Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
BMC Cancer ; 18(1): 99, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29370782

RESUMEN

BACKGROUND: In menopausal women, one of the critical risk factors for breast cancer is obesity/adiposity. It is evident from various studies that leptin, a 16 kDa protein hormone overproduced in obese people, plays the critical role in neovascularization and tumorigenesis in breast and other organs. However, the mechanisms by which obesity influences the breast carcinogenesis remained unclear. In this study, by analyzing different estrogen receptor-α (ER-α)-positive and ER-α-negative BC cell lines, we defined the role of CCN5 in the leptin-mediated regulation of growth and invasive capacity. METHODS: We analyzed the effect of leptin on cell viability of ER-α-positive MCF-7 and ZR-75-1 cell lines and ER-α-negative MDA-MB-231 cell line. Additionally, we also determined the effect of leptin on the epithelial-mesenchymal transition (EMT) bio-markers, in vitro invasion and sphere-formation of MCF-7 and ZR-75-1 cell lines. To understand the mechanism, we determined the impact of leptin on CCN5 expression and the functional role of CCN5 in these cells by the treatment of human recombinant CCN5 protein(hrCCN5). Moreover, we also determined the role of JAK-STAT and AKT in the regulation of leptin-induced suppression of CCN5 in BC cells. RESULTS: Present studies demonstrate that leptin can induce cell viability, EMT, sphere-forming ability and migration of MCF-7 and ZR-75-1 cell lines. Furthermore, these studies found that leptin suppresses the expression of CCN5 at the transcriptional level. Although the CCN5 suppression has no impact on the constitutive proliferation of MCF-7 and ZR-75-1 cells, it is critical for leptin-induced viability and necessary for EMT, induction of in vitro migration and sphere formation, as the hrCCN5 treatment significantly inhibits the leptin-induced viability, EMT, migration and sphere-forming ability of these cells. Mechanistically, CCN5-suppression by leptin is mediated via activating JAK/AKT/STAT-signaling pathways. CONCLUSIONS: These studies suggest that CCN5 serves as a gatekeeper for leptin-dependent growth and progression of luminal-type (ER-positive) BC cells. Leptin may thus need to destroy the CCN5-barrier to promote BC growth and progression via activating JAK/AKT/STAT signaling. Therefore, these observations suggest a therapeutic potency of CCN5 by restoration or treatment in obese-related luminal-type BC growth and progression.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas CCN de Señalización Intercelular/genética , Receptor alfa de Estrógeno/genética , Leptina/genética , Obesidad/genética , Proteínas Represoras/genética , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas CCN de Señalización Intercelular/metabolismo , Carcinogénesis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Humanos , Quinasas Janus/genética , Leptina/metabolismo , Células MCF-7 , Menopausia/genética , Invasividad Neoplásica , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Represoras/metabolismo , Factores de Transcripción STAT
2.
Mol Cancer ; 10: 8, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21232118

RESUMEN

BACKGROUND: Despite recent advances in outlining the mechanisms involved in pancreatic carcinogenesis, precise molecular pathways and cellular lineage specification remains incompletely understood. RESULTS: We show here that Cyr61/CCN1 play a critical role in pancreatic carcinogenesis through the induction of EMT and stemness. Cyr61 mRNA and protein were detected in the early precursor lesions and their expression intensified with disease progression. Cyr61/CCN1 expression was also detected in different pancreatic cancer cell lines. The aggressive cell lines, in which the expressions of mesenchymal/stem cell molecular markers are predominant; exhibit more Cyr61/CCN1 expression. Cyr61 expression is exorbitantly higher in cancer stem/tumor initiating Panc-1-side-population (SP) cells. Upon Cyr61/CCN1 silencing, the aggressive behaviors are reduced by obliterating interlinking pathobiological events such as reversing the EMT, blocking the expression of stem-cell-like traits and inhibiting migration. In contrast, addition of Cyr61 protein in culture medium augments EMT and stemness features in relatively less aggressive BxPC3 pancreatic cancer cells. Using a xenograft model we demonstrated that cyr61/CCN1 silencing in Panc-1-SP cells reverses the stemness features and tumor initiating potency of these cells. Moreover, our results imply a miRNA-based mechanism for the regulation of aggressive behaviors of pancreatic cancer cells by Cyr61/CCN1. CONCLUSIONS: In conclusion, the discovery of the involvement of Cyr61/CCN1 in pancreatic carcinogenesis may represent an important marker for PDAC and suggests Cyr61/CCN1 can be a potential cancer therapeutic target.


Asunto(s)
Adenocarcinoma/patología , Proteína 61 Rica en Cisteína/biosíntesis , Transición Epitelial-Mesenquimal , Neoplasias Pancreáticas/patología , Adenocarcinoma/metabolismo , Animales , Biomarcadores de Tumor , Movimiento Celular , Proteína 61 Rica en Cisteína/genética , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , MicroARNs/biosíntesis , MicroARNs/genética , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/metabolismo , Comunicación Paracrina , Interferencia de ARN , Células de Población Lateral , Regulación hacia Arriba
3.
Mol Cancer ; 9: 209, 2010 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-20687910

RESUMEN

BACKGROUND: New blood vessel formation, or angiogenic switch, is an essential event in the development of solid tumors and their metastatic growth. Tumor blood vessel formation and remodeling is a complex and multi-step processes. The differentiation and recruitment of mural cells including vascular smooth muscle cells and pericytes are essential steps in tumor angiogenesis. However, the role of tumor cells in differentiation and recruitment of mural cells has not yet been fully elucidated. This study focuses on the role of human tumor cells in governing the differentiation of mouse mesenchymal stem cells (MSCs) to pericytes and their recruitment in the tumor angiogenesis process. RESULTS: We show that C3H/10T1/2 mouse embryonic mesenchymal stem cells, under the influence of different tumor cell-derived conditioned media, differentiate into mature pericytes. These differentiated pericytes, in turn, are recruited to bind with capillary-like networks formed by endothelial cells on the matrigel under in vitro conditions and recruited to bind with blood vessels on gel-foam under in vivo conditions. The degree of recruitment of pericytes into in vitro neo-angiogenesis is tumor cell phenotype specific. Interestingly, invasive cells recruit less pericytes as compared to non-invasive cells. We identified tumor cell-secreted platelet-derived growth factor-B (PDGF-B) as a crucial factor controlling the differentiation and recruitment processes through an interaction with neuropilin-1 (NRP-1) in mesenchymal stem cells. CONCLUSION: These new insights into the roles of tumor cell-secreted PDGF-B-NRP-1 signaling in MSCs-fate determination may help to develop new antiangiogenic strategies to prevent the tumor growth and metastasis and result in more effective cancer therapies.


Asunto(s)
Células Madre Mesenquimatosas/citología , Neuropilina-1/fisiología , Pericitos/citología , Proteínas Proto-Oncogénicas c-sis/fisiología , Animales , Diferenciación Celular , Línea Celular , Medios de Cultivo Condicionados , Ratones , Ratones Endogámicos C3H
4.
Mol Cancer Ther ; 8(2): 315-23, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19208826

RESUMEN

Crocetin, a carotenoid compound derived from saffron, has long been used as a traditional ancient medicine against different human diseases including cancer. The aim of the series of experiments was to systematically determine whether crocetin significantly affects pancreatic cancer growth both in vitro and/or in vivo. For the in vitro studies, first, MIA-PaCa-2 cells were treated with crocetin and in these sets of experiments, a proliferation assay using H(3)-thymidine incorporation and flow cytometric analysis suggested that crocetin inhibited proliferation. Next, cell cycle proteins were investigated. Cdc-2, Cdc-25C, Cyclin-B1, and epidermal growth factor receptor were altered significantly by crocetin. To further confirm the findings of inhibition of proliferation, H(3)-thymidine incorporation in BxPC-3, Capan-1, and ASPC-1 pancreatic cancer cells was also significantly inhibited by crocetin treatment. For the in vivo studies, MIA-PaCa-2 as highly aggressive cells than other pancreatic cancer cells used in this study were injected into the right hind leg of the athymic nude mice and crocetin was given orally after the development of a palpable tumor. The in vivo results showed significant regression in tumor growth with inhibition of proliferation as determined by proliferating cell nuclear antigen and epidermal growth factor receptor expression in the crocetin-treated animals compared with the controls. Both the in vitro pancreatic cancer cells and in vivo athymic nude mice tumor, apoptosis was significantly stimulated as indicated by Bax/Bcl-2 ratio. This study indicates that crocetin has a significant antitumorigenic effect in both in vitro and in vivo on pancreatic cancer.


Asunto(s)
Carotenoides/farmacología , Neoplasias Pancreáticas/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carotenoides/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Receptores ErbB/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Vitamina A/análogos & derivados
5.
Cancer Res ; 67(4): 1520-6, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17308090

RESUMEN

Previously, we have shown that the expression of Wnt-1-induced signaling protein-2 (WISP-2), also known as CCN5, can be regulated by multiple stimulants in estrogen receptor (ER)-positive breast tumor cells to exert their mitogenic action in these cells. Here, we show that insulin-like growth factor-1 (IGF-1), a strong mitogen, enhanced the expression of the WISP-2/CCN5 gene parallel with the induction of proliferation of ER-positive breast tumor cells. An additive effect was also seen in combination with estrogen. Perturbation of IGF-1-induced WISP-2/CCN5 expression by WISP-2-specific RNA interference impaired the mitogenic action of IGF-1 on ER-positive breast tumor cells. Furthermore, the studies have shown that the multiple molecular cross-talks and side-talks among IGF-1R, ER-alpha, and phosphatidylinositol 3-kinase (PI3K)/Akt signaling molecules are required to induce WISP-2/CCN5 mRNA by IGF-1 in ER-positive, noninvasive breast tumor cells. Because a pure anti-ER ICI 182,780 is not only able to suppress the up-regulation of WISP-2/CCN5 mRNA expression by IGF-1, it also suppresses the PI3K/Akt activity induced by IGF-1 in MCF-7 cells; we anticipate that the membrane ER receptor may participate in this event. Collectively, these studies propose for the first time that WISP-2/CCN5 is an integral signaling molecule in mitogenic action of IGF-1 axis in ER-positive human breast tumor cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Proteínas de Neoplasias/biosíntesis , Receptores de Estrógenos/metabolismo , Factores de Transcripción/biosíntesis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas CCN de Señalización Intercelular , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Sinergismo Farmacológico , Estradiol/farmacología , Humanos , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de Neoplasias/genética , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor IGF Tipo 1/metabolismo , Receptores de Estrógenos/biosíntesis , Proteínas Represoras , Transducción de Señal , Factores de Transcripción/genética , Regulación hacia Arriba/efectos de los fármacos
6.
Mol Cancer Ther ; 18(4): 788-800, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30787177

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) develops extrinsic- and intrinsic-resistant phenotypes to prevent chemotherapies from entering into the cells by promoting desmoplastic reactions (DR) and metabolic malfunctions of the drugs. It is well established that these responses are also associated with pancreatic cancer cells' gemcitabine resistance. However, the mechanism by which these resistant pathways function in the pancreatic cancer cells remains poorly understood. In these studies, we show that CYR61/CCN1 signaling plays a vital role in making pancreatic cancer cells resistant to gemcitabine in vitro and also in a tumor xenograft model. We proved that the catastrophic effect of gemcitabine could significantly be increased in gemcitabine-resistant PDAC cells when CYR61/CCN1 is depleted, while this effect can be suppressed in gemcitabine-sensitive neoplastic cells by treating them with CYR61/CCN1 recombinant protein. Ironically, nontransformed pancreatic cells, which are sensitive to gemcitabine, cannot be resistant to gemcitabine by CYR61/CCN1 protein treatment, showing a unique feature of CYR61/CCN signaling that only influences PDAC cells to become resistant. Furthermore, we demonstrated that CYR61/CCN1 suppresses the expression of the gemcitabine-activating enzyme deoxycytidine kinase (dCK) while it induces the expression of a DR-promoting factor CTGF (connective tissue growth factor) in pancreatic cancer cells in vitro and in vivo Thus, the previously described mechanisms (dCK and CTGF pathways) for gemcitabine resistance may be two novel targets for CYR61/CCN1 to protect pancreatic cancer cells from gemcitabine. Collectively, these studies reveal a novel paradigm in which CYR61/CCN1regulates both extrinsic and intrinsic gemcitabine resistance in PDAC cells by employing unique signaling pathways.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Proteína 61 Rica en Cisteína/metabolismo , Desoxicitidina Quinasa/metabolismo , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/genética , Neoplasias Pancreáticas/metabolismo , Fenotipo , Animales , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Proteína 61 Rica en Cisteína/genética , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Femenino , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/patología , Transducción de Señal/efectos de los fármacos , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
7.
Sci Rep ; 7(1): 1220, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28450698

RESUMEN

Breast cancer progression and relapse is conceivably due to tumor initiating cells (TICs)/cancer stem cells. EMT (epithelial-mesenchymal-transition)-signaling regulates TICs' turnover. However, the mechanisms associated with this episode are unclear. We show that, in triple-negative-breast cancer (TNBC) cells enriched with TICs, CCN5 significantly blocks cellular growth via apoptosis, reversing EMT-signaling and impairing mammosphere formation, thereby blocking the tumor-forming ability and invasive capacity of these cells. To corroborate these findings, we isolated tumor-initiating side populations (SP) and non-side population (NSP or main population) from MCF-7 cell line, and evaluated the impact of CCN5 on these subpopulations. CCN5 was overexpressed in the NSP but downregulated in the SP. Characteristically, NSP cells are ER-α positive and epithelial type with little tumorigenic potency, while SP cells are very similar to triple-negative ones that do not express ER-α- and Her-2 and are highly tumorigenic in xenograft models. The overexpression of CCN5 in SP results in EMT reversion, ER-α upregulation and delays in tumor growth in xenograft models. We reasoned that CCN5 distinguishes SP and NSP and could reprogram SP to NSP transition, thereby delaying tumor growth in the xenograft model. Collectively, we reveal how CCN5-signaling underlies the driving force to prevent TNBC growth and progression.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas CCN de Señalización Intercelular/metabolismo , Proliferación Celular , Transición Epitelial-Mesenquimal , Proteínas Represoras/metabolismo , Humanos , Células MCF-7 , Modelos Biológicos
8.
Mol Cancer Res ; 3(3): 151-62, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15798095

RESUMEN

Epidermal growth factor (EGF) is a mitogen for estrogen receptor (ER)-positive breast tumor cells, and it has been proven that EGF occasionally mimicked estrogen action and cross-talks with ER-alpha to exert its activity. Therefore, the present study was undertaken to explore whether EGF is able to modulate the expression of Wnt-1-induced signaling protein-2/connective tissue growth factor/cysteine-rich 61/nephroblastoma overexpressed 5 (WISP-2/CCN5), an estrogen-responsive gene, in normal and transformed cell lines of the human breast and, if so, whether this induction is critical for EGF mitogenesis and what downstream signaling pathways are associated with this event. Here, we show that EGF-induced WISP-2 expression in ER- and EGF receptor-positive noninvasive MCF-7 breast tumor cells was dose and time dependent and that expression was modulated at transcription level. A synergism was seen in combination with estrogen. Moreover, small interfering RNA-mediated inhibition of WISP-2/CCN5 activity in MCF-7 cells resulted in abrogation of proliferation by EGF. The multiple molecular cross-talks, including the interactions between phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase signaling pathways and two diverse receptors (i.e., ER-alpha and EGFR), were essential in the event of EGF-induced WISP-2/CCN5 up-regulation in MCF-7 cells. Moreover, EGF action on WISP-2/CCN5 is restricted to ER- and EGFR-positive noninvasive breast tumor cells, and this effect of EGF cannot be instigated in ER-alpha-negative and EGFR-positive normal or invasive breast tumor cells by introducing ER-alpha. Finally, regulation of phosphorylation of ER-alpha and EGFR may play critical roles in EGF-induced transcriptional activation of WISP-2 gene in breast tumor cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Factor de Crecimiento Epidérmico/fisiología , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Proteínas de Neoplasias/biosíntesis , Factores de Transcripción/biosíntesis , Northern Blotting , Western Blotting , Butadienos/farmacología , Proteínas CCN de Señalización Intercelular , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular , Clonación Molecular , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Sistema de Señalización de MAP Quinasas , Microscopía Confocal , Microscopía Fluorescente , Invasividad Neoplásica , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , ARN/química , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Fracciones Subcelulares , Factores de Tiempo , Transcripción Genética , Activación Transcripcional , Transfección , Regulación hacia Arriba
9.
Front Biosci ; 10: 1763-7, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15769665

RESUMEN

Expression of epidermal growth factor receptors (EGFR) is exaggerated in pancreatic adenocarcinoma and activation of EGFR appears to have an important role in the growth and differentiation of this and in other tumors. Therefore, blockade or inactivation of EGFR by monoclonal antibodies or by tyrosine kinase inhibitors has significant potential as an effective anti-cancer therapy. One of the very recent significant developments in the field of molecular biology involves the use of antisense of EGFR or EGFR gene silencing in pancreatic cancer cells as a potential targeted therapy for patients with pancreatic adenocarcinoma.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Receptores ErbB/antagonistas & inhibidores , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico
10.
J Environ Biol ; 25(3): 359-63, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15847349

RESUMEN

The inland freshwater resources are being increasingly subjected to heavy stress as a result of indiscriminate dumping of industrial wastes, domestic sewage and agricultural run-off causing deterioration of the water quality and adverse impact on aquatic biota. Pesticides drained to the aquatic environment are primarily of agricultural origin. Phosphamidon (widely used organophosphate pesticide in paddy field) significantly reduced dissolved oxygen (DO) at 1.8 mg/l exposure and reduced alkalinity at 0.9 and 1.8 mg/l. Hardness also reduced gradually but not significantly. Free carbondioxide was increased significantly at 1.8 mg/l of the insecticide compared to control. The insecticide had no influence on pH and temperature. There was maximum reduction of phytoplankton and zooplankton population at 1.8 mg/l of phosphamidon. Though gradual reduction of plankton community was also noticed at different lower concentrations of pesticides but in case of phytoplankton an abrupt reduction (about 50% of the control) was observed. The normal behaviour and feeding rate of air breathing teleost, Channa punctatus was also hampered. Therefore, phosphamidon even at low concentrations may create disorders in the aquatic ecosystem.


Asunto(s)
Agua Dulce/química , Insecticidas/toxicidad , Compuestos Organofosforados/toxicidad , Fosfamidón/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Ecosistema , Monitoreo del Ambiente , Concentración de Iones de Hidrógeno , Residuos Industriales , Oxígeno/metabolismo , Fitoplancton/efectos de los fármacos , Fitoplancton/fisiología , Aguas del Alcantarillado/química , Temperatura , Zooplancton/efectos de los fármacos , Zooplancton/fisiología
11.
Sci Rep ; 4: 4995, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24833309

RESUMEN

The complex signaling networks between cancer cells and adjacent endothelial cells make it challenging to unravel how cancer cells send extracellular messages to promote aberrant vascularization or tumor angiogenesis. Here, in vitro and in vivo models show that pancreatic cancer cell generated unique microenvironments can underlie endothelial cell migration and tumor angiogenesis. Mechanistically, we find that pancreatic cancer cell secreted CCN1/Cyr61 matricellular protein rewires the microenvironment to promote endothelial cell migration and tumor angiogenesis. This event can be overcome by Sonic Hedgehog (SHh) antibody treatment. Collectively, these studies identify a novel CCN1 signaling program in pancreatic cancer cells which activates SHh through autocrine-paracrine circuits to promote endothelial cell migration and tumor angiogenesis and suggests that CCN1 signaling of pancreatic cancer cells is vital for the regulation of tumor angiogenesis. Thus CCN1 signaling could be an ideal target for tumor vascular disruption in pancreatic cancer.


Asunto(s)
Movimiento Celular/fisiología , Proteína 61 Rica en Cisteína/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Neovascularización Patológica/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Animales , Línea Celular , Línea Celular Tumoral , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Neovascularización Patológica/metabolismo , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología
12.
J Cell Commun Signal ; 7(2): 129-40, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23292944

RESUMEN

Adipocytes play a vital role in glucose metabolism. 3T3 L1 pre adipocytes after differentiation to adipocytes serve as excellent in vitro models and are useful tools in understanding the glucose metabolism. The traditional approaches adopted in pre adipocyte differentiation are lengthy exercises involving the usage of IBMX and Dexamethasone. Any effort to shorten the time of differentiation and quality expression of functional differentiation in 3T3 L1 cells in terms of enhanced Insulin sensitivity has an advantage in the drug discovery process. Thus, there is a need to develop a new effective method of differentiating the pre adipocytes to adipocytes and to use such methods for developing efficacious therapeutic molecules. We observed that a combination of Dexamethasone and Troglitazone generated differentiated adipocytes over fewer days as compared to the combination of IBMX and Dexamethasone which constitutes the standard protocol followed in our laboratory. The experiments conducted to compare the quality of differentiation yielded by various differentiating agents indicated that the lipid droplet accumulation increased by 112 % and the GLUT4 mediated glucose uptake by 137 % in cells differentiated with Troglitazone and Dexamethasone than in cells differentiated traditionally. The comparative studies conducted for evaluating efficient measurable glucose uptake by GOPOD assay, radioactive (3)H-2-deoxy-D-glucose assay and by non-radioactive 6-NBDG (fluorescent analog of glucose) indicated that the non-radioactive method using 6-NBDG showed a higher signal to noise ratio than the conventional indirect glucose uptake method (GOPOD assay) and the radioactive (3)H-2-deoxy-D-glucose uptake method. Differentiated 3T3 L1 cells when triggered with 2.5 ng/mL of Insulin showed 3.3 fold more glucose uptake in non-radioactive method over the radioactive (3)H-2-deoxy-D-glucose uptake method. The results of this study have suggested that a combination of Dexamethasone and Troglitazone for 3T3 L1 cell differentiation helps in better quality differentiation over a short period of time with increased sensitivity to Insulin. The application of these findings for developing new methods of screening novel Insulin mimetics and for evaluating the immunological responses has been discussed.

13.
Mol Cancer Ther ; 9(3): 523-34, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20197389

RESUMEN

The purpose of this study was to evaluate whether 2-methoxyestradiol (2-ME(2)), a promising anticancer agent, modulates Barrett's esophageal adenocarcinoma (BEAC) cell growth and behavior through a cellular pathway involving beta-catenin in partnership with E-cadherin, which seems to play a critical role in the induction of antitumor responses in cancer cells. We found that 2-ME(2) markedly reduced the BEAC cell proliferation through regulating apoptotic machinery such as Bcl-2 and Bax. It may nullify the aggressive behavior of the cells by reducing the migratory behavior. Expressions of beta-catenin and E-cadherin and binding of these two proteins is activated in a 2-ME(2)-dependent fashion in Bic-1 cells. Moreover, overexpressions of these two proteins may be due to the stabilization of these proteins by 2-ME(2). We found that 2-ME(2)-induced antimigratory effects are mediated through the beta-catenin-E-cadherin signaling pathways. In view of these results, we determined whether 2-ME(2) reduces BEAC tumor growth. Administration of 2-ME2 significantly decreased the growth of BEAC cells xenografted on the flank of nude mice. The evidence presented points out that the effect of 2-ME(2) on beta-catenin-orchestrated signal transduction plausibly plays a multifaceted functional role to inhibit the proliferation and cell migration of 2-ME(2)-treated malignant cells and it could be a potential candidate in novel treatment strategies for Barrett's esophageal adenocarcinoma.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Cadherinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Esofágicas/patología , Estradiol/análogos & derivados , beta Catenina/metabolismo , 2-Metoxiestradiol , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Esófago de Barrett/tratamiento farmacológico , Esófago de Barrett/metabolismo , Cadherinas/fisiología , Transformación Celular Neoplásica , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Estradiol/farmacología , Estradiol/uso terapéutico , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/fisiología
14.
Cancer Res ; 68(12): 4580-7, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559502

RESUMEN

CCN5/WISP-2 is overexpressed in noninvasive breast cancer cells and tissue samples, whereas its expression is minimal or undetected in invasive conditions. CCN5/WISP-2 has been considered as an antiinvasive gene because CCN5/WISP-2 silencing augments the invasive phenotypes in vitro. However, the mechanism of silencing of CCN5 during the progression of the disease has been elusive. Because p53 mutations are associated with breast cancer progression and have been shown to correlate inversely with CCN5/WISP-2 expression in other cancer cell types, the objective of this study was to explore whether p53 mutants suppress CCN5 expression in breast tumor cells resulting in the progression of this disease. We found CCN5 expression is inversely correlated with the mutational activation of p53 in human breast tumor cells. The ectopic expression of p53 mutants in ER-positive noninvasive breast tumor cells silenced the CCN5/WISP-2 expression and enhanced invasive phenotypes, including the induction of morphologic changes from the epithelial-to-mesenchymal type along with the alterations of hallmark proteins of these cell types and an augmentation of the migration of these cells. The suppression of CCN5 by the p53 mutants can be nullified by estrogen signaling in these cells through the transcriptional activation of the CCN5 gene. Moreover, the invasive changes can be imitated by blocking the CCN5/WISP-2 expression through RNA interference or can be reversed by the addition of CCN5/WISP-2 recombinant protein in the culture. Thus, these studies suggest that CCN5 inactivation could be an essential molecular event for p53 mutant-induced invasive phenotypes.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Mutación/genética , Interferencia de ARN , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Western Blotting , Neoplasias de la Mama/metabolismo , Proteínas CCN de Señalización Intercelular , Carcinoma Intraductal no Infiltrante/metabolismo , Carcinoma Intraductal no Infiltrante/patología , Diferenciación Celular , Movimiento Celular , Cloranfenicol O-Acetiltransferasa/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Invasividad Neoplásica , Fenotipo , Regiones Promotoras Genéticas , Receptores de Estrógenos/metabolismo , Proteínas Represoras , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Transfección , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
15.
Biochemistry ; 47(11): 3345-51, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18284215

RESUMEN

Vascular smooth muscle cells (SMCs), one of the major cell types of the vascular wall, play a critical role in the process of angiogenesis under both physiological and pathophysiological conditions, including the cancer microenvironment. Previous studies have shown that VEGF-A 165 augments vascular SMC migration via VEGFR2 (KDR/Flk1) pathways. In this study, we found that VEGF-A 165 (recombinant protein or breast tumor cell-secreted) is also capable of inducing migration of VEGFR2-negative human aortic smooth muscle cells (hAOSMCs), and this induction is mediated through a molecular cross-talk of neuropilin-1 (NRP-1), VEGFR1 (Flt-1), and phosphoinositide 3-kinase (PI3K)/Akt signaling kinase. We found that VEGF-A 165 induces hAOSMC migration parallel with the induction of NRP-1 and VEGFR1 expressions and their associations along with the activation of PI3K/Akt. Neutralization of VEGF action by its antibody or inhibition of VEGF-induced PI3K/Akt kinase activation by wortmannin, a PI3K/Akt specific inhibitor, results in inhibition of VEGF-induced hAOSMC migration. Moreover, RNAi-mediated elimination of the NRP-1 expression or blocking of the activity of VEGFR1 by its antibody in hAOSMCs impairs the VEGF-A 165-induced migration of these cells as well as activation of PI3K/Akt kinase. Collectively, these results establish, for the first time, a mechanistic link among VEGF-A 165, NRP-1, VEGFR1, and PI3K/Akt in the regulation of migration of human vascular smooth muscle cells that eventually could be involved in the angiogenic switch.


Asunto(s)
Aorta/citología , Movimiento Celular/fisiología , Músculo Liso Vascular/citología , Neuropilina-1/metabolismo , Fragmentos de Péptidos/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Anticuerpos Bloqueadores/farmacología , Aorta/química , Aorta/fisiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Sinergismo Farmacológico , Silenciador del Gen , Humanos , Músculo Liso Vascular/química , Músculo Liso Vascular/fisiología , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/biosíntesis , Neuropilina-1/fisiología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Proteína Oncogénica v-akt/fisiología , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/inmunología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología
16.
Biochemistry ; 45(35): 10698-709, 2006 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-16939222

RESUMEN

PMA and active phorbol esters stimulate the proliferation of various tumor cells, including ER-positive human breast tumor cell lines. However, the specific signaling pathways involved in the PMA-induced mitogenic effect on breast tumor cells have not been fully elucidated. In the present study, we explored the mechanisms associated with the mitogenic influence of PMA on breast tumor cells. Following an acute exposure (i.e., within 2 to 6 h) to PMA (50 nM), a mitogenic effect was observed on WISP-2/CCN5-positive breast tumor cell lines, including MCF-7, ZR-75-1 and SKBR-3 cells, and induction of WISP-2/CCN5 mRNA expression paralleled the observed mitogenic proliferation. This effect was undetected in WISP-2/CCN5 negative MDA-MB-231 breast tumor cells or human mammary epithelial cells with or without ER-alpha transfection. The mitogenic effect of PMA was perturbed by short hairpin RNA (shRNA)-mediated inhibition of WISP-2/CCN5 signaling in MCF-7 cells. Moreover, the upregulation of WISP-2/CCN5 by PMA is not ER dependent but is instead mediated through a complex PKCalpha-MAPK/ERK and SAPK/JNK signaling pathway, which leads to growth stimulation of MCF-7 breast tumor cells. These series of experiments provide the first evidence that WISP-2/CCN5 is a novel signaling molecule that critically participates in the mitogenic action of PMA on noninvasive, WISP-2/CCN5-positive breast tumor cells through PKCalpha-dependent, multiple molecular signal transduction pathways.


Asunto(s)
Neoplasias de la Mama/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Neoplasias/metabolismo , Ésteres del Forbol/farmacología , Proteína Quinasa C-alfa/metabolismo , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo , Mama/citología , Proteínas CCN de Señalización Intercelular , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Células Epiteliales , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Sistema de Señalización de MAP Quinasas , Modelos Biológicos , Proteínas de Neoplasias/fisiología , Proteína Quinasa C-alfa/fisiología , Receptores de Estrógenos/metabolismo , Proteínas Represoras , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/fisiología , Transfección , Regulación hacia Arriba
17.
Mol Carcinog ; 45(11): 871-80, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16847823

RESUMEN

Motility of vascular smooth muscle cells (SMCs) is an essential step for both normal and pathologic angiogenesis. We report here that breast tumor cells, such as MCF-7 and MDA-MB-231, can modulate this SMC migration. We present evidence that the tumor cell-derived platelet-derived growth factor (PDGF) is the key regulator of vascular SMCs motility induced by breast cancer cells. PDGF significantly upregulates neuropilin-1 (NRP-1) mRNA expression and protein production in aortic smooth muscle cells (AOSMCs) and depletion of NRP-1 production by AOSMCs with specific short hairpin RNA (shRNA) prevents the PDGF-dependent migration of vascular SMCs. Moreover, we demonstrate that PDGF physically interacts with NRP-1. We propose that tumor-derived PDGF and NRP-1 of AOSMCs function as a relay system that promotes motility of vascular SMCs.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular/fisiología , Músculo Liso Vascular/citología , Neuropilina-1/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Secuencia de Bases , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Cultivadas , Medios de Cultivo Condicionados , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA