Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Dev Biol ; 55(4-5): 477-82, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21732274

RESUMEN

Cancer stem cells (CSCs) or tumor initiating cells were identified and characterized as a unique subpopulation with stem cell features in many types of cancer. Current CSC studies provide novel insights regarding tumor initiation, progression, angiogenesis, resistance to therapy and interplay with the tumor micro-environment. A cancer stem cell niche has been proposed based on these findings. The niche provides the soil for CSC self-renewal and maintenance, stimulating essential signaling pathways in CSCs and leading to secretion of factors that promote angiogenesis and long term growth of CSCs. We present evidence which has emerged over the past 5 years indicating interaction of CSCs with angiogenesis in the proposed "vascular niche". Based on these findings, targeting the "cancer stem cell niche" by combining an individualized anti-CSC approach with treatment of their microenvironment may represent a novel therapeutic strategy against solid tumor systems.


Asunto(s)
Neoplasias/irrigación sanguínea , Neoplasias/patología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Animales , Humanos , Modelos Biológicos , Neoplasias/terapia , Transducción de Señal/fisiología , Nicho de Células Madre/patología
2.
Int J Gynecol Cancer ; 21(4): 717-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21543933

RESUMEN

BACKGROUND: The clinicopathological significance of the local spontaneous immune reaction in vulvar squamous cell carcinoma remains unclear. The purpose of this study was to clarify the role of the subtypes of tumor-infiltrating lymphocytes, both individually and synergistically. METHODS: Seventy-six patients with verified histopathological data and complete clinical history were included into the study. We collected 76 paraffin-embedded samples of the primary tumor. The presence of CD4+ and CD8+ T cells was evaluated by immunohistochemistry and compared with commonly recognized prognostic factors. The primary end point analyzed was the overall survival. RESULTS: CD4+ and CD8+ T cells were detected both within the nests of carcinoma and in the stroma, but only the infiltration within cancer cell nests was further analyzed. There was significant positive correlation (Spearman rho test R = 0.282, P = 0.014) between the number of intratumoral CD4+ and CD8+ T cells. No correlation was observed between the number of tumor-infiltrating CD4+ and CD8+ T cells and the patients' survival. Patients were classified into the following 4 groups (CD4+/CD8+, CD4⁻/CD8⁻ CD4+/CD8⁻, CD4⁻/CD8+), but none of them correlated with overall survival. CONCLUSIONS: These data support the statement that CD4⁻ and CD8+ T cells cooperate within cancer cell nests, but this spontaneous immune reaction is an individual feature not influencing the prognosis. Intratumoral CD4+ T cells might control or reflect the immune responses against cancer cells, whereas CD8+ T cells do not seem to work as sufficient effectors in tumor tissues.


Asunto(s)
Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Carcinoma de Células Escamosas/diagnóstico , Linfocitos Infiltrantes de Tumor/patología , Neoplasias de la Vulva/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Femenino , Humanos , Inmunohistoquímica , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/metabolismo , Persona de Mediana Edad , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Valor Predictivo de las Pruebas , Pronóstico , Nicho de Células Madre/inmunología , Nicho de Células Madre/patología , Análisis de Supervivencia , Neoplasias de la Vulva/inmunología , Neoplasias de la Vulva/mortalidad , Neoplasias de la Vulva/patología
3.
Glia ; 59(8): 1148-54, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21547954

RESUMEN

Neuro-oncology research has rediscovered a complexity of nervous system cancers through the incorporation of cellular heterogeneity into tumor models with cellular subsets displaying stem-cell characteristics. Self-renewing cancer stem cells (CSCs) can propagate tumors and yield nontumorigenic tumor bulk cells that display a more differentiated phenotype. The ability to prospectively isolate and interrogate CSCs is defining molecular mechanisms responsible for the tumor maintenance and growth. The clinical relevance of CSCs has been supported by their resistance to cytotoxic therapies and their promotion of tumor angiogenesis. Although the field of CSC biology is relatively young, continued elucidation of the features of these cells holds promise for the development of novel patient therapies. © 2011 Wiley-Liss, Inc.


Asunto(s)
Glioma/patología , Células Madre Neoplásicas/fisiología , Animales , Modelos Animales de Enfermedad , Glioma/etiología , Humanos , Ratones , Modelos Biológicos , Células Madre Neoplásicas/patología , Nicho de Células Madre/patología , Nicho de Células Madre/fisiopatología
4.
Haematologica ; 96(7): 1041-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21459792

RESUMEN

The field of hematopoietic oncology has traditionally focused on the study of hematopoietic cell autonomous genetic events in an effort to understand malignant transformation and develop therapeutics. Although highly rewarding in both aspects, this cell autonomous approach has failed to fully satisfy our need to understand tumor cell behavior and related clinical observations. In recent years, it has been increasingly recognized that the tumor microenvironment plays a pivotal role in cancer initiation and progression. This review will discuss recent experimental evidence in support of this view derived from investigations in both epithelial and hematopoietic systems. Based on this, conceptual views and therapeutic implications will be provided on the emerging role of the bone marrow microenvironment in leukemogenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Sistema Hematopoyético/metabolismo , Nicho de Células Madre/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Sistema Hematopoyético/patología , Humanos , Leucemia/metabolismo , Leucemia/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Nicho de Células Madre/patología , Microambiente Tumoral/genética
5.
Nat Rev Cancer ; 11(4): 278-88, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21412254

RESUMEN

Testicular germ cell tumours (TGCTs) of adults and adolescents are putatively derived from primordial germ cells or gonocytes. Recently reported genome-wide association studies implicate six gene loci that predispose to TGCT development. Remarkably, the functions of proteins encoded by genes within these regions bridge our understanding between the pathways involved in primordial germ cell physiology, male germ cell development and the molecular pathology of TGCTs. Furthermore, this improved understanding of the mechanisms underlying TGCT development and dissemination has clinical relevance for the management of patients with these tumours.


Asunto(s)
Predisposición Genética a la Enfermedad , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/patología , Nicho de Células Madre/patología , Neoplasias Testiculares/genética , Neoplasias Testiculares/patología , Adolescente , Adulto , Humanos , Masculino
6.
Ann Neurol ; 69(5): 878-91, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21391234

RESUMEN

OBJECTIVE: The subventricular zone (SVZ) of the brain constitutes a niche for neural stem and progenitor cells that can initiate repair after central nervous system (CNS) injury. In a relapsing-remitting model of experimental autoimmune encephalomyelitis (EAE), the neural stem cells (NSCs) become activated and initiate regeneration during acute disease, but lose this ability during the chronic phases of disease. We hypothesized that chronic microglia activation contributes to the failure of the NSC repair potential in the SVZ. METHODS: Using bromodeoxyuridine injections at different time points during EAE, we quantified the number of proliferating and differentiating progenitors, and evaluated the structure of the SVZ by electron microscopy. In vivo minocycline treatment during EAE was used to address the effect of microglia inactivation on SVZ dysfunction. RESULTS: In vivo treatment with minocycline, an inhibitor of microglia activation, increases stem cell proliferation in both naive and EAE animals. Minocycline treatment decreases cortical and periventricular pathology in the chronic phase of EAE, improving the proliferation of Sox2 stem cells and NG2 oligodendrocyte precursors cells originating in the SVZ and their differentiation into mature oligodendrocytes. INTERPRETATION: These data suggest that failure of repair observed during chronic EAE correlates with microglia activation and that treatments targeting chronic microglial activation have the potential for enhancing repair in the CNS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Células-Madre Neurales/fisiología , Nicho de Células Madre/patología , Animales , Antibacterianos/farmacología , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/fisiopatología , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Minociclina/farmacología , Esclerosis Múltiple , Proteína Proteolipídica de la Mielina/efectos adversos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/ultraestructura , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Fragmentos de Péptidos/efectos adversos , Prevención Secundaria , Nicho de Células Madre/efectos de los fármacos , Factores de Tiempo
7.
Nat Rev Neurosci ; 12(2): 88-104, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21248788

RESUMEN

Astrocyte-like cells, which act as stem cells in the adult brain, reside in a few restricted stem cell niches. However, following brain injury, glia outside these niches acquire or reactivate stem cell potential as part of reactive gliosis. Recent studies have begun to uncover the molecular pathways involved in this process. A comparison of molecular pathways activated after injury with those involved in the normal neural stem cell niches highlights strategies that could overcome the inhibition of neurogenesis outside the stem cell niche and instruct parenchymal glia towards a neurogenic fate. This new view on reactive glia therefore suggests a widespread endogenous source of cells with stem cell potential, which might potentially be harnessed for local repair strategies.


Asunto(s)
Astrocitos/patología , Lesiones Encefálicas/patología , Encéfalo/patología , Gliosis/patología , Células-Madre Neurales/patología , Neurogénesis , Nicho de Células Madre/patología , Adulto , Animales , Proliferación Celular , Humanos
8.
BMC Syst Biol ; 5: 4, 2011 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-21214945

RESUMEN

BACKGROUND: Multilevelness is a defining characteristic of complex systems. For example, in the intestinal tissue the epithelial lining is organized into crypts that are maintained by a niche of stem cells. The behavior of the system 'as a whole' is considered to emerge from the functioning and interactions of its parts. What we are seeking here is a conceptual framework to demonstrate how the "fate" of intestinal crypts is an emergent property that inherently arises from the complex yet robust underlying biology of stem cells. RESULTS: We establish a conceptual framework in which to formalize cross-level principles in the context of tissue organization. To this end we provide a definition for stemness, which is the propensity of a cell lineage to contribute to a tissue fate. We do not consider stemness a property of a cell but link it to the process in which a cell lineage contributes towards tissue (mal)function. We furthermore show that the only logically feasible relationship between the stemness of cell lineages and the emergent fate of their tissue, which satisfies the given criteria, is one of dominance from a particular lineage. CONCLUSIONS: The dominance theorem, conceived and proven in this paper, provides support for the concepts of niche succession and monoclonal conversion in intestinal crypts as bottom-up relations, while crypt fission is postulated to be a top-down principle.


Asunto(s)
Diferenciación Celular , Modelos Biológicos , Nicho de Células Madre , Neoplasias Colorrectales/patología , Humanos , Nicho de Células Madre/citología , Nicho de Células Madre/patología , Células Madre/citología , Células Madre/patología
9.
Blood ; 117(14): 3893-902, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21224474

RESUMEN

Angiogenesis alleviates hypoxic stress in ischemic tissues or during tumor progression. In addition to endothelial cell proliferation and migration, the angiogenic process requires bone marrow-derived cell (BMDC) recruitment to sites of neovascularization. However, the mechanism of communication between hypoxic tissues and the BM remains unknown. Using 2 models of hypoxia-induced angiogenesis (ischemic hindlimb surgery and subcutaneous tumor growth), we show that platelet infusion promotes BMDC mobilization into the circulation, BMDC recruitment into growing neovasculature, tumor vascularization, and blood flow restoration in ischemic limbs, whereas platelet depletion inhibits these effects. Thus, platelets are required for BMDC recruitment into ischemia-induced vasculature. Secretion of platelet α-granules, but neither dense granules nor platelet aggregation is crucial for BMDC homing and subsequent angiogenesis, as determined using VAMP-8(-/-), Pearl, and integrin Beta 3(-/-) platelets. Finally, platelets sequester tumor-derived promoters of angiogenesis and BMDC mobilization, which are counterbalanced by the antiangiogenic factor thrombospondin-1. A lack of thrombospondin-1 in platelets leads to an imbalance in proangiogenic and antiangiogenic factors and accelerates tumor growth and vascularization. Our data demonstrate that platelets stimulate BMDC homing in a VAMP-8-dependent manner, revealing a previously unknown role for platelets as key mediators between hypoxic tissues and the bone marrow during angiogenesis.


Asunto(s)
Plaquetas/metabolismo , Plaquetas/fisiología , Células de la Médula Ósea/fisiología , Movimiento Celular , Neovascularización Patológica/etiología , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Procesos de Crecimiento Celular/genética , Movimiento Celular/fisiología , Femenino , Hipoxia/patología , Hipoxia/fisiopatología , Integrina beta3/genética , Integrina beta3/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Estrés Oxidativo/fisiología , Proteínas R-SNARE/genética , Proteínas R-SNARE/fisiología , Nicho de Células Madre/metabolismo , Nicho de Células Madre/patología , Trombospondina 1/genética , Trombospondina 1/fisiología , Células Tumorales Cultivadas
10.
Cancer Res ; 71(4): 1465-73, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21159660

RESUMEN

Fes is a protein tyrosine kinase with cell autonomous oncogenic activities that are well established in cell culture and animal models, but its involvement in human cancer has been unclear. Abundant expression of Fes in vascular endothelial cells and myeloid cell lineages prompted us to explore roles for Fes in the tumor microenvironment. In an orthotopic mouse model of breast cancer, we found that loss of Fes in the host correlated with reductions in engrafted tumor growth rates, metastasis, and circulating tumor cells. The tumor microenvironment in Fes-deficient mice also showed reduced vascularity and fewer macrophages. In co-culture with tumor cells, Fes-deficient macrophages also poorly promoted tumor cell invasive behavior. Taken together, our observations argue that Fes inhibition might provide therapeutic benefits in breast cancer, in part by attenuating tumor-associated angiogenesis and the metastasis-promoting functions of tumor-associated macrophages.


Asunto(s)
Macrófagos/metabolismo , Neoplasias/patología , Células Neoplásicas Circulantes/metabolismo , Neovascularización Patológica/genética , Proteínas Proto-Oncogénicas c-fes/genética , Nicho de Células Madre/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular , Femenino , Humanos , Macrófagos/patología , Ratones , Ratones Noqueados , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/genética , Neoplasias/metabolismo , Células Neoplásicas Circulantes/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-fes/metabolismo , Nicho de Células Madre/patología , Trasplante Heterólogo , Células Tumorales Cultivadas
11.
Fertil Steril ; 95(1): 338-41, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20850729

RESUMEN

Expression of the pluripotency factors SOX-2, OCT-4, KLF-4, and NANOG was analyzed by quantitative real-time polymerase chain reaction, immunohistochemistry, and immunofluorescence microscopy in the endometrium, myometrium, and endometriotic tissue of 36 patients. Aberrant expression of SOX-2 may indicate a stem cell origin of endometriosis, whereas the presence of all progenitor markers in endometrial tissue marks the endometrium as a potential source for induced pluripotent stem cell generation.


Asunto(s)
Biomarcadores/metabolismo , Endometriosis , Endometrio/fisiología , Células Madre Pluripotentes/fisiología , Factores de Transcripción SOXB1/genética , Adulto , Endometriosis/genética , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/patología , Femenino , Expresión Génica/fisiología , Proteínas de Homeodominio/genética , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Persona de Mediana Edad , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/metabolismo , Nicho de Células Madre/patología , Nicho de Células Madre/fisiología , Células del Estroma/citología , Células del Estroma/fisiología
12.
J Theor Biol ; 269(1): 138-49, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-20969880

RESUMEN

Recent research in cancer biology has suggested the hypothesis that tumors are initiated and driven by a small group of cancer stem cells (CSCs). Furthermore, cancer stem cell niches have been found to be essential in determining fates of CSCs, and several signaling pathways have been proven to play a crucial role in cellular behavior, which could be two important factors in cancer development. To better understand the progression, heterogeneity and treatment response of breast cancer, especially in the context of CSCs, we propose a mathematical model based on the cell compartment method. In this model, three compartments of cellular subpopulations are constructed: CSCs, progenitor cells (PCs), and terminal differentiated cells (TCs). Moreover, (1) the cancer stem cell niche is, considered by modeling its effect on division patterns (symmetric or asymmetric) of CSCs, and (2) the EGFR signaling pathway is integrated by modeling its role in cell proliferation, apoptosis. Our simulation results indicate that (1) a higher probability for symmetric division of CSC may result in a faster expansion of tumor population, and for a larger number of niches, the tumor grows at a slower rate, but the final tumor volume is larger; (2) higher EGFR expression correlates to tumors with larger volumes while a saturation function is observed, and (3) treatments that inhibit tyrosine kinase activity of EGFR may not only repress the tumor volume, but also decrease the CSCs percentages by shifting CSCs from symmetric divisions to asymmetric divisions. These findings suggest that therapies should be designed to effectively control or eliminate the symmetric division of CSCs and to reduce or destroy the CSC niches.


Asunto(s)
Receptores ErbB/metabolismo , Modelos Biológicos , Neoplasias/patología , Neoplasias/terapia , Células Madre Neoplásicas/patología , Lesiones Precancerosas/patología , Nicho de Células Madre/patología , División Celular , Proliferación Celular , Simulación por Computador , Humanos , Recurrencia Local de Neoplasia/patología , Neoplasias/enzimología , Células Madre Neoplásicas/enzimología , Lesiones Precancerosas/enzimología , Lesiones Precancerosas/terapia , Transducción de Señal , Carga Tumoral
13.
Semin Oncol ; 37 Suppl 2: S2-14, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21111245

RESUMEN

Some of the most common cancer types, including breast cancer, prostate cancer, and lung cancer, show a predilection to metastasize to bone. The molecular basis of this preferential growth of cancer cells in the bone microenvironment has been an area of active investigation. Although the precise molecular mechanisms underlying this process remain to be elucidated, it is now increasingly being recognized that the unique characteristics of the bone niche provide homing signals to cancer cells, and create a microenvironment conducive for the cancer cells to colonize. Concomitantly, cancer cells release several regulatory factors that result in abnormal bone destruction and/or formation. This complex bidirectional interplay between tumor cells and bone microenvironment establishes a "vicious cycle" that leads to a selective growth advantage for the cancer cells. The molecular insights gained on the underpinnings of bone metastasis in recent years have also provided us with avenues to devise innovative approaches for therapeutic intervention. The goal of this review is to describe our current understanding of molecular pathophysiology of cancer metastases to bone, as well as its therapeutic implications.


Asunto(s)
Neoplasias Óseas/secundario , Carcinoma/secundario , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/patología , Nicho de Células Madre/patología , Microambiente Tumoral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Neoplasias Óseas/clasificación , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Remodelación Ósea/genética , Remodelación Ósea/fisiología , Huesos/metabolismo , Huesos/patología , Carcinoma/clasificación , Carcinoma/tratamiento farmacológico , Carcinoma/metabolismo , Humanos , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Nicho de Células Madre/metabolismo , Microambiente Tumoral/efectos de los fármacos
14.
Clin Cancer Res ; 16(24): 5928-35, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20962028

RESUMEN

Hypoxia is a common condition found in a wide range of solid tumors and is often associated with poor prognosis. Hypoxia increases tumor glycolysis, angiogenesis, and other survival responses, as well as invasion and metastasis by activating relevant gene expressions through hypoxia-inducible factors (HIF). HIF-1α and HIF-2α undergo oxygen-dependent regulation, and their overexpression is frequently associated with metastasis and poor clinical outcomes. Recent studies show that each step of the metastasis process, from the initial epithelial-mesenchymal transition to the ultimate organotropic colonization, can potentially be regulated by hypoxia, suggesting a master regulator role of hypoxia and HIFs in metastasis. Furthermore, modulation of cancer stem cell self-renewal by HIFs may also contribute to the hypoxia-regulated metastasis program. The hypoxia-induced metastatic phenotype may be one of the reasons for the modest efficacy of antiangiogenic therapies and may well explain the recent provocative findings that antiangiogenic therapy increased metastasis in preclinical models. Multiple approaches to targeting hypoxia and HIFs, including HIF inhibitors, hypoxia-activated bioreductive prodrugs, and gene therapies may become effective treatments to prevent or reduce metastasis.


Asunto(s)
Factor 1 Inducible por Hipoxia/fisiología , Hipoxia/complicaciones , Metástasis de la Neoplasia/genética , Animales , Antineoplásicos/uso terapéutico , Movimiento Celular/genética , Movimiento Celular/fisiología , Transición Epitelial-Mesenquimal/fisiología , Terapia Genética/métodos , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Hipoxia/patología , Hipoxia/fisiopatología , Factor 1 Inducible por Hipoxia/genética , Modelos Biológicos , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Nicho de Células Madre/metabolismo , Nicho de Células Madre/patología
15.
Rom J Morphol Embryol ; 51(3): 427-35, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20809016

RESUMEN

Similar to normal organs arising from normal stem cells, cancers can be viewed as organs composed of heterogeneous cellular populations arising from cancer cells with indefinite proliferation abilities. The continuous malignant progression is maintained by the proliferation of cancer stem cells and not the progeny that undergo limited proliferation before terminally differentiating. Effective therapy must eradicate malignant cells with unlimited clonogenic expansion within the primary tumor bulk. Thus, resolving both the specific cell of origin for prostate cancer and the interactions between the cells and the surrounding microenvironment within the cancer stem cell niche are crucial to appropriately define rational targets for therapeutic intervention and cure prostate cancer.


Asunto(s)
Comunicación Celular , Lesiones Precancerosas/patología , Neoplasias de la Próstata/patología , Resistencia a Antineoplásicos , Humanos , Masculino , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/patología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/embriología , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/terapia , Nicho de Células Madre/patología
16.
Clin Cancer Res ; 16(23): 5618-23, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20861166

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) is commonly caused by the overexpression of oncogenic transcription factors in developing T cells. In a mouse model of one such oncogene, LMO2, the cellular effect is to induce self-renewal of committed T cells in the thymus, which persist long-term while acquiring additional mutations and eventually giving rise to leukemia. These precancerous stem cells (pre-CSC) are intrinsically resistant to radiotherapy, implying that they may be refractory to conventional cancer therapies. However, they depend on an aberrantly expressed stem cell-like self-renewal program for their maintenance, in addition to a specialized thymic microenvironmental niche. Here, we discuss potential approaches for targeting pre-CSCs in T-ALL by using therapies directed at oncogenic transcription factors themselves, downstream self-renewal pathways, and the supportive cell niche.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Metaloproteínas/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Humanos , Proteínas con Dominio LIM , Metaloproteínas/genética , Metaloproteínas/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Nicho de Células Madre/metabolismo , Nicho de Células Madre/patología
17.
Cell Cycle ; 9(15): 3012-21, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20714216

RESUMEN

Glioblastoma, the most frequent and aggressive malignant brain tumor, has a very poor prognosis of approximately 1-year. The associated aggressive phenotype and therapeutic resistance of glioblastoma is postulated to be due to putative brain tumor stem-like cells (BTSC). The best hope for improved therapy lies in the ability to understand the molecular biology that controls BTSC behavior. The tumor vascular microenvironment of brain tumors has emerged as important regulators of BTSC behavior. Emerging data have identified the vascular microenvironment as home to a multitude of cell types engaged in various signaling that work collectively to foster a supportive environment for BTSCs. Characterization of the signaling pathways and intercellular communication between resident cell types in the microvascular niche of brain tumors is critical to the identification of potential BTSC-specific targets for therapy.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Progresión de la Enfermedad , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Espacio Extracelular/metabolismo , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Transducción de Señal , Nicho de Células Madre/metabolismo , Nicho de Células Madre/patología
18.
Semin Cancer Biol ; 20(3): 178-85, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20637871

RESUMEN

Poor prognosis of acute leukemia with current treatments is mainly due to the relapse of the disease following chemotherapy. In the last decade, an emerging concept has proposed that the leukemia stem cells (LSCs) and their interactions with the BM microenvironment are the major cause of the acute leukemia relapse. Adhesion to the stromal niche is crucial for LSCs as it directly supports self-renewal, proliferation, arrest of differentiation and protects from damaging chemo-agents. One of the key players in this crosstalk between leukemic cells and the BM stroma niche is the chemokine SDF-1. SDF-1 regulates the process of homing and engraftment of LSCs into the BM and inhibition of its receptor CXCR4 induces leukemic cell mobilization into the circulation. However, besides its chemotactic and adhesive functions, SDF-1 is also a pleiotropic cytokine that regulates leukemic cell proliferation as well as their program of differentiation. CXCR4 antagonists are used in combination with chemotherapy in preclinical and clinical studies, which demonstrate that blocking CXCR4 is a novel promising approach of therapy. In this review, we focus on the multifaceted SDF-1/CXCR4 axis in acute leukemia and discuss how targeting this pathway could provide potential interest to eradicate the LSCs.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Quimiocina CXCL12/antagonistas & inhibidores , Leucemia/tratamiento farmacológico , Receptores CXCR4/antagonistas & inhibidores , Enfermedad Aguda , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Médula Ósea/patología , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/fisiología , Humanos , Leucemia/metabolismo , Modelos Biológicos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Receptores CXCR4/metabolismo , Receptores CXCR4/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/metabolismo , Nicho de Células Madre/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología
19.
Tissue Eng Part C Methods ; 16(6): 1523-31, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20586611

RESUMEN

Human mesenchymal stem cells form the supportive structure in which the functional cells of a differentiated tissue reside. We describe the creation of ectopic niches within polyurethane scaffolds coated with human mesenchymal stem cells. When implanted subcutaneously in NOD/SCID mice, these niches supported engraftment of primary human acute myeloid leukemia cells. The scaffolds showed vascularization and presence of osteoclasts and adipocytes, suggestive of an organizing human bone marrow microenvironment in the murine host. The chemokine stromal-derived factor-1 (SDF-1 or CXCL12) and its receptor CXCR4 are critical for homing and migration of acute myeloid leukemia. We found that a CXCR4 antagonist could disrupt homing to the ectopic niches, possibly by modulation of the mesenchymal stroma. We believe that these scaffold niches provide a new and powerful tool to study the leukemia stem cell microenvironment and may be useful for identification of novel drug targets.


Asunto(s)
Coristoma/patología , Leucemia/patología , Células Madre Mesenquimatosas/citología , Nicho de Células Madre/patología , Andamios del Tejido , Animales , Células Cultivadas , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Modelos Animales de Enfermedad , Humanos , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/patología , Nicho de Células Madre/citología , Andamios del Tejido/química , Trasplante Heterólogo , Microambiente Tumoral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...