Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncotarget ; 12(18): 1737-1748, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34504647

RESUMEN

Aerobic exercise is receiving increased recognition in oncology for its multiple purported benefits. Exercise is known to induce physiologic adaptations that improve patient quality-of-life parameters as well as all-cause mortality. There also is a growing body of evidence that exercise may directly alter the tumor microenvironment to influence tumor growth, metastasis, and response to anticancer therapies. Furthermore, the physiologic adaptations to exercise in normal tissues may protect against treatment-associated toxicity and allow for greater treatment tolerance. However, the exercise prescription required to induce these beneficial tumor-related outcomes remains unclear. This study characterized the aerobic adaptations to voluntary wheel running in normal tissues and the tumor microenvironment. Female, retired breeder BALB/c mice and syngeneic breast adenocarcinoma cells were utilized in primary tumor and metastasis models. Aerobic exercise was found to induce numerous adaptations across various tissues in these mice, although primary tumor growth and metastasis were largely unaffected. However, intratumoral hypoxia and global metabolism were altered in the tumors of exercising hosts relative to non-wheel running controls. Doxorubicin chemotherapy also was found to be more efficacious at delaying tumor growth with adjuvant aerobic exercise. Additionally, doxorubicin-induced cardiac toxicity was ameliorated in exercising hosts relative to non-wheel running controls. Taken together, these data suggest that the normal tissue and tumor microenvironment adaptations to aerobic exercise can improve doxorubicin efficacy while simultaneously limiting its toxicity.

2.
Tissue Eng Part A ; 27(7-8): 438-453, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33059528

RESUMEN

A challenge in cancer research is the lack of physiologically responsive in vitro models that enable tracking of cancer cells in tissue-like environments. A model that enables real-time investigation of cancer cell migration, fate, and function during angiogenesis does not exist. Current models, such as 2D or 3D in vitro culturing, can contain multiple cell types, but they do not incorporate the complexity of intact microvascular networks. The objective of this study was to establish a tumor microvasculature model by demonstrating the feasibility of bioprinting cancer cells onto excised mouse tissue. Inkjet-printed DiI+ breast cancer cells on mesometrium tissues from C57Bl/6 mice demonstrated cancer cells' motility and proliferation through time-lapse imaging. Colocalization of DAPI+ nuclei confirmed that DiI+ cancer cells remained intact postprinting. Printed DiI+ 4T1 cells also remained viable after printing on Day 0 and after culture on Day 5. Time-lapse imaging over 5 days enabled tracking of cell migration and proliferation. The number of cells and cell area were significantly increased over time. After culture, cancer cell clusters were colocalized with angiogenic microvessels. The number of vascular islands, defined as disconnected endothelial cell segments, was increased for tissues with bioprinted cancer cells, which suggests that the early stages of angiogenesis were influenced by the presence of cancer cells. Bioprinting cathepsin L knockdown 4T1 cancer cells on wild-type tissues or nontarget 4T1 cells on NG2 knockout tissues served to validate the use of the model for probing tumor cell versus microenvironment changes. These results establish the potential for bioprinting cancer cells onto live mouse tissues to investigate cancer microvascular dynamics within a physiologically relevant microenvironment. Impact statement To keep advancing the cancer biology field, tissue engineering has been focusing on developing in vitro tumor biomimetic models that more closely resemble the native microenvironment. We introduce a novel methodology of bioprinting exogenous cancer cells onto mouse tissue that contains multiple cells and systems within native physiology to investigate cancer cell migration and interactions with nearby microvascular networks. This study corroborates the manipulation of different exogenous cells and host microenvironments that impact cancer cell dynamics in a physiologically relevant tissue. Overall, it is a new approach for delineating the effects of the microenvironment on cancer cells and vice versa.


Asunto(s)
Bioimpresión , Neoplasias , Animales , Ratones , Microvasos , Neovascularización Patológica , Impresión Tridimensional , Ingeniería de Tejidos
3.
PLoS One ; 14(5): e0215584, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31050686

RESUMEN

Exercise has long been known to be beneficial to human health. Studies aimed at understanding the effects of exercise specifically focus on predetermined exercise intensities defined by measuring the aerobic capacity of each individual. Many disease models involving animal training often establish aerobic capacity by using the maximal lactate steady state (MLSS), a widely used method in humans that has frequently been used in rodent studies. The MLSS is defined as the highest exercise intensity at which blood lactate concentration remains constant and is roughly equivalent to 70-80% of maximal aerobic capacity. Due to our up-coming experiments investigating the effect of different exercise intensities in specific strains of tumor-bearing mice, the aim of the present study was to determine the MLSS in athymic nude (NCr nu/nu and NMRI), CDF1, and C3H mice by treadmill running at increasing speeds. However, despite thorough exercise acclimation and the use of different exercise protocols and aversive stimuli, less than half of the experiments across strains pointed towards an established MLSS. Moreover, gently prodding the mice during low to moderate intensity running caused a 30-121% (p<0.05) increase in blood lactate concentration compared to running without stimulation, further questioning the use of lactate as a measure of exercise intensity. Overall, MLSS is difficult to determine and large variations of blood lactate levels were observed depending on the exercise protocol, mice handling strategy and strain. This should be considered when planning experiments in mice using forced exercise protocols.


Asunto(s)
Tolerancia al Ejercicio/fisiología , Ácido Láctico/sangre , Carrera/fisiología , Animales , Femenino , Masculino , Ratones , Modelos Animales , Condicionamiento Físico Animal/fisiología
4.
Bioconjug Chem ; 29(8): 2793-2805, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30011185

RESUMEN

Low tumor accumulation following systemic delivery remains a key challenge for advancing many cancer nanomedicines. One obstacle in engineering nanoparticles for high tumor accumulation is a lack of techniques to monitor their stability and mobility in situ. One way to monitor the stability and mobility of magnetic nanoparticles biological fluids in situ is through dynamic magnetic susceptibility measurements (DMS), which under certain conditions provide a measure of the particle's rotational diffusivity. For magnetic nanoparticles modified to have commonly used biomedical surface coatings, we describe a systematic comparison of DMS measurements in whole blood and tumor tissue explants. DMS measurements clearly demonstrated that stability and mobility changed over time and from one medium to another for each different coating. It was found that nanoparticles coated with covalently grafted, dense layers of PEG were the only ones to show good stability and mobility in all settings tested. These studies illustrate the utility of DMS measurements to estimate the stability and mobility of nanoparticles in situ, and which can provide insights that lead to engineering better nanoparticles for in vivo use.


Asunto(s)
Magnetismo , Nanopartículas , Sangre , Humanos , Neoplasias/metabolismo , Propiedades de Superficie
5.
J Clin Invest ; 126(11): 4174-4186, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27721239

RESUMEN

Carcinoma cells can acquire increased motility and invasiveness through epithelial-to-mesenchymal transition (EMT). However, the significance of EMT in cancer metastasis has been controversial, and the exact fates and functions of EMT cancer cells in vivo remain inadequately understood. Here, we tracked epithelial cancer cells that underwent inducible or spontaneous EMT in various tumor transplantation models. Unlike epithelial cells, the majority of EMT cancer cells were specifically located in the perivascular space and closely associated with blood vessels. EMT markedly activated multiple pericyte markers in carcinoma cells, in particular PDGFR-ß and N-cadherin, which enabled EMT cells to be chemoattracted towards and physically interact with endothelium. In tumor xenografts generated from carcinoma cells that were prone to spontaneous EMT, a substantial fraction of the pericytes associated with tumor vasculature were derived from EMT cancer cells. Depletion of such EMT cells in transplanted tumors diminished pericyte coverage, impaired vascular integrity, and attenuated tumor growth. These findings suggest that EMT confers key pericyte attributes on cancer cells. The resulting EMT cells phenotypically and functionally resemble pericytes and are indispensable for vascular stabilization and sustained tumor growth. This study thus proposes a previously unrecognized role for EMT in cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Transición Epitelial-Mesenquimal , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Pericitos/metabolismo , Células A549 , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Humanos , Células MCF-7 , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/patología , Pericitos/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
6.
Int J Cancer ; 138(11): 2665-77, 2016 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26757413

RESUMEN

It is estimated that approximately 90% of patients with advanced prostate cancer develop bone metastases; an occurrence that results in a substantial reduction in the quality of life and a drastic worsening of prognosis. The development of novel therapeutic strategies that impair the metastatic process and associated skeletal adversities is therefore critical to improving prostate cancer patient survival. Recognition of the importance of Cathepsin L (CTSL) to metastatic dissemination of cancer cells has led to the development of several CTSL inhibition strategies. The present investigation employed intra-cardiac injection of human PC-3ML prostate cancer cells into nude mice to examine tumor cell dissemination in a preclinical bone metastasis model. CTSL knockdown confirmed the validity of targeting this protease and subsequent intervention studies with the small molecule CTSL inhibitor KGP94 resulted in a significant reduction in metastatic tumor burden in the bone and an improvement in overall survival. CTSL inhibition by KGP94 also led to a significant impairment of tumor initiated angiogenesis. Furthermore, KGP94 treatment decreased osteoclast formation and bone resorptive function, thus, perturbing the reciprocal interactions between tumor cells and osteoclasts within the bone microenvironment which typically result in bone loss and aggressive growth of metastases. These functional effects were accompanied by a significant downregulation of NFκB signaling activity and expression of osteoclastogenesis related NFκB target genes. Collectively, these data indicate that the CTSL inhibitor KGP94 has the potential to alleviate metastatic disease progression and associated skeletal morbidities and hence may have utility in the treatment of advanced prostate cancer patients.


Asunto(s)
Neoplasias Óseas/genética , Catepsina L/genética , Osteoclastos/metabolismo , Neoplasias de la Próstata/genética , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Resorción Ósea/genética , Resorción Ósea/patología , Catepsina L/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Osteoclastos/patología , Neoplasias de la Próstata/patología , Tiosemicarbazonas/administración & dosificación , Tiourea/administración & dosificación , Tiourea/análogos & derivados , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Res ; 13(3): 461-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25373480

RESUMEN

UNLABELLED: Epigenetic alteration is a hallmark of all cancers. Such alterations lead to modulation of fundamental cancer-related functions, such as proliferation, migration, and invasion. In particular, methylation of Histone H3 Lysine 4 (H3K4), a histone mark generally associated with transcriptional activation, is altered during progression of several human cancers. While the depletion of H3K4 demethylases promotes breast cancer metastasis, the effect of H3K4 methyltransferases on metastasis is not clear. Nevertheless, gene duplications in the human SETD1A (hSETD1A) H3K4 methyltransferase are present in almost half of breast cancers. Herein, expression analysis determined that hSETD1A is upregulated in multiple metastatic human breast cancer cell lines and clinical tumor specimens. Ablation of hSETD1A in breast cancer cells led to a decrease in migration and invasion in vitro and to a decrease in metastasis in nude mice. Furthermore, a group of matrix metalloproteinases (including MMP2, MMP9, MMP12, MMP13, and MMP17) were identified which were downregulated upon depletion of hSETD1A and demonstrated a decrease in H3K4me3 at their proximal promoters based on chromatin immunoprecipitation analysis. These results provide evidence for a functional and mechanistic link among hSETD1A, MMPs, and metastasis in breast cancer, thereby supporting an oncogenic role for hSETD1A in cancer. IMPLICATIONS: This study reveals that hSETD1A controls tumor metastasis by activating MMP expression and provides an epigenetic link among hSETD1A, MMPs, and metastasis of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular , Epigénesis Genética , Femenino , Histonas/metabolismo , Humanos , Células MCF-7 , Metaloproteinasas de la Matriz/genética , Metilación , Ratones , Metástasis de la Neoplasia , Regulación hacia Arriba
8.
Neoplasia ; 15(4): 454-60, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23555190

RESUMEN

Adenomatous polyposis coli (APC) is a multifunctional protein having diverse cellular functions including cell migration, cell-cell adhesion, cell cycle control, chromosomal segregation, and apoptosis. Recently, we found a new role of APC in base excision repair (BER) and showed that it interacts with DNA polymerase ß and 5'-flap endonuclease 1 and interferes in BER. Previously, we have also reported that cigarette smoke condensate (CSC) increases expression of APC and enhances the growth of normal human breast epithelial (MCF10A) cells in vitro. In the present study, using APC overexpression and knockdown systems, we have examined the molecular mechanisms by which CSC and its major component, Benzo[α]pyrene, enhances APC-mediated accumulation of abasic DNA lesions, which is cytotoxic and mutagenic in nature, leading to enhanced neoplastic transformation of MCF10A cells in an orthotopic xenograft model.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Neoplasias de la Mama/genética , Mama/patología , Transformación Celular Neoplásica/metabolismo , Daño del ADN , Células Epiteliales/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Ácido Apurínico/genética , Benzo(a)pireno/toxicidad , Neoplasias de la Mama/etiología , Neoplasias de la Mama/patología , Carcinógenos/toxicidad , Línea Celular , Transformación Celular Neoplásica/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , ARN Interferente Pequeño/genética , Humo/efectos adversos , Nicotiana/química
9.
Blood ; 121(4): 707-15, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23212515

RESUMEN

VEGF is a key regulator of normal and pathologic angiogenesis. Although many trans-activating factors of VEGF have been described, the transcriptional repression of VEGF remains much less understood. We have previously reported the identification of a SCAN domain-containing C2H2 zinc finger protein, ZNF24, that represses the transcription of VEGF. In the present study, we identify the mechanism by which ZNF24 represses VEGF transcription. Using reporter gene and electrophoretic mobility shift assays, we identify an 11-bp fragment of the proximal VEGF promoter as the ZNF24-binding site that is essential for ZNF24-mediated repression. We demonstrate in 2 in vivo models the potent inhibitory effect of ZNF24 on the vasculature. Expression of human ZNF24 induced in vivo vascular defects consistent with those induced by VEGF knockdown using a transgenic zebrafish model. These defects could be rescued by VEGF overexpression. Overexpression of ZNF24 in human breast cancer cells also inhibited tumor angiogenesis in an in vivo tumor model. Analyses of human breast cancer tissues showed that ZNF24 and VEGF levels were inversely correlated in malignant compared with normal tissues. These data demonstrate that ZNF24 represses VEGF transcription through direct binding to an 11-bp fragment of the VEGF proximal promoter and that it functions as a negative regulator of tumor growth by inhibiting angiogenesis.


Asunto(s)
Vasos Sanguíneos/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Sitios de Unión , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Represoras/genética , Activación Transcripcional , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra
10.
Clin Exp Metastasis ; 29(3): 253-61, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22286523

RESUMEN

c-Met tyrosine kinase hyperactivation is strongly associated with tumor metastasis. In a prior study we showed that BMS-777607, a novel selective small molecule Met kinase inhibitor, potently suppressed ligand-mediated functions in prostate cancer cells. Herein we evaluated the impact of this agent on the potential of the highly metastatic murine KHT sarcoma that carries constitutive activated c-Met. MET gene knockdown was found to reduce spontaneous cell scatter and motility, suggesting a c-Met-dependent disseminating ability in KHT cells. Furthermore, BMS-777607 treatment potently inhibited KHT cell scatter, motility and invasion at doses in the nanomolar range. In contrast, cell proliferation and clonogenicity were modestly affected by BMS-777607. At the molecular level, BMS-777607 potently blocked phosphorylation of c-Met and downstream pathways over the same dose range that impacted metastasis-associated cell functions. In vivo, daily treatment with BMS-777607 (25 mg/kg/day) over the course of the study significantly decreased the number of KHT lung tumor nodules (28.3 ± 14.9%, P < 0.001) without apparent systemic toxicity. While treatment for short intervals (day 1 or 4) clearly reduced the foci number, delaying the initiation of BMS-777607 treatment until 8 days after tumor cell injection failed to show any reduction, implying that impairment of the initiation phases of the secondary growth via c-Met targeting is required to constrain the formation of macroscopic metastases. Together, the present findings demonstrate that the disruption of c-Met signaling by BMS-777607 significantly impairs the metastatic phenotype, suggesting that this agent may have therapeutic utility in targeting cancer metastasis.


Asunto(s)
Aminopiridinas/uso terapéutico , Metástasis de la Neoplasia/prevención & control , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Piridonas/uso terapéutico , Sarcoma Experimental/tratamiento farmacológico , Aminopiridinas/farmacología , Animales , Línea Celular Tumoral , Movimiento Celular , Femenino , Humanos , Ratones , Ratones Endogámicos C3H , Invasividad Neoplásica , Piridonas/farmacología , Sarcoma Experimental/patología , Transducción de Señal/efectos de los fármacos
11.
Proc Natl Acad Sci U S A ; 108(37): 15231-6, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21896759

RESUMEN

Angiogenesis is meticulously controlled by a fine balance between positive and negative regulatory activities. Vascular endothelial growth factor (VEGF) is a predominant angiogenic factor and its dosage is precisely regulated during normal vascular formation. In cancer, VEGF is commonly overproduced, resulting in abnormal neovascularization. VEGF is induced in response to various stimuli including hypoxia; however, very little is known about the mechanisms that confine its induction to ensure proper angiogenesis. Chromatin insulation is a key transcription mechanism that prevents promiscuous gene activation by interfering with the action of enhancers. Here we show that the chromatin insulator-binding factor CTCF binds to the proximal promoter of VEGF. Consistent with the enhancer-blocking mode of chromatin insulators, CTCF has little effect on basal expression of VEGF but specifically affects its activation by enhancers. CTCF knockdown cells are sensitized for induction of VEGF and exhibit elevated proangiogenic potential. Cancer-derived CTCF missense mutants are mostly defective in blocking enhancers at the VEGF locus. Moreover, during mouse retinal development, depletion of CTCF causes excess angiogenesis. Therefore, CTCF-mediated chromatin insulation acts as a crucial safeguard against hyperactivation of angiogenesis.


Asunto(s)
Cromatina/metabolismo , Elementos Aisladores/genética , Neovascularización Patológica/genética , Proteínas Represoras/metabolismo , Dedos de Zinc/genética , Animales , Factor de Unión a CCCTC , Línea Celular , Elementos de Facilitación Genéticos/genética , Genes Reporteros/genética , Humanos , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Retina/crecimiento & desarrollo , Retina/patología , Transcripción Genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Microvasc Res ; 81(1): 44-51, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20974154

RESUMEN

Unlike normal blood vessels, the unique characteristics of an expanding, disorganized and leaky tumor vascular network can be targeted for therapeutic gain by vascular disrupting agents (VDAs), which promote rapid and selective collapse of tumor vessels, causing extensive secondary cancer cell death. A hallmark observation following VDA treatment is the survival of neoplastic cells at the tumor periphery. However, comparative studies with the second generation tubulin-binding VDA OXi4503 indicate that the viable rim of tumor tissue remaining following treatment with this agent is significantly smaller than that seen for the lead VDA, combretastatin. OXi4503 is the cis-isomer of CA1P and it has been speculated that this agent's increased antitumor efficacy may be due to its reported metabolism to orthoquinone intermediates leading to the formation of cytotoxic free radicals. To examine this possibility in situ, KHT sarcoma-bearing mice were treated with either the cis- or trans-isomer of CA1P. Since both isomers can form quinone intermediates but only the cis-isomer binds tubulin, such a comparison allows the effects of vascular collapse to be evaluated independently from those caused by the reactive hydroxyl groups. The results showed that the cis-isomer (OXi4503) significantly impaired tumor blood flow leading to secondary tumor cell death and >95% tumor necrosis 24h post drug exposure. Treatment with the trans-isomer had no effect on these parameters. However, the combination of the trans-isomer with combretastatin increased the antitumor efficacy of the latter agent to near that of OXi4503. These findings indicate that while the predominant in vivo effect of OXi4503 is clearly due to microtubule collapse and vascular shut-down, the formation of toxic free radicals likely contributes to its enhanced potency.


Asunto(s)
Antineoplásicos/farmacología , Difosfatos/farmacología , Difosfatos/uso terapéutico , Radicales Libres/metabolismo , Microtúbulos/efectos de los fármacos , Sarcoma Experimental/tratamiento farmacológico , Estilbenos/farmacología , Estilbenos/uso terapéutico , Moduladores de Tubulina/farmacología , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Difosfatos/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C3H , Microtúbulos/patología , Necrosis/patología , Neovascularización Fisiológica/efectos de los fármacos , Flujo Sanguíneo Regional/efectos de los fármacos , Sarcoma Experimental/irrigación sanguínea , Sarcoma Experimental/patología , Estilbenos/metabolismo , Moduladores de Tubulina/metabolismo , Moduladores de Tubulina/uso terapéutico , Ensayo de Tumor de Célula Madre
13.
Anticancer Res ; 30(11): 4405-13, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21115886

RESUMEN

BACKGROUND: Src, a non-receptor tyrosine kinase frequently overexpressed and highly activated in malignancies, has been associated with a poor patient prognosis. The aim of the present studies was to examine the impact of an Src inhibitor (saracatinib) on a highly metastatic murine sarcoma cell line (KHT). MATERIALS AND METHODS: Phosphorylation of Src and downstream effectors was determined using Western immunoblotting. Cell cycle was analyzed by flow cytometry using propidium iodide DNA staining, migration and invasion in modified Boyden chambers, activated MMP-9 by gel zymography, and visualization of pSrc and pFAK by confocal immunofluorescence. The number of KHT lung nodules in saracatinib-treated mice was compared to controls. RESULTS: Saracatinib inhibited major pathways in the metastatic cascade in vitro, including Src and FAK activation. Functions required for metastasis, such as migration and invasion, were reduced when cells were exposed to 0.5 µM and 1.0 µM saracatinib, respectively (p<0.0001). Pretreatment of KHT cells with either 1 µM or 5 µM saracatinib prior to tail vein injection decreased lung colonies in mice from 13.0 to 5.0 (p<0.05) and less than 1.0 (p<0.01), respectively. CONCLUSION: These findings suggest that Src inhibition by saracatinib may reduce the metastatic activity of tumor cells.


Asunto(s)
Benzodioxoles/farmacología , Modelos Animales de Enfermedad , Fibrosarcoma/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Quinazolinas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Animales , Apoptosis , Western Blotting , Adhesión Celular , Ciclo Celular , Movimiento Celular , Proliferación Celular , Femenino , Fibrosarcoma/patología , Técnica del Anticuerpo Fluorescente , Quinasa 1 de Adhesión Focal/metabolismo , Ratones , Ratones Endogámicos C3H , Fosforilación , Sarcoma Experimental/tratamiento farmacológico , Sarcoma Experimental/patología , Tasa de Supervivencia , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...