Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Tipo de estudio
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 2517, 2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-32054925

RESUMEN

Triple-negative breast cancer (TNBC) is among the most aggressive forms of breast cancer with limited therapeutic options. TAK1 is implicated in aggressive behavior of TNBC, while means are not fully understood. Here, we report that pharmacological blockade of TAK1 signaling hampered ribosome biogenesis (RBG) by reducing expression of RBG regulators such as RRS1, while not changing expression of ribosomal core proteins. Notably, TAK1 blockade upregulated expression of p53 target genes in cell lines carrying wild type (wt) TP53 but not in p53-mutant cells, suggesting involvement of ribosomal stress in the response. Accordingly, p53 activation by blockade of TAK1 was prevented by depletion of ribosomal protein RPL11. Further, siRNA-mediated depletion of TAK1 or RELA resulted in RPL11-dependent activation of p53 signaling. Knockdown of RRS1 was sufficient to disrupt nucleolar structures and resulted in activation of p53. TCGA data showed that TNBCs express high levels of RBG regulators, and elevated RRS1 levels correlate with unfavorable prognosis. Cytotoxicity data showed that TNBC cell lines are more sensitive to TAK1 inhibitor compared to luminal and HER2+ cell lines. These results show that TAK1 regulates p53 activation by controlling RBG factors, and the TAK1-ribosome axis is a potential therapeutic target in TNBC.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Ribosomas/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Femenino , Humanos
2.
Oncotarget ; 8(37): 61969-61981, 2017 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-28977919

RESUMEN

The breast carcinoma microenvironment strikingly influences cancer progression and response to therapy. Various cell types in the carcinoma microenvironment show significant activity of p38 mitogen-activated protein kinase (MAPK), although the role of p38MAPK in breast cancer progression is still poorly understood. The present study examined the contribution of tumor p38MAPK to breast carcinoma microenvironment and metastatic capacity. Inactivation of p38MAPK signaling in metastatic breast carcinoma cells was achieved by forced expression of the kinase-inactive mutant of p38/MAPK14 (a dominant-negative p38, dn-p38). Disruption of tumor p38MAPK signaling reduced growth and metastases of breast carcinoma xenografts. Importantly, dn-p38 markedly decreased tumor blood-vessel density and lumen sizes. Mechanistic studies revealed that p38 controls expression of pro-angiogenic extracellular factors such as matrix protein Fibronectin and cytokines VEGFA, IL8, and HBEGF. Tumor-associated fibroblasts enhanced tumor growth and vasculature as well as increased expression of the pro-angiogenic factors. These effects were blunted by dn-p38. Metadata analysis showed elevated expression of p38 target genes in breast cancers and this was an unfavorable marker of disease recurrence and poor-outcome. Thus, our study demonstrates that tumor p38MAPK signaling promotes breast carcinoma growth, invasive and metastatic capacities. Importantly, p38 enhances carcinoma vascularization by facilitating expression and deposition of pro-angiogenic factors. These results argue that p38MAPK is a valuable target for anticancer therapy affecting tumor vasculature. Anti-p38 drugs may provide new therapeutic strategies against breast cancer, including metastatic disease.

3.
Oncotarget ; 8(22): 35592-35608, 2017 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-28423685

RESUMEN

Advance-stage breast carcinomas include significant amounts of fibroblasts and infiltrating immune cells which have been implicated in tumor growth, recurrence, and response to therapy. The present study investigated the contribution of fibroblasts to tumor growth using direct tumor-fibroblast co-cultures and tumor xenograft models. Our findings revealed that fibroblasts enhance breast carcinoma growth by promoting the tumor vasculature via the MMP9-dependent mechanism. In tumor-fibroblast co-cultures, fibroblasts increased expression of TGF-ß, TNF, and IL-1ß cytokines in tumor cells. These cytokines cooperatively induced expression of matrix metalloproteinase MMP9 in tumor cells. Knockdown of MMP9 by shRNA significantly reduced tumor vascularization induced by fibroblasts. Mechanistically, our findings argue that expression of MMP9 in tumor cellsis regulated by crosstalk of TGF-ß with TNF and/or IL-1ß cytokines. The mechanism of this cooperative response did not involve cross-activation of the canonical signaling pathways as TGF-ß did not activate RELA/p65 signaling, while TNF did not affect SMAD signaling. Instead, TGF-ß and TNF cytokines co-stimulated MAP kinases and expression of JUN and JUNB, AP1 transcription factor subunits, which together with RELA/p65 were essential for the regulation of MMP9. Depletion of JUN and JUNB or RELA in tumor cells blocked the cooperative induction of MMP9 by the cytokines. Thus, our studies uncovered a previously unappreciated role of tumor-fibroblast interactions in the stimulation of tumor angiogenesis, and an essential role of the MAPK-AP1 axis in the cooperative up-regulation of the angiogenic driver MMP9 by cytokine crosstalk.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Citocinas/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neovascularización Patológica/metabolismo , Animales , Apoptosis , Comunicación Celular , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Humanos , Mediadores de Inflamación , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Ratas , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cell Cycle ; 12(21): 3377-89, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24036928

RESUMEN

Cell adhesion to the extracellular matrix is an essential element of various biological processes. TGF-ß cytokines regulate the matrix components and cell-matrix adhesions. The present study investigates the molecular organization of TGF-ß-induced matrix adhesions. The study demonstrates that in various mouse and human epithelial cells TGF-ß induces cellular structures containing 2 matrix adhesions bridged by a stretch of actin fibers. These structures are similar to ventral stress fibers (VSFs). Suppression of integrin-ß5 by RNA interference reduces VSFs in majority of cells (> 75%), while overexpression of integrin-ß5 fragments revealed a critical role of a distinct sequence in the cytoplasmic domain of integrin-ß5 in the VSF structures. In addition, the integrity of actin fibers and Src kinase activity contribute to integrin-ß5-mediated signaling and VSF formation. TGF-ß-Smad signaling upregulates actin-regulatory proteins, such as caldesmon, zyxin, and zyxin-binding protein Csrp1 in mouse and human epithelial cells. Suppression of zyxin markedly inhibits formation of VSFs in response to TGF-ß and integrin-ß5. Zyxin is localized at actin fibers and matrix adhesions of VSFs and might bridge integrin-ß5-mediated adhesions to actin fibers. These findings provide a platform for defining the molecular mechanism regulating the organization and activities of VSFs in response to TGF-ß.


Asunto(s)
Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Cadenas beta de Integrinas/metabolismo , Fibras de Estrés/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Zixina/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/ultraestructura , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/ultraestructura , Femenino , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/ultraestructura , Regulación de la Expresión Génica , Humanos , Cadenas beta de Integrinas/genética , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fibras de Estrés/efectos de los fármacos , Fibras de Estrés/ultraestructura , Zixina/genética
5.
Chem Commun (Camb) ; 49(98): 11482-4, 2013 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23900594

RESUMEN

A novel Lewis acid-catalyzed domino (3+2)-cyclodimerization of 2-arylcyclopropane-1,1-diesters and related stepwise cross-reaction of two different cyclopropanes were developed. These processes provide efficient and highly stereoselective access to polyoxygenated indanes and cyclopentannulated heteroarene derivatives, which display significant cytotoxicity against several lines of cancer cells (IC50 of 10(-6)-10(-5) M) while being non-toxic for normal cells.


Asunto(s)
Hidrocarburos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dimerización , Humanos , Hidrocarburos/farmacología , Ácidos de Lewis/química , Estereoisomerismo
6.
Mol Cancer Res ; 9(8): 1042-53, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21700681

RESUMEN

Advanced-stage breast cancers frequently metastasize to the bones and cause bone destruction, but the underlying mechanism is not fully understood. This study presents evidence that TGF-ß-activated protein kinase 1 (TAK1) signaling in tumor cells promotes bone destruction by metastatic breast carcinoma cells, controlling expression of prometastatic factors including matrix metalloproteinase (MMP) 9 and COX2. Suppression of TAK1 signaling by dominant-negative TAK1 (dn-TAK1) in breast carcinoma MDA-MB-231 cells impairs bone colonization by carcinoma cells and bone osteolysis in the intracardiac injection model. Mechanistic studies showed that inhibition of TAK1 by dn-TAK1 or siRNA blocked expression of factors implicated in bone metastasis, such as MMP-9, COX2/PTGS2, parathyroid hormone-related protein (PTHrP) and interleukin 8 (IL-8), but did not affect activation of p38MAPK by TGF-ß. TAK1 signaling is mediated by TAK1-binding partners TAB1, TAB2, and TAB3. Carcinoma cells express elevated mRNA levels of TAB2 and TAB3, whereas the TAB1 expression is noticeably low. Accordingly, depletion of TAB2 by siRNA reduced expression of MMP-9 and COX2. Together, these studies show that the TAK1-TAB2-TAB3 signaling axis is critical for carcinoma-induced bone lesions, mediating expression of proinvasive and osteolytic factors. These findings identify the TAK1-TAB2 axis as a potential therapeutic target in bone metastasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma/metabolismo , Carcinoma/patología , Quinasas Quinasa Quinasa PAM/metabolismo , Animales , Línea Celular Tumoral , Ciclooxigenasa 2/metabolismo , Femenino , Humanos , Interleucina-8/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones SCID , Neoplasias de la Próstata/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA