Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Genet ; 17(8): e1009094, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34398873

RESUMEN

The systematic identification of genetic events driving cellular transformation and tumor progression in the absence of a highly recurrent oncogenic driver mutation is a challenge in cutaneous oncology. In cutaneous squamous cell carcinoma (cuSCC), the high UV-induced mutational burden poses a hurdle to achieve a complete molecular landscape of this disease. Here, we utilized the Sleeping Beauty transposon mutagenesis system to statistically define drivers of keratinocyte transformation and cuSCC progression in vivo in the absence of UV-IR, and identified both known tumor suppressor genes and novel oncogenic drivers of cuSCC. Functional analysis confirms an oncogenic role for the ZMIZ genes, and tumor suppressive roles for KMT2C, CREBBP and NCOA2, in the initiation or progression of human cuSCC. Taken together, our in vivo screen demonstrates an extremely heterogeneous genetic landscape of cuSCC initiation and progression, which can be harnessed to better understand skin oncogenic etiology and prioritize therapeutic candidates.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Transformación Celular Neoplásica/genética , Queratinocitos/patología , Mutagénesis Insercional/métodos , Análisis de Secuencia de ADN/métodos , Neoplasias Cutáneas/genética , Proteína de Unión a CREB/genética , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica/patología , Elementos Transponibles de ADN , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Coactivador 2 del Receptor Nuclear/genética , Neoplasias Cutáneas/patología
2.
Nat Biotechnol ; 34(9): 962-72, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27479497

RESUMEN

A central challenge in oncology is how to kill tumors containing heterogeneous cell populations defined by different combinations of mutated genes. Identifying these mutated genes and understanding how they cooperate requires single-cell analysis, but current single-cell analytic methods, such as PCR-based strategies or whole-exome sequencing, are biased, lack sequencing depth or are cost prohibitive. Transposon-based mutagenesis allows the identification of early cancer drivers, but current sequencing methods have limitations that prevent single-cell analysis. We report a liquid-phase, capture-based sequencing and bioinformatics pipeline, Sleeping Beauty (SB) capture hybridization sequencing (SBCapSeq), that facilitates sequencing of transposon insertion sites from single tumor cells in a SB mouse model of myeloid leukemia (ML). SBCapSeq analysis of just 26 cells from one tumor revealed the tumor's major clonal subpopulations, enabled detection of clonal insertion events not detected by other sequencing methods and led to the identification of dominant subclones, each containing a unique pair of interacting gene drivers along with three to six cooperating cancer genes with SB-driven expression changes.


Asunto(s)
ADN de Neoplasias/genética , Genes Relacionados con las Neoplasias/genética , Hibridación in Situ/métodos , Leucemia Mieloide/genética , Mutagénesis Insercional/genética , Análisis de Secuencia de ADN/métodos , Algoritmos , Animales , Biomarcadores de Tumor/genética , Elementos Transponibles de ADN , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Leucemia Mieloide/patología , Masculino , Ratones , Proteínas de Neoplasias/genética , Programas Informáticos , Transposasas/genética
3.
Mol Ther ; 24(1): 34-40, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26316391

RESUMEN

Progression of many cancers is associated with tumor infiltration by mesenchymal stromal cells (MSC). Adipose stromal cells (ASC) are MSC that serve as adipocyte progenitors and endothelium-supporting cells in white adipose tissue (WAT). Clinical and animal model studies indicate that ASC mobilized from WAT are recruited by tumors. Direct evidence for ASC function in tumor microenvironment has been lacking due to unavailability of approaches to specifically inactivate these cells. Here, we investigate the effects of a proteolysis-resistant targeted hunter-killer peptide D-WAT composed of a cyclic domain CSWKYWFGEC homing to ASC and of a proapoptotic domain KLAKLAK2. Using mouse bone marrow transplantation models, we show that D-WAT treatment specifically depletes tumor stromal and perivascular cells without directly killing malignant cells or tumor-infiltrating leukocytes. In several mouse carcinoma models, targeted ASC cytoablation reduced tumor vascularity and cell proliferation resulting in hemorrhaging, necrosis, and suppressed tumor growth. We also validated a D-WAT derivative with a proapoptotic domain KFAKFAK2 that was found to have an improved cytoablative activity. Our results for the first time demonstrate that ASC, recruited as a component of tumor microenvironment, support cancer progression. We propose that drugs targeting ASC can be developed as a combination therapy complementing conventional cancer treatments.


Asunto(s)
Tejido Adiposo/citología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Células Madre Mesenquimatosas/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Péptidos/administración & dosificación , Animales , Trasplante de Médula Ósea , Carcinoma Pulmonar de Lewis/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Melanoma Experimental/patología , Células Madre Mesenquimatosas/citología , Ratones , Péptidos/farmacología , Microambiente Tumoral/efectos de los fármacos
4.
Nat Commun ; 4: 2472, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24045463

RESUMEN

The presence of brown adipose tissue responsible for thermogenic energy dissipation has been revealed in adult humans and has high clinical importance. Owing to limitations of current methods for brown adipose tissue detection, analysing the abundance and localization of brown adipose tissue in the body has remained challenging. Here we screen a combinatorial peptide library in mice and characterize a peptide (with the sequence CPATAERPC) that selectively binds to the vascular endothelium of brown adipose tissue, but not of intraperitoneal white adipose tissue. We show that in addition to brown adipose tissue, this peptide probe also recognizes the vasculature of brown adipose tissue-like depots of subcutaneous white adipose tissue. Our results indicate that the CPATAERPC peptide localizes to brown adipose tissue even in the absence of sympathetic nervous system stimulation. Finally, we demonstrate that this probe can be used to identify brown adipose tissue depots in mice by whole-body near-infrared fluorescence imaging.


Asunto(s)
Tejido Adiposo Pardo/ultraestructura , Endotelio Vascular/ultraestructura , Sondas Moleculares/metabolismo , Imagen Óptica/métodos , Péptidos/metabolismo , Imagen de Cuerpo Entero/métodos , Tejido Adiposo Pardo/metabolismo , Animales , Endotelio Vascular/metabolismo , Masculino , Ratones , Sondas Moleculares/química , Biblioteca de Péptidos , Péptidos/química , Grasa Subcutánea/metabolismo , Grasa Subcutánea/ultraestructura
5.
Cancer Res ; 72(20): 5198-208, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23071132

RESUMEN

Epidemiologic studies associate cancer with obesity, but the pathophysiologic connections remain obscure. In this study, we show that obesity facilitates tumor growth in mice irrespective of concurrent diet, suggesting a direct effect of excess white adipose tissue (WAT). When transplanted into mice, adipose stromal cells (ASC) can serve as perivascular adipocyte progenitors that promote tumor growth, perhaps helping explain the obesity-cancer link. In developing this hypothesis, we showed that ASCs are expanded in obesity and that they traffic from endogenous WAT to tumors in several mouse models of cancer. Strikingly, a comparison of circulating and tumor-infiltrating cell populations in lean, and obese mice revealed that cancer induces a six-fold increase of ASC frequency in the systemic circulation. We obtained evidence that ASCs mobilized in this way can be recruited into tumors, where they can be incorporated into blood vessels as pericytes and they can differentiate into adipocytes in an obesity-dependent manner. Extending this evidence, we found that increased tumor vascularization (reflected by changes in tumor vascular morphology and a two-fold increase in vascular density) was associated with intratumoral adipocytes and elevated proliferation of neighboring malignant cells. Taken together, our results suggest that ASCs recruited from endogenous adipose tissue can be recruited by tumors to potentiate the supportive properties of the tumor microenvironment.


Asunto(s)
Adipocitos/patología , Tejido Adiposo/patología , Pericitos/patología , Células Madre/patología , Células del Estroma/patología , Microambiente Tumoral , Animales , Trasplante de Médula Ósea , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Obesidad/patología
6.
Clin Cancer Res ; 18(3): 771-82, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22167410

RESUMEN

PURPOSE: Adipose tissue contains a population of tumor-tropic mesenchymal progenitors, termed adipose stromal cells (ASC), which engraft in neighboring tumors to form supportive tumor stroma. We hypothesized that intra-abdominal visceral adipose tissue may contain a uniquely tumor-promoting population of ASC to account for the relationship between excess visceral adipose tissue and mortality of intra-abdominal cancers. EXPERIMENTAL DESIGN: To investigate this, we isolated and characterized ASC from intra-abdominal omental adipose tissue (O-ASC) and characterized their effects on endometrial cancer progression as compared with subcutaneous adipose-derived mesenchymal stromal cells (SC-ASC), bone marrow-derived mesenchymal stromal cells (BM-MSC), and lung fibroblasts. To model chronic recruitment of ASC by tumors, cells were injected metronomically into mice bearing Hec1a xenografts. RESULTS: O-ASC expressed cell surface markers characteristic of BM-MSC and differentiated into mesenchymal lineages. Coculture with O-ASC increased endometrial cancer cell proliferation in vitro. Tumor tropism of O-ASC and SC-ASC for human Hec1a endometrial tumor xenografts was comparable, but O-ASC more potently promoted tumor growth. Compared with tumors in SC-ASC-injected mice, tumors in O-ASC-injected mice contained higher numbers of large tortuous desmin-positive blood vessels, which correlated with decreased central tumor necrosis and increased tumor cell proliferation. O-ASC exhibited enhanced motility as compared with SC-ASC in response to Hec1a-secreted factors. CONCLUSIONS: Visceral adipose tissue contains a population of multipotent MSCs that promote endometrial tumor growth more potently than MSCs from subcutaneous adipose tissue. We propose that O-ASCs recruited to tumors express specific factors that enhance tumor vascularization, promoting survival and proliferation of tumor cells.


Asunto(s)
Tejido Adiposo/citología , Neoplasias Endometriales/patología , Células Madre Mesenquimatosas/citología , Epiplón/citología , Células del Estroma/citología , Animales , Diferenciación Celular , Femenino , Citometría de Flujo , Humanos , Ratones , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Grasa Subcutánea/citología , Trasplante Heterólogo
7.
Cell Stem Cell ; 9(1): 74-86, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21683670

RESUMEN

Adipose stromal cells (ASCs) serve as mesenchymal progenitors in white adipose tissue (WAT). Intercellular interactions involving ASCs have remained obscure. By merging phage display technology with fluorescence-activated cell sorting (FACS), we screened a combinatorial library for peptides that target mouse ASCs in vivo. We isolated peptide CSWKYWFGEC that specifically homes to ASCs, used it as bait to purify the corresponding ASC surface receptor, and identified it as a previously unreported cleavage product of decorin (DCN) lacking the glycanation site (termed ΔDCN). We demonstrate that ΔDCN is differentially expressed on ASC surface. In a screen for ΔDCN-binding proteins, we identified resistin, an adipokine for which the receptor has been unknown. Expression of ΔDCN in 3T3-L1 cells promoted proliferation and migration but suppressed lipid accumulation upon adipogenesis induction, which was resistin dependent. We conclude that ΔDCN serves as a functional receptor of resistin in adipocyte progenitors and may regulate WAT expansion.


Asunto(s)
Tejido Adiposo/citología , Membrana Celular/metabolismo , Decorina/metabolismo , Receptores de Superficie Celular/metabolismo , Resistina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Secuencia de Aminoácidos , Animales , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Reproducibilidad de los Resultados
8.
Cancer Res ; 69(12): 5259-66, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19491274

RESUMEN

The connection between obesity and accelerated cancer progression has been established, but the mediating mechanisms are not well understood. We have shown that stromal cells from white adipose tissue (WAT) cooperate with the endothelium to promote blood vessel formation through the secretion of soluble trophic factors. Here, we hypothesize that WAT directly mediates cancer progression by serving as a source of cells that migrate to tumors and promote neovascularization. To test this hypothesis, we have evaluated the recruitment of WAT-derived cells by tumors and the effect of their engraftment on tumor growth by integrating a transgenic mouse strain engineered for expansion of traceable cells with established allograft and xenograft cancer models. Our studies show that entry of adipose stromal and endothelial cells into systemic circulation leads to their homing to and engraftment into tumor stroma and vasculature, respectively. We show that recruitment of adipose stromal cells by tumors is sufficient to promote tumor growth. Finally, we show that migration of stromal and vascular progenitor cells from WAT grafts to tumors is also associated with acceleration of cancer progression. These results provide a biological insight for the clinical association between obesity and cancer, thus outlining potential avenues for preventive and therapeutic strategies.


Asunto(s)
Tejido Adiposo/patología , Modelos Animales de Enfermedad , Neoplasias Experimentales/patología , Animales , Progresión de la Enfermedad , Ratones , Ratones Transgénicos
9.
PLoS One ; 4(2): e4513, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19229337

RESUMEN

Rx is a paired-like homeobox gene that is required for vertebrate eye formation. Mice lacking Rx function do not develop eyes or the posterior pituitary. To determine whether Rx is required cell autonomously in these tissues, we generated embryonic chimeras consisting of wild type and Rx-/- cells. We found that in the eye, Rx-deficient cells cannot participate in the formation of the neuroretina, retina pigment epithelium and the distal part of the optic stalk. In addition, in the ventral forebrain, Rx function is required cell autonomously for the formation of the posterior pituitary. Interestingly, Rx-/- and wild type cells segregate before the morphogenesis of these two tissues begins. Our observations suggest that Rx function is not only required for the morphogenesis of the retina and posterior pituitary, but also prior to morphogenesis, for the sorting out of cells to form distinct fields of retinal/pituitary cells.


Asunto(s)
Proteínas del Ojo/fisiología , Proteínas de Homeodominio/fisiología , Morfogénesis , Neurohipófisis/crecimiento & desarrollo , Retina/crecimiento & desarrollo , Animales , Movimiento Celular , Quimera , Embrión de Mamíferos , Ratones , Ratones Noqueados , Neurohipófisis/embriología , Retina/embriología
10.
Genesis ; 44(8): 361-3, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16850473

RESUMEN

Rx is a homeobox-containing gene that is critical for vertebrate eye development. Its expression domain delineates a field of cells from which the retina and the ventral hypothalamus develop. The 5' upstream regulatory sequences of the medaka fish Rx gene are functionally conserved during evolution to a degree that they direct gene expression into the Rx-expressing field of cells in mice. Using these sequences, we made a Cre line that can be used for inactivation of gene expression in the developing retina.


Asunto(s)
Proteínas del Ojo/genética , Proteínas del Ojo/fisiología , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Retina/embriología , Alelos , Animales , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Integrasas/genética , Operón Lac , Ratones , Ratones Transgénicos , Oryzias/embriología , Oryzias/genética
11.
Mol Cell Biol ; 25(20): 8854-63, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16199865

RESUMEN

During mouse eye development, the correct formation of the lens occurs as a result of reciprocal interactions between the neuroectoderm that forms the retina and surface ectoderm that forms the lens. Although many transcription factors required for early lens development have been identified, the mechanism and genetic interactions mediated by them remain poorly understood. Foxe3 encodes a winged helix-forkhead transcription factor that is initially expressed in the developing brain and in the lens placode and later restricted exclusively to the anterior lens epithelium. Here, we show that targeted disruption of Foxe3 results in abnormal development of the eye. Cells of the anterior lens epithelium show a decreased rate of proliferation, resulting in a smaller than normal lens. The anterior lens epithelium does not properly separate from the cornea and frequently forms an unusual, multilayered tissue. Because of the abnormal differentiation, lens fiber cells do not form properly, and the morphogenesis of the lens is greatly affected. The abnormally differentiated lens cells remain irregular in shape, and the lens becomes vacuolated. The defects in lens development correlate with changes in the expression of growth and differentiation factor genes, including DNase II-like acid DNase, Prox1, p57, and PDGFalpha receptor. As a result of abnormal lens development, the cornea and the retina are also affected. While Foxe3 is also expressed in a distinct region of the embryonic brain, we have not observed abnormal development of the brain in Foxe3(-/-) animals.


Asunto(s)
Factores de Transcripción Forkhead/deficiencia , Cristalino/anomalías , Animales , Secuencia de Bases , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular , Proliferación Celular , Forma de la Célula , ADN/genética , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Cápsula del Cristalino/anomalías , Cápsula del Cristalino/embriología , Cápsula del Cristalino/metabolismo , Cápsula del Cristalino/patología , Cristalino/embriología , Cristalino/metabolismo , Cristalino/patología , Ratones , Ratones Noqueados , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA