Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cancer Cell Int ; 21(1): 507, 2021 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-34548097

RESUMEN

BACKGROUND: The Discoidin Domain Receptor 1 (DDR1) is one of the two members of a unique family of receptor tyrosine kinase receptors that signal in response to collagen, which has been implicated in cancer progression. Here, we examined the expression of DDR1 in prostate cancer (PCa), and assessed its potential value as a prognostic marker, as a function of grade, stage and other clinicopathologic parameters. METHODS: We investigated the association between the expression level and subcellular localization of DDR1 protein and PCa aggressiveness by immunohistochemistry, using tissue microarrays (TMAs) encompassing 200 cases of PCa with various Gleason scores (GS) and pathologic stages with matched normal tissue, and a highly specific monoclonal antibody. RESULTS: DDR1 was found to be localized in the membrane, cytoplasm, and nuclear compartments of both normal and cancerous prostate epithelial cells. Analyses of DDR1 expression in low GS (≤ 7[3 + 4]) vs high GS (≥ 7[4 + 3]) tissues showed no differences in nuclear or cytoplasmic DDR1in either cancerous or adjacent normal tissue cores. However, relative to normal-matched tissue, the percentage of cases with higher membranous DDR1 expression was significantly lower in high vs. low GS cancers. Although nuclear localization of DDR1 was consistently detected in our tissue samples and also in cultured human PCa and normal prostate-derived cell lines, its presence in that site could not be associated with disease aggressiveness. No associations between DDR1 expression and overall survival or biochemical recurrence were found in this cohort of patients. CONCLUSION: The data obtained through multivariate logistic regression model analysis suggest that the level of membranous DDR1 expression status may represent a potential biomarker of utility for better determination of PCa aggressiveness.

2.
J Transl Med ; 16(1): 148, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29859097

RESUMEN

BACKGROUND: Discoidin domain receptor 1 (DDR1) is a collagen-activated receptor tyrosine kinase extensively implicated in diseases such as cancer, atherosclerosis and fibrosis. Multiple preclinical studies, performed using either a gene deletion or a gene silencing approaches, have shown this receptor being a major driver target of fibrosis and glomerulosclerosis. METHODS: The present study investigated the role and relevance of DDR1 in human crescentic glomerulonephritis (GN). Detailed DDR1 expression was first characterized in detail in human GN biopsies using a novel selective anti-DDR1 antibody using immunohistochemistry. Subsequently the protective role of DDR1 was investigated using a highly selective, novel, small molecule inhibitor in a nephrotoxic serum (NTS) GN model in a prophylactic regime and in the NEP25 GN mouse model using a therapeutic intervention regime. RESULTS: DDR1 expression was shown to be mainly limited to renal epithelium. In humans, DDR1 is highly induced in injured podocytes, in bridging cells expressing both parietal epithelial cell (PEC) and podocyte markers and in a subset of PECs forming the cellular crescents in human GN. Pharmacological inhibition of DDR1 in NTS improved both renal function and histological parameters. These results, obtained using a prophylactic regime, were confirmed in the NEP25 GN mouse model using a therapeutic intervention regime. Gene expression analysis of NTS showed that pharmacological blockade of DDR1 specifically reverted fibrotic and inflammatory gene networks and modulated expression of the glomerular cell gene signature, further validating DDR1 as a major mediator of cell fate in podocytes and PECs. CONCLUSIONS: Together, these results suggest that DDR1 inhibition might be an attractive and promising pharmacological intervention for the treatment of GN, predominantly by targeting the renal epithelium.


Asunto(s)
Receptor con Dominio Discoidina 1/antagonistas & inhibidores , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/prevención & control , Adulto , Anciano , Anciano de 80 o más Años , Animales , Receptor con Dominio Discoidina 1/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/patología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Glomerulonefritis/genética , Glomerulonefritis/patología , Humanos , Inflamación/patología , Riñón/patología , Masculino , Ratones , Persona de Mediana Edad , Fenotipo
3.
J Transl Med ; 14: 72, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26975354

RESUMEN

BACKGROUND: Characterization of genes linked to bone metastasis is critical for identification of novel prognostic or predictive biomarkers and potential therapeutic targets in metastatic castrate-resistant prostate cancer (mCRPC). Although bone marrow core biopsies (BMBx) can be obtained for gene profiling, the procedure itself is invasive and uncommon practice in mCRPC patients. Conversely, circulating tumor cells (CTCs), which are likely to stem from bone metastases, can be isolated from blood. The goals of this exploratory study were to establish a sensitive methodology to analyze gene expression in BMBx and CTCs, and to determine whether the presence or absence of detectable gene expression is concordant in matching samples from mCRPC patients. METHODS: The CellSearch(®) platform was used to enrich and enumerate CTCs. Low numbers of PC3 prostate cancer (PCa) cells were spiked into normal blood to assess cell recovery rate. RNA extracted from recovered PC3 cells was amplified using an Eberwine-based procedure to obtain antisense mRNA (aRNA), and assess the linearity of the RNA amplification method. In this pilot study, RNAs extracted from CTCs and PCa cells microdissected from formalin-fixed paraffin-embedded BMBx, were amplified to obtain aRNA and assess the expression of eight genes functionally relevant to PCa bone metastasis using RT-PCR. RESULTS: RNAs were successfully extracted from as few as 1-5 PCa cells in blood samples. The relative expression levels of reference genes were maintained after RNA amplification. The integrity of the amplified RNA was also demonstrated by RT-PCR analysis using primer sets that target the 5'-end, middle, and 3'-end of reference mRNA. We found that in 21 out of 28 comparisons, the presence or absence of detectable gene expression in CTCs and PCa cells microdissected from single bone lesions of the same patients was concordant. CONCLUSIONS: This exploratory analysis suggests that aRNA amplification through in vitro transcription may be useful as a method to detect gene expression in small numbers of CTCs and tumor cells microdissected from bone metastatic lesions. In some cases, gene expression in CTCs and BMBxs was not concordant, raising questions about using CTC gene expression to make clinical decisions.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células Neoplásicas Circulantes/patología , Neoplasias de la Próstata Resistentes a la Castración/patología , Anciano , Anciano de 80 o más Años , Biopsia , Médula Ósea/patología , Línea Celular Tumoral , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata Resistentes a la Castración/genética , ARN sin Sentido/genética , ARN sin Sentido/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Reproducibilidad de los Resultados
4.
Int J Cancer ; 136(1): 11-20, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24798488

RESUMEN

Loss of BRCA2 function stimulates prostate cancer (PCa) cell invasion and is associated with more aggressive and metastatic tumors in PCa patients. Concurrently, the receptor tyrosine kinase c-kit is highly expressed in skeletal metastases of PCa patients and induced in PCa cells placed into the bone microenvironment in experimental models. However, the precise requirement of c-kit for intraosseous growth of PCa and its relation to BRCA2 expression remain unexplored. Here, we show that c-kit expression promotes migration and invasion of PCa cells. Alongside, we found that c-kit expression in PCa cells parallels BRCA2 downregulation. Gene rescue experiments with human BRCA2 transgene in c-kit-transfected PCa cells resulted in reduction of c-kit protein expression and migration and invasion, suggesting a functional significance of BRCA2 downregulation by c-kit. The inverse association between c-kit and BRCA2 gene expressions in PCa cells was confirmed using laser capture microdissection in experimental intraosseous tumors and bone metastases of PCa patients. Inhibition of bone-induced c-kit expression in PCa cells transduced with lentiviral short hairpin RNA reduced intraosseous tumor incidence and growth. Overall, our results provide evidence of a novel pathway that links bone-induced c-kit expression in PCa cells to BRCA2 downregulation and supports bone metastasis.


Asunto(s)
Neoplasias Óseas/enzimología , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Neoplasias Óseas/secundario , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo
5.
Am J Pathol ; 180(3): 1017-1027, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22209699

RESUMEN

Platelet-derived growth factor (PDGF) family members are potent growth factors that regulate cell proliferation, migration, and transformation. Clinical studies have shown that both PDGF receptor ß (ß-PDGFR) and its ligand PDGF D are up-regulated in primary prostate cancers and bone metastases, whereas PDGF B, a classic ligand for ß-PDGFR, is not frequently detected in clinical samples. In this study, we examined the role of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN) in the regulation of PDGF expression levels using both a prostate-specific, conditional PTEN-knockout mouse model and mouse prostate epithelial cell lines established from these mice. We found an increase in PDGF D and ß-PDGFR expression levels in PTEN-null tumor cells, accompanied by a decrease in PDGF B expression. Among Akt isoforms, increased Akt3 expression was most prominent in mouse PTEN-null cells, and phosphatidylinositol 3-kinase/Akt activity was essential for the maintenance of increased PDGF D and ß-PDGFR expression. In vitro deletion of PTEN resulted in a PDGF ligand switch from PDGF B to PDGF D in normal mouse prostate epithelial cells, further demonstrating that PTEN regulates this ligand switch. Similar associations between PTEN status and PDGF isoforms were noted in human prostate cancer cell lines. Taken together, these results suggest a mechanism by which loss of PTEN may promote prostate cancer progression via PDGF D/ß-PDGFR signal transduction.


Asunto(s)
Fosfohidrolasa PTEN/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Neoplasias de la Próstata/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Humanos , Ligandos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/fisiología , Células Tumorales Cultivadas , Regulación hacia Arriba
6.
Oncoscience ; 10: 69-80, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38033786

RESUMEN

During the last decade, blood sampling of cancer patients aimed at analyzing the presence of cells, membrane-bound vesicles, or molecules released by primary tumors or metastatic growths emerged as an alternative to traditional tissue biopsies. The advent of this minimally invasive approach, known as blood-based liquid biopsy, began to play a pivotal role in the management of diverse cancers, establishing itself as a vital component of precision medicine. Here, we discuss three blood-based liquid biopsies, namely circulating tumor cells (CTCs), circulating tumor DNA (ctDNA) and tumor-derived exosomes, as they relate to prostate cancer (PCa) management. The advances achieved in the molecular characterization of these types of liquid biopsies and their potential to predict recurrence, improve responses to certain treatments, and evaluate prognosis, in PCa patients, are highlighted herein. While there is currently full clinical validation for only one CTC-based and one ctDNA-based liquid biopsy for patients with metastatic castration-resistant PCa, the adoption of additional methods is anticipated as they undergo standardization and achieve analytical and clinical validation. Advantages and disadvantages of different blood-based liquid biopsy approaches in the context of PCa are outlined herein, while also considering potential synergies through combinatory strategies.

7.
Prostate ; 72(12): 1328-38, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22213159

RESUMEN

BACKGROUND: The major cause of death in prostate cancer (PCa) cases is due to distant metastatic lesions, with the bone being the most prevalent site for secondary colonization. Utilization of small molecule inhibitors to treat bone metastatic PCa have had limited success either as monotherapies or in combination with other chemotherapeutics due to intolerable toxicities. In the current study, we developed a clinically relevant in vivo intraosseous tumor model overexpressing the platelet-derived growth factor D (PDGF D) to test the efficacy of a newly characterized vascular endothelial growth factor receptor (VEGFR)/PDGFR inhibitor, cediranib (also called AZD2171). METHODS: An intratibial-injection model was established utilizing DU145 cells with or without increased PDGF D expression. Tumor-bearing mice were treated by daily gavage administration of cediranib and/or weekly i.p. injection of docetaxel for 7 weeks. Tibiae were monitored by in vivo/ex vivo X-rays and histomorphometry analysis was performed to estimate tumor volume and tumor-associated trabecular bone growth. RESULTS: Cediranib reduced intraosseous growth of prostate tumors as well as tumor-associated bone responses. When compared to the standard chemotherapeutic agent docetaxel, cediranib exhibited a stronger inhibition of tumor-associated bone response. The efficacy of cediranib was further enhanced when the drug was co-administered with docetaxel. Importantly, the therapeutic benefits of cediranib and docetaxel are more prominent in intraosseous prostate tumors overexpressing PDGF D. CONCLUSION: These novel findings support the utilization of cediranib, either alone or in combination with docetaxel, to treat bone metastatic PCa exhibiting PDGF D expression.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Inhibidores de Crecimiento/uso terapéutico , Linfocinas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Quinazolinas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Humanos , Linfocinas/antagonistas & inhibidores , Masculino , Ratones , Ratones SCID , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Neoplasias de la Próstata/metabolismo , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Bosn J Basic Med Sci ; 22(5): 683-698, 2022 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-35490363

RESUMEN

c-kit is a classical proto-oncogene that encodes a receptor tyrosine kinase (RTK) that responds to stem cell factor (SCF). C-KIT signaling is a critical regulator of cell proliferation, survival, and migration and is implicated in several physiological processes, including pigmentation, hematopoiesis and gut movement. Accumulating evidence suggests that dysregulated c-KIT function, caused by either overexpression or mutations in c-kit, promotes tumor development and progression in various human cancers. In this review, we discuss the most important structural and biological features of c-KIT, as well as insights into the activation of intracellular signaling pathways following SCF binding to this RTK. We then illustrate how different c-kit alterations are associated with specific human cancers and describe recent studies that highlight the contribution of c-KIT to cancer stemness, epithelial-mesenchymal transition and progression to metastatic disease in different experimental models. The impact of tyrosine kinase inhibitors in treating c-KIT-positive tumors and limitations due to their propensity to develop drug resistance are summarized. Finally, we appraise the potential of novel therapeutic approaches targeting c-KIT more selectively while minimizing toxicity to normal tissue.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proliferación Celular , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/genética , Factor de Células Madre/metabolismo
9.
Sci Rep ; 10(1): 2309, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32047176

RESUMEN

The Discoidin Domain Receptors (DDRs) constitute a unique set of receptor tyrosine kinases that signal in response to collagen. Using an inducible expression system in human HT1080 fibrosarcoma cells, we investigated the role of DDR1b and DDR2 on primary tumour growth and experimental lung metastases. Neither DDR1b nor DDR2 expression altered tumour growth at the primary site. However, implantation of DDR1b- or DDR2-expressing HT1080 cells with collagen I significantly accelerated tumour growth rate, an effect that could not be observed with collagen I in the absence of DDR induction. Interestingly, DDR1b, but not DDR2, completely hindered the ability of HT1080 cells to form lung colonies after intravenous inoculation, suggesting a differential role for DDR1b in primary tumour growth and lung colonization. Analyses of tumour extracts revealed specific alterations in Hippo pathway core components, as a function of DDR and collagen expression, that were associated with stimulation of tumour growth by DDRs and collagen I. Collectively, these findings identified divergent effects of DDRs on primary tumour growth and experimental lung metastasis in the HT1080 xenograft model and highlight the critical role of fibrillar collagen and DDRs in supporting the growth of tumours thriving within a collagen-rich stroma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Colágeno Tipo I/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Receptor con Dominio Discoidina 2/metabolismo , Colágenos Fibrilares/metabolismo , Fibrosarcoma/patología , Neoplasias Pulmonares/prevención & control , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Receptor con Dominio Discoidina 1/genética , Receptor con Dominio Discoidina 2/genética , Femenino , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Fosforilación , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Cancer Res ; 6(3): 446-57, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18337451

RESUMEN

Chemokines and their receptors function in migration and homing of cells to target tissues. Recent evidence suggests that cancer cells use a chemokine receptor axis for metastasis formation at secondary sites. Previously, we showed that binding of the chemokine CXCL12 to its receptor CXCR4 mediated signaling events resulting in matrix metalloproteinase-9 expression in prostate cancer bone metastasis. A variety of methods, including lipid raft isolation, stable overexpression of CXCR4, cellular adhesion, invasion assays, and the severe combined immunodeficient-human bone tumor growth model were used. We found that (a) CXCR4 and HER2 coexist in lipid rafts of prostate cancer cells; (b) the CXCL12/CXCR4 axis results in transactivation of the HER2 receptor in lipid rafts of prostate cancer cells; (c) Src kinase mediates CXCL12/CXCR4 transactivation of HER2 in prostate cancer cells; (d) a pan-HER inhibitor desensitizes CXCR4-induced transactivation and subsequent matrix metalloproteinase-9 secretion and invasion; (e) lipid raft-disrupting agents inhibited raft-associated CXCL12/CXCR4 transactivation of the HER2 and cellular invasion; (f) overexpression of CXCR4 in prostate cancer cells leads to increased HER2 phosphorylation and migratory properties of prostate cancer cells; and (g) CXCR4 overexpression enhances bone tumor growth and osteolysis. These data suggest that lipid rafts on the cell membrane are the key site for CXCL12/CXCR4-induced HER2 receptor transactivation. This transactivation contributes to enhanced invasive signals and metastatic growth in the bone microenvironment.


Asunto(s)
Neoplasias Óseas/secundario , Quimiocina CXCL12/fisiología , Microdominios de Membrana/fisiología , Metástasis de la Neoplasia/patología , Neoplasias de la Próstata/patología , Receptor ErbB-2/metabolismo , Receptores CXCR4/fisiología , Biotinilación , Neoplasias Óseas/patología , División Celular , Línea Celular Tumoral , Membrana Celular/fisiología , Quimiocina CXCL12/genética , Humanos , Masculino , Invasividad Neoplásica , Receptor Cross-Talk , Receptores CXCR4/genética , Activación Transcripcional , Transfección
11.
ACS Chem Biol ; 14(1): 37-49, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30452219

RESUMEN

The importance of Discoidin Domain Receptor 1 (DDR1) in renal fibrosis has been shown via gene knockout and use of antisense oligonucleotides; however, these techniques act via a reduction of DDR1 protein, while we prove the therapeutic potential of inhibiting DDR1 phosphorylation with a small molecule. To date, efforts to generate a selective small-molecule to specifically modulate the activity of DDR1 in an in vivo model have been unsuccessful. We performed parallel DNA encoded library screens against DDR1 and DDR2, and discovered a chemical series that is highly selective for DDR1 over DDR2. Structure-guided optimization efforts yielded the potent DDR1 inhibitor 2.45, which possesses excellent kinome selectivity (including 64-fold selectivity over DDR2 in a biochemical assay), a clean in vitro safety profile, and favorable pharmacokinetic and physicochemical properties. As desired, compound 2.45 modulates DDR1 phosphorylation in vitro as well as prevents collagen-induced activation of renal epithelial cells expressing DDR1. Compound 2.45 preserves renal function and reduces tissue damage in Col4a3-/- mice (the preclinical mouse model of Alport syndrome) when employing a therapeutic dosing regime, indicating the real therapeutic value of selectively inhibiting DDR1 phosphorylation in vivo. Our results may have wider significance as Col4a3-/- mice also represent a model for chronic kidney disease, a disease which affects 10% of the global population.


Asunto(s)
ADN/genética , Receptor con Dominio Discoidina 1/antagonistas & inhibidores , Riñón/fisiopatología , Nefritis Hereditaria/genética , Animales , Autoantígenos/genética , Autoantígenos/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Pruebas de Función Renal , Ratones , Ratones Noqueados , Nefritis Hereditaria/fisiopatología , Fosforilación , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo
12.
Int J Cancer ; 122(11): 2482-90, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18324629

RESUMEN

At the cellular level, the process of bone metastasis involves many steps. Circulating cancer cells enter the marrow, proliferate, induce neovascularization, and ultimately expand into a clinically detectable, often symptomatic, metastatic deposit. Although the initial establishment and later expansion of the metastatic deposit in bone require tumor cells to possess invasive capability, the exact proteases responsible for this phenotype are not well known. The objective of our study was to take an unbiased approach to determine which proteases were expressed and functional during the initial interactions between prostate cancer cells and bone marrow stromal (BMS) cells. We found that the combination of human prostate cancer PC3 and BMS cells stimulates the invasive ability of cancer cells through type I collagen. The use of inhibitors for each of the major protease families indicated that 1 or more MMPs was/were responsible for the BMS-induced invasion. Gene profiling and semiquantitative RT-PCR analysis revealed an increased expression of several MMP genes because of PC3/BMS cell interaction. However, only MMP-12 showed an increase in protein expression. Downregulation of MMP-12 expression in PC3 cells by siRNA inhibited the enhanced invasion induced by PC3/BMS cell interaction. In vivo, MMP-12 was found to be primarily expressed by prostate cancer cells growing in bone. Our data suggest that BMS cells induce MMP-12 expression in prostate cancer cells, which results in invasive cells capable of degradation of type I collagen.


Asunto(s)
Células de la Médula Ósea/patología , Colágeno Tipo I/metabolismo , Metaloproteinasa 12 de la Matriz/metabolismo , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Células del Estroma/patología , Western Blotting , Línea Celular Tumoral , Técnicas de Cocultivo , Regulación hacia Abajo , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Queratinas/análisis , Masculino , Metaloproteinasas de la Matriz/metabolismo , Invasividad Neoplásica , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Regulación hacia Arriba
13.
Oncol Rep ; 19(5): 1205-11, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18425377

RESUMEN

Despite its effectiveness as an antineoplastic drug, doxorubicin (DOX) is usually associated with cardiotoxicity. Lovastatin (LOV), a hypolipidemic agent used in the clinic, has been demonstrated to have antitumoral and antimetastatic effects in murine models. Since the two agents arrest tumor cells in different phases of the cell cycle and induce apoptosis, the goal of this study was to examine the efficacy of a combination therapy with LOV and low doses of DOX, in an attempt to obtain an improved antitumoral effect devoid of toxicity, by using a rat B-cell lymphoma and a mouse mammary tumor. In the two models, the combined treatment showed a synergistic antitumoral effect, which is mainly ascribed to an increased apoptotic response elicited by a LOV/DOX combination than either agent alone. The therapeutic benefit demonstrated by the combination treatment is further emphasized by the lack of toxicity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Doxorrubicina/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lovastatina/administración & dosificación , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas
14.
Prostate ; 67(1): 107-14, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17075820

RESUMEN

BACKGROUND: Experimental bone metastases are typically analyzed when the skeletal tumor burden is large enough to be detected by imaging or histology. By this time, the bone microenvironment is usually destroyed, preventing useful analysis of tumor-bone interactions. METHODS: Small intraosseous tumors generated by intratibial injection of C4-2B prostate cancer cells transfected with green fluorescent protein (GFP) were assessed using in vivo and ex vivo fluorescence imaging, radiography, histology, and fluorometric analysis of bone lysates. RESULTS: Ex vivo fluorescence imaging and fluorometric analysis were capable of detecting tiny bone tumors as early as 10 days after injection. Ex vivo fluorescence imaging allowed simple quantification of small skeletal tumor burden and was useful in measuring the effect of systemic therapy. CONCLUSIONS: Ex vivo fluorescence imaging is a sensitive and easy method to quantify small skeletal tumor burden. This technique allows investigation of tumor-bone interactions while the bone microanatomy is still intact.


Asunto(s)
Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/patología , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Animales , Neoplasias Óseas/secundario , Línea Celular Tumoral , Humanos , Masculino , Ratones , Ratones SCID , Radiografía , Espectrometría por Rayos X/métodos , Espectrometría por Rayos X/normas
15.
BMC Cancer ; 7: 184, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17908302

RESUMEN

BACKGROUND: Despite significant advancement in breast cancer therapy, there is a great need for a better understanding of the mechanisms involved in breast carcinogenesis and progression, as well as of the role of epigenetic contributions from stromal cells in mammary tumorigenesis. In this study, we isolated and characterized murine mammary tumor-derived epithelial and myofibroblast cell lines, and investigated the in vitro and in vivo effect of cellular soluble factors produced by the epithelial cell line on tumor cells. METHODS: Morphology, immunophenotype, cytogenetics, invasiveness, and tumorigenicity of epithelial (LM-234ep) and myofibroblast (LM-234mf) cell lines isolated from two murine mammary adenocarcinomas with common ancestor were studied. The in vitro effects of LM-234ep conditioned medium on proliferation, cell cycle distribution, and expression of cell cycle proteins, were investigated in LM-234mf cells, mouse melanoma cells (B16-F10), and human cervical adenocarcinoma cells (HeLa). The in vivo anti-tumor activity of LM-234ep conditioned media was evaluated in subcutaneous tumors formed in nude mice by B16-F10 and HeLa cells. RESULTS: LM-234ep cells were found to be cytokeratin positive and hipertriploid, whereas LM-234mf cells were alpha-smooth muscle actin positive and hypohexaploid. Chromosome aberrations were found in both cases. Only LM-234mf revealed to be invasive in vitro and to secrete active MMP-2, though neither of the cell types were able to produce progressing tumors. LM-234ep-derived factors were able to inhibit the in vitro growth of LM-234mf, B16-F10, and HeLa cells, inducing cell cycle arrest in G0/G1 phase. The administration of LM-234ep conditioned medium inhibited the growth of B16-F10 and HeLa tumors in nude mice. CONCLUSION: Our data suggest the existence of epithelial cell variants with tumor suppressive properties within mammary tumors. To our knowledge, this is the first report showing antiproliferative and antineoplastic activities induced by tumor-derived epithelial cells.


Asunto(s)
Adenocarcinoma/metabolismo , Proliferación Celular , Células Epiteliales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Actinas/metabolismo , Adenocarcinoma/genética , Animales , Pruebas de Carcinogenicidad , Línea Celular Tumoral , Aberraciones Cromosómicas , Medios de Cultivo Condicionados , Femenino , Gelatinasas/metabolismo , Humanos , Queratinas/metabolismo , Neoplasias Mamarias Experimentales/genética , Melanoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Comunicación Paracrina , Neoplasias del Cuello Uterino/metabolismo
16.
Urol Oncol ; 25(5): 407-11, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17826661

RESUMEN

The arrival of cancer cells in the marrow upsets the delicate homeostatic nature of the bone microenvironment. Cell surface or secreted factors brought in by cancer cells perturb the web-like communication network between different bone cell types and bone matrix. Chemokines not only attract cancer cells from the circulation into the marrow, they also stimulate a cell signaling process leading to attachment, invasion, and further stimulation of bone matrix turnover. Cancer cell surface-associated proteases have also been associated with tumor growth and bone matrix turnover. Recent data indicate that autocrine proteolytic shedding of cell surface chemokines further promotes osteoclastogenesis. Proteases also contribute to autocrine and paracrine shedding of growth factors, another mechanism of promoting growth and expansion of the metastatic deposit. Studies of the bone microenvironment have thus revealed multiple potential targets of intervention with regard to the expanding metastatic deposit.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Quimiocinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Neovascularización Patológica/metabolismo , Neoplasias de la Próstata/patología , Animales , Células de la Médula Ósea/metabolismo , Neoplasias Óseas/metabolismo , Comunicación Celular , División Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Células Neoplásicas Circulantes/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteólisis/metabolismo , Neoplasias de la Próstata/metabolismo , Serina Endopeptidasas/metabolismo , Células del Estroma/metabolismo
17.
Cancer Res ; 77(4): 886-896, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27923833

RESUMEN

Maspin (SerpinB5) is an epithelial-specific tumor suppressor gene product that displays context-dependent cellular functions. Maspin-deficient mouse models created to date have not definitively established maspin functions critical for cancer suppression. In this study, we generated a mouse strain in which exon 4 of the Maspin gene was deleted, confirming its essential role in development but also enabling a breeding scheme to bypass embryonic lethality. Phenotypic characterization of this viable strain established that maspin deficiency was associated with a reduction in maximum body weight and a variety of context-dependent epithelial abnormalities. Specifically, maspin-deficient mice exhibited pulmonary adenocarcinoma, myoepithelial hyperplasia of the mammary gland, hyperplasia of luminal cells of dorsolateral and anterior prostate, and atrophy of luminal cells of ventral prostate and stratum spinosum of epidermis. These cancer phenotypes were accompanied by increased inflammatory stroma. These mice also displayed the autoimmune disorder alopecia aerate. Overall, our findings defined context-specific tumor suppressor roles for maspin in a clinically relevant model to study maspin functions in cancer and other pathologies. Cancer Res; 77(4); 886-96. ©2017 AACR.


Asunto(s)
Desarrollo Embrionario , Serpinas/fisiología , Proteínas Supresoras de Tumor/fisiología , Alopecia Areata/etiología , Animales , Femenino , Histona Desacetilasa 1/fisiología , Masculino , Glándulas Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Próstata/patología , Serpinas/genética
18.
Clin Exp Metastasis ; 23(7-8): 335-44, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17136575

RESUMEN

Matrix metalloproteinases (MMPs) have been associated with initiation, progression and vascularization of a number of tumors. However, clinical trials using MMP inhibitors failed to meet expectations. Previously, we demonstrated the potential importance of MMP-9 activity in experimental prostate cancer bone tumor tissue. However, the particular roles of host- and tumor-derived MMP-9 remains to be defined. Herein, we examined the role of host MMP-9 in subcutaneous and intraosseous growth of the human androgen independent prostate cancer cell line PC3 in MMP-9 deficient mice. In the subcutaneous model, the tumor incidence in the control (RAG-1(ko/ko)) and experimental (RAG-1(ko/ko) /MMP-9(ko/ko)) group was 100%, with similar tumor growth kinetics and microvascular densities. In the intraosseous tumor model, the tumor incidence was higher in RAG-1(ko/ko) /MMP-9(ko/ko mice than in RAG-1(ko/ko) mice (67% and 39%, respectively), though no statistical differences were found. The intraosseous tumor areas were similar in both groups, and the number of tumor-associated osteoclasts did not differ significantly. However, the microvascular density of intraosseous tumors was higher in RAG-1(ko/ko) than in RAG-1(ko/ko)/MMP-9(ko/ko) mice, though no changes in tumor growth could be detected. In an in vitro assay, we found that bone marrow (BM) cells increased the invasiveness of PC3 cells, and that this enhancement was independent of MMP-9 expression by marrow cells. Our results with the RAG-1 model suggest that host-derived MMP-9 is neither necessary nor sufficient for subcutaneous or intraosseous PC3 tumor growth, osteoclastic response, or in vitro invasiveness of tumor cells.


Asunto(s)
Neoplasias Óseas/secundario , Metaloproteinasa 9 de la Matriz/fisiología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/patología , Animales , Movimiento Celular , Proteínas de Homeodominio/fisiología , Humanos , Masculino , Ratones , Invasividad Neoplásica
19.
Cancer Res ; 64(5): 1722-9, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14996732

RESUMEN

The platelet-derived growth factor (PDGF) proteins are potent stimulators of cell proliferation/transformation and play a major role in cell-cell communication. For over two decades, PDGFs were thought to exist as three dimeric polypeptides (the homodimers AA and BB and the heterodimer AB). Recently, however, the PDGF C and D chains were discovered in a BLAST search of the expressed sequence tag databases. The PDGF CC and DD dimers have a unique two-domain structure with an NH(2)-terminal CUB (compliment subcomponents C1r/C1s, Uegf, and Bmp1) domain and a COOH-terminal PDGF/vascular endothelial growth factor domain. Whereas secreted PDGF AA, BB, and AB readily activate their cell surface receptors, it was suggested that extracellular proteolytic removal of the CUB domain is required for the PDGF/vascular endothelial growth factor domain of PDGF CC and DD to activate PDGF receptors. In the present study, we examined the processing of latent PDGF D into its active form and the effects of PDGF D expression on prostate cancer progression. We show that LNCaP cells auto-activate latent PDGF DD into the active PDGF domain, which can induce phosphorylation of the beta-PDGF receptor and stimulates LNCaP cell proliferation in an autocrine manner. Additionally, LNCaP-PDGF D-conditioned medium induces migration of the prostate fibroblast cell line 1532-FTX, indicating LNCaP-processed PDGF DD is active in a paracrine manner as well. In a severe combined immunodeficient mouse model, PDGF DD expression accelerates early onset of prostate tumor growth and drastically enhances prostate carcinoma cell interaction with surrounding stromal cells. These demonstrate a potential oncogenic activity of PDGF DD in the development and/or progression of prostate cancer.


Asunto(s)
Linfocinas , Factor de Crecimiento Derivado de Plaquetas/fisiología , Neoplasias de la Próstata/patología , Comunicación Celular , División Celular , Movimiento Celular , Progresión de la Enfermedad , Humanos , Masculino , Neoplasias de la Próstata/etiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Células del Estroma/fisiología
20.
Bosn J Basic Med Sci ; 16(1): 8-13, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26773172

RESUMEN

Despite substantial similarities in embryological, cellular and molecular biology features, human and mouse prostates differ in organ morphology and tissue architecture. Thus, a clear understanding of the anatomy and histology of the mouse prostate is essential for the identification of urogenital phenotypes in genetically engineered mice, as well as for the study of the etiology, development, and treatment of human prostatic diseases for which mouse models are used. The purpose of this manuscript is to provide a brief guide for the dissection of the mouse prostate and the identification of its different lobes and histology, to both basic researchers and medical pathologists who are unfamiliar with mouse tissues.


Asunto(s)
Próstata/anatomía & histología , Próstata/patología , Animales , Modelos Animales de Enfermedad , Perros , Eosina Amarillenta-(YS)/química , Hematoxilina/química , Histología , Humanos , Masculino , Ratones , Modelos Animales , Fenotipo , Próstata/cirugía , Ratas , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA