion of stromal and epithelial tissue and a lower proportion of adipose tissue. Epithelial expression levels of Ki-67, oestrogen receptor (ER) and progesterone receptor (PR) were not associated with mammographic density. Epithelial Ki-67 was associated with a greater proportion of epithelial tissue, and epithelial PR was associated with a greater proportion of stromal and a lower proportion of adipose tissue. Epithelial ER was not associated with any tissues. In contrast, expression of ER in the stroma was significantly associated with a greater proportion of stroma, and negatively associated with the amount of adipose tissue. High mammographic density is associated with higher amount of stroma and epithelium and less amount of fat, but is not associated with a change in epithelial proliferation or receptor status. Increased expressions of both epithelial PR and stromal ER are associated with a greater proportion of stroma, suggesting hormonal involvement in regulating breast tissue composition.


Asunto(s)
Mama/diagnóstico por imagen , Mama/patología , Mamografía/métodos , Receptores de Estrógenos/metabolismo , Células del Estroma/metabolismo , Adulto , Anciano , Biopsia con Aguja Gruesa , Mama/metabolismo , Densidad de la Mama , Proliferación Celular , Femenino , Humanos , Persona de Mediana Edad , Ultrasonografía Mamaria
3.
Semin Cancer Biol ; 25: 61-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24560651

RESUMEN

Prognostication is an integral part of cancer diagnostic and helps oncologists to guide treatment decisions and therapy intensity. Accumulating evidence suggest that the stroma compartment also contains independent prognostic information, best exemplified by the impact of immune cells and cells of the vasculature on cancer progression. Similarly, strong experimental evidence exist that stromal fibroblasts, often designated cancer associated fibroblasts (CAFs), are actively involved in tumorigenesis. Thus, it can be anticipated that the molecular repertoire of CAFs is likewise important for the clinical behavior of the tumor. In this review we present recent studies addressing the prognostic impact of CAFs, with the focus on human lung and breast cancer. Several single markers have been suggested, either CAF specific or CAF derived, that in immunohistochemical studies have demonstrated independent association with survival. This includes members of the platelet derived growth factor receptor (PDGFR) family, CAF-markers like podoplanin and fibroblast activation protein (FAP) as well as transcription factors (FoxF1) and secreted factors (matrix metalloproteinases (MMPs), SPARC). However, most studies are based on explorative evaluations on single patient cohorts and require further validation. Using a more comprehensive approach, microarray studies have been employed to create gene expression signatures that detect an activated fibroblast state. These "stroma signatures" have been applied to identify specific CAF features associated with prognosis in several independent data sets of breast and lung cancer patients. Early studies in breast cancer have also demonstrated that fibroblast features influence therapy response. Thus, many strategies have been used to present encouraging proof-of-concept findings that CAFs could be exploited for prognostication. However, these studies also highlight the difficulties to conclusively define an "activated stroma" and to identify the individual factors involved in clinically relevant tumor-stroma interactions.


Asunto(s)
Neoplasias de la Mama/patología , Fibroblastos/fisiología , Neoplasias Pulmonares/patología , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Supervivencia Celular , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Pronóstico , Resultado del Tratamiento , Microambiente Tumoral
4.
Am J Pathol ; 182(6): 2037-47, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583284

RESUMEN

In this study, we describe a novel gene expression signature of platelet-derived growth factor (PDGF)-activated fibroblasts, which is able to identify breast cancers with a PDGF-stimulated fibroblast stroma and displays an independent and strong prognostic significance. Global gene expression was compared between PDGF-stimulated human fibroblasts and cultured resting fibroblasts. The most differentially expressed genes were reduced to a gene expression signature of 113 genes. The biological significance and prognostic capacity of this signature were investigated using four independent clinical breast cancer data sets. Concomitant high expression of PDGFß receptor and its cognate ligands is associated with a high PDGF signature score. This supports the notion that the signature detects tumors with PDGF-activated stroma. Subsequent analyses indicated significant associations between high PDGF signature score and clinical characteristics, including human epidermal growth factor receptor 2 positivity, estrogen receptor negativity, high tumor grade, and large tumor size. A high PDGF signature score is associated with shorter survival in univariate analysis. Furthermore, the high PDGF signature score acts as a significant marker of poor prognosis in multivariate survival analyses, including classic prognostic markers, Ki-67 status, a proliferation gene signature, or other recently described stroma-derived gene expression signatures.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células Cultivadas , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/fisiología , Genes Relacionados con las Neoplasias , Humanos , Estimación de Kaplan-Meier , Ligandos , Persona de Mediana Edad , Clasificación del Tumor , Proteínas de Neoplasias/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-sis/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Células del Estroma/metabolismo
5.
Future Oncol ; 10(9): 1695-708, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25145436

RESUMEN

PDGF receptors (PDGFRs) exert cell type-specific effects in many different tumor types. They are emerging as key regulators of mesenchymal cells of the tumor microenvironment, and of many common malignancies, such as cancer of the breast, colon and prostate. In some tumor types PDGFRs are genetically activated and are thus directly involved in stimulation of malignant cell growth. Recent studies have uncovered clinically relevant variations in stromal PDGFR expression. High stromal PDGFRß expression or activation is associated with poor prognosis in breast and prostate cancer. Indications of prognostic significance of stromal PDGFRß expression in various GI tract tumor types also exist. The prognostic significance of PDGFRα and ß in malignant cells of common epithelial tumor types should be further studied. Collectively data suggest that continued characterization of PDGFR expression in human tumors should present opportunities for improved accuracy in prognosis and also allow novel biomarker-based clinical studies exploring the efficacy of PDGFR-directed tumor therapies.


Asunto(s)
Biomarcadores de Tumor/fisiología , Neoplasias/metabolismo , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Humanos , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/patología , Pericitos/metabolismo , Pronóstico , Transducción de Señal
6.
Int J Cancer ; 128(8): 1981-8, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20589679

RESUMEN

Platelet-derived growth factor receptor (PDGFR) signaling has been implicated in the pathogenesis of glioblastomas and represents a target for the tyrosine kinase inhibitor imatinib. To examine the prognostic or predictive role of PDGFRs in recurrent glioblastomas, expression was examined in tumor samples of 101 patients of CSTI571BDE40, a randomized trial comparing hydroxyurea monotherapy and a combination of hydroxyurea and imatinib. Furthermore, PDGFRα phosphorylation was investigated using in situ proximity ligation assay. PDGFRα protein was expressed in 33% of tumors and was associated with male sex, young age, presence of R132H mutated isocitrate dehydrogenase 1 protein and short median survival (142 vs. 187 days, p = 0.028). Tumor PDGFRα phosphorylation was also associated with short survival (p = 0.030). The subset of patients with PDGFRα positive glioblastoma did not have longer survival on treatment with hydroxyurea and imatinib compared with hydroxyurea monotherapy. In conclusion, both PDGFRα protein expression and phosphorylation status had a prognostic role in recurrent glioblastomas but did not define a group that showed benefit from the combination therapy consisting of hydroxyurea and imatinib.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Benzamidas , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Resistencia a Antineoplásicos , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Hidroxiurea/administración & dosificación , Mesilato de Imatinib , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Fosforilación , Piperazinas/administración & dosificación , Pronóstico , Pirimidinas/administración & dosificación , Tasa de Supervivencia
7.
Am J Pathol ; 175(1): 334-41, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19498003

RESUMEN

This study systematically analyzes platelet-derived growth factor (PDGF) receptor expression in six types of common tumors as well as examines associations between PDGF beta-receptor status and clinicopathological characteristics in breast cancer. PDGF receptor expression was determined by immunohistochemistry on tumor tissue microarrays. Breast tumor data were combined with prognostic factors and related to outcome endpoints. PDGF alpha- and beta-receptors were independently expressed, at variable frequencies, in the tumor stroma of all tested tumor types. There was a significant association between PDGF beta-receptor expression on fibroblasts and perivascular cells in individual colon and prostate tumors. In breast cancer, high stromal PDGF beta-receptor expression was significantly associated with high histopathological grade, estrogen receptor negativity, and high HER2 expression. High stromal PDGF beta-receptor expression was correlated with significantly shorter recurrence-free and breast cancer-specific survival. The prognostic significance of stromal PDGF beta-receptor expression was particularly prominent in tumors from premenopausal women. Stromal PDGF alpha- and beta-receptor expression is a common, but variable and independent, property of solid tumors. In breast cancer, stromal PDGF beta-receptor expression significantly correlates with less favorable clinicopathological parameters and shorter survival. These findings highlight the prognostic significance of stromal markers and should be considered in ongoing clinical development of PDGF receptor inhibitors.


Asunto(s)
Neoplasias de la Mama/metabolismo , Tejido Conectivo/metabolismo , Fibroblastos/metabolismo , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-sis/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Neoplasias del Colon/metabolismo , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Premenopausia , Pronóstico , Neoplasias de la Próstata/metabolismo , Receptor ErbB-2/biosíntesis , Receptores de Estrógenos/biosíntesis , Análisis de Matrices Tisulares
8.
Am J Pathol ; 175(4): 1631-7, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19717644

RESUMEN

Choroid plexus tumors are intraventricular neoplasms predominantly affecting young children. In contrast to choroid plexus papillomas, choroid plexus carcinomas progress frequently, necessitating the development of adjuvant treatment concepts. Platelet derived growth factor (PDGF) signaling has been shown to support growth in a variety of tumors. The finding of PDGF receptor expression in choroid plexus tumors prompted us to elucidate PDGF receptor activation state using a novel method, in situ proximity ligation assay, on formalin-fixed, paraffin-embedded, archival samples of 19 choroid plexus tumors. As assessed by in situ proximity ligation assay, the proportion of phosphorylated PDGF receptor alpha was low in choroid plexus papillomas and choroid plexus carcinomas, whereas phosphorylated PDGF receptor beta was found to be significantly higher in choroid plexus carcinomas. In the immortalized choroid plexus epithelial cell line Z310 expressing PDGF receptor beta, PDGF-BB exhibited a time- and dose-dependent proliferative response, which was significantly attenuated by imatinib (gleevec). In conclusion, our findings suggest that PDGF receptor beta is functionally involved in the biology of choroid plexus tumors and may represent a molecular target for therapy. In addition, this study demonstrates the feasibility and usefulness of in situ proximity ligation assay for monitoring receptor tyrosine kinase activation in formalin-fixed, paraffin-embedded, archival tissues.


Asunto(s)
Neoplasias del Plexo Coroideo/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Línea Celular , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Neoplasias del Plexo Coroideo/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Inmunohistoquímica , Lactante , Masculino , Papiloma/metabolismo , Papiloma/patología , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas
9.
Int J Cancer ; 124(5): 1227-34, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19058199

RESUMEN

Imatinib is currently in early clinical trials as targeted therapy for relapsed neuroblastomas and other childhood solid tumors expressing platelet-derived growth factor receptors (PDGFR) or c-Kit. Short-term treatment with imatinib in clinically achievable concentrations is ineffective in neuroblastoma in vitro. However, clinically, imatinib is administered daily over long time periods. The effects of combining imatinib with chemotherapy in neuroblastoma are unknown. Here, a panel of neuroblastoma cell lines (n = 5) were studied, representing tumors with different biological (MYCN-amplification +/-) and clinical (drug resistance) features. Using a protracted low-dose treatment schedule (1-3 weeks; 0.5-5microM) imatinib dose-dependently inhibited proliferation and clonogenic survival for all tested cell lines with IC50 <2.5microM. In contrast, short-term treatment (<96 hrs) was ineffective. Low-dose imatinib was synergistic in combination with doxorubicin and caused increased G2/M- and S-phase arrest and apoptosis as evidenced by enhanced caspase-3 activation and sub-G1 DNA accumulation. A significant but less pronounced effect was observed when imatinib was combined with etoposide or vincristine, as opposed to cisplatin, melphalan, or irinotecan. All cell lines expressed PDGFRbeta, whereas no protein expression of PDGFRalpha was detected in MYCN amplified cell lines. PDGF-BB caused PDGFRbeta phosphorylation and partially rescued neuroblastoma cells from doxorubicin-induced apoptosis, in an imatinib-sensitive manner. In vivo, treatment with imatinib in combination with doxorubicin induced a significant growth inhibition of established neuroblastoma xenografts. These findings suggest clinical testing of imatinib in combination with selected chemotherapeutic drugs, in particular doxorubicin, in children with high-risk neuroblastoma.


Asunto(s)
Antineoplásicos/administración & dosificación , Neuroblastoma/tratamiento farmacológico , Piperazinas/administración & dosificación , Pirimidinas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Becaplermina , Benzamidas , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Esquema de Medicación , Humanos , Mesilato de Imatinib , Masculino , Neuroblastoma/patología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-sis , Ratas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología
10.
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
11.
Int J Cancer ; 122(7): 1548-56, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18033683

RESUMEN

Platelet-derived growth factors (PDGF) play a major role in pericyte recruitment in tumor capillaries. Pericytes are required for proper vessel development, and contribute to tumor angiogenesis by promoting stabilization and maturation of newly formed vessels. To investigate the effects of pericyte coverage on tumor vessel morphology and function in vivo, tumors derived from B16 melanoma cells transfected with either control plasmid (B16/ctr) or plasmid encoding full-length PDGF-BB (B16/PDGF), the latter previously shown to have enhanced blood vessel pericyte coverage and an increased tumor growth rate, were assessed using histopathological methods, Hoechst 33342-based perfusion analyses, and two noninvasive susceptibility magnetic resonance imaging (MRI) methods. Susceptibility-contrast MRI, incorporating the use of ultrasmall superparamagnetic iron oxide particles, revealed a significant (p < 0.05) reduction in vessel size index (R(v)) of B16/PDGF tumors, and which was validated histologically by the presence of significantly smaller (p < 0.001), more punctate blood vessels identified by fluorescence microscopy of the perfusion marker Hoechst 33342. Intrinsic-susceptibility MRI was used to measure the transverse MRI relaxation rate R(2)*, sensitive to changes in endogenous paramagnetic [deoxyhaemoglobin], and used to probe for vascular maturation and function. Hypercapnia (5% CO(2)/95% air) induced a negligible Delta R(2)* response in the B16/ctr and B16/PDGF tumors. In contrast, hyperoxia (5% CO(2)/95% O(2)) induced a significantly greater R(2)* reduction in the B16/PDGF tumors (p < 0.02). Together the susceptibility MRI-derived biomarkers reveal novel pericyte-dependent changes in the morphology and function of the perfused tumor vasculature in vivo.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Biomarcadores de Tumor/metabolismo , Imagen por Resonancia Magnética , Melanoma Experimental/irrigación sanguínea , Neovascularización Patológica/metabolismo , Pericitos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Becaplermina , Capilares/patología , Hipercapnia/metabolismo , Hiperoxia/metabolismo , Inmunohistoquímica , Masculino , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Proteínas Proto-Oncogénicas c-sis , Factores de Tiempo
12.
FASEB J ; 21(2): 523-34, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17158785

RESUMEN

Growth factor-dependent tissue remodeling, such as restenosis, is believed to be predominantly regulated by changes in expression of receptor-tyrosine-kinases (RTKs) and their ligands. As endogenous antagonists of RTKs, protein-tyrosine-phosphatases (PTPs) are additional candidate regulators of these processes. Using laser-capture-microdissection and quantitative RT-polymerase chain reaction (qRT-PCR), we investigated the layer-specific expression of the four platelet-derived growth factor (PDGF) isoforms, the PDGF-alpha and beta receptors, and five PTPs implied in control of PDGF-receptor signaling 8 and 14 days after balloon injury of the rat carotid. Results were correlated with analyses of PDGF-beta receptor phosphorylation and vascular smooth muscle cell (VSMC) proliferation in vivo. The expression levels of all components, as well as receptor activation and VSMC proliferation, showed specific changes, which varied between media and neointima. Interestingly, PTP expression--particularly, DEP-1 levels--appeared to be the dominating factor determining receptor-phosphorylation and VSMC proliferation. In support of these findings, cultured DEP-1(-/-) cells displayed increased PDGF-dependent cell signaling. Hyperactivation of PDGF-induced signaling was also observed after siRNA-down-regulation of DEP-1 in VSMCs. The results indicate a previously unrecognized role of PDGF-receptor-targeting PTPs in controlling neointima formation. In more general terms, the observations indicate transcriptional regulation of PTPs as an important mechanism for controlling onset and termination of RTK-dependent tissue remodeling.


Asunto(s)
Músculo Liso Vascular/metabolismo , Proteínas Tirosina Fosfatasas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Túnica Íntima/metabolismo , Animales , Traumatismos de las Arterias Carótidas/fisiopatología , Proliferación Celular , Quimiotaxis , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/citología , Fibroblastos/metabolismo , Expresión Génica , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Músculo Liso Vascular/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo , Túnica Íntima/patología
13.
Cancer Res ; 65(17): 7824-31, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16140951

RESUMEN

Whereas radioimmunotherapy of hematologic malignancies has evolved into a viable treatment option, the responses of solid tumors to radioimmunotherapy are discouraging. The likely cause of this problem is the interstitial hypertension inherent to all solid tumors. Remarkable improvements in tumor responses to radioimmunotherapy were discovered after the inclusion of STI571 in the therapy regimen. A combination of the tumor stroma-reactive STI571, a potent platelet-derived growth factor receptor-beta (PDGFr-beta) antagonist, and the tumor-seeking radiolabeled antibody B72.3 yielded long-lasting growth arrest of the human colorectal adenocarcinoma LS174T grown as s.c. xenografts in athymic mice. The interaction of STI571 with the stromal PDGFr-beta reduced tumor interstitial fluid pressure (P(IF)) by >50% and in so doing improved the uptake of B72.3. The attenuation of P(IF) also had a positive effect on the homogeneity of antibody distribution. These effects were dose-dependent and under optimized dosing conditions allowed for a 2.45 times increase in the tumor uptake of B72.3 as determined in the biodistribution studies. Single-photon emission computed tomography imaging studies substantiated these results and indicated that the homogeneity of the radioisotope distribution was also much improved when compared with the control mice. The increased uptake of radioimmunotherapy into the tumor resulted in >400% increase in the tumor absorbed radiation doses in STI571 + radioimmunotherapy-treated mice compared with PBS + radioimmunotherapy-treated mice. The improved antibody uptake in response to the attenuation of tumor P(IF) was identified as the primary reason for the growth arrest of the STI571 + radioimmunotherapy-treated tumors. Two related causes were also identified: (a) the improved homogeneity of monoclonal antibody distribution in tumor and (b) the increased tumor radiosensitivity resulting from the improved tumor oxygenation.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias Colorrectales/terapia , Piperazinas/farmacología , Pirimidinas/farmacología , Radioinmunoterapia/métodos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Adenocarcinoma/enzimología , Adenocarcinoma/metabolismo , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/metabolismo , Anticuerpos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Benzamidas , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Mesilato de Imatinib , Inmunotoxinas/farmacocinética , Inmunotoxinas/farmacología , Radioisótopos de Yodo/farmacología , Ratones , Ratones Desnudos , Inhibidores de Proteínas Quinasas/farmacología , Porcinos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto , Radioisótopos de Itrio/farmacología
14.
J Pathol Clin Res ; 3(1): 38-43, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28138400

RESUMEN

Cancer-associated fibroblasts (CAFs) regulate tumour growth, metastasis and response to treatment. Recent studies indicate the existence of functionally distinct CAF subsets. Suggested mechanisms whereby CAFs can impact on treatment response include paracrine signalling affecting cancer cell drug sensitivity and effects on tumour drug uptake. PDGFRß is an important regulator of fibroblasts. Experimental studies have linked PDGFRß-positive fibroblasts to metastasis and also to reduced tumour drug uptake. This study has investigated the potential role of PDGFRß-positive fibroblasts in response to adjuvant tamoxifen treatment of breast cancer. Analyses of two breast cancer collections from randomised studies analysing adjuvant tamoxifen treatment in early breast cancer demonstrated significant benefit of tamoxifen in the group with low stromal PDGFRß, which was not observed in the group with high stromal PDGFRß. In general terms these findings provide novel evidence, derived from analyses of randomised clinical studies, of response-predictive capacity of a marker-defined subset of CAFs and, more specifically, identify stromal PDGFRß as a marker related to tamoxifen benefit in early breast cancer.

15.
Neuro Oncol ; 17(7): 953-64, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25534822

RESUMEN

BACKGROUND: Platelet-derived growth factor receptor A is altered by amplification and/or mutation in diffuse intrinsic pontine glioma (DIPG). We explored in vitro on new DIPG models the efficacy of dasatinib, a multi-tyrosine kinase inhibitor targeting this receptor. METHODS: Gene expression profiles were generated from 41 DIPGs biopsied at diagnosis and compared with the signature associated with sensitivity/resistance to dasatinib. A panel of 12 new DIPG cell lines were established from biopsy at diagnosis, serially passaged, and characterized by gene expression analyses. Effects of dasatinib (1-10 µM) on proliferation, invasion, and cytotoxicity were determined on 4 of these cell lines using live-cell imaging and flow cytometry assays. Downstream signaling and receptor tyrosine kinases (RTKs) were assessed by western blot and phospho-RTK array. The effect of the combination with the c-Met inhibitor cabozantinib was studied on cellular growth and invasion analyzed by the Chou-Talaly method. RESULTS: DIPG primary tumors and cell lines exhibited the gene expression signature of sensitivity to dasatinib. Dasatinib reduced proliferation (half-maximal inhibitory concentration = 10-100 nM) and invasion (30%-60% reduction) at 100 nM in 4/4 cultures and induced apoptosis in 1 of 4 DIPG cell lines. Activity of downstream effectors of dasatinib targets including activin receptor 1 was strongly reduced. Since multiple RTKs were activated simultaneously in DIPG cell lines, including c-Met, which can be also amplified in DIPG, the benefit of the combination of dasatinib with cabozantinib was explored for its synergistic effects on proliferation and migration/invasion in these cell lines. CONCLUSION: Dasatinib exhibits antitumor effects in vitro that could be increased by the combination with another RTK inhibitor targeting c-Met.


Asunto(s)
Anilidas/farmacología , Antineoplásicos/farmacología , Neoplasias del Tronco Encefálico/metabolismo , Dasatinib/farmacología , Glioma/metabolismo , Piridinas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Anilidas/uso terapéutico , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Niño , Preescolar , Dasatinib/uso terapéutico , Femenino , Glioma/tratamiento farmacológico , Humanos , Masculino , Invasividad Neoplásica/prevención & control , Puente/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/metabolismo , Piridinas/uso terapéutico , Transducción de Señal/efectos de los fármacos
16.
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
17.
PLoS One ; 5(5): e10747, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20505768

RESUMEN

BACKGROUND: The identification of new prognostic markers for prostate cancer is highly warranted, since it is difficult to identify patients requiring curative treatment. Data from both experimental models and clinical samples have identified important functions of PDGFRbeta on pericytes and fibroblasts in the tumor stroma. METHODOLOGY/PRINCIPAL FINDINGS: In this study the prognostic significance of PDGFRbeta in prostate cancer stroma, and in matched non-malignant tissue, was evaluated with immunohistochemistry. PDGFRbeta expression was analyzed in normal and tumor stroma from more than 300 prostate cancer patients. High PDGFRbeta expression in tumor stroma was associated with large tumor size, advanced stage, high Gleason score and high vessel density. Perivascular PDGFRbeta staining in tumors was also correlated with high Gleason score. Correlations were also observed between PDGFRbeta status in tumor stroma and non-malignant stroma. Similarly, high PDGFRbeta expression in adjacent non-malignant tissue stroma correlated with large tumor size, advanced stage, high Gleason score and proliferation in non-malignant epithelium. Interestingly, high levels of PDGFRbeta in the stroma of tumor and non-malignant tissue were associated with shorter cancer specific survival in prostate cancer patients. CONCLUSIONS/SIGNIFICANCE: The study revealed a number of novel associations between stromal PDGFRbeta expression in prostate tumors and several important clinical characteristics, including survival.


Asunto(s)
Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Actinas/metabolismo , Biomarcadores de Tumor/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Humanos , Masculino , Pronóstico , Modelos de Riesgos Proporcionales , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/patología , Análisis de Regresión , Coloración y Etiquetado , Células del Estroma/metabolismo , Células del Estroma/patología , Análisis de Supervivencia
18.
Cancer Res ; 70(7): 2644-54, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20233876

RESUMEN

Cancer-associated fibroblasts (CAF) attract increasing attention as potential cancer drug targets due to their ability to stimulate, for example, tumor growth, invasion, angiogenesis, and metastasis. However, the molecular mechanisms causing the tumor-promoting properties of CAFs remain poorly understood. Forkhead box F1 (FoxF1) is a mesenchymal target of hedgehog signaling, known to regulate mesenchymal-epithelial interactions during lung development. Studies with FoxF1 gain- and loss-of-function fibroblasts revealed that FoxF1 regulates the contractility of fibroblasts, their production of hepatocyte growth factor and fibroblast growth factor-2, and their stimulation of lung cancer cell migration. FoxF1 status of fibroblasts was also shown to control the ability of fibroblasts to stimulate xenografted tumor growth. FoxF1 was expressed in CAFs of human lung cancer and associated with activation of hedgehog signaling. These observations suggest that hedgehog-dependent FoxF1 is a clinically relevant lung CAF-inducing factor, and support experimentally the general concept that CAF properties can be induced by activation of developmentally important transcription factors.


Asunto(s)
Fibroblastos/patología , Factores de Transcripción Forkhead/fisiología , Neoplasias Pulmonares/patología , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Fibroblastos/metabolismo , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Proteínas Hedgehog/metabolismo , Factor de Crecimiento de Hepatocito/biosíntesis , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Células 3T3 NIH , Transducción de Señal , Transfección
19.
Mod Pathol ; 21(3): 265-70, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18157090

RESUMEN

Platelet-derived growth factor (PDGF) receptor signaling has been implicated in the development of glial tumors, but not yet been examined in choroid plexus carcinomas, pediatric tumors with dismal prognosis for which novel treatment options would be desirable. Therefore, protein expression of PDGF receptors alpha and beta as well as amplification status of the respective genes, PDGFRA and PDGFRB, were examined in a series of 22 patients harboring choroid plexus carcinoma using immunohistochemistry and chromogenic in situ hybridization (CISH). The majority of choroid plexus carcinomas expressed PDGF receptors with 6 cases (27%) displaying high staining scores for PDGF receptor alpha and 13 cases (59%) showing high staining scores for PDGF receptor beta. Correspondingly, copy-number gains of PDGFRA were observed in 8 cases out of 12 cases available for CISH and 1 case displayed amplification (six or more signals per nucleus). The proportion of choroid plexus carcinomas with amplification of PDGFRB was even higher (5/12 cases). PDGFRB amplification status and PDGF receptor beta protein expression scores were significantly correlated (P=0.01, Spearman). Expression status of PDGF receptor alpha or PDGF receptor beta was not significantly associated with progression-free survival. To conclude, expression and amplification of PDGF receptors, particularly PDGF receptor beta, are frequent in choroid plexus carcinomas, providing a first rationale for the development of treatments targeting PDGF receptor signaling in these rare malignant pediatric tumors.


Asunto(s)
Carcinoma/metabolismo , Neoplasias del Plexo Coroideo/metabolismo , Amplificación de Genes , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Biomarcadores de Tumor , Carcinoma/genética , Preescolar , Neoplasias del Plexo Coroideo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Lactante , Masculino , Pronóstico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética
20.
Mol Cell Proteomics ; 6(9): 1500-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17565975

RESUMEN

Improved methods are needed for in situ characterization of post-translational modifications in cell lines and tissues. For example, it is desirable to monitor the phosphorylation status of individual receptor tyrosine kinases in samples from human tumors treated with inhibitors to evaluate therapeutic responses. Unfortunately the leading methods for observing the dynamics of tissue post-translational modifications in situ, immunohistochemistry and immunofluorescence, exhibit limited sensitivity and selectivity. Proximity ligation assay is a novel method that offers improved selectivity through the requirement of dual recognition and increased sensitivity by including DNA amplification as a component of detection of the target molecule. Here we therefore established a generalized in situ proximity ligation assay to investigate phosphorylation of platelet-derived growth factor receptor beta (PDGFRbeta) in cells stimulated with platelet-derived growth factor BB. Antibodies specific for immunoglobulins from different species, modified by attachment of DNA strands, were used as secondary proximity probes together with a pair of primary antibodies from the corresponding species. Dual recognition of receptors and phosphorylated sites by the primary antibodies in combination with the secondary proximity probes was used to generate circular DNA strands; this was followed by signal amplification by replicating the DNA circles via rolling circle amplification. We detected tyrosine phosphorylated PDGFRbeta in human embryonic kidney cells stably overexpressing human influenza hemagglutinin-tagged human PDGFRbeta in porcine aortic endothelial cells transfected with the beta-receptor, but not in cells transfected with the alpha-receptor, and also in immortalized human foreskin fibroblasts, BJ hTert, endogenously expressing the PDGFRbeta. We furthermore visualized tyrosine phosphorylated PDGFRbeta in tissue sections from fresh frozen human scar tissue undergoing wound healing. The method should be of great value to study signal transduction, screen for effects of pharmacological agents, and enhance the diagnostic potential in histopathology.


Asunto(s)
Proteómica/métodos , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Actinas/metabolismo , Línea Celular , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunoglobulinas/química , Inmunohistoquímica/métodos , Riñón/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Transducción de Señal , Tirosina/química , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda