Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Metastasis Rev ; 42(1): 323-334, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36754910

RESUMEN

Cancer plasticity is now a recognized new hallmark of cancer which is due to disturbances of cell differentiation programs. It is manifested not only in various forms like the best-known epithelial-mesenchymal transition (EMT) but also in vasculogenic and megakaryocytic mimicries regulated by EMT-specific or less-specific transcription factors such as HIF1a or STAT1/2. Studies in the past decades provided ample data that cancer plasticity can be manifested also in the expression of a vast array of immune cell genes; best-known examples are PDL1/CD274, CD47, or IDO, and we termed it immunogenic mimicry (IGM). However, unlike other types of plasticities which are epigenetically regulated, expression of IGM genes are frequently due to gene amplifications. It is important that the majority of the IGM genes are regulated by interferons (IFNs) suggesting that their protein expressions are regulated by the immune microenvironment. Most of the IGM genes have been shown to be involved in immune escape of cancers broadening the repertoire of these mechanisms and offering novel targets for immunotherapeutics.


Asunto(s)
Neoplasias , Neovascularización Patológica , Humanos , Neovascularización Patológica/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Transición Epitelial-Mesenquimal/genética , Adaptación Fisiológica , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Microambiente Tumoral/genética
2.
Adv Exp Med Biol ; 1139: 105-114, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31134497

RESUMEN

Metastatic melanoma continues to present a significant challenge-with a cure rate of less than 10% and a median survival of 6-9 months. Despite noteworthy advances in the field, the heterogeneity of melanoma tumors, comprised of cell subpopulations expressing a cancer stem cell (CSC) phenotype concomitant with drug resistance markers presents a formidable challenge in the design of current therapies. Particularly vexing is the ability of distinct subpopulations of melanoma cells to resist standard-of-care treatments, resulting in relapse and progression to metastasis. Recent studies have provided new information and insights into the expression and function of CSC markers associated with the aggressive melanoma phenotype, such as the embryonic morphogen Nodal and CD133, together with a drug resistance marker ABCA1. This chapter highlights major findings that demonstrate the promise of targeting Nodal as a viable option to pursue in combination with standard-of-care therapy. In recognizing that aggressive melanoma tumors utilize multiple mechanisms to survive, we must consider a more strategic approach to effectively target heterogeneity, tumor cell plasticity, and functional adaptation and resistance to current therapies-to eliminate relapse, disease progression, and metastasis.


Asunto(s)
Plasticidad de la Célula , Melanoma/patología , Células Madre Neoplásicas/citología , Biomarcadores de Tumor , Humanos , Recurrencia Local de Neoplasia
3.
Cancer Metastasis Rev ; 35(1): 21-39, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26951550

RESUMEN

The transforming growth factor beta (TGFß) superfamily member Nodal is an established regulator of early embryonic development, with primary roles in endoderm induction, left-right asymmetry, and primitive streak formation. Nodal signals through TGFß family receptors at the plasma membrane and induces signaling cascades leading to diverse transcriptional regulation. While conceptually simple, the regulation of Nodal and its molecular effects are profoundly complex and context dependent. Pioneering work by developmental biologists has characterized the signaling pathways, regulatory components, and provided detailed insight into the mechanisms by which Nodal mediates changes at the cellular and organismal levels. Nodal is also an important factor in maintaining pluripotency of embryonic stem cells through regulation of core transcriptional programs. Collectively, this work has led to an appreciation for Nodal as a powerful morphogen capable of orchestrating multiple cellular phenotypes. Although Nodal is not active in most adult tissues, its reexpression and signaling have been linked to multiple types of human cancer, and Nodal has emerged as a driver of tumor growth and cellular plasticity. In vitro and in vivo experimental evidence has demonstrated that inhibition of Nodal signaling reduces cancer cell aggressive characteristics, while clinical data have established associations with Nodal expression and patient outcomes. As a result, there is great interest in the potential targeting of Nodal activity in a therapeutic setting for cancer patients that may provide new avenues for suppressing tumor growth and metastasis. In this review, we evaluate our current understanding of the complexities of Nodal function in cancer and highlight recent experimental evidence that sheds light on the therapeutic potential of its inhibition.


Asunto(s)
Terapia Molecular Dirigida , Neoplasias/genética , Proteína Nodal/genética , Factor de Crecimiento Transformador beta/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/patología , Neoplasias/terapia , Proteína Nodal/biosíntesis , Transducción de Señal
5.
Nat Rev Cancer ; 7(4): 246-55, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17384580

RESUMEN

Aggressive tumour cells share many characteristics with embryonic progenitors, contributing to the conundrum of tumour cell plasticity. Recent studies using embryonic models of human stem cells, the zebrafish and the chick have shown the reversion of the metastatic phenotype of aggressive melanoma cells, and revealed the convergence of embryonic and tumorigenic signalling pathways, which may help to identify new targets for therapeutic intervention. This Review will summarize the embryonic models used to reverse the metastatic melanoma phenotype, and highlight the prominent signalling pathways that have emerged as noteworthy targets for future consideration.


Asunto(s)
Embrión de Mamíferos/citología , Embrión no Mamífero , Melanoma/patología , Metástasis de la Neoplasia/patología , Animales , Comunicación Celular , Diferenciación Celular , Línea Celular Tumoral , Movimiento Celular , Embrión de Pollo , Células Madre Embrionarias , Humanos , Melanoma/metabolismo , Modelos Animales , Trasplante de Neoplasias , Cresta Neural , Proteína Nodal , Transducción de Señal , Factor de Crecimiento Transformador beta/fisiología , Pez Cebra/embriología
6.
Int J Mol Sci ; 17(3): 418, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27011171

RESUMEN

Expression of Nodal, a Transforming Growth Factor-beta (TGF-ß) related growth factor, is associated with aggressive melanoma. Nodal expression in adult dysplastic nevi may predict the development of aggressive melanoma in some patients. A subset of pediatric patients diagnosed with giant or large congenital melanocytic nevi (LCMN) has shown increased risk for development of melanoma. Here, we investigate whether Nodal expression can help identify the rare cases of LCMN that develop melanoma and shed light on why the majority of these patients do not. Immunohistochemistry (IHC) staining results show varying degree of Nodal expression in pediatric dysplastic nevi and LCMN. Moreover, median scores from Nodal IHC expression analysis were not significantly different between these two groups. Additionally, none of the LCMN patients in this study developed melanoma, regardless of Nodal IHC levels. Co-culture experiments revealed reduced tumor growth and lower levels of Nodal and its signaling molecules P-SMAD2 and P-ERK1/2 when melanoma cells were grown in vivo or in vitro with normal melanocytes. The same was observed in melanoma cells cultured with melanocyte conditioned media containing pigmented melanocyte derived melanosomes (MDM). Since MDM contain molecules capable of inactivating radical oxygen species, to investigate potential anti-oxidant effect of MDM on Nodal expression and signaling in melanoma, melanoma cells were treated with either N-acetyl-l-cysteine (NAC), a component of the anti-oxidant glutathione or synthetic melanin, which in addition to providing pigmentation can also exert free radical scavenging activity. Melanoma cells treated with NAC or synthetic melanin showed reduced levels of Nodal, P-SMAD2 and P-ERK1/2 compared to untreated melanoma cells. Thus, the potential role for Nodal in melanoma development in LCMN is less evident than in adult dysplastic nevi possibly due to melanocyte cross-talk in LCMN capable of offsetting or delaying the pro-melanoma effects of Nodal via anti-oxidant effects of MDM.


Asunto(s)
Melanocitos/metabolismo , Melanoma/metabolismo , Proteína Nodal/metabolismo , Transducción de Señal , Acetilcisteína/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Niño , Femenino , Humanos , Melaninas/farmacología , Melanocitos/efectos de los fármacos , Melanoma/congénito , Melanoma/patología , Ratones , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Nodal/genética , Proteína Smad2/metabolismo
7.
Semin Cancer Biol ; 29: 40-50, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25073112

RESUMEN

The Ras-ERK pathway is deregulated in approximately a third of human cancers, particularly those of epithelial origin. In aggressive, triple-negative, basal-like breast cancers, most tumors display increased MEK and ERK phosphorylation and exhibit a gene expression profile characteristic of Kras or EGFR mutant tumors; however, Ras family genetic mutations are uncommon in triple-negative breast cancer and EGFR mutations account for only a subset of these tumors. Therefore, the upstream events that activate MAPK signaling and promote tumor aggression in triple-negative breast cancers remain poorly defined. We have previously shown that a secreted TGF-ß family signaling ligand, Nodal, is expressed in breast cancer in correlation with disease progression. Here we highlight key findings demonstrating that Nodal is required in aggressive human breast cancer cells to activate ERK signaling and downstream tumorigenic phenotypes both in vitro and in vivo. Experimental knockdown of Nodal signaling downregulates ERK activity, resulting in loss of c-myc, upregulation of p27, G1 cell cycle arrest, increased apoptosis and decreased tumorigenicity. The data suggest that ERK activation by Nodal signaling regulates c-myc and p27 proteins post-translationally and that this cascade is essential for aggressive breast tumor behavior in vivo. As the MAPK pathway is an important target for treating triple-negative breast cancers, upstream Nodal signaling may represent a promising target for breast cancer diagnosis and combined therapies aimed at blocking ERK pathway activation.


Asunto(s)
Transformación Celular Neoplásica/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Proteína Nodal/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Apoptosis/genética , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteína Nodal/genética , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Neoplasias de la Mama Triple Negativas/genética
8.
Int J Cancer ; 136(5): E242-51, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25204799

RESUMEN

The significant role of the embryonic morphogen Nodal in maintaining the pluripotency of embryonic stem cells is well documented. Interestingly, the recent discovery of Nodal's re-expression in several aggressive and metastatic cancers has highlighted its critical role in self renewal and maintenance of the stem cell-like characteristics of tumor cells, such as melanoma. However, the key TGFß/Nodal signaling component(s) governing Nodal's effects in metastatic melanoma remain mostly unknown. By employing receptor profiling at the mRNA and protein level(s), we made the novel discovery that embryonic stem cells and metastatic melanoma cells share a similar repertoire of Type I serine/threonine kinase receptors, but diverge in their Type II receptor expression. Ligand:receptor crosslinking and native gel binding assays indicate that metastatic melanoma cells employ the heterodimeric TGFß receptor I/TGFß receptor II (TGFßRI/TGFßRII) for signal transduction, whereas embryonic stem cells use the Activin receptors I and II (ACTRI/ACTRII). This unexpected receptor usage by tumor cells was tested by: neutralizing antibody to block its function; and transfecting the dominant negative receptor to compete with the endogenous receptor for ligand binding. Furthermore, a direct biological role for TGFßRII was found to underlie vasculogenic mimicry (VM), an endothelial phenotype contributing to vascular perfusion and associated with the functional plasticity of aggressive melanoma. Collectively, these findings reveal the divergence in Nodal signaling between embryonic stem cells and metastatic melanoma that can impact new therapeutic strategies targeting the re-emergence of embryonic pathways.


Asunto(s)
Células Madre Embrionarias/metabolismo , Melanoma/metabolismo , Proteína Nodal/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Activinas/genética , Activinas/metabolismo , Western Blotting , Células Cultivadas , Células Madre Embrionarias/citología , Humanos , Melanoma/genética , Melanoma/patología , Proteína Nodal/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/secundario , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
9.
Connect Tissue Res ; 56(5): 364-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26327334

RESUMEN

Cripto-1 (CR-1) is a multifunctional embryonic protein that is re-expressed during inflammation, wound repair, and malignant transformation. CR-1 can function either as a tethered co-receptor or shed as a free ligand underpinning its flexible role in cell physiology. CR-1 has been shown to mediate cell growth, migration, invasion, and induce epithelial to mesenchymal transition (EMT). The main signaling pathways mediating CR-1 effects include Nodal-dependent (Smad2/3) and Nodal-independent (Src/p44/42/Akt) signaling transduction pathways. In addition, there are several naturally occurring binding partner proteins (BPPs) for CR-1 that can either agonize or antagonize its bioactivity. We will review the collective role of CR-1 as an extracellular protein, discuss caveats to consider in developing a quantitation assay, define possible mechanistic avenues applicable for drug discovery, and report on our experimental approaches to overcome these problematic issues.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Proteínas Ligadas a GPI/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal/fisiología , Autoanticuerpos/inmunología , Factor de Crecimiento Epidérmico/fisiología , Transición Epitelial-Mesenquimal/inmunología , Espacio Extracelular/metabolismo , Humanos , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/metabolismo
10.
Int J Mol Sci ; 16(9): 21342-62, 2015 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-26370966

RESUMEN

Nodal is a potent embryonic morphogen belonging to the TGF-ß superfamily. Typically, it also binds to the ALK4/ActRIIB receptor complex in the presence of the co-receptor Cripto-1. Nodal expression is physiologically restricted to embryonic tissues and human embryonic stem cells, is absent in normal cells but re-emerges in several human cancers, including melanoma, breast, and colon cancer. Our aim was to obtain mAbs able to recognize Nodal on a major CBR (Cripto-Binding-Region) site and to block the Cripto-1-mediated signalling. To achieve this, antibodies were raised against hNodal(44-67) and mAbs generated by the hybridoma technology. We have selected one mAb, named 3D1, which strongly associates with full-length rhNodal (KD 1.4 nM) and recognizes the endogenous protein in a panel of human melanoma cell lines by western blot and FACS analyses. 3D1 inhibits the Nodal-Cripto-1 binding and blocks Smad2/3 phosphorylation. Data suggest that inhibition of the Nodal-Cripto-1 axis is a valid therapeutic approach against melanoma and 3D1 is a promising and interesting agent for blocking Nodal-Cripto mediated tumor development. These findings increase the interest for Nodal as both a diagnostic and prognostic marker and as a potential new target for therapeutic intervention.


Asunto(s)
Anticuerpos Monoclonales/química , Modelos Moleculares , Proteína Nodal/química , Estructura Secundaria de Proteína , Secuencia de Aminoácidos , Anticuerpos Monoclonales/farmacología , Mapeo Epitopo/métodos , Epítopos/química , Epítopos/metabolismo , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/metabolismo , Factores de Diferenciación de Crecimiento/química , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteína Nodal/antagonistas & inhibidores , Proteína Nodal/metabolismo , Péptidos/síntesis química , Péptidos/química , Péptidos/aislamiento & purificación , Péptidos/metabolismo , Unión Proteica
11.
Cancer Metastasis Rev ; 31(3-4): 529-51, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22752408

RESUMEN

Maspin, a non-inhibitory member of the serine protease inhibitor superfamily, has been characterized as a tumor suppressor gene in multiple cancer types. Among the established anti-tumor effects of Maspin are the inhibition of cancer cell invasion, attachment to extracellular matrices, increased sensitivity to apoptosis, and inhibition of angiogenesis. However, while significant experimental data support the role of Maspin as a tumor suppressor, clinical data regarding the prognostic implications of Maspin expression have led to conflicting results. This highlights the need for a better understanding of the context dependencies of Maspin in normal biology and how these are perturbed in the context of cancer. In this review, we outline the regulation and roles of Maspin in normal and developmental biology while discussing novel evidence and emerging theories related to its functions in cancer. We provide insight into the immense therapeutic potential of Maspin and the challenges related to its successful clinical translation.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Serpinas/fisiología , Serpinas/uso terapéutico , Animales , Apoptosis , Resistencia a Antineoplásicos , Epigénesis Genética , Humanos , Integrinas/fisiología , Neovascularización Fisiológica , Óxido Nítrico/fisiología , Unión Proteica , Proteínas Recombinantes/uso terapéutico , Serpinas/química , Serpinas/genética , Tamoxifeno/farmacología , Proteína p53 Supresora de Tumor/fisiología
12.
Am J Pathol ; 181(4): 1115-25, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22944600

RESUMEN

In 1999, The American Journal of Pathology published an article entitled "Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry," by Maniotis and colleagues, which ignited a spirited debate for several years and earned distinction as a citation classic. Tumor cell vasculogenic mimicry (VM) refers to the plasticity of aggressive cancer cells forming de novo vascular networks, which thereby contribute to perfusion of rapidly growing tumors, transporting fluid from leaky vessels, and/or connecting with the constitutional endothelial-lined vasculature. The tumor cells capable of VM share a plastic, transendothelial phenotype, which may be induced by hypoxia. Since VM was introduced as a novel paradigm for melanoma tumor perfusion, many studies have contributed new findings illuminating the underlying molecular pathways supporting VM in a variety of tumors, including carcinomas, sarcomas, glioblastomas, astrocytomas, and melanomas. Facilitating the functional plasticity of tumor cell VM are key proteins associated with vascular, stem cell, and hypoxia-related signaling pathways, each deserving serious consideration as potential therapeutic targets and diagnostic indicators of the aggressive, metastatic phenotype.


Asunto(s)
Imitación Molecular , Neoplasias/irrigación sanguínea , Neoplasias/terapia , Investigación Biomédica Traslacional , Animales , Humanos , Metástasis de la Neoplasia , Neoplasias/patología , Células Madre Neoplásicas/patología , Transducción de Señal , Microambiente Tumoral
13.
Nat Med ; 12(8): 925-32, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16892036

RESUMEN

Bidirectional cellular communication is integral to both cancer progression and embryological development. In addition, aggressive tumor cells are phenotypically plastic, sharing many properties with embryonic cells. Owing to the similarities between these two types of cells, the developing zebrafish can be used as a biosensor for tumor-derived signals. Using this system, we show that aggressive melanoma cells secrete Nodal (a potent embryonic morphogen) and consequently can induce ectopic formation of the embryonic axis. We further show that Nodal is present in human metastatic tumors, but not in normal skin, and thus may be involved in melanoma pathogenesis. Inhibition of Nodal signaling reduces melanoma cell invasiveness, colony formation and tumorigenicity. Nodal inhibition also promotes the reversion of melanoma cells toward a melanocytic phenotype. These data suggest that Nodal signaling has a key role in melanoma cell plasticity and tumorigenicity, thereby providing a previously unknown molecular target for regulating tumor progression.


Asunto(s)
Melanoma/patología , Proteínas de la Membrana/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Blástula/trasplante , Línea Celular Tumoral , Embrión no Mamífero , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Melanocitos/metabolismo , Melanocitos/patología , Proteínas de la Membrana/antagonistas & inhibidores , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Oligonucleótidos Antisentido/farmacología , Trasplante Heterólogo , Proteínas de Pez Cebra/antagonistas & inhibidores
14.
Nat Rev Cancer ; 3(6): 411-21, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12778131

RESUMEN

The gene-expression profile of aggressive cutaneous and uveal melanoma cells resembles that of an undifferentiated, embryonic-like cell. The plasticity of certain types of cancer cell could explain their ability to mimic the activities of endothelial cells and to participate in processes such as neovascularization and the formation of a fluid-conducting, matrix-rich meshwork. This ability has been termed 'vasculogenic mimicry'. How does vasculogenic mimicry contribute to tumour progression, and can it be targeted by therapeutic agents?


Asunto(s)
Endotelio Vascular/patología , Melanoma/irrigación sanguínea , Neovascularización Patológica/etiología , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias de la Úvea/irrigación sanguínea , Animales , Humanos , Melanoma/patología , Neoplasias Cutáneas/patología , Neoplasias de la Úvea/patología
15.
Pathol Oncol Res ; 29: 1611038, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37351538

RESUMEN

CVM-1118 (foslinanib) is a phosphoric ester compound selected from 2-phenyl-4-quinolone derivatives. The NCI 60 cancer panel screening showed CVM-1125, the major active metabolite of CVM-1118, to exhibit growth inhibitory and cytotoxic effects at nanomolar range. CVM-1118 possesses multiple bioactivities, including inducing cellular apoptosis, cell cycle arrest at G2/M, as well as inhibiting vasculogenic mimicry (VM) formation. The TNF receptor associated protein 1 (TRAP1) was identified as the binding target of CVM-1125 using nematic protein organization technique (NPOT) interactome analysis. Further studies demonstrated CVM-1125 reduced the protein level of TRAP1 and impeded its downstream signaling by reduction of cellular succinate levels and destabilization of HIF-1α. The pharmacogenomic biomarkers associated with CVM-1118 were also examined by Whole Genome CRISPR Knock-Out Screening. Two hits (STK11 and NF2) were confirmed with higher sensitivity to the drug in cell knock-down experiments. Biological assays indicate that the mechanism of action of CVM-1118 is via targeting TRAP1 to induce mitochondrial apoptosis, suppress tumor cell growth, and inhibit vasculogenic mimicry formation. Most importantly, the loss-of-function mutations of STK11 and NF2 are potential biomarkers of CVM-1118 which can be applied in the selection of cancer patients for CVM-1118 treatment. CVM-1118 is currently in its Phase 2a clinical development.


Asunto(s)
Apoptosis , Neovascularización Patológica , Humanos , Factor 1 Asociado a Receptor de TNF/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Biomarcadores , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/metabolismo
16.
Breast Cancer Res ; 14(3): R75, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22577960

RESUMEN

INTRODUCTION: The re-emergence of the tumour growth factor-beta (TGF-beta)-related embryonic morphogen Nodal has recently been reported in several different human cancers. In this study, we examined the expression of Nodal in a series of benign and malignant human breast tissues to determine the clinical significance of this expression and whether Nodal could represent a potential therapeutic target in breast cancer. METHODS: Tissue sections from 431 therapeutically naive patients diagnosed with benign or malignant breast disease were stained for Nodal by immunohistochemistry and analysed in a blinded manner. The degree of Nodal staining was subsequently correlated with available clinical data, such as diagnoses and disease stage. These tissue findings were further explored in breast cancer cell lines MDA-MB-231 and MDA-MB-468 treated with a Nodal blocking antibody to determine biological effects for target validation. RESULTS: A variable degree of Nodal staining was detected in all samples. The intensity of Nodal staining was significantly greater in undifferentiated, advanced stage, invasive breast cancer compared with benign breast disease or early stage breast cancer. Treatment of human breast cancer cells in vitro with Nodal blocking antibody significantly reduced proliferation and colony-forming ability in soft agar, concomitant with increased apoptosis. CONCLUSIONS: These data suggest a potential role for Nodal as a biomarker for disease progression and a promising target for anti-Nodal therapy in breast cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Proteína Nodal/metabolismo , Adulto , Anciano , Anticuerpos Bloqueadores/inmunología , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Proteína Nodal/inmunología , Pronóstico
17.
Sarcoma ; 2012: 820254, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22448124

RESUMEN

Chondrosarcomas are among the most malignant skeletal tumors. Dedifferentiated chondrosarcoma is a highly aggressive subtype of chondrosarcoma, with lung metastases developing within a few months of diagnosis in 90% of patients. In this paper we performed comparative analyses of the transcriptomes of five individual metastatic lung lesions that were surgically resected from a patient with dedifferentiated chondrosarcoma. We document for the first time a high heterogeneity of gene expression profiles among the individual lung metastases. Moreover, we reveal a signature of "multifunctional" genes that are expressed in all metastatic lung lesions. Also, for the first time, we document the occurrence of massive macrophage infiltration in dedifferentiated chondrosarcoma lung metastases.

18.
Lab Invest ; 91(6): 819-24, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21464823

RESUMEN

As the frequency of melanoma diagnosis increases, current treatment strategies are still struggling to significantly impact patient survival. Some promise has been shown in treating certain melanomas by targeting activated signaling pathways resulting from specific mutations in proteins, such as BRAF and NRAS. Recently, the identification of embryonic signaling pathways in melanoma has helped us better understand certain biological characteristics, such as cellular heterogeneity and phenotypic plasticity, and has provided novel insight pertinent to diagnosis and therapy. For instance, our studies have shown that the TGF-ß family member, Nodal, is expressed in melanoma and is responsible, at least in part, for tumor cell plasticity and aggressiveness. Since the majority of normal adult tissues do not express Nodal, we reason that this embryonic morphogen could be used to identify and target aggressive melanoma cells. We have also identified that molecular cross-talk between the Notch and Nodal pathways may represent a mechanism responsible for the overexpression of Nodal in melanoma. Further exploitation of the relationship between embryonic signaling pathways and cancer pathogenesis could lead to novel approaches for diagnosis and therapy in cancers, such as melanoma.


Asunto(s)
Embrión de Mamíferos/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Melanoma/metabolismo , Proteína Nodal/metabolismo , Fenotipo , Transducción de Señal/fisiología , Línea Celular Tumoral , Embrión de Mamíferos/metabolismo , Humanos , Melanoma/diagnóstico , Melanoma/terapia , Modelos Biológicos , Receptores Notch/metabolismo , Células Madre/citología
19.
Prostate ; 71(11): 1198-209, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21656830

RESUMEN

BACKGROUND: Nodal is a member of the transforming growth factor ß (TGFß) superfamily that directs embryonic patterning and promotes the plasticity and tumorigenicity of tumor cells, but its role in the prostate is unknown. The goal of this study was to characterize the expression and function of Nodal in prostate cancer and determine whether, like other TGFß ligands, it modulates androgen receptor (AR) activity. METHODS: Nodal expression was investigated using immunohistochemistry of tissue microarrays and Western blots of prostate cell lines. The functional role of Nodal was examined using Matrigel and soft agar growth assays. Cross-talk between Nodal and AR signaling was assessed with luciferase reporter assays and expression of endogenous androgen regulated genes. RESULTS: Significantly increased Nodal expression was observed in cancer compared with benign prostate specimens. Nodal was only expressed by DU145 and PC3 cells. All cell lines expressed Nodal's co-receptor, Cripto-1, but lacked Lefty, a critical negative regulator of Nodal signaling. Recombinant human Nodal triggered downstream Smad2 phosphorylation in DU145 and LNCaP cells, and stable transfection of pre-pro-Nodal enhanced the growth of LNCaP cells in Matrigel and soft agar. Finally, Nodal attenuated AR signaling, reducing the activity of a PSA promoter construct in luciferase assays and down-regulating the endogenous expression of androgen regulated genes. CONCLUSIONS: An aberrant Nodal signaling pathway is re-expressed and functionally active in prostate cancer cells.


Asunto(s)
Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/fisiología , Proteína Nodal/fisiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal/fisiología , Humanos , Masculino , Proteína Nodal/metabolismo , Neoplasias de la Próstata/embriología , Receptores Androgénicos/fisiología , Factor de Crecimiento Transformador beta/biosíntesis , Células Tumorales Cultivadas
20.
Stem Cells ; 28(8): 1303-14, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20549704

RESUMEN

Deregulation of stem cells is associated with the generation and progression of malignant tumors. In addition, genes that are associated with early embryogenesis are frequently expressed in cancer. Cripto-1 (CR-1), a glycosylphosphatidylinositol-linked glycoprotein, is expressed during early embryogenesis and in various human carcinomas. We demonstrated that human embryonal carcinoma (EC) cells are heterogeneous for CR-1 expression and consist of two distinct subpopulations: a CR-1(High) and a CR-1(Low) population. By segregating CR-1(High) and CR-1(Low) populations of NTERA2/D1 EC cells by fluorescence-activated cell sorting, we demonstrated that CR-1(High) cells were more tumorigenic than CR-1(Low) cells by an in vitro tumor sphere assay and by in vivo xenograft formation. The CR-1(High) population was enriched in mRNA expression for the pluripotent embryonic stem (ES) cell genes Oct4, Sox2, and Nanog. CR-1 expression in NTERA2/D1 cells was regulated by a Smad2/3-dependent autocrine loop, by the ES cell-related transcription factors Oct4/Nanog, and partially by the DNA methylation status of the promoter region. These results demonstrate that CR-1 expression is enriched in an undifferentiated, tumorigenic subpopulation and is regulated by key regulators of pluripotent stem cells.


Asunto(s)
Células Madre de Carcinoma Embrionario/citología , Células Madre de Carcinoma Embrionario/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Metilación de ADN , Factor de Crecimiento Epidérmico/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas Ligadas a GPI , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular , Glicoproteínas de Membrana/genética , Ratones , Ratones Desnudos , Proteína Homeótica Nanog , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA