Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Proc Natl Acad Sci U S A ; 105(11): 4329-34, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18334633

RESUMEN

Embryonic stem cells sustain a microenvironment that facilitates a balance of self-renewal and differentiation. Aggressive cancer cells, expressing a multipotent, embryonic cell-like phenotype, engage in a dynamic reciprocity with a microenvironment that promotes plasticity and tumorigenicity. However, the cancer-associated milieu lacks the appropriate regulatory mechanisms to maintain a normal cellular phenotype. Previous work from our laboratory reported that aggressive melanoma and breast carcinoma express the embryonic morphogen Nodal, which is essential for human embryonic stem cell (hESC) pluripotency. Based on the aberrant expression of this embryonic plasticity gene by tumor cells, this current study tested whether these cells could respond to regulatory cues controlling the Nodal signaling pathway, which might be sequestered within the microenvironment of hESCs, resulting in the suppression of the tumorigenic phenotype. Specifically, we discovered that metastatic tumor cells do not express the inhibitor to Nodal, Lefty, allowing them to overexpress this embryonic morphogen in an unregulated manner. However, exposure of the tumor cells to a hESC microenvironment (containing Lefty) leads to a dramatic down-regulation in their Nodal expression concomitant with a reduction in clonogenicity and tumorigenesis accompanied by an increase in apoptosis. Furthermore, this ability to suppress the tumorigenic phenotype is directly associated with the secretion of Lefty, exclusive to hESCs, because it is not detected in other stem cell types, normal cell types, or trophoblasts. The tumor-suppressive effects of the hESC microenvironment, by neutralizing the expression of Nodal in aggressive tumor cells, provide previously unexplored therapeutic modalities for cancer treatment.


Asunto(s)
Células Madre Embrionarias/metabolismo , Neoplasias/genética , Neoplasias/patología , Técnicas de Cultivo de Célula , Células Cultivadas , Humanos , Proteína Nodal , Fenotipo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Ensayo de Tumor de Célula Madre
9.
J Cell Physiol ; 225(2): 390-3, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20568225

RESUMEN

Studies are beginning to emerge that demonstrate intriguing differences between human-induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs). Here, we investigated the expression of key members of the Nodal embryonic signaling pathway, critical to the maintenance of pluripotency in hESCs. Western blot and real-time RT-PCR analyses reveal slightly lower levels of Nodal (a TGF-beta family member) and Cripto-1 (Nodal's co-receptor) and a dramatic decrease in Lefty (Nodal's inhibitor and TGF-beta family member) in hiPSCs compared with hESCs. The noteworthy drop in hiPSC's Lefty expression correlated with an increase in the methylation of Lefty B CpG island. Based on these findings, we addressed a more fundamental question related to the consequences of epigenetically reprogramming hiPSCs, especially with respect to maintaining a stable ESC phenotype. A global comparative analysis of 365 microRNAs (miRs) in two hiPSC versus four hESC lines ultimately identified 10 highly expressed miRs in hiPCSs with >10-fold difference, which have been shown to be cancer related. These data demonstrate cancer hallmarks expressed by hiPSCs, which will require further assessment for their impact on future therapies..


Asunto(s)
Biomarcadores de Tumor/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/metabolismo , Biomarcadores de Tumor/genética , Western Blotting , Línea Celular , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Pluripotentes/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Cancers (Basel) ; 11(3)2019 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-30857267

RESUMEN

Aggressive cancer cells are characterized by their capacity to proliferate indefinitely and to propagate a heterogeneous tumor comprised of subpopulations with varying degrees of metastatic propensity and drug resistance properties. Particularly daunting is the challenge we face in the field of oncology of effectively targeting heterogeneous tumor cells expressing a variety of markers, especially those associated with a stem cell phenotype. This dilemma is especially relevant in breast cancer, where therapy is based on traditional classification schemes, including histological criteria, differentiation status, and classical receptor markers. However, not all patients respond in a similar manner to standard-of-care therapy, thereby necessitating the need to identify and evaluate novel biomarkers associated with the difficult-to-target stem cell phenotype and drug resistance. Findings related to the convergence of embryonic and tumorigenic signaling pathways have identified the embryonic morphogen Nodal as a promising new oncofetal target that is reactivated only in aggressive cancers, but not in normal tissues. The work presented in this paper confirms previous studies demonstrating the importance of Nodal as a cancer stem cell molecule associated with aggressive breast cancer, and advances the field by providing new findings showing that Nodal is not targeted by standard-of-care therapy in breast cancer patients. Most noteworthy is the linkage found between Nodal expression and the drug resistance marker ATP-binding cassette member 1 (ABCA1), which may provide new insights into developing combinatorial approaches to overcome drug resistance and disease recurrence.

11.
Stem Cell Rev ; 3(1): 68-78, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17873384

RESUMEN

As our understanding of embryonic stem cell biology becomes more sophisticated, the similarities between multipotent cancer cells and these totipotent precursors are increasingly striking. Both multipotent cancer cells and embryonic stem cells possess the ability to self-renew, epigenetically alter their neighboring cellular architecture, and populate a tissue mass with a phenotypically heterogeneous composition of cells. While the molecular signature of these cell types continues to be elucidated, new insights are emerging related to the convergence of embryonic and tumorigenic signaling pathways. Understanding the molecular underpinnings of these two stem cell phenotypes may lead to new therapeutic targets for the elusive cancer cell. While still in its infancy, the potential of adapting embryonic stem cells, and more specifically the factors they produce, is enormous for clinical application. Here we outline evidence that demonstrates the inductive influence of embryonic stem cells and their microenvironment to reprogram cancer cells to exhibit a more benign phenotype, with profound implications for differentiation therapy.


Asunto(s)
Células Madre Embrionarias/fisiología , Neoplasias/terapia , Células Madre Neoplásicas/patología , Transducción de Señal/fisiología , Animales , Reprogramación Celular/fisiología , Embrión no Mamífero , Marcación de Gen , Humanos , Modelos Biológicos , Células Madre Multipotentes/citología , Invasividad Neoplásica , Neoplasias/patología , Pez Cebra
12.
Cancer Res ; 65(21): 9851-60, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16267008

RESUMEN

Malignant melanoma continues to remain a significant health threat, with death often occurring as a result of metastasis. The metastatic phenotype typically is characterized by augmented tumor cell invasion and migration in addition to tumor cell plasticity as shown by vasculogenic mimicry. Therefore, understanding the molecular mechanisms that promote an aggressive phenotype is essential to predicting the likelihood of metastasis at a stage when intervention may be possible. This study focuses on the role of focal adhesion kinase (FAK), a cytoplasmic tyrosine kinase important for many cellular processes, including cell survival, invasion, and migration. We found FAK to be phosphorylated on its key tyrosine residues, Tyr397 and Tyr576, in only aggressive uveal and cutaneous melanoma cells, which correlates with their increased invasion, migration, and vasculogenic mimicry plasticity. Additionally, we confirmed the presence of FAK phosphorylated on Tyr397 and Tyr576 in both cutaneous and uveal melanoma tumors in situ. Examination of a functional role for FAK in aggressive melanoma revealed that disruption of FAK-mediated signal transduction pathways, through the expression of FAK-related nonkinase (FRNK), results in a decrease in melanoma cell invasion, migration, and inhibition of vasculogenic mimicry. Moreover, we found that FRNK expression resulted in a down-regulation of Erk1/2 phosphorylation resulting in a decrease in urokinase activity. Collectively, these data suggest a new mechanism involved in promoting the aggressive melanoma phenotype through FAK-mediated signal transduction pathways, thus providing new insights into possible therapeutic intervention strategies.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Melanoma/enzimología , Melanoma/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasas de la Matriz Asociadas a la Membrana , Melanoma/genética , Metaloendopeptidasas/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Transfección , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Neoplasias de la Úvea/enzimología , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/patología
13.
Cancer Res ; 65(22): 10164-9, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16288000

RESUMEN

The clinical management of cutaneous melanoma would benefit significantly from a better understanding of the molecular changes that occur during melanocytic progression to a melanoma phenotype. To gain unique insights into this process, we developed a three-dimensional in vitro model that allows observations of normal human melanocytes interacting with a metastatic melanoma matrix to determine whether these normal cells could be reprogrammed by inductive cues in the tumor cell microenvironment. The results show the epigenetic transdifferentiation of the normal melanocytic phenotype to that of an aggressive melanoma-like cell with commensurate increased migratory and invasive ability with no detectable genomic alterations. Removal of the transdifferentiated melanocytes from the inductive metastatic melanoma microenvironment results in a reversion to their normal phenotype. However, a normal melanocyte microenvironment had no epigenetic influence on the phenotype of metastatic melanoma cells. This novel approach identifies specific genes involved in the transdifferentiation of melanocytes to a more aggressive phenotype, which may offer significant therapeutic value.


Asunto(s)
Transformación Celular Neoplásica/genética , Melanocitos/patología , Melanocitos/fisiología , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Diferenciación Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/secundario , Familia de Multigenes , Hibridación de Ácido Nucleico , Neoplasias Cutáneas/secundario
14.
Curr Mol Biol Rep ; 3(3): 159-164, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29152453

RESUMEN

PURPOSE OF REVIEW: Cancer is a major public health problem worldwide. In aggressive cancers, which are heterogeneous in nature, there exists a paucity of targetable molecules that can be used to predict outcome and response to therapy in patients, especially those in the high risk category with a propensity to relapse following chemotherapy. This review addresses the challenges pertinent to treating aggressive cancer cells with inherent stem cell properties, with a special focus on triple-negative breast cancer (TNBC). RECENT FINDINGS: Plasticity underlies the cancer stem cell (CSC) phenotype in aggressive cancers like TNBC. Progenitors and CSCs implement similar signaling pathways to sustain growth, and the convergence of embryonic and tumorigenic signaling pathways has led to the discovery of novel oncofetal targets, rigorously regulated during normal development, but aberrantly reactivated in aggressive forms of cancer. SUMMARY: Translational studies have shown that Nodal, an embryonic morphogen, is reactivated in aggressive cancers, but not in normal tissues, and underlies tumor growth, invasion, metastasis and drug resistance. Front-line therapies do not inhibit Nodal, but when a combinatorial approach is used with an agent such as doxorubicin followed by anti-Nodal antibody therapy, significant decreases in cell growth and viability occur. These findings are of special interest in the development of new therapeutic interventions that target the stem cell properties of cancer cells to overcome drug resistance and metastasis.

15.
Cancer Biol Ther ; 5(2): 228-33, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16481735

RESUMEN

The formation of matrix-rich, vasculogenic-like networks, termed vasculogenic mimicry (VM), is a unique process characteristic of highly aggressive melanoma cells found to express genes previously thought to be exclusively associated with endothelial and epithelial cells. This study contributes new observations demonstrating that VE-cadherin can regulate the expression of EphA2 at the cell membrane by mediating its ability to become phosphorylated through interactions with its membrane bound ligand, ephrin-A1. VE-cadherin and EphA2 were also found to be colocalized in cell-cell adhesion junctions, both in vitro and in vivo. Immunoprecipitation studies revealed that EphA2 and VE-cadherin could interact, directly and/or indirectly, during VM. Furthermore, there was no change in the colocalization of EphA2 and VE-cadherin at cell-cell adhesion sites when EphA2 was phosphorylated on tyrosine residues. Although transient knockout of EphA2 expression did not alter VE-cadherin localization, transient knockout of VE-cadherin expression resulted in the reorganization of EphA2 on the cells' surface, an accumulation of EphA2 in the cytoplasm, and subsequent dephosphorylation of EphA2. Collectively, these results suggest that VE-cadherin and EphA2 act in a coordinated manner as a key regulatory element in the process of melanoma VM and illuminate a novel signaling pathway that could be potentially exploited for therapeutic intervention.


Asunto(s)
Cadherinas/metabolismo , Melanoma/irrigación sanguínea , Neovascularización Patológica/metabolismo , Receptor EphA2/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Antígenos CD , Cadherinas/análisis , Adhesión Celular , Membrana Celular/química , Membrana Celular/metabolismo , Citoplasma/química , Citoplasma/metabolismo , Efrina-A1/metabolismo , Humanos , Uniones Intercelulares/química , Uniones Intercelulares/metabolismo , Melanoma/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacología , Fosforilación , Receptor EphA2/análisis , Receptor EphA2/genética , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Células Tumorales Cultivadas , Tirosina/metabolismo
16.
Cancer Res ; 63(16): 4757-62, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12941789

RESUMEN

Vasculogenic mimicry (VM) describes the unique ability of highly aggressive melanoma tumor cells to express endothelial cell-associated genes (such as EphA2 and VE-cadherin) and form vasculogenic-like networks when cultured on a three-dimensional matrix. VM has been described in several types of aggressive tumors, including melanoma, prostate, breast, and ovarian carcinomas. However, the molecular underpinnings of this phenomenon remain somewhat elusive. In this study, we examined key molecular mechanisms underlying VM in aggressive human cutaneous and uveal melanoma. The data reveal that phosphoinositide 3-kinase (PI3K) is an important regulator of VM, specifically affecting membrane type 1 matrix metalloproteinase (MT1-MMP) and matrix metalloproteinase-2 (MMP-2) activity, critical in the formation of vasculogenic-like networks. Using specific inhibitors of PI3K, melanoma VM was abrogated coincident with decreased MMP-2 and MT1-MMP activity. Furthermore, inhibition of PI3K blocked the cleavage of laminin 5 gamma 2 chain, resulting in decreased levels of the gamma 2' and gamma 2x promigratory fragments. Collectively, these results indicate that PI3K is an important regulator of melanoma VM directly affecting the cooperative interactions of MMP-2, MT1-MMP, and laminin 5 gamma 2 chain and, thus, the remodeling of the tumor cell microenvironment. PI3K may represent an excellent target for therapeutic intervention of a novel signaling cascade underlying VM.


Asunto(s)
Metaloproteinasa 2 de la Matriz/metabolismo , Melanoma/irrigación sanguínea , Melanoma/enzimología , Metaloendopeptidasas/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Humanos , Laminina/metabolismo , Metaloproteinasas de la Matriz Asociadas a la Membrana , Melanoma/patología , Subunidades de Proteína , Células Tumorales Cultivadas
17.
Cancer Res ; 63(17): 5381-9, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500372

RESUMEN

Vasculogenic mimicry (VM), the formation of matrix-rich vascular-like networks in three-dimensional culture corresponding with the expression of vascular cell-associated genes, and the lining of matrix-rich networks in situ, has been observed in highly aggressive and malignant melanoma. However, little is known about the molecular underpinnings of this phenomenon. On the basis of gene profiling, protein detection, and immunohistochemistry, aggressive relative to poorly aggressive melanoma showed up-regulation of tissue factor (TF), TF pathway inhibitor 1 (TFPI-1) and 2 (TFPI-2), critical genes that initiate and regulate the coagulation pathways. The procoagulant function of TF on highly aggressive melanoma is shown to be regulated by TFPI-1 but not by TFPI-2. Thus, aggressive melanoma exhibits endothelial cell-like anticoagulant mechanisms that may contribute to the fluid-conducting potential of melanoma cell-lined networks, as studied by correlative in vivo Doppler flow measurements. Antibody inhibition experiments reveal that TFPI-2 is required for VM in vitro, but plasmin is an unlikely target protease of TFPI-2. Blockade of TFPI-2 suppressed matrix metalloproteinase-2 activation, and, therefore, TFPI-2 appears to regulate an essential pathway of VM. TFPI-2 is synthesized by endothelial and tumor cells, which deposit TFPI-2 into extracellular matrices. Culturing poorly aggressive melanoma cells on three-dimensional matrix containing recombinant TFPI-2 produces some of the phenotypic changes associated with aggressive, vasculogenic melanoma cells. Thus, TFPI-2 contributes to VM plasticity, whereas TFPI-1 has anticoagulant functions of relevance for perfusion of VM channels formed by TF-expressing melanoma cells.


Asunto(s)
Glicoproteínas/fisiología , Lipoproteínas/fisiología , Melanoma/irrigación sanguínea , Animales , Células CHO , Cricetinae , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Humanos , Lipoproteínas/biosíntesis , Lipoproteínas/genética , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Tromboplastina/antagonistas & inhibidores , Tromboplastina/fisiología , Trasplante Heterólogo , Neoplasias de la Úvea/irrigación sanguínea , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/patología
18.
Cancer Res ; 62(3): 665-8, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11830517

RESUMEN

On the basis of the ability of aggressive melanoma cells to participate in vasculogenic mimicry, particularly their expression of endothelial-associated genes, we examined the plasticity of human metastatic cutaneous melanoma cells with respect to vascular function. Fluorescently labeled metastatic melanoma cells were challenged to an ischemic microenvironment surgically induced in the hind limbs of nude mice. The data reveal the capability of these melanoma cells to express cell-fate determination molecules, normally expressed during embryonic vasculogenesis, and to participate in the neovascularization of circulation-deficient muscle. These results demonstrate the powerful influence of the microenvironment on the transendothelial differentiation of aggressive melanoma cells, and may provide new perspectives on tumor cell plasticity that could be exploited for novel therapeutic strategies.


Asunto(s)
Endotelio Vascular/patología , Melanoma/secundario , Neovascularización Patológica/patología , Neoplasias Cutáneas/secundario , Animales , Diferenciación Celular/fisiología , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/patología , Melanoma/irrigación sanguínea , Melanoma/patología , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Trasplante Heterólogo
19.
Pharmacol Ther ; 159: 83-92, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26808163

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 the journal's distinction of a "citation classic" (Maniotis et al., 1999). Tumor cell vasculogenic mimicry (VM), also known as vascular mimicry, describes the plasticity of aggressive cancer cells forming de novo vascular networks and is associated with the malignant phenotype and poor clinical outcome. The tumor cells capable of VM share the commonality of a stem cell-like, transendothelial phenotype, which may be induced by hypoxia. Since its introduction 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. Of special significance is the lack of effectiveness of angiogenesis inhibitors on tumor cell VM, suggesting a selective resistance by this phenotype to conventional therapy. Facilitating the functional plasticity of tumor cell VM are key proteins associated with vascular, stem cell, extracellular matrix, and hypoxia-related signaling pathways--each deserving serious consideration as potential therapeutic targets and diagnostic indicators of the aggressive, metastatic phenotype. This review highlights seminal findings pertinent to VM, including the effects of a novel, small molecular compound, CVM-1118, currently under clinical development to target VM, and illuminates important molecular pathways involved in the suppression of this plastic, aggressive phenotype, using melanoma as a model.


Asunto(s)
Melanoma/patología , Animales , Plasticidad de la Célula , Humanos , Neovascularización Patológica , Transducción de Señal
20.
Nat Commun ; 7: 13322, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27827359

RESUMEN

Small cell lung cancer (SCLC) is characterized by prevalent circulating tumour cells (CTCs), early metastasis and poor prognosis. We show that SCLC patients (37/38) have rare CTC subpopulations co-expressing vascular endothelial-cadherin (VE-cadherin) and cytokeratins consistent with vasculogenic mimicry (VM), a process whereby tumour cells form 'endothelial-like' vessels. Single-cell genomic analysis reveals characteristic SCLC genomic changes in both VE-cadherin-positive and -negative CTCs. Higher levels of VM are associated with worse overall survival in 41 limited-stage patients' biopsies (P<0.025). VM vessels are also observed in 9/10 CTC patient-derived explants (CDX), where molecular analysis of fractionated VE-cadherin-positive cells uncovered copy-number alterations and mutated TP53, confirming human tumour origin. VE-cadherin is required for VM in NCI-H446 SCLC xenografts, where VM decreases tumour latency and, despite increased cisplatin intra-tumour delivery, decreases cisplatin efficacy. The functional significance of VM in SCLC suggests VM regulation may provide new targets for therapeutic intervention.


Asunto(s)
Variaciones en el Número de Copia de ADN , Neoplasias Pulmonares/patología , Células Neoplásicas Circulantes/metabolismo , Neovascularización Patológica/patología , Carcinoma Pulmonar de Células Pequeñas/patología , Animales , Antígenos CD/metabolismo , Biopsia , Cadherinas/metabolismo , Línea Celular Tumoral , Estudios de Cohortes , Femenino , Humanos , Queratinas/metabolismo , Pulmón/patología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Masculino , Ratones , Persona de Mediana Edad , Mutación , Neovascularización Patológica/genética , Análisis de la Célula Individual , Carcinoma Pulmonar de Células Pequeñas/irrigación sanguínea , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/mortalidad , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA