Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem J ; 477(17): 3131-3145, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32766732

RESUMEN

The Hedgehog-regulated transcription factors GLI1 and GLI2 play overlapping roles in development and disease; however, the mechanisms underlying their interplay remain elusive. We report for the first time that GLI1 and GLI2 physically and functionally interact in cancer cells. GLI1 and GLI2 were shown to co-immunoprecipitate in PANC1 pancreatic cancer cells and RMS13 rhabdomyosarcoma cells. Mapping analysis demonstrated that the zinc finger domains of both proteins are required for their heteromerization. RNAi knockdown of either GLI1 or GLI2 inhibited expression of many well-characterized GLI target genes (BCL2, MYCN, PTCH2, IL7 and CCND1) in PANC1 cells, whereas PTCH1 expression was only inhibited by GLI1 depletion. qPCR screening of a large set of putative canonical and non-canonical Hedgehog/GLI targets identified further genes (e.g. E2F1, BMP1, CDK2) strongly down-regulated by GLI1 and/or GLI2 depletion in PANC1 cells, and demonstrated that ANO1, AQP1 and SOCS1 are up-regulated by knockdown of either GLI1 or GLI2. Chromatin immunoprecipitation showed that GLI1 and GLI2 occupied the same regions at the BCL2, MYCN and CCND1 promoters. Furthermore, depletion of GLI1 inhibited GLI2 occupancy at these promoters, suggesting that GLI1/GLI2 interaction is required for the recruitment of GLI2 to these sites. Together, these findings indicate that GLI1 and GLI2 co-ordinately regulate the transcription of some genes, and provide mechanistic insight into the roles of GLI proteins in carcinogenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/metabolismo , Rabdomiosarcoma/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína Gli2 con Dedos de Zinc/metabolismo , Línea Celular Tumoral , Proteínas Hedgehog/genética , Humanos , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Multimerización de Proteína , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Proteína con Dedos de Zinc GLI1/genética , Proteína Gli2 con Dedos de Zinc/genética
2.
J Investig Dermatol Symp Proc ; 19(2): S87-S88, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30471761

RESUMEN

Hedgehog (HH) signaling plays an important role both during embryonic development and adult life. It is involved in the regulation of cell differentiation, cell proliferation and tissue polarity, as well as in the maintenance of stem cells, tissue repair, and regeneration (Briscoe and Therond, 2013; Jiang and Hui, 2008). Three ligands, Indian, Sonic, and Desert HH, can activate this pathway. Binding of HH ligands to their receptor, PTCH1 (Figure 1) lift its inhibition on SMO, resulting in activation and nuclear translocation of GLI transcription factors (Javelaud et al., 2012). The vertebrate GLI gene family is composed of three distinct genes GLI1, GLI2, and GLI3, encoding Krüppel-like transcription factors. GLI proteins exhibit distinct regulations, biochemical properties, and target genes. GLI3 acts as the main repressor of the pathway in the absence of HH ligands, whereas, in their presence, GLI2 is the main HH effector that drives the expression of GLI1 (Briscoe and Therond, 2013).

3.
Life Sci Alliance ; 7(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38834194

RESUMEN

Vinculin is a cytoskeletal linker strengthening cell adhesion. The Shigella IpaA invasion effector binds to vinculin to promote vinculin supra-activation associated with head-domain-mediated oligomerization. Our study investigates the impact of mutations of vinculin D1D2 subdomains' residues predicted to interact with IpaA VBS3. These mutations affected the rate of D1D2 trimer formation with distinct effects on monomer disappearance, consistent with structural modeling of a closed and open D1D2 conformer induced by IpaA. Notably, mutations targeting the closed D1D2 conformer significantly reduced Shigella invasion of host cells as opposed to mutations targeting the open D1D2 conformer and later stages of vinculin head-domain oligomerization. In contrast, all mutations affected the formation of focal adhesions (FAs), supporting the involvement of vinculin supra-activation in this process. Our findings suggest that IpaA-induced vinculin supra-activation primarily reinforces matrix adhesion in infected cells, rather than promoting bacterial invasion. Consistently, shear stress studies pointed to a key role for IpaA-induced vinculin supra-activation in accelerating and strengthening cell-matrix adhesion.


Asunto(s)
Adhesión Celular , Adhesiones Focales , Vinculina , Vinculina/metabolismo , Vinculina/genética , Humanos , Adhesiones Focales/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Mutación , Interacciones Huésped-Patógeno , Células HeLa , Unión Proteica , Shigella/metabolismo , Shigella/genética , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/genética , Disentería Bacilar/microbiología , Disentería Bacilar/metabolismo
4.
J Biol Chem ; 287(22): 17996-8004, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22496449

RESUMEN

The melanocyte-specific transcription factor M-MITF is involved in numerous aspects of melanoblast lineage biology including pigmentation, survival, and migration. It plays complex roles at all stages of melanoma progression and metastasis. We established previously that GLI2, a Kruppel-like transcription factor that acts downstream of Hedgehog signaling, is a direct transcriptional target of the TGF-ß/SMAD pathway and contributes to melanoma progression, exerting antagonistic activities against M-MITF to control melanoma cell invasiveness. Herein, we dissected the molecular mechanisms underlying both TGF-ß and GLI2-driven M-MITF gene repression. Using transient cell transfection experiments with M-MITF promoter constructs, chromatin immunoprecipitation, site-directed mutagenesis, and electrophoretic mobility shift assays, we identified a GLI2 binding site within the -334/-296 region of the M-MITF promoter, critical for GLI2-driven transcriptional repression. This region is, however, not needed for inhibition of M-MITF promoter activity by TGF-ß. We determined that TGF-ß rapidly repressed protein kinase A activity, thus reducing both phospho-cAMP-response element-binding protein (CREB) levels and CREB-dependent transcription of the M-MITF promoter. Increased GLI2 binding to its cognate cis-element, associated with reduced CREB-dependent transcription, allowed maximal inhibition of the M-MITF promoter via two distinct mechanisms.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/fisiología , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Proteínas Nucleares/fisiología , Factor de Crecimiento Transformador beta/fisiología , Secuencia de Bases , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cartilla de ADN , ADN de Neoplasias/genética , Progresión de la Enfermedad , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica , Humanos , Factor de Transcripción Asociado a Microftalmía/fisiología , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Transcripción Genética , Proteína Gli2 con Dedos de Zinc
5.
Mol Cancer ; 10(1): 2, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21211030

RESUMEN

BACKGROUND: SKI and SnoN proteins have been shown to inhibit TGF-ß signaling, acting both as transcriptional co-repressors in the cell nucleus, and as sequestrators of SMAD proteins in the cytoplasm. TGF-ß, on the other hand, induces rapid, proteasome-mediated, degradation of both proteins. How elevated SKI and SnoN protein levels co-exist with active autocrine TGF-ß signaling in cancer cells is yet to be understood. RESULTS: In this study, we found elevated SKI and SnoN protein levels in a panel of melanoma cell lines, as compared to normal melanocytes. There was no correlation between SKI protein content and the capacity of melanoma cells to invade Matrigel™, to form subcutaneous tumors, or to metastasize to bone after intracardiac inoculation into nude mice. Nor did we find a correlation between SKI expression and histopathological staging of human melanoma. TGF-ß induced a rapid and dose-dependent degradation of SKI protein, associated with SMAD3/4 specific transcriptional response and induction of pro-metastatic target genes, partially prevented by pharmacologic blockade of proteasome activity. SKI knockdown in 1205Lu melanoma cells did not alter their invasive capacity or transcriptional responses to TGF-ß, and did not allow p21 expression in response to TGF-ß or reveal any growth inhibitory activity of TGF-ß. CONCLUSIONS: Despite high expression in melanoma cells, the role of SKI in melanoma remains elusive: SKI does not efficiently interfere with the pro-oncogenic activities of TGF-ß, unless stabilized by proteasome blockade. Its highly labile nature makes it an unlikely target for therapeutic intervention.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Melanoma/patología , Proteínas Proto-Oncogénicas/metabolismo , Neoplasias Cutáneas/patología , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Humanos , Leupeptinas/farmacología , Melanoma/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Inhibidores de Proteasoma , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Neoplasias Cutáneas/metabolismo , Activación Transcripcional , Regulación hacia Arriba
6.
Sci Rep ; 10(1): 14491, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32879407

RESUMEN

GLI1 expression is broadly accepted as a marker of Hedgehog pathway activation in tumors. Efficacy of Hedgehog inhibitors is essentially limited to tumors bearing activating mutations of the pathway. GLI2, a critical Hedgehog effector, is necessary for GLI1 expression and is a direct transcriptional target of TGF-ß/SMAD signaling. We examined the expression correlations of GLI1/2 with TGFB and HH genes in 152 distinct transcriptome datasets totaling over 23,500 patients and representing 37 types of neoplasms. Their prognostic value was measured in over 15,000 clinically annotated tumor samples from 26 tumor types. In most tumor types, GLI1 and GLI2 follow a similar pattern of expression and are equally correlated with HH and TGFB genes. However, GLI1/2 broadly share prognostic value with TGFB genes and a mesenchymal/EMT signature, not with HH genes. Our results provide a likely explanation for the frequent failure of anti-Hedgehog therapies in tumors, as they suggest a key role for TGF-ß, not Hedgehog, ligands, in tumors with elevated GLI1/2-expression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/genética , Neoplasias/diagnóstico , Proteínas Nucleares/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína Gli2 con Dedos de Zinc/genética , Biología Computacional , Perfilación de la Expresión Génica , Humanos , Ligandos , Análisis Multivariante , Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Modelos de Riesgos Proporcionales , Factores de Riesgo , Transducción de Señal/genética , Transcriptoma
7.
Cancer Res ; 67(5): 2317-24, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17332363

RESUMEN

Melanoma has a propensity to metastasize to bone, where it is exposed to high concentrations of transforming growth factor-beta (TGF-beta). Because TGF-beta promotes bone metastases from other solid tumors, such as breast cancer, we tested the role of TGF-beta in melanoma metastases to bone. 1205Lu melanoma cells, stably transfected to overexpress the natural TGF-beta/Smad signaling inhibitor Smad7, were studied in an experimental model of bone metastasis whereby tumor cells are inoculated into the left cardiac ventricle of nude mice. All mice bearing parental and mock-transfected 1205Lu cells developed osteolytic bone metastases 5 weeks post-tumor inoculation. Mice bearing 1205Lu-Smad7 tumors had significantly less osteolysis on radiographs and longer survival compared with parental and mock-transfected 1205Lu mice. To determine if the reduced bone metastases observed in mice bearing 1205Lu-Smad7 clones was due to reduced expression of TGF-beta target genes known to enhance metastases to bone from breast cancer cells, we analyzed gene expression of osteolytic factors, parathyroid hormone-related protein (PTHrP) and interleukin-11 (IL-11), the chemotactic receptor CXCR4, and osteopontin in 1205Lu cells. Quantitative reverse transcription-PCR analysis indicated that PTHrP, IL-11, CXCR4, and osteopontin mRNA steady-state levels were robustly increased in response to TGF-beta and that Smad7 and the TbetaRI small-molecule inhibitor, SB431542, prevented such induction. In addition, 1205Lu-Smad7 bone metastases expressed significantly lower levels of IL-11, connective tissue growth factor, and PTHrP. These data suggest that TGF-beta promotes osteolytic bone metastases due to melanoma by stimulating the expression of prometastatic factors via the Smad pathway. Blockade of TGF-beta signaling may be an effective treatment for melanoma metastasis to bone.


Asunto(s)
Neoplasias Óseas/secundario , Melanoma/genética , Melanoma/patología , Proteína smad7/genética , Animales , Neoplasias Óseas/genética , Femenino , Humanos , Melanoma/metabolismo , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/genética , Proteína smad7/metabolismo , Análisis de Supervivencia , Transfección , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas
8.
J Dermatol Sci ; 94(3): 321-329, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31208857

RESUMEN

BACKGROUND: Tyrosinase-Related Protein 2 (TRP2) is an enzyme involved in melanogenesis, that also exerts proliferative, anti-apoptotic and immunogenic functions in melanoma cells. TRP2 transcription is regulated by the melanocytic master transcription factor MITF. GLI2, a transcription factor that acts downstream of Hedgehog signaling, is also a direct transcriptional target of the TGF-ß/SMAD pathway that contributes to melanoma progression and exerts transcriptional antagonistic activities against MITF. OBJECTIVES: To characterize the molecular events responsible for TGF-ß and GLI2 repression of TRP2 expression. METHODS: In silico promoter analysis, transient cell transfection experiments with 5'-end TRP2 promoter deletion constructs, chromatin immuno-precipitation, and site-directed promoter mutagenesis were used to dissect the molecular mechanisms of TRP2 gene regulation by TGF-ß and GLI2. RESULTS: We demonstrate that TGF-ß and GLI2-specific TRP2 repression involves direct mechanisms that occur in addition to MITF downregulation by TGF-ß and GLI2. We identify two functional GLI2 binding sites within the TRP2 promoter that are critical for TGF-ß and GLI2 responsiveness, one of them overlapping a CREB binding site. GLI2 and CREB competing for the same cis-element is associated with opposite transcriptional outcome. CONCLUSION: Our results further refine the understanding of how TGF-ß and GLI2 control the phenotypic plasticity of melanoma cells. In particular, we identify critical GLI2-binding cis-elements within the TRP2 promoter region that allow for its transcriptional repression independently from MITF concomitant downregulation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Oxidorreductasas Intramoleculares/genética , Melanoma/genética , Proteínas Nucleares/metabolismo , Neoplasias Cutáneas/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteína Gli2 con Dedos de Zinc/metabolismo , Línea Celular Tumoral , Biología Computacional , Humanos , Melanoma/patología , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas/genética , Neoplasias Cutáneas/patología , Transcripción Genética
9.
Oncogene ; 24(37): 5742-50, 2005 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-16123807

RESUMEN

Transforming growth factor-beta (TGF-beta) superfamily members signal via membrane-bound heteromeric serine-threonine kinase receptor complexes. Upon ligand-binding, receptor activation leads to phosphorylation of cytoplasmic protein substrates of the Smad family. Following phosphorylation and oligomerization, the latter move into the nucleus to act as transcription factors to regulate target gene expression. TGF-beta responses are not solely the result of the activation Smad cascade, but are highly cell-type specific and dependent upon interactions of Smad signaling with a variety of other intracellular signaling mechanisms, initiated or not by TGF-beta itself, that may either potentiate, synergize, or antagonize, the rather linear TGF-beta/Smad pathway. These include, (a), regulation of Smad activity by mitogen-activated protein kinases (MAPKs), (b), nuclear interaction of activated Smads with transcriptional cofactors, whether coactivators or corepressors, that may be themselves be regulated by diverse signaling mechanisms, and (c), negative feedback loops exerted by inhibitory Smads, transcriptional targets of the Smad cascade. This review focuses on how MAPKs modulate the outcome of Smad activation by TGF-beta, and how cross-signaling mechanisms between the Smad and MAPK pathways may take place and affect cell fate in the context of carcinogenesis.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias/etiología , Transducción de Señal/fisiología , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Activación Enzimática , Humanos , Proteínas Smad , Factores de Transcripción/fisiología
10.
Oncogene ; 24(51): 7624-9, 2005 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-16007121

RESUMEN

We previously identified constitutive Smad signaling in human melanoma cells despite resistance to transforming growth factor-beta (TGF-beta) control of cell proliferation. This led us to investigate the effect of inhibitory Smad7 overexpression on melanoma cell behavior. Using the highly metastatic cell line, 1205-Lu, we thus generated melanoma cell clones constitutively expressing Smad7, and their mock-transfected counterparts. Stable expression of Smad7 resulted in an inhibition of constitutive Smad2/3 phosphorylation, and in a reduced TGF-beta response of Smad3/Smad4-driven gene transactivation, as measured using transfected Smad3/4-specific reporter gene constructs. Smad7 overexpression, however, did not alter their proliferative capacity and resistance to TGF-beta-driven growth inhibition. On the other hand, expression of Smad7 efficiently reduced the capacity of human melanoma cells to invade Matrigel in Boyden migration chambers, while not affecting their motility and adhesion to collagen and laminin. Gelatin zymography identified reduced MMP-2 and MMP-9 secretion by Smad7-expressing melanoma cells as compared with their control counterparts. Smad7-expressing melanoma cells exhibited a dramatically reduced capacity to form colonies under anchorage-independent culture conditions, and, when injected subcutaneously into nude mice, were largely delayed in their ability to form tumors. These results suggest that TGF-beta production by melanoma cells not only affects the tumor environment but also directly contributes to tumor cell aggressiveness through autocrine activation of Smad signaling.


Asunto(s)
Melanoma/patología , Neoplasias Cutáneas/patología , Proteína smad7/biosíntesis , Animales , Adhesión Celular , Movimiento Celular , Perfilación de la Expresión Génica , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Melanoma/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Proteína smad7/fisiología , Factor de Crecimiento Transformador beta , Células Tumorales Cultivadas
11.
Dev Cell ; 32(5): 640-51, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25758862

RESUMEN

Cell-cell contacts inhibit cell growth and proliferation in part by activating the Hippo pathway that drives the phosphorylation and nuclear exclusion of the transcriptional coactivators YAP and TAZ. Cell density and Hippo signaling have also been reported to block transforming growth factor ß (TGF-ß) responses, based on the ability of phospho-YAP/TAZ to sequester TGF-ß-activated SMAD complexes in the cytoplasm. Herein, we provide evidence that epithelial cell polarization interferes with TGF-ß signaling well upstream and independent of cytoplasmic YAP/TAZ. Rather, polarized basolateral presentation of TGF-ß receptors I and II deprives apically delivered TGF-ß of access to its receptors. Basolateral ligand delivery nonetheless remains entirely effective to induce TGF-ß responses. These data demonstrate that cell-type-specific inhibition of TGF-ß signaling by cell density is restricted to polarized epithelial cells and reflects the polarized distribution of TGF-ß receptors, which thus affects SMAD activation irrespective of Hippo pathway activation.


Asunto(s)
Citoplasma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Aciltransferasas , Western Blotting , Recuento de Células , Proteínas de Ciclo Celular , Proliferación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Vía de Señalización Hippo , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Proteínas Nucleares/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/genética
12.
Int J Biochem Cell Biol ; 36(7): 1161-5, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15109563

RESUMEN

Since its discovery in the early 1980s, transforming growth factor-beta (TGF-beta) has emerged as a family of growth factors involved in essential physiological processes, including embryonic development, differentiation, tissue repair and cell growth control. Knockout experiments for the three mammalian isoforms of TGF-betas in mice have demonstrated their importance in regulating inflammation and tissue repair. Also, TGF-beta has been implicated in the pathogenesis of human diseases, including tissue fibrosis and carcinogenesis where, in the latter case, it may exert both tumor suppressor and pro-oncogenic activities depending on the stage of the tumor. Cellular signaling by TGF-beta family members is initiated by the assembly of specific cell surface serine/threonine kinase type receptors that activate transcription factors of the Smad family.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Proteínas de Unión al ADN/fisiología , Enfermedad/etiología , Humanos , Ratones , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Isoformas de Proteínas/uso terapéutico , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Proteínas Smad , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/uso terapéutico
13.
FEBS Lett ; 578(1-2): 111-5, 2004 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-15581626

RESUMEN

Repression of activation of c-Jun N-terminal kinase (JNK) participates in the anti-apoptotic effect of nuclear factor-kappaB (NF-kappaB) in TNFalpha-treated Ewing sarcoma cells. As oxidative stress is one of the most prominent activators of JNK, we investigated the relationship between TNFalpha-induced NF-kappaB activation and the control of oxidative stress. Inhibition of NF-kappaB activation resulted in an increase in TNFalpha-induced ROS production, lipid peroxidation and protein oxidation. Those ROS and lipid peroxides were both involved in TNFalpha-induced apoptosis, whereas only ROS elevation triggered sustained JNK activation. TNFalpha increased the level of two antioxidant enzymes, thioredoxin and manganese superoxide dismutase by an NF-kappaB-dependent mechanism. Inhibition of expression or activity of these enzymes sensitized cells to TNFalpha-induced apoptosis, indicating their functional role in protection from cell death. Thus, agents that inhibit activities of these enzymes may prove helpful in the treatment of Ewing tumors.


Asunto(s)
Apoptosis/fisiología , FN-kappa B/metabolismo , Estrés Oxidativo , Sarcoma de Ewing/metabolismo , Superóxido Dismutasa/metabolismo , Tiorredoxinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adolescente , Antioxidantes/metabolismo , Caspasa 3 , Caspasas/metabolismo , Niño , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Peroxidación de Lípido , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas
14.
J Dermatol Sci ; 35(2): 83-92, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15265520

RESUMEN

Members of the transforming growth factor-beta (TGF-beta) superfamily are pleiotropic cytokines that have the ability to regulate numerous cell functions, including proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and production of extracellular matrix, allowing them to play an important role during embryonic development and for maintenance of tissue homeostasis. Three TGF-beta isoforms have been identified in mammals. They propagate their signal via a signal transduction network involving receptor serine/threonine kinases at the cell surface and their substrates, the SMAD proteins. Upon phosphorylation and oligomerization, the latter move into the nucleus to regulate transcription of target genes. This review will summarize recent advances in the understanding of the mechanisms underlying SMAD modulation of extracellular matrix gene expression in the context of wound healing and tissue fibrosis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Matriz Extracelular/fisiología , Regulación de la Expresión Génica , Transducción de Señal/fisiología , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Cicatrización de Heridas/fisiología , Animales , Humanos , Proteínas Smad
15.
Pharmacol Ther ; 137(2): 183-99, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23063491

RESUMEN

Recent advances in the field of cancer therapeutics come from the development of drugs that specifically recognize validated oncogenic or pro-metastatic targets. The latter may be mutated proteins with altered function, such as kinases that become constitutively active, or critical components of growth factor signaling pathways, whose deregulation leads to aberrant malignant cell proliferation and dissemination to metastatic sites. We herein focus on the description of the overlapping activities of two important developmental pathways often exacerbated in cancer, namely Transforming Growth Factor-ß (TGF-ß) and Hedgehog (HH) signaling, with a special emphasis on the unifying oncogenic role played by GLI1/2 transcription factors. The latter are the main effectors of the canonical HH pathway, yet are direct target genes of TGF-ß/SMAD signal transduction. While tumor-suppressor in healthy and pre-malignant tissues, TGF-ß is often expressed at high levels in tumors and contributes to tumor growth, escape from immune surveillance, invasion and metastasis. HH signaling regulates cell proliferation, differentiation and apoptosis, and aberrant HH signaling is found in a variety of cancers. We discuss the current knowledge on HH and TGF-ß implication in cancer including cancer stem cell biology, as well as the current state, both successes and failures, of targeted therapeutics aimed at blocking either of these pathways in the pre-clinical and clinical settings.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Hedgehog/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador alfa/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Ensayos Clínicos como Asunto , Proteínas Hedgehog/genética , Humanos , Neoplasias/metabolismo , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Factor de Crecimiento Transformador alfa/genética
16.
Ann Dermatol ; 25(2): 135-44, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23717002

RESUMEN

Transforming growth factor-ß (TGF-ß) is a pleiotropic growth factor with broad tissue distribution that plays critical roles during embryonic development, normal tissue homeostasis, and cancer. While its cytostatic activity on normal epithelial cells initially defined TGF-ß signaling as a tumor suppressor pathway, there is ample evidence indicating that TGF-ß is a potent pro-tumorigenic agent, acting via autocrine and paracrine mechanisms to promote peri-tumoral angiogenesis, together with tumor cell migration, immune escape, and dissemination to metastatic sites. This review summarizes the current knowledge on the implication of TGF-ß signaling in melanoma.

17.
Pigment Cell Melanoma Res ; 26(6): 861-73, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23890107

RESUMEN

In melanoma cells, high expression of the transcription factor GLI2 is associated with increased invasive potential and loss of E-cadherin expression, an event reminiscent of the epithelial-to-mesenchymal transition (EMT). Herein, we provide evidence that GLI2 represses E-cadherin gene (CDH1) expression in melanoma cells via distinct mechanisms, enhancing transcription of the EMT-activator ZEB1 and cooperative repression of CDH1 gene transcription via direct binding of both GLI2 and ZEB1 to two closely positioned Kruppel-like factor-binding sites within the CDH1 promoter. GLI2 silencing rescued CDH1 expression except in melanoma cell lines in which the CDH1 promoter was hypermethylated. Proximity ligation assays identified GLI2-ZEB1 complexes in melanoma cell nuclei, proportional to endogenous GLI2 and ZEB1 expression, and whose accumulation was enhanced by the classical EMT inducer TGF-ß. These data identify GLI2 as a critical modulator of the cadherin switch in melanoma, a molecular process that is critical for metastatic spread of the disease.


Asunto(s)
Cadherinas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Melanoma/genética , Proteínas Nucleares/metabolismo , Neoplasias Cutáneas/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Antígenos CD , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/patología , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Silenciador del Gen/efectos de los fármacos , Humanos , Melanoma/patología , Modelos Biológicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Proteínas Represoras/metabolismo , Neoplasias Cutáneas/patología , Factores de Transcripción de la Familia Snail , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , Proteína Gli2 con Dedos de Zinc
18.
FEBS Lett ; 586(14): 2016-25, 2012 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-22609357

RESUMEN

Hedgehog (HH) and TGF-ß signals control various aspects of embryonic development and cancer progression. While their canonical signal transduction cascades have been well characterized, there is increasing evidence that these pathways are able to exert overlapping activities that challenge efficient therapeutic targeting. We herein review the current knowledge on HH signaling and summarize the recent findings on the crosstalks between the HH and TGF-ß pathways in cancer.


Asunto(s)
Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Transición Epitelial-Mesenquimal , Fibrosis/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ligandos , Ratones , Modelos Biológicos , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Procesamiento Postranscripcional del ARN , Ratas , Transducción de Señal , Transactivadores/metabolismo , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
19.
Pigment Cell Melanoma Res ; 25(3): 343-53, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22336146

RESUMEN

There is growing evidence that the metastatic spread of melanoma is driven not by a linear increase in tumorigenic aggressiveness, but rather by switching back and forth between two different phenotypes of metastatic potential. In vitro these phenotypes are respectively defined by the characteristics of strong proliferation/weak invasiveness and weak proliferation/strong invasiveness. Melanoma cell phenotype is tightly linked to gene expression. Taking advantage of this, we have developed a gene expression-based tool for predicting phenotype called Heuristic Online Phenotype Prediction. We demonstrate the predictive utility of this tool by comparing phenotype-specific signatures with measurements of characteristics of melanoma phenotype-specific biology in different melanoma cell lines and short-term cultures. We further show that 86% of 536 tested melanoma lines and short-term cultures are significantly associated with the phenotypes we describe. These findings reinforce the concept that a two-state system, as described by the phenotype switching model, underlies melanoma progression.


Asunto(s)
Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Perfilación de la Expresión Génica , Melanoma/clasificación , Neoplasias Cutáneas/clasificación , Proliferación Celular , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Estudios de Asociación Genética , Humanos , Melanoma/genética , Melanoma/patología , Análisis por Micromatrices , Invasividad Neoplásica , Fenotipo , Reproducibilidad de los Resultados , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Distribución Tisular , Células Tumorales Cultivadas/clasificación
20.
Cancer Res ; 72(23): 6247-56, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23002206

RESUMEN

TGF-ß derived from bone fuels melanoma bone metastases by inducing tumor secretion of prometastatic factors that act on bone cells to change the skeletal microenvironment. Halofuginone is a plant alkaloid derivative that blocks TGF-ß signaling with antiangiogenic and antiproliferative properties. Here, we show for the first time that halofuginone therapy decreases development and progression of bone metastasis caused by melanoma cells through the inhibition of TGF-ß signaling. Halofuginone treatment of human melanoma cells inhibited cell proliferation, phosphorylation of SMAD proteins in response to TGF-ß, and TGF-ß-induced SMAD-driven transcription. In addition, halofuginone reduced expression of TGF-ß target genes that enhance bone metastases, including PTHrP, CTGF, CXCR4, and IL11. Also, cell apoptosis was increased in response to halofuginone. In nude mice inoculated with 1205 Lu melanoma cells, a preventive protocol with halofuginone inhibited bone metastasis. The beneficial effects of halofuginone treatment were comparable with those observed with other anti-TGF-ß strategies, including systemic administration of SD208, a small-molecule inhibitor of TGF-ß receptor I kinase, or forced overexpression of Smad7, a negative regulator of TGF-ß signaling. Furthermore, mice with established bone metastases treated with halofuginone had significantly less osteolysis than mice receiving placebo assessed by radiography. Thus, halofuginone is also effective in reducing the progression of melanoma bone metastases. Moreover, halofuginone treatment reduced melanoma metastasis to the brain, showing the potential of this novel treatment against cancer metastasis.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/prevención & control , Neoplasias Óseas/secundario , Melanoma/tratamiento farmacológico , Piperidinas/farmacología , Quinazolinonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Melanoma/metabolismo , Melanoma/patología , Melanoma/secundario , Ratones , Ratones Desnudos , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA