Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(4)2021 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-33567533

RESUMEN

Transformed epithelial cells can activate programs of epithelial plasticity and switch from a sessile, epithelial phenotype to a motile, mesenchymal phenotype. This process is linked to the acquisition of an invasive phenotype and the formation of distant metastases. The development of compounds that block the acquisition of an invasive phenotype or revert the invasive mesenchymal phenotype into a more differentiated epithelial phenotype represent a promising anticancer strategy. In a high-throughput assay based on E-cadherin (re)induction and the inhibition of tumor cell invasion, 44,475 low molecular weight (LMW) compounds were screened. The screening resulted in the identification of candidate compounds from the PROAM02 class. Selected LMW compounds activated E-cadherin promoter activity and inhibited cancer cell invasion in multiple metastatic human cancer cell lines. The intraperitoneal administration of selected LMW compounds reduced the tumor burden in human prostate and breast cancer in vivo mouse models. Moreover, selected LMW compounds decreased the intra-bone growth of xenografted human prostate cancer cells. This study describes the identification of the PROAM02 class of small molecules that can be exploited to reduce cancer cell invasion and metastases. Further clinical evaluation of selected candidate inhibitors is warranted to address their safety, bioavailability and antitumor efficacy in the management of patients with aggressive cancers.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Descubrimiento de Drogas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata/patología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Prostate ; 75(8): 815-24, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25663076

RESUMEN

BACKGROUND: The inflammatory tumor microenvironment, and more specifically the tumor-associated macrophages, plays an essential role in the development and progression of prostate cancer towards metastatic bone disease. Tumors are often characterized by a leaky vasculature, which - combined with the prolonged circulation kinetics of liposomes - leads to efficient tumor localization of these drug carriers, via the so-called enhanced permeability and retention (EPR) -effect. In this study, we evaluated the utility of targeted, liposomal drug delivery of the glucocorticoid dexamethasone in a model of prostate cancer bone metastases. METHODS: Tumor-bearing Balb-c nu/nu mice were treated intravenously with 0.2-1.0-5.0 mg/kg/week free- and liposomal DEX for 3-4 weeks and tumor growth was monitored by bioluminescent imaging. RESULTS: Intravenously administered liposomes localize efficiently to bone metastases in vivo and treatment of established bone metastases with (liposomal) dexamethasone resulted in a significant inhibition of tumor growth up to 26 days after initiation of treatment. Furthermore, 1.0 mg/kg liposomal dexamethasone significantly outperformed 1.0 mg/kg free dexamethasone, and was found to be well-tolerated at clinically-relevant dosages that display potent anti-tumor efficacy. CONCLUSIONS: Liposomal delivery of the glucocorticoid dexamethasone inhibits the growth of malignant bone lesions. We believe that liposomal encapsulation of dexamethasone offers a promising new treatment option for advanced, metastatic prostate cancer which supports further clinical evaluation.


Asunto(s)
Antineoplásicos Hormonales/administración & dosificación , Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Dexametasona/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Humanos , Liposomas , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Próstata/patología , Ratas , Ratas Sprague-Dawley
3.
Mol Cancer ; 13: 10, 2014 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-24438191

RESUMEN

BACKGROUND: Tumor cell migration and metastasis require dynamic rearrangements of the actin cytoskeleton. Interestingly, the F-actin cross-linking and stabilizing protein L-plastin, originally described as a leukocyte specific protein, is aberrantly expressed in several non-hematopoietic malignant tumors. Therefore, it has been discussed as a tumor marker. However, systematic in vivo analyses of the functional relevance of L-plastin for tumor cell metastasis were so far lacking. METHODS: We investigated the relevance of L-plastin expression and phosphorylation by ectopical expression of L-plastin in human melanoma cells (MV3) and knock-down of endogenous L-plastin in prostate cancer (PC3M). The growth and metastatic potential of tumor cells expressing no L-plastin, phosphorylatable or non-phosphorylatable L-plastin was analyzed in a preclinical mouse model after subcutaneous and intracardial injection of the tumor cells. RESULTS: Knock-down of endogenous L-plastin in human prostate carcinoma cells led to reduced tumor cell growth and metastasis. Vice versa, and in line with these findings, ectopic expression of L-plastin in L-plastin negative melanoma cells significantly increased the number of metastases. Strikingly, the metastasis promoting effect of L-plastin was not observed if a non-phosphorylatable L-plastin mutant was expressed. CONCLUSIONS: Our data provide the first in vivo evidence that expression of L-plastin promotes tumor metastasis and, importantly, that this effect depends on an additionally required phosphorylation of L-plastin. In conclusion, these findings imply that for determining the importance of tumor-associated proteins like L-plastin a characterization of posttranslational modifications is indispensable.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Melanoma/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Citoesqueleto/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Inmunohistoquímica , Masculino , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Fosforilación , Neoplasias de la Próstata/patología , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Am J Pathol ; 179(5): 2559-68, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21907176

RESUMEN

Integrins participate in multiple cellular processes, including cell adhesion, migration, proliferation, survival, and the activation of growth factor receptors. Recent studies have shown that expression of αv integrins is elevated in the prostate cancer stem/progenitor cell subpopulation compared with more differentiated, committed precursors. Here, we examine the functional role of αv integrin receptor expression in the acquisition of a metastatic stem/progenitor phenotype in human prostate cancer. Stable knockdown of αv integrins expression in PC-3M-Pro4 prostate cancer cells coincided with a significant decrease of prostate cancer stem/progenitor cell characteristics (α2 integrin, CD44, and ALDH(hi)) and decreased expression of invasion-associated genes Snail, Snail2, and Twist. Consistent with these observations, αv-knockdown strongly inhibited the clonogenic and migratory potentials of human prostate cancer cells in vitro and significantly decreased tumorigenicity and metastatic ability in preclinical models of orthotopic growth and bone metastasis. Our data indicate that integrin αv expression is functionally involved in the maintenance of a highly migratory, mesenchymal cellular phenotype as well as the acquisition of a stem/progenitor phenotype in human prostate cancer cells with metastasis-initiating capacity.


Asunto(s)
Integrina alfaV/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Movimiento Celular , Transformación Celular Neoplásica/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Fenotipo , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba
5.
Recent Results Cancer Res ; 192: 1-31, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22307368

RESUMEN

In this chapter currently available preclinical models of tumor progression and bone metastasis, including genetically engineered mice that develop primary and metastatic carcinomas and transplantable animal models, will be described. Understanding the multistep process of incurable bone metastasis is pivotal to the development of new therapeutic strategies. Novel technologies for imaging molecules or pathologic processes in cancers and their surrounding stroma have emerged rapidly and have greatly facilitated cancer research, in particular the cellular behavior of osteotropic tumors and their response to new and existing therapeutic agents. Optical imaging, in particular, has become an important tool in preclinical bone metastasis models, clinical trials and medical practice. Advances in experimental and clinical imaging will-in the long run-result in significant improvements in diagnosis, tumor localization, enhanced drug delivery and treatment.


Asunto(s)
Neoplasias Óseas/secundario , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Animales , Antineoplásicos/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Humanos , Ratones
6.
Future Oncol ; 8(4): 415-30, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22515445

RESUMEN

Bone metastasis is a complex process that ultimately leads to devastating metastatic bone disease. It is therefore of key interest to unravel the mechanisms underlying the multistep process of skeletal metastasis and cancer-induced bone disease, and to develop better treatment and management of patients with this devastating disease. Fortunately, novel technologies are rapidly emerging that allow real-time imaging of molecules, pathogenic processes, drug delivery and drug response in preclinical in vivo models. The outcome of these experimental studies will facilitate clinical cancer research by improving the detection of cancer cell invasion, metastasis and therapy response.


Asunto(s)
Neoplasias Óseas/diagnóstico , Neoplasias Óseas/secundario , Diagnóstico por Imagen/métodos , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Imagen Molecular/métodos , Metástasis de la Neoplasia/diagnóstico
7.
Exp Cell Res ; 317(10): 1411-21, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21402068

RESUMEN

The canonical Wnt signaling pathway influences the differentiation of mesenchymal cell lineages in a quantitative and qualitative fashion depending on the dose of ß-catenin signaling. Adenomatous polyposis coli (Apc) is the critical intracellular regulator of ß-catenin turnover. To better understand the molecular mechanisms underlying the role of Apc in regulating the differentiation capacity of skeletal progenitor cells, we have knocked down Apc in the murine mesenchymal stem cell-like KS483 cells by stable expression of Apc-specific small interfering RNA. In routine culture, KSFrt-Apc(si) cells displayed a mesenchymal-like spindle shape morphology, exhibited markedly decreased proliferation and increased apoptosis. Apc knockdown resulted in upregulation of the Wnt/ß-catenin and the BMP/Smad signaling pathways, but osteogenic differentiation was completely inhibited. This effect could be rescued by adding high concentrations of BMP-7 to the differentiation medium. Furthermore, KSFrt-Apc(si) cells showed no potential to differentiate into chondrocytes or adipocytes. These results demonstrate that Apc is essential for the proliferation, survival and differentiation of KS483 cells. Apc knockdown blocks the osteogenic differentiation of skeletal progenitor cells, a process that can be overruled by high BMP signaling.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/citología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Adipocitos/citología , Adipocitos/metabolismo , Animales , Apoptosis , Western Blotting , Proteína Morfogenética Ósea 7/genética , Proliferación Celular , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Técnica del Anticuerpo Fluorescente , Ratones , Osteoblastos/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteínas Wnt/genética , beta Catenina/genética
8.
Cancer Gene Ther ; 29(6): 793-802, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34135475

RESUMEN

Treatment of castration-resistant prostate cancer remains a challenging clinical problem. Despite the promising effects of immunotherapy in other solid cancers, prostate cancer has remained largely unresponsive. Oncolytic viruses represent a promising therapeutic avenue, as oncolytic virus treatment combines tumour cell lysis with activation of the immune system and mounting of effective anti-tumour responses. Mammalian Orthoreoviruses are non-pathogenic human viruses with a preference of lytic replication in human tumour cells. In this study, we evaluated the oncolytic efficacy of the bioselected oncolytic reovirus mutant jin-3 in multiple human prostate cancer models. The jin-3 reovirus displayed efficient infection, replication, and anti-cancer responses in 2D and 3D prostate cancer models, as well as in ex vivo cultured human tumour slices. In addition, the jin-3 reovirus markedly reduced the viability and growth of human cancer cell lines and patient-derived xenografts. The infection induced the expression of mediators of immunogenic cell death, interferon-stimulated genes, and inflammatory cytokines. Taken together, our data demonstrate that the reovirus mutant jin-3 displays tumour tropism, and induces potent oncolytic and immunomodulatory responses in human prostate cancer models. Therefore, jin-3 reovirus represents an attractive candidate for further development as oncolytic agent for treatment of patients with aggressive localised or advanced prostate cancer.


Asunto(s)
Orthoreovirus Mamífero 3 , Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias de la Próstata , Reoviridae , Animales , Línea Celular Tumoral , Humanos , Masculino , Mamíferos , Virus Oncolíticos/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Reoviridae/genética
9.
Nat Rev Urol ; 18(1): 33-45, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33173206

RESUMEN

Preclinical knowledge of dysregulated pathways and potential biomarkers for urological cancers has undergone limited translation into the clinic. Moreover, the low approval rate of new anticancer drugs and the heterogeneous drug responses in patients indicate that current preclinical models do not always reflect the complexity of malignant disease. Patient-derived tumour models used in preclinical uro-oncology research include 3D culture systems, organotypic tissue slices and patient-derived xenograft models. Technological innovations have enabled major improvements in the capacity of these tumour models to reproduce the clinical complexity of urological cancers. Each type of patient-derived model has inherent advantages and limitations that can be exploited, either alone or in combination, to gather specific knowledge on clinical challenges and address unmet clinical needs. Nevertheless, few opportunities exist for patients with urological cancers to benefit from personalized therapeutic approaches. Clinical validation of experimental data is needed to facilitate the translation and implementation of preclinical knowledge into treatment decision making.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Modelos Animales de Enfermedad , Medicina de Precisión/métodos , Neoplasias Urológicas/terapia , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Antineoplásicos/uso terapéutico , Línea Celular Transformada , Ensayos Clínicos como Asunto/métodos , Humanos , Neoplasias Urológicas/patología
10.
Eur J Cancer ; 146: 11-20, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33561783

RESUMEN

With a 5-year recurrence rate of 30-78%, urothelial cell carcinoma (UCC) rates amongst the highest of all solid malignancies. Consequently, after transurethral resection, patients are subjugated to life-long endoscopic surveillance. A multimodal near-infrared (NIR) fluorescence-based imaging strategy can improve diagnosis, resection and surveillance, hence increasing quality of life. METHODS: Expression of urokinase plasminogen activator receptor (uPAR) and epithelial cell adhesion molecule (EpCAM) are determined on paraffin-embedded human UCC using immunohistochemistry and on UCC cell lines by flow cytometry. MNPR-101, a humanised monoclonal antibody targeting uPAR is conjugated to IRDye800CW and binding is validated in vitro using surface plasmon resonance and cell-based binding assays. In vivo NIR fluorescence and photoacoustic three-dimensional (3D) imaging are performed with subcutaneously growing human UM-UC-3luc2 cells in BALB/c-nude mice. The translational potential is confirmed in a metastasising UM-UC-3luc2 orthotopic mouse model. Infliximab-IRDye800CW and rituximab-IRDye800CW are used as controls. RESULTS: UCCs show prominent uPAR expression at the tumour-stroma interface and EpCAM on epithelial cells. uPAR and EpCAM are expressed by 6/7 and 4/7 UCC cell lines, respectively. In vitro, MNPR-101-IRDye800CW has a picomolar affinity for domain 2-3 of uPAR. In vivo fluorescence imaging with MNPR-101-IRDye800CW, specifically delineates both subcutaneous and orthotopic tumours with tumour-to-background ratios reaching as high as 6.8, differing significantly from controls (p < 0.0001). Photoacoustic 3D in depth imaging confirms the homogenous distribution of MNPR-101-IRDye800CW through the tumour. CONCLUSIONS: MNPR-101-IRDye800CW is suitable for multimodal imaging of UCC, awaiting clinical translation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Imagen Molecular/métodos , Imagen Óptica/métodos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Cirugía Asistida por Computador/métodos , Neoplasias de la Vejiga Urinaria/diagnóstico , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/cirugía , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Oncol ; 14(12): 3121-3134, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32896947

RESUMEN

More effective therapy for patients with either muscle-invasive or high-risk non-muscle-invasive urothelial carcinoma of the bladder (UCB) is an unmet clinical need. For this, drug repositioning of clinically approved drugs represents an interesting approach. By repurposing existing drugs, alternative anticancer therapies can be introduced in the clinic relatively fast, because the safety and dosing of these clinically approved pharmacological agents are generally well known. Cationic amphiphilic drugs (CADs) dose-dependently decreased the viability of a panel of human UCB lines in vitro. CADs induced lysosomal puncta formation, a hallmark of lysosomal leakage. Intravesical instillation of the CAD penfluridol in an orthotopic mouse xenograft model of human UCB resulted in significantly reduced intravesical tumor growth and metastatic progression. Furthermore, treatment of patient-derived ex vivo cultured human UCB tissue caused significant partial or complete antitumor responses in 97% of the explanted tumor tissues. In conclusion, penfluridol represents a promising treatment option for bladder cancer patients and warrants further clinical evaluation.


Asunto(s)
Antineoplásicos/uso terapéutico , Tensoactivos/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Cationes , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Clonales , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Penfluridol/farmacología , Penfluridol/uso terapéutico , Tensoactivos/farmacología , Neoplasias de la Vejiga Urinaria/patología , Urotelio/efectos de los fármacos , Urotelio/patología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
BMC Dev Biol ; 9: 26, 2009 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-19356224

RESUMEN

BACKGROUND: During skeletogenesis, protein levels of beta-catenin in the canonical Wnt signaling pathway determine lineage commitment of skeletal precursor cells to osteoblasts and chondrocytes. Adenomatous polyposis coli (Apc) is a key controller of beta-catenin turnover by down-regulating intracellular levels of beta-catenin. RESULTS: To investigate whether Apc is involved in lineage commitment of skeletal precursor cells, we generated conditional knockout mice lacking functional Apc in Col2a1-expressing cells. In contrast to other models in which an oncogenic variant of beta-catenin was used, our approach resulted in the accumulation of wild type beta-catenin protein due to functional loss of Apc. Conditional homozygous Apc mutant mice died perinatally showing greatly impaired skeletogenesis. All endochondral bones were misshaped and lacked structural integrity. Lack of functional Apc resulted in a pleiotropic skeletal cell phenotype. The majority of the precursor cells lacking Apc failed to differentiate into chondrocytes or osteoblasts. However, skeletal precursor cells in the proximal ribs were able to escape the noxious effect of functional loss of Apc resulting in formation of highly active osteoblasts. Inactivation of Apc in chondrocytes was associated with dedifferentiation of these cells. CONCLUSION: Our data indicate that a tight Apc-mediated control of beta-catenin levels is essential for differentiation of skeletal precursors as well as for the maintenance of a chondrocytic phenotype in a spatio-temporal regulated manner.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Huesos/metabolismo , Embrión de Mamíferos/metabolismo , beta Catenina/genética , Animales , Huesos/citología , Huesos/embriología , Diferenciación Celular/genética , Condrocitos/citología , Condrocitos/metabolismo , Condrogénesis/genética , Colágeno Tipo II/genética , Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Ratones , Ratones Noqueados , Ratones Transgénicos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/genética , Fenotipo , Factores de Tiempo
13.
Cancer Res ; 67(18): 8742-51, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875715

RESUMEN

Bone morphogenetic protein 7 (BMP7) counteracts the physiological epithelial-to-mesenchymal transition (EMT), a process that is indicative of epithelial plasticity. Because EMT is involved in cancer, we investigated whether BMP7 plays a role in breast cancer growth and metastasis. In this study, we show that decreased BMP7 expression in primary breast cancer is significantly associated with the formation of clinically overt bone metastases in patients with > or = 10 years of follow-up. In line with these clinical observations, BMP7 expression is inversely related to tumorigenicity and invasive behavior of human breast cancer cell lines. Moreover, BMP7 decreased the expression of vimentin, a mesenchymal marker associated with invasiveness and poor prognosis, in human MDA-MB-231 (MDA-231)-B/Luc(+) breast cancer cells under basal and transforming growth factor-beta (TGF-beta)-stimulated conditions. In addition, exogenous addition of BMP7 to TGF-beta-stimulated MDA-231 cells inhibited Smad-mediated TGF-beta signaling. Furthermore, in a well-established bone metastasis model using whole-body bioluminescent reporter imaging, stable overexpression of BMP7 in MDA-231 cells inhibited de novo formation and progression of osteolytic bone metastases and, hence, their metastatic capability. In line with these observations, daily i.v. administration of BMP7 (100 mug/kg/d) significantly inhibited orthotopic and intrabone growth of MDA-231-B/Luc(+) cells in nude mice. Our data suggest that decreased BMP7 expression during carcinogenesis in the human breast contributes to the acquisition of a bone metastatic phenotype. Because exogenous BMP7 can still counteract the breast cancer growth at the primary site and in bone, BMP7 may represent a novel therapeutic molecule for repression of local and bone metastatic growth of breast cancer.


Asunto(s)
Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Animales , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Células Epiteliales/patología , Femenino , Humanos , Mesodermo/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Estudios Retrospectivos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Methods Mol Biol ; 1786: 67-79, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29786787

RESUMEN

Prostate cancer is the most common malignancy diagnosed in men in the western world. The development of distant metastases and therapy resistance are major clinical problems in the management of prostate cancer patients. In order for prostate cancer to metastasize to distant sites in the human body, prostate cancer cells have to migrate and invade neighboring tissue. Cancer cells can acquire a migratory and invasive phenotype in several ways, including single cell and collective migration. As a requisite for migration, epithelial prostate cancer cells often need to acquire a motile, mesenchymal-like phenotype. This way prostate cancer cells often lose polarity and epithelial characteristics (e.g., expression of E-cadherin homotypic adhesion receptor), and acquire mesenchymal phenotype (for example, cytoskeletal rearrangements, enhanced expression of proteolytic enzymes and other repertory of integrins). This process is referred to as epithelial-to-mesenchymal transition (EMT). Cellular invasion, one of the hallmarks of cancer, is characterized by the movement of cells through a three-dimensional matrix, resulting in remodeling of the cellular environment. Cellular invasion requires adhesion, proteolysis of the extracellular matrix, and migration of cells. Studying the migratory and invasive ability of cells in vitro represents a useful tool to assess the aggressiveness of solid cancers, including those of the prostate.This chapter provides a comprehensive description of the Transwell migration assay, a commonly used technique to investigate the migratory behavior of prostate cancer cells in vitro. Furthermore, we will provide an overview of the adaptations to the Transwell migration protocol to study the invasive capacity of prostate cancer cells, i.e., the Transwell invasion assay. Finally, we will present a detailed description of the procedures required to stain the Transwell filter inserts and quantify the migration and/or invasion.


Asunto(s)
Técnicas de Cultivo de Célula , Movimiento Celular , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Humanos , Masculino , Invasividad Neoplásica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo
15.
Methods Mol Biol ; 1786: 81-102, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29786788

RESUMEN

Current treatments of advanced prostate cancer only marginally increase overall survival and can be regarded as predominantly palliative. Hence, there is an urgent need for novel therapeutic strategies for the treatment of primary tumors and, more importantly perhaps, for the prevention of tumor progression and metastasis formation. Clinically relevant preclinical models are therefore urgently needed. An ideal, clinically relevant preclinical model would mimic the genetic and phenotypic changes that occur at the different stages of human prostate cancer progression and subsequent metastasis. In this chapter, transplantable xenograft prostate cancer models are described, in which human prostate cancer cells are transplanted into host animals (e.g., immune-deficient mice). Cancer cells can be administered to the small laboratory animals in various ways, including inoculation of the prostate tumor cells subcutaneously, at the anatomical site of origin (orthotopically), or at the metastatic site. In addition, we describe imaging methods suitable for small laboratory animals with emphasis on optical imaging (bioluminescence and fluorescence).


Asunto(s)
Trasplante de Neoplasias , Imagen Óptica , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Imagen de Cuerpo Entero , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Mediciones Luminiscentes/métodos , Masculino , Ratones , Imagen Multimodal/métodos , Imagen Óptica/métodos , Imagen de Cuerpo Entero/métodos
16.
Front Oncol ; 8: 400, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30333957

RESUMEN

Urological malignancies, including prostate and bladder carcinoma, represent a major clinical problem due to the frequent occurrence of therapy resistance and the formation of incurable distant metastases. As a result, there is an urgent need for versatile and predictive disease models for the assessment of the individualized drug response in urological malignancies. Compound testing on ex vivo cultured patient-derived tumor tissues could represent a promising approach. In this study, we have optimized an ex vivo culture system of explanted human prostate and bladder tumors derived from clinical specimens and human cancer cell lines xenografted in mice. The explanted and cultured tumor slices remained viable and tissue architecture could be maintained for up to 10 days of culture. Treatment of ex vivo cultured human prostate and bladder cancer tissues with docetaxel and gemcitabine, respectively, resulted in a dose-dependent anti-tumor response. The dose-dependent decrease in tumor cells upon administration of the chemotherapeutic agents was preceded by an induction of apoptosis. The implementation and optimization of the tissue slice technology may facilitate the assessment of anti-tumor efficacies of existing and candidate pharmacological agents in the complex multicellular neoplastic tissues from prostate and bladder cancer patients. Our model represents a versatile "near-patient" tool to determine tumor-targeted and/or stroma-mediated anti-neoplastic responses, thus contributing to the field of personalized therapeutics.

17.
J Bone Miner Res ; 22(1): 19-28, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17032150

RESUMEN

UNLABELLED: Sclerostin is an osteocyte-derived negative regulator of bone formation. It inhibits BMP-stimulated bone formation both in vitro and in vivo but has no direct effect on BMP signaling. Instead, sclerostin inhibits Wnt signaling that is required for BMP-stimulated osteoblastic differentiation. INTRODUCTION: Sclerostin is a member of the Dan family of glycoproteins of which many members have been reported to antagonize BMP activity. Sclerostin has been shown to inhibit BMP-stimulated bone formation, but its mechanism of action seems to be different from classical BMP antagonists. In this study, we investigated the mechanism by which sclerostin inhibits BMP-stimulated bone formation. MATERIALS AND METHODS: DNA electroporation of calf muscle of mice using expression plasmids for BMP and sclerostin was used to study the effect of sclerostin on BMP-induced bone formation in vivo. Transcriptional profiling using microarrays of osteoblastic cells treated with BMP in the absence or presence of sclerostin was used to find specific growth factor signaling pathways affected by sclerostin. The affected pathways were further studied using growth factor-specific reporter constructs. RESULTS: BMP-induced ectopic bone formation in calf muscle of mice was prevented by co-expression of sclerostin in vivo. Transcriptional profiling analysis of osteoblastic cultures indicated that sclerostin specifically affects BMP and Wnt signaling out of many other growth signaling pathways. Sclerostin, however, did not inhibit stimulation of direct BMP target genes. Furthermore, we did not obtain any evidence for sclerostin acting as a direct BMP antagonist using a BMP-specific reporter construct. In contrast, sclerostin shared many characteristics with the Wnt antagonist dickkopf-1 in antagonizing BMP-stimulated bone formation and BMP- and Wnt-induced Wnt reporter construct activation. CONCLUSIONS: Sclerostin inhibits BMP-stimulated bone formation but does not affect BMP signaling. Instead, it antagonizes Wnt signaling in osteoblastic cells. High bone mass in sclerosteosis and van Buchem disease may, therefore, result from increased Wnt signaling.


Asunto(s)
Desarrollo Óseo/fisiología , Proteínas Morfogenéticas Óseas/fisiología , Marcadores Genéticos/fisiología , Proteínas Wnt/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Desarrollo Óseo/efectos de los fármacos , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/farmacología , Células Cultivadas , Electroporación , Regulación de la Expresión Génica , Genes Reporteros , Glicoproteínas , Humanos , Péptidos y Proteínas de Señalización Intercelular , Mesodermo/citología , Mesodermo/fisiología , Ratones , Músculo Esquelético/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Recombinantes/farmacología , Transducción de Señal , Transfección
18.
Clin Exp Metastasis ; 24(8): 609-17, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18008174

RESUMEN

The skeleton is the second most frequent site of metastasis. However, only a restricted number of solid cancers, especially those of the breast and prostate, are responsible for the majority of the bone metastases. Metastatic bone disease is a major cause of morbidity, characterised by severe pain and high incidence of skeletal and haematopoietic complications (fractures, spinal cord compression and bone marrow aplasia) requiring hospitalisation. Despite the frequency of skeletal metastases, the molecular mechanisms for their propensity to colonise bone are poorly understood and treatment options are often unsatisfactory. TGF-beta and the signalling pathway it controls appears to play major roles in the pathogenesis of many carcinomas, both in their early stages, when TGF-beta acts to arrest growth of many cell types, and later in cancer progression when it contributes, paradoxically, to the phenotype of tumour invasiveness. Here we discuss some novel insights of the TGF-beta superfamily-including BMPs and their antagonists-in the formation of bone metastasis. Increasing evidence suggests that the TGF-beta superfamily is involved in bone homing, tumour dormancy, and development of micrometastases into overt bone metastases. The established role of TGF-beta/BMPs and their antagonists in epithelial plasticity during embryonic development closely resembles neoplastic processes at the primary site as well as in (bone) metastasis. For instance, the tumour-stroma interactions occurring in the tissue of cancer origin, including epithelium-to-mesenchyme transition (EMT), bear similarities with the role of bone matrix-derived TGF-beta in skeletal metastasis formation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Neoplasias Óseas/secundario , Células Madre Neoplásicas/citología , Factor de Crecimiento Transformador beta/metabolismo , Proteína Morfogenética Ósea 7 , Progresión de la Enfermedad , Humanos , Unión Proteica
19.
Invest Ophthalmol Vis Sci ; 48(11): 4882-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17962434

RESUMEN

PURPOSE: Bone morphogenetic protein-7 (BMP7), a member of the TGF-beta superfamily, is essential for early ocular morphogenesis, and lack of BMP7 causes epithelial development disturbances in the eye. In the present study, the association of tumorigenicity and malignant behavior of human uveal melanoma with BMP7 expression and the possibility that overexpression of BMP7 in uveal melanoma affects intraocular tumor growth in vivo were investigated. METHODS: To establish the role of BMP7 in uveal melanoma progression, the human OCM-1 cell line was stably transfected to overexpress BMP7 (OCM-1 FRT/BMP7) using targeted homologous recombination. RESULTS: Transcriptional profiling revealed low or no detectable expression of BMP7 in primary tumor tissue of patients with uveal melanoma. In line with these clinical observations, BMP7 mRNA levels were low or not detectable in cultured human uveal melanoma cell lines, when compared with normal cultured melanocytes. Inoculation of OCM-1 FRT/BMP7 cells into the anterior chamber of the eye of nude mice inhibited tumor progression significantly, compared with progression in the control cell line (no BMP7 expression). CONCLUSIONS: Collectively, the data provide novel evidence that decreased BMP7 expression contributes to progression of uveal melanoma. Furthermore, BMP7 may represent a novel therapeutic molecule for repression of tumor growth in uveal melanoma.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Melanoma/patología , Neoplasias de la Úvea/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Cámara Anterior/patología , Proteína Morfogenética Ósea 7 , Proliferación Celular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Masculino , Melanoma/genética , Melanoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Transfección , Células Tumorales Cultivadas , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/metabolismo
20.
PLoS One ; 12(11): e0188228, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29145505

RESUMEN

Prostate cancer research is hampered by the lack of in vivo preclinical models that accurately reflect patient tumour biology and the clinical heterogeneity of human prostate cancer. To overcome these limitations we propagated and characterised a new collection of patient-derived prostate cancer xenografts. Tumour fragments from 147 unsupervised, surgical prostate samples were implanted subcutaneously into immunodeficient Rag2-/-γC-/- mice within 24 hours of surgery. Histologic and molecular characterisation of xenografts was compared with patient characteristics, including androgen-deprivation therapy, and exome sequencing. Xenografts were established from 47 of 147 (32%) implanted primary prostate cancers. Only 14% passaged successfully resulting in 20 stable lines; derived from 20 independent patient samples. Surprisingly, only three of the 20 lines (15%) were confirmed as prostate cancer; one line comprised of mouse stroma, and 16 were verified as human donor-derived lymphoid neoplasms. PCR for Epstein-Barr Virus (EBV) nuclear antigen, together with exome sequencing revealed that the lymphomas were exclusively EBV-associated. Genomic analysis determined that 14 of the 16 EBV+ lines had unique monoclonal or oligoclonal immunoglobulin heavy chain gene rearrangements, confirming their B-cell origin. We conclude that the generation of xenografts from tumour fragments can commonly result in B-cell lymphoma from patients carrying latent EBV. We recommend routine screening, of primary outgrowths, for latent EBV to avoid this phenomenon.


Asunto(s)
Herpesvirus Humano 4/patogenicidad , Linfoma/virología , Neoplasias de la Próstata/virología , Anciano , Xenoinjertos , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA