Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 116(4): 479-488, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-28103615

RESUMEN

BACKGROUND: The prognosis of triple-negative breast cancer (TNBC) is still difficult to establish. Some TNBC benefit from radiotherapy (RT) and are cured, while in other patients metastases appear during the first 3 years after treatment. In this study, an animal model of TNBC was used to determine whether the expression of the cell membrane protease MT1-MMP in cancer cells was associated with radiation-stimulated development of lung metastases. METHODS: Using invasion chambers, irradiated fibroblasts were used as chemoattractants to assess the invasiveness of TNBC D2A1 cell lines showing downregulated expression of MT1-MMP, which were compared with D2A1-wt (wild-type) and D2A1 shMT1-mock (empty vector) cell lines. In a mouse model, a mammary gland was irradiated followed by the implantation of the downregulated MT1-MMP D2A1, D2A1-wt or D2A1 shMT1-mock cell lines. Migration of D2A1 cells in the mammary gland, number of circulating tumour cells and development of lung metastases were assessed. RESULTS: The reduction of MT1-MMP expression decreased the invasiveness of D2A1 cells and blocked the radiation enhancement of cancer cell invasion. In BALB/c mice, irradiation of the mammary gland has stimulated the invasion of cancer cells, which was associated with a higher number of circulating tumour cells and of lung metastases. These adverse effects of radiation were prevented by downregulating the MT1-MMP. CONCLUSIONS: This study shows that the MT1-MMP is necessary for the radiation enhancement of lung metastasis development, and that its expression level and/or localisation could be evaluated as a biomarker for predicting the early recurrence observed in some TNBC patients.


Asunto(s)
Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Metaloproteinasa 14 de la Matriz/genética , Neoplasias Inducidas por Radiación/patología , Neoplasias de la Mama Triple Negativas/patología , Células 3T3 , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Neoplasias Inducidas por Radiación/genética , Neoplasias de la Mama Triple Negativas/genética
2.
BMC Cancer ; 17(1): 157, 2017 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-28235401

RESUMEN

BACKGROUND: Suppressor of cytokine signaling 1 (SOCS1) is considered a tumor suppressor due to frequent epigenetic and micro-RNA-mediated repression of its gene expression in diverse cancers. In prostate cancer (PCa), elevated expression of miR-30d that targets SOCS1 mRNA is associated with increased risk of disease recurrence. SOCS1 can mediate its tumor suppressor functions by diverse mechanisms such as inhibiting the JAK-STAT signaling pathway, promoting the tumor suppressor functions of p53, attenuating MET receptor tyrosine kinase signaling and blocking the oncogenic potential of the cell cycle inhibitor p21CIP1 (p21). Here, we studied the expression of SOCS1 and the downstream targets of its putative tumor suppressor functions (p53, MET and p21) in human PCa specimens to evaluate their significance as markers of disease prognosis. METHODS: Tissue microarrays were constructed of 78 archived prostatectomy specimens that were grouped according to the recommendations of the International Society of Urological Pathology (ISUP) based on the Gleason patterns. SOCS1, p53, MET and p21 protein expression were evaluated by immunohistochemical staining alongside the common prostate cancer-related markers Ki67, prostein and androgen receptor. Statistical correlations between the staining intensities of these markers and ISUP grade groups, local invasion or lymph node metastasis were evaluated. RESULTS: SOCS1 showed diffuse staining in the prostatic epithelium. SOCS1 staining intensity correlated inversely with the ISUP grade groups (ρ = -0.4687, p <0.0001) and Ki67 (ρ = -0.2444, p = 0.031), and positively with prostein (ρ = 0.3511, p = 0.0016). Changes in SOCS1 levels did not significantly associate with those of p53, MET or p21. However, p21 positively correlated with androgen receptor expression (ρ = -0.1388, p = 0.0003). A subset of patients with regional lymph node metastasis, although small in number, showed reduced SOCS1 expression and increased expression of MET and p21. CONCLUSIONS: Our findings suggest that evaluating SOCS1 and p21 protein expression in prostatectomy specimens may have a prognostic value in identifying the aggressive disease. Hence, prospective studies with larger numbers of metastatic PCa specimens incorporating clinical correlates such as disease-free and overall survival are warranted.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas/genética , Genes Supresores de Tumor , Humanos , Masculino , Pronóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo
3.
Biochem J ; 473(11): 1617-27, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27048591

RESUMEN

Met receptor tyrosine kinase (RTK) is known to bind to the three distinct protein isoforms encoded by the ShcA (Shc) gene. Structure-function studies have unveiled critical roles for p52Shc-dependent signalling pathways in Met-regulated biological functions. The molecular basis of the interaction between the Met and p52Shc proteins is well-defined, but not for the longest protein isoform, p66Shc. In the present study, co-immunoprecipitation assays were performed in human embryonic kidney 293 (HEK293) cells, transiently co-transfected with Met and p66Shc mutants, in order to define the molecular determinants involved in mediating Met-p66Shc interaction. Our results show that p66Shc interacts constitutively with the receptor Met, and the Grb2 (growth factor receptor-bound protein-2) and Gab1 (Grb2-associated binder-1) adaptor proteins. Although its phosphotyrosine-binding domain (PTB) and Src homology 2 (SH2) domains co-ordinate p66Shc binding to non-activated Met receptor, these phosphotyrosine-binding modules, and its collagen homology domain 2 (CH2) region, exert negative constraints. In contrast, p66Shc interaction with the activated Met depends mainly on the integrity of its PTB domain, and to a lesser extent of its SH2 domain. Even though not required for the recruitment of p66Shc, tyrosine phosphorylation of p66Shc by activated Met enhances these interactions by mechanisms not reliant on the integrity of the Met multisubstrate-binding site. In turn, this increases phosphotyrosine-dependent p66Shc-Grb2-Gab1 complex formation away from the receptor, while blocking Grb2 and Gab1 recruitment to activated Met. In conclusion, we identify, for the first time, a novel non-canonical dynamic mode of interaction between Met and the p66 protein isoform of Shc and its effects on rewiring binding effector complexes according to the activation state of the receptor.


Asunto(s)
Proteínas Proto-Oncogénicas c-met/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Dominio BTB-POZ/genética , Dominio BTB-POZ/fisiología , Sitios de Unión/genética , Sitios de Unión/fisiología , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Mutación/genética , Fosforilación/genética , Fosforilación/fisiología , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Dominios Homologos src/genética , Dominios Homologos src/fisiología
4.
Cytokine ; 82: 87-94, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26811119

RESUMEN

The Suppressor Of Cytokine Signaling 1 (SOCS1) has been extensively investigated in immune cells where it works as a potent inhibitor of inflammation by negative feedback regulation of the cytokine-activated JAK-STAT signaling pathways. SOCS1 is also recognized as a tumor suppressor in numerous cancers and its critical functional relevance in non-immune cells, including epithelial cells, has just begun to emerge. Most notably, conflicting results from clinical and experimental studies suggest that SOCS1 may function as either a tumor suppressor or a tumor promoter, in a cell context-dependent manner. Here, we present an overview of the mechanisms underlying SOCS1 function as a tumor suppressor and discuss the emerging evidences of SOCS1 activity as an oncogene.


Asunto(s)
Neoplasias , Oncogenes , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de Tumor , Animales , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
5.
BMC Cancer ; 16: 361, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27282478

RESUMEN

BACKGROUND: Some triple negative breast cancer (TNBC) patients are at higher risk of recurrence in the first three years after treatment. This rapid relapse has been suggested to be associated with inflammatory mediators induced by radiation in healthy tissues that stimulate cancer cell migration and metastasis formation. In this study, the ability of chloroquine (CQ) to inhibit radiation-stimulated development of metastasis was assessed. METHODS: The capacity of CQ to prevent radiation-enhancement of cancer cell invasion was assessed in vitro with the TNBC cell lines D2A1, 4T1 and MDA-MB-231 and the non-TNBC cell lines MC7-L1, and MCF-7. In Balb/c mice, a single mammary gland was irradiated with four daily doses of 6 Gy. After the last irradiation, irradiated and control mammary glands were implanted with D2A1 cells. Mice were treated with CQ (vehicle, 40 or 60 mg/kg) 3 h before each irradiation and then every 72 h for 3 weeks. Migration of D2A1 cells in the mammary gland, the number of circulating tumor cells and lung metastasis were quantified, and also the expression of some inflammatory mediators. RESULTS: Irradiated fibroblasts have increased the invasiveness of the TNBC cell lines only, a stimulation that was prevented by CQ. On the other hand, invasiveness of the non-TNBC cell lines, which was not enhanced by irradiated fibroblasts, was also not significantly modified by CQ. In Balb/c mice, treatment with CQ prevented the stimulation of D2A1 TNBC cell migration in the pre-irradiated mammary gland, and reduced the number of circulating tumor cells and lung metastases. This protective effect of CQ was associated with a reduced expression of the inflammatory mediators interleukin-1ß, interleukin-6, and cyclooxygenase-2, while the levels of matrix metalloproteinases-2 and -9 were not modified. CQ also promoted a blocking of autophagy. CONCLUSION: CQ prevented radiation-enhancement of TNBC cell invasion and reduced the number of lung metastases in a mouse model.


Asunto(s)
Cloroquina/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Células Neoplásicas Circulantes/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/radioterapia , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Cloroquina/farmacología , Ciclooxigenasa 2/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interleucinas/metabolismo , Neoplasias Pulmonares/secundario , Células MCF-7 , Metaloproteinasas de la Matriz/metabolismo , Ratones , Metástasis de la Neoplasia , Resultado del Tratamiento , Neoplasias de la Mama Triple Negativas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Biol Chem ; 289(9): 5687-703, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24407288

RESUMEN

Signals downstream of growth factor receptors play an important role in mammary carcinogenesis. Recently, we demonstrated that the small GTPases ARF1 and ARF6 were shown to be activated downstream of the epidermal growth factor receptor (EGFR) and act as a key regulator of growth, migration, and invasion of breast cancer cells. However, the mechanism via which the EGFR recruits and activates ARF1 and ARF6 to transmit signals has yet to be fully elucidated. Here, we identify adaptor proteins Grb2 and p66Shc as important regulators mediating ARF activation. We demonstrate that ARF1 can be found in complex with Grb2 and p66Shc upon EGF stimulation of the basal-like breast cancer MDA-MB-231 cell line. However, we report that these two adaptors regulate ARF1 activation differently, with Grb2 promoting ARF1 activation and p66Shc blocking this response. Furthermore, we show that Grb2 is essential for the recruitment of ARF1 to the EGFR, whereas p66Shc hindered ARF1 receptor recruitment. We demonstrate that the negative regulatory role of p66Shc stemmed from its ability to block the recruitment of Grb2/ARF1 to the EGFR. Conversely, p66Shc potentiates ARF6 activation as well as the recruitment of this ARF isoform to the EGFR. Interestingly, we demonstrate that Grb2 is also required for the activation and receptor recruitment of ARF6. Additionally, we show an important role for p66Shc in modulating ARF activation, cell growth, and migration in HER2-positive breast cancer cells. Together, our results highlight a central role for adaptor proteins p66Shc and Grb2 in the regulation of ARF1 and ARF6 activation in invasive breast cancer cells.


Asunto(s)
Factor 1 de Ribosilacion-ADP/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Neoplasias de la Mama/metabolismo , Proteína Adaptadora GRB2/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Transducción de Señal , Factor 1 de Ribosilacion-ADP/genética , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Activación Enzimática/genética , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Proteína Adaptadora GRB2/genética , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas Adaptadoras de la Señalización Shc/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
7.
BMC Cancer ; 14: 240, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24708867

RESUMEN

BACKGROUND: Deregulation of receptor tyrosine kinases (RTK) contributes to the initiation and progression of intestinal-derived epithelial cancers, including colorectal cancer (CRC). However, the roles of the proximal signaling molecules engaged by RTKs in different oncogenic functions of CRC remain unclear. METHODS: Herein, the functional impact of expressing variant forms of the oncogenic Met receptor (Tpr-Met) that selectively recruit the adaptor proteins Grb2 or Shc was investigated in a model derived from normal intestinal epithelial cells (IEC-6). An RNA interference (RNAi) approach was used to define the requirement of Grb2 or Shc in Tpr-Met-transformed IEC-6 cells. Since Grb2 and Shc couple RTKs to the activation of the Ras/MEK/Erk and PI3K/Akt pathways, Erk and Akt phosphorylation/activation states were monitored in transformed IEC-6 cells, and a pharmacological approach was employed to provide insights into the roles of these pathways in oncogenic processes evoked by activated Met, and downstream of Grb2 and Shc. RESULTS: We show, for the first time, that constitutive activation of either Grb2 or Shc signals in IEC-6 cells, promotes morphological transformation associated with down-regulation of E-cadherin, as well as increased cell growth, loss of growth contact inhibition, anchorage-independent growth, and resistance to serum deprivation and anoikis. Oncogenic activation of Met was revealed to induce morphological transformation, E-cadherin down-regulation, and protection against anoikis by mechanisms dependent on Grb2, while Shc was shown to be partly required for enhanced cell growth. The coupling of activated Met to the Ras/MEK/Erk and PI3K/Akt pathways, and the sustained engagement of Grb2 or Shc in IECs, was shown to trigger negative feedback, limiting the extent of activation of these pathways. Nonetheless, morphological alterations and E-cadherin down-regulation induced by the oncogenic Tpr-Met, and by Grb2 or Shc signals, were blocked by MEK, but not PI3K, inhibitors while the enhanced growth and resistance to anoikis induced by Tpr-Met were nearly abolished by co-treatment with both inhibitors. CONCLUSION: Overall, these results identify Grb2 and Shc as central signaling effectors of Met-driven progression of intestinal epithelial-derived cancers. Notably, they suggest that Grb2 may represent a promising target for the design of novel CRC therapies.


Asunto(s)
Neoplasias Colorrectales/genética , Proteína Adaptadora GRB2/biosíntesis , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Adaptadoras de la Señalización Shc/biosíntesis , Cadherinas/metabolismo , Línea Celular , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Células Epiteliales/metabolismo , Proteína Adaptadora GRB2/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Intestinos/patología , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Transducción de Señal/genética
8.
Biosci Rep ; 44(6)2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38836326

RESUMEN

Cumulative research findings support the idea that endocytic trafficking is crucial in regulating receptor signaling and associated diseases. Specifically, strong evidence points to the involvement of sorting nexins (SNXs), particularly SNX1 and SNX2, in the signaling and trafficking of the receptor tyrosine kinase (RTK) MET in colorectal cancer (CRC). Activation of hepatocyte growth factor (HGF) receptor MET is a key driver of CRC progression. In the present study, we utilized human HCT116 CRC cells with SNX1 and SNX2 genes knocked out to demonstrate that their absence leads to a delay in MET entering early endosomes. This delay results in increased phosphorylation of both MET and AKT upon HGF stimulation, while ERK1/2 (extracellular signal-regulated kinases 1 and 2) phosphorylation remains unaffected. Despite these changes, HGF-induced cell proliferation, scattering, and migration remain similar between the parental and the SNX1/2 knockout cells. However, in the absence of SNX1 and SNX2, these cells exhibit increased resistance to TRAIL-induced apoptosis. This research underscores the intricate relationship between intracellular trafficking, receptor signaling, and cellular responses and demonstrates for the first time that the modulation of MET trafficking by SNX1 and SNX2 is critical for receptor signaling that may exacerbate the disease.


Asunto(s)
Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales , Factor de Crecimiento de Hepatocito , Proteínas Proto-Oncogénicas c-met , Nexinas de Clasificación , Humanos , Nexinas de Clasificación/metabolismo , Nexinas de Clasificación/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/genética , Células HCT116 , Factor de Crecimiento de Hepatocito/metabolismo , Transducción de Señal , Fosforilación , Endosomas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transporte de Proteínas
9.
J Hepatol ; 55(6): 1300-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21703184

RESUMEN

BACKGROUND & AIMS: Frequent repression of the Socs1 (suppressor of cytokine signaling 1) gene in hepatocellular carcinoma (HCC) and increased susceptibility of SOCS1-deficient mice to hepatocarcinogens suggest a tumor suppressor role for SOCS1 in the liver, but the underlying mechanisms remain unclear. Here we investigated the role of SOCS1 in regulating hepatocyte proliferation following partial hepatectomy and HGF stimulation. METHODS: Because Socs1(-/-) mice die prematurely due to deregulated IFNγ signaling, we used Socs1(-/-)Ifng(-/-) mice to study the role of SOCS1 in liver regeneration following partial hepatectomy. We examined the activation of signaling molecules downstream of IL-6 and hepatocyte growth factor (HGF) receptors in the regenerating liver, primary hepatocytes, and in human hepatoma cells. We examined the interaction between SOCS1 and the HGF receptor c-Met by reciprocal immunoprecipitation. RESULTS: Socs1(-/-)Ifng(-/-) mice displayed accelerated liver regeneration with increased DNA synthesis compared to Ifng(-/-) and wild type mice. The regenerating liver of Socs1(-/-)Ifng(-/-) mice did not show increased IL-6 signaling, but displayed earlier phosphorylation of Gab1, a signaling adaptor downstream of c-Met. Following HGF stimulation, hepatocytes from Socs1(-/-)Ifng(-/-) mice displayed increased phosphorylation of c-Met and Gab1, cell migration and proliferation. Accordingly, SOCS1 overexpression attenuated HGF-induced phosphorylation of c-Met, Gab1, and ERK1/2 in hepatoma cells, and decreased their proliferation and migration. SOCS1 interacted with the Tpr-Met, an oncogenic form of the Met receptor. CONCLUSIONS: SOCS1 attenuates c-Met signaling and thus negative regulation of HGF signaling could be an important mechanism underlying the anti-tumor role of SOCS1 in the liver.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Hepatocitos/fisiología , Regeneración Hepática/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN/genética , Humanos , Interferón gamma/deficiencia , Interferón gamma/genética , Interleucina-6/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Oncogénica tpr-met/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/deficiencia , Proteínas Supresoras de la Señalización de Citocinas/genética
10.
Am J Physiol Gastrointest Liver Physiol ; 299(3): G677-86, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20539003

RESUMEN

The deregulation of Met/hepatocyte growth factor (HGF) receptor tyrosine kinase signaling constitutes a common event in colorectal cancers. However, the physiopathological functions of such a deregulation remain poorly understood. In the present study, we investigated the role of the deregulation of Met receptor in the neoplastic transformation of intestinal epithelial cells. To do so, the normal, well-established and characterized rat intestinal epithelial IEC-6 cells were transduced with a retrovirus carrying the oncogenic constitutive active form of Met receptor, Tpr-Met. Herein, we show that compared with control IEC-6 cells, Tpr-Met-IEC-6 cells exhibit enhanced proliferation, loss of growth-contact inhibition, cell morphological alterations, actin cytoskeletal reorganization, loss of E-cadherin expression and anchorage-independent growth. Moreover, Tpr-Met-IEC-6 cells are conferred the capacity to produce the proangiogenic factor VEGF and to reduce the potent antiangiogenic factor thrombospondin-1. Of significance, Tpr-Met-IEC-6 cells are endowed with the ability to elicit angiogenic responses and to form tumors and metastases in vivo. Hence, our study demonstrates for the first time that the sole oncogenic engagement of Met receptor in normal intestinal epithelial cells is sufficient to induce a wide array of cancerous biological processes that are fundamental to the initiation and malignant progression of colorectal cancers.


Asunto(s)
Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular , Línea Celular , Proliferación Celular , Citoesqueleto , Mucosa Intestinal , Metástasis de la Neoplasia , Neoplasias/metabolismo , Proteína Oncogénica tpr-met , Proteínas Proto-Oncogénicas c-met/genética , Ratas , Trombospondina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Int J Cancer ; 125(7): 1575-86, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19462441

RESUMEN

Constitutive activation of the MAP kinase kinase MEK1 induces oncogenic transformation in intestinal epithelial cells. Loss of cell-cell adhesion followed by the dissociation of epithelial structures is a prerequisite for increased cell motility and tumor invasion. This phenotypic switch is designated epithelial-to-mesenchymal transition (EMT). EMT also plays an important role in determining the dissemination of tumors. However, the role of MEK1 in intestinal EMT, tumor invasion and metastasis has not been elucidated. To determine the functions of activated MEK1 in intestinal tumorigenesis, we established intestinal epithelial cell lines that overexpress wild-type MEK1 (wtMEK) or activated MEK1 (caMEK). Our results indicate that expression of caMEK is sufficient to induce EMT as confirmed with the induction of N-cadherin, vimentin, Snail1 and Snail2, whereas a reduction in E-cadherin, occludin, ZO-1 and cortical F-actin was noted. The Snail1 and Snail2 promoter analyses revealed that Egr-1 and Fra-1, an AP-1 protein, are responsible for MEK1-induced Snail1 and Snail2 expression, respectively. Cells expressing activated MEK1 clearly acquired an invasive capacity when compared to wtMEK-expressing cells. Zymography studies confirmed elevated levels of MMP2 and MMP9 activities in media of caMEK-expressing cells. Importantly, cells expressing activated MEK1 induced tumors with short latency in correlation with their ability to induce experimental metastasis in vivo and to express factors known to promote colorectal cancer cell metastasis. In conclusion, our results demonstrate, for the first time, that constitutive activation of MEK1 in intestinal epithelial cells is sufficient to induce an EMT associated with tumor invasion and metastasis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Células Epiteliales/patología , MAP Quinasa Quinasa 1/metabolismo , Mesodermo/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Animales , Inmunoprecipitación de Cromatina , Neoplasias Colorrectales/metabolismo , Células Epiteliales/enzimología , Técnica del Anticuerpo Fluorescente , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Intestinos , MAP Quinasa Quinasa 1/genética , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Mesodermo/enzimología , Ratones , Ratones Desnudos , Mutación , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
12.
Mol Biol Cell ; 17(9): 3717-28, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16775003

RESUMEN

We have shown previously that either Grb2- or Shc-mediated signaling from the oncogenic Met receptor Tpr-Met is sufficient to trigger cell cycle progression in Xenopus oocytes. However, direct binding of these adaptors to Tpr-Met is dispensable, implying that another Met binding partner mediates these responses. In this study, we show that overexpression of Grb2-associated binder 1 (Gab1) promotes cell cycle progression when Tpr-Met is expressed at suboptimal levels. This response requires that Gab1 possess an intact Met-binding motif, the pleckstrin homology domain, and the binding sites for phosphatidylinositol 3-kinase and tyrosine phosphatase SHP-2, but not the Grb2 and CrkII/phospholipase Cgamma binding sites. Importantly, we establish that Gab1-mediated signals are critical for cell cycle transition promoted by the oncogenic Met and fibroblast growth factor receptors, but not by progesterone, the natural inducer of cell cycle transition in Xenopus oocytes. Moreover, Gab1 is essential for Tpr-Met-mediated morphological transformation and proliferation of fibroblasts. This study provides the first evidence that Gab1 is a key binding partner of the Met receptor for induction of cell cycle progression, proliferation, and oncogenic morphological transformation. This study identifies Gab1 and its associated signaling partners as potential therapeutic targets to impair proliferation or transformation of cancer cells in human malignancies harboring a deregulated Met receptor.


Asunto(s)
Ciclo Celular , Transformación Celular Neoplásica , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proliferación Celular , Células Cultivadas , Secuencia Conservada , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Proteína Oncogénica tpr-met/farmacología , Oocitos/citología , Oocitos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/química , Progesterona/farmacología , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
13.
Cancers (Basel) ; 11(9)2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31546971

RESUMEN

Hypoxia is a common characteristic of advanced solid tumors and a potent driver of tumor invasion and metastasis. Recent evidence suggests the involvement of autotaxin (ATX) and lysophosphatidic acid receptors (LPARs) in cancer cell invasion promoted by the hypoxic tumor microenvironment; however, the transcriptional and/or spatiotemporal control of this process remain unexplored. Herein, we investigated whether hypoxia promotes cell invasion by affecting the main enzymes involved in its production (ATX) and degradation (lipid phosphate phosphatases, LPP1 and LPP3). We report that hypoxia not only modulates the expression levels of lysophosphatidic acid (LPA) regulatory enzymes but also induces their significant spatial segregation in a variety of cancers. While LPP3 expression was downregulated by hypoxia, ATX and LPP1 were asymmetrically redistributed to the leading edge and to the trailing edge, respectively. This was associated with the opposing roles of ATX and LPPs in cell invasion. The regulated expression and compartmentalization of these enzymes of opposing function can provide an effective way to control the generation of an LPA gradient that drives cellular invasion and migration in the hypoxic zones of tumors.

14.
Carcinogenesis ; 29(3): 647-55, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18192688

RESUMEN

Hepatocyte growth factor receptor (Met) plays an important role in the progression of multiple cancer types. The overexpression of Met in DLD-1 colon carcinoma cells with kirsten rat sarcoma oncogene homolog (KRAS) oncogene activation resulted in enhanced subcutaneous and orthotopic tumor growth rate and increased metastatic potential. To elucidate the mechanism of this effect, we stably expressed kinase-inactive Met(K1110A), Src homology 2 (SH2)-binding domain-inactive Met(Y1349/1356F), growth factor receptor-bound protein 2 (Grb2) non-binding Met(N1358H) and mutant receptors with ability to selectively recruit signaling proteins Grb2, src homology domain c-terminal adaptor homolog (Shc), phospholipase c-gamma (PLCgamma) and p85 phosphatidyl inositol 3 kinase. As subcutaneous implants, DLD-1 cells that expressed the majority of these receptor constructs failed to recapitulate the tumor growth-enhancing effect of the wild-type Met receptor. The Grb2- and Shc-recruiting Met mutants demonstrated slight but consistent tumor-suppressive activity, whereas the expression of N1358H mutant stimulated tumor growth rate comparable with the wild-type receptor. This suggests that direct Grb2/Shc binding does not contribute to the tumor progression activity of Met receptor. The tumors expressing Grb2- and Shc-recruiting Met receptors demonstrated a marked loss in Grb2-associated adaptor protein 1 (Gab1) protein levels, which was not observed in the cell lines, consistent with a post-translationally regulated process. Moreover, a moderate level of Gab1 overexpression stimulated tumor growth. The findings suggest a delicate balance for intact Y1349/1356 SH2-binding domain to mediate the tumor progression activity of the coactivated Met-rat sarcoma oncogene homolog (RAS) pathways. Selectivity for specific adaptor protein involvement may be the key that determines the tissue- and cell-type specificity of Met-mediated tumorigenicity in human cancers.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Neoplasias Colorrectales/patología , Proteína Adaptadora GRB2/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Secuencia de Bases , Biomarcadores de Tumor/metabolismo , Línea Celular , Neoplasias Colorrectales/metabolismo , Cartilla de ADN , Progresión de la Enfermedad , Humanos , Proteínas Proto-Oncogénicas c-met
15.
Mol Cell Biol ; 25(21): 9632-45, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16227611

RESUMEN

The Met receptor tyrosine kinase (RTK) regulates epithelial remodeling, dispersal, and invasion and is deregulated in many human cancers. It is now accepted that impaired down-regulation, as well as sustained activation, of RTKs could contribute to their deregulation. Down-regulation of the Met receptor involves ligand-induced internalization, ubiquitination by Cbl ubiquitin ligases, and lysosomal degradation. Here we report that a ubiquitination-deficient Met receptor mutant (Y1003F) is tumorigenic in vivo. The Met Y1003F mutant is internalized, and undergoes endosomal trafficking with kinetics similar to the wild-type Met receptor, yet is inefficiently targeted for degradation. This results in sustained activation of Met Y1003F and downstream signals involving the Ras-mitogen-activated protein kinase pathway, cell transformation, and tumorigenesis. Although Met Y1003F undergoes endosomal trafficking and localizes with the cargo-sorting protein Hrs, it is unable to induce phosphorylation of Hrs. Fusion of monoubiquitin to Met Y1003F is sufficient to decrease Met receptor stability and prevent sustained MEK1/2 activation. In addition, this rescues Hrs tyrosine phosphorylation and decreases transformation in a focus-forming assay. These results demonstrate that Cbl-dependent ubiquitination is dispensable for Met internalization but is critical to target the Met receptor to components of the lysosomal sorting machinery and to suppress its inherent transforming activity.


Asunto(s)
Transformación Celular Neoplásica , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-met/fisiología , Ubiquitina/metabolismo , Animales , Línea Celular , Perros , Complejos de Clasificación Endosomal Requeridos para el Transporte , Endosomas/fisiología , Activación Enzimática , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Mutación , Fosforilación , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal , Proteínas ras/fisiología
16.
Methods Mol Biol ; 1765: 27-42, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29589299

RESUMEN

Colorectal cancer (CRC) is a progressive disorder associated with an accumulation of multiple heterogeneous genetic alterations in intestinal epithelial cells (IEC). However, when these cells undergo neoplastic transformation and become cancerous and metastatic, they invariably acquire hallmarks conferring them the ability to hyperproliferate, escape growth-inhibitory and death-inducing cues, and promote angiogenesis as well as epithelial-to-mesenchymal transformation (EMT), fostering their invasive dissemination from primary tumor into distant tissues. Compelling clinical and experimental evidence suggest that aberrant engagement of cell surface growth factor receptor tyrosine kinase (RTK) signaling, like that of the hepatocyte growth factor (HGF)/MET receptor, underlies CRC metastatic progression by promoting these cancer hallmarks. To date, though, the use of RTK-targeting agents has been viewed as a promising approach for the treatment of metastatic CRC, clinical success has been modest.Our vision is that the prospect of designing RTK-based, improved and innovative CRC therapies and prognostic markers likely rests on a comprehensive understanding of the biological processes and underlying regulatory molecular mechanisms by which deregulation of RTK signaling governs IEC's neoplastic transformation and their transition from noninvasive to metastatic and malignant cells. Herein, we describe our scheme for defining the full scope of oncogenic MET-driven cancer biological processes, in cellulo and in vivo, as well as the individual contribution of MET-binding effectors in a nontransformed IEC model, the IEC-6 cell line.


Asunto(s)
Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/patología , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Ratas , Transducción de Señal
17.
Environ Sci Pollut Res Int ; 25(8): 7647-7661, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29285699

RESUMEN

In this study, avocado seed was successfully used as raw material for producing activated carbons by conventional pyrolysis. In order to determine the best condition to produce the activated carbons, a 22 full-factorial design of experiment (DOE) with three central points was employed by varying the temperature and time of pyrolysis. The two evaluated factors (temperature and time of pyrolysis) strongly influenced the SBET, pore volumes, hydrophobicity-hydrophilicity ratio (HI) and functional groups values; both factors had a negative effect over SBET, pore volumes and functional groups which means that increasing the values of factors leads to decrease of these responses; on the other hand, with regards to HI, both factors caused a positive effect which means that increasing their values, the HI has an enhancement over its values. The produced activated carbon exhibited high specific surface areas in the range of 1122-1584 m2 g-1. Surface characterisation revealed that avocado seed activated carbons (ASACs) have hydrophilic surfaces and have predominantly acidic groups on their surfaces. The prepared ASACs were employed in the adsorption of 25 emerging organic compounds such as 10 pharmaceuticals and 15 phenolic compounds which presented high uptake values for all emerging pollutants. It was observed that the activated carbon prepared at higher temperature of pyrolysis (700 °C), which generated less total functional groups and presented higher HI, was the activated carbon with higher sorption capacity for uptaking emerging organic contaminants. Based on results of this work, it is possible to conclude that avocado seed can be employed as a raw material to produce high surface area and very efficient activated carbons in relation to treatment of polluted waters with emerging organic pollutants.


Asunto(s)
Carbono/química , Carbón Orgánico/química , Persea , Fenoles/química , Semillas/metabolismo , Adsorción , Compuestos Orgánicos , Semillas/química , Temperatura
18.
Int J Radiat Biol ; 93(5): 507-516, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27935337

RESUMEN

PURPOSE: Radiotherapy increases the level of inflammatory cytokines, some of which are known to promote metastasis. In a mouse model of triple negative breast cancer (TNBC), we determined whether irradiation of the mammary tumor increases the level of key cytokines and favors the development of lung metastases. MATERIALS AND METHODS: D2A1 TNBC cells were implanted in the mammary glands of a Balb/c mouse and then 7 days old tumors were irradiated (4 × 6 Gy). The cytokines IL-1ß, IL-4, IL-6, IL-10, IL-17 and MIP-2 were quantified in plasma before, midway and after irradiation. The effect of tumor irradiation on the invasion of cancer cells, the number of circulating tumor cells (CTC) and lung metastases were also measured. RESULTS: TNBC tumor irradiation significantly increased the plasma level of IL-1ß, which was associated with a greater number of CTC (3.5-fold) and lung metastases (2.3-fold), compared to sham-irradiated animals. Enhancement of D2A1 cell invasion in mammary gland was associated with an increase of the matrix metalloproteinases-2 and -9 activity (MMP-2, -9). The ability of IL-1ß to stimulate the invasiveness of irradiated D2A1 cells was confirmed by in vitro invasion chamber assays. CONCLUSION: Irradiation targeting a D2A1 tumor and its microenvironment increased the level of the inflammatory cytokine IL-1ß and was associated with the promotion of cancer cell invasion and lung metastasis development.


Asunto(s)
Citocinas/inmunología , Interleucina-1beta/biosíntesis , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/radioterapia , Animales , Línea Celular Tumoral , Femenino , Mediadores de Inflamación/inmunología , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/radioterapia , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Metástasis de la Neoplasia , Dosificación Radioterapéutica
19.
World J Gastroenterol ; 23(36): 6639-6649, 2017 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-29085209

RESUMEN

AIM: To investigate the role of suppressor of cytokine signaling 1 (SOCS1) in regulating MET-mediated invasive potential of hepatocellular carcinoma (HCC) cells. METHODS: Stable derivatives of mouse (Hepa1-6) and human (hep3B, HepG2) HCC cell lines expressing SOCS1 or control vector were evaluated for their ability to migrate towards hepatocyte growth factor (HGF) in the transwell migration assay, invade extracellular matrix in response to HGF stimulation in a 3-D invasion assay by confocal microscopy, and to undergo anchorage-independent proliferation in semisolid agar. Following intravenous and intrasplenic inoculation into NOD.scid.gamma mice, the ability of Hepa cells to form othotopic tumors was evaluated. Following HGF stimulation of Hepa and Hep3B cells, expression of proteins implicated in epithelial-to-mesenchymal transition was evaluated by western blot and qRT-PCR. RESULTS: SOCS1 expression in mouse and human HCC cells inhibited HGF-induced migration through matrigel. In the 3-D invasion assay, HGF stimulation induced invasion of HCC cells across type-I collagen matrix, and SOCS1 expression significantly reduced the depth of invasion. SOCS1 expression also reduced the number and size of colonies formed by anchorage-independent growth in semisolid agar. Following intravenous inoculation, control Hepa cell formed large tumor nodules that obliterated the liver whereas the SOCS1-expressing Hepa cells formed significantly smaller nodules. Tumors formed by SOCS1-expressing cells showed reduced phosphorylation of STAT3 and ERK that was accompanied by reduced levels of MET protein expression. HGF stimulated Hepa cells expressing SOCS1 showed increased expression of E-cadherin and decreased expression of EGR1, SNAI1 and ZEB1. Comparable results were obtained with Hep3B cells. SOCS1 expressing HCC cells also showed reduced levels of EGR1 and SNAI1 transcripts. CONCLUSION: Our findings indicate that loss of SOCS1-dependent control over epithelial-to-mesenchymal transition may contribute to MET-mediated migration, invasion and metastatic growth of HCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Animales , Movimiento Celular , Transición Epitelial-Mesenquimal , Células Hep G2 , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Invasividad Neoplásica/patología , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cell Death Discov ; 3: 16100, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28179995

RESUMEN

The unfolding of apoptosis involves the cleavage of hundreds of proteins by the caspase family of cysteinyl peptidases. Among those substrates are proteins involved in intracellular vesicle trafficking with a net outcome of shutting down the crucial processes governing protein transport to organelles and to the plasma membrane. However, because of the intertwining of receptor trafficking and signaling, cleavage of specific proteins may lead to unintended consequences. Here we show that in apoptosis, sorting nexin 1 and 2 (SNX1 and SNX2), two proteins involved in endosomal sorting, are cleaved by initiator caspases and also by executioner caspase-6 in the case of SNX2. Moreover, SNX1 is cleaved at multiple sites, including following glutamate residues. Cleavage of SNX2 results in a loss of association with the endosome-to-trans-Golgi network transport protein Vps35 and in a delocalization from endosomes of its associated partner Vps26. We also demonstrate that SNX2 depletion causes an increase in hepatocyte growth factor receptor tyrosine phosphorylation and Erk1/2 signaling in cells. Finally, we show that SNX2 mRNA and protein levels are decreased in colorectal carcinoma and that lower SNX2 gene expression correlates with an increase in cancer patient mortality. Our study reveals the importance to characterize the cleavage fragments produced by caspases of specific death substrates given their potential implication in the mechanism of regulation of physiological (signaling/trafficking) pathways or in the dysfunction leading to pathogenesis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA