Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 256
Filtrar
1.
Cell Rep ; 21(1): 274-288, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28978480

RESUMEN

The small GTPase RhoA is involved in a variety of fundamental processes in normal tissue. Spatiotemporal control of RhoA is thought to govern mechanosensing, growth, and motility of cells, while its deregulation is associated with disease development. Here, we describe the generation of a RhoA-fluorescence resonance energy transfer (FRET) biosensor mouse and its utility for monitoring real-time activity of RhoA in a variety of native tissues in vivo. We assess changes in RhoA activity during mechanosensing of osteocytes within the bone and during neutrophil migration. We also demonstrate spatiotemporal order of RhoA activity within crypt cells of the small intestine and during different stages of mammary gestation. Subsequently, we reveal co-option of RhoA activity in both invasive breast and pancreatic cancers, and we assess drug targeting in these disease settings, illustrating the potential for utilizing this mouse to study RhoA activity in vivo in real time.


Asunto(s)
Técnicas Biosensibles , Transferencia Resonante de Energía de Fluorescencia/métodos , Microscopía Intravital/métodos , Imagen de Lapso de Tiempo/métodos , Proteínas de Unión al GTP rho/genética , Animales , Antineoplásicos/farmacología , Huesos/citología , Huesos/metabolismo , Movimiento Celular/efectos de los fármacos , Dasatinib/farmacología , Clorhidrato de Erlotinib/farmacología , Femenino , Transferencia Resonante de Energía de Fluorescencia/instrumentación , Regulación de la Expresión Génica , Intestino Delgado/metabolismo , Intestino Delgado/ultraestructura , Microscopía Intravital/instrumentación , Glándulas Mamarias Animales/irrigación sanguínea , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/ultraestructura , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/ultraestructura , Mecanotransducción Celular , Ratones , Ratones Transgénicos , Neutrófilos/metabolismo , Neutrófilos/ultraestructura , Osteocitos/metabolismo , Osteocitos/ultraestructura , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/ultraestructura , Imagen de Lapso de Tiempo/instrumentación , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
2.
Nat Med ; 23(7): 890-898, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28604702

RESUMEN

The extracellular matrix (ECM) is a master regulator of cellular phenotype and behavior. It has a crucial role in both normal tissue homeostasis and disease pathology. Here we present a fast and efficient approach to enhance the study of ECM composition and structure. Termed in situ decellularization of tissues (ISDoT), it allows whole organs to be decellularized, leaving native ECM architecture intact. These three-dimensional decellularized tissues can be studied using high-resolution fluorescence and second harmonic imaging, and can be used for quantitative proteomic interrogation of the ECM. Our method is superior to other methods tested in its ability to preserve the structural integrity of the ECM, facilitate high-resolution imaging and quantitatively detect ECM proteins. In particular, we performed high-resolution sub-micron imaging of matrix topography in normal tissue and over the course of primary tumor development and progression to metastasis in mice, providing the first detailed imaging of the metastatic niche. These data show that cancer-driven ECM remodeling is organ specific, and that it is accompanied by comprehensive changes in ECM composition and topological structure. We also describe differing patterns of basement-membrane organization surrounding different types of blood vessels in healthy and diseased tissues. The ISDoT procedure allows for the study of native ECM structure under normal and pathological conditions in unprecedented detail.


Asunto(s)
Membrana Basal/ultraestructura , Neoplasias de la Mama/ultraestructura , Matriz Extracelular/ultraestructura , Neoplasias Mamarias Experimentales/ultraestructura , Proteómica , Microambiente Tumoral , Animales , Membrana Basal/metabolismo , Neoplasias de la Mama/metabolismo , Matriz Extracelular/metabolismo , Femenino , Humanos , Imagenología Tridimensional , Lactancia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/ultraestructura , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/ultraestructura , Metástasis Linfática , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/ultraestructura , Neoplasias Mamarias Experimentales/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/ultraestructura , Ratones , Imagen Óptica , Nervios Periféricos/metabolismo , Nervios Periféricos/ultraestructura , Neoplasias de la Lengua/metabolismo , Neoplasias de la Lengua/ultraestructura
3.
Microsc Microanal ; 22(5): 1056-1061, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27653152

RESUMEN

Although the rat model of mammary tumors chemically induced by N-methyl-N-nitrosourea (MNU) has been frequently used by several research teams, there is a lack of ultrastructural studies in this field. The main aim of this work was to perform an ultrastructural characterization of MNU-induced mammary tumors in female rats. Some alterations previously reported in human mammary tumors, such as nucleus size and shape, accumulation of heterochromatin in the perinuclear region, and interdigitating cytoplasmic processes between cancer cells were also observed in MNU-induced mammary tumors. Although a low number of samples were analyzed by transmission electron microscopy in the present study, we consider that it may contribute to a better understanding of MNU-induced mammary carcinogenesis in a rat model. The ultrastructural characteristics of the two most frequently diagnosed mammary carcinomas described in the present work can be useful to differentiate them from other histological patterns. In addition, the loss of cytoplasm in neoplastic cells and formation of vacuoles were described.


Asunto(s)
Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/ultraestructura , Metilnitrosourea , Microscopía Electrónica de Transmisión , Animales , Femenino , Humanos , Ratas , Ratas Sprague-Dawley
4.
Eur J Pharmacol ; 765: 171-8, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26297304

RESUMEN

Several studies have assumed that myoepithelial cells (MECs) loss may contribute to epithelial tumor induction and/or progression. We adopted an in vitro assay and a syngeneic mice breast cancer model with histological and molecular characteristics resembling human lesions to evaluate tumor suppression effects of MECs. Flow cytometric, cell viability, blood chemistry, transmission electron microscope, immunohistochemistry and qRT-PCR assays were performed at the end of the study. We demonstrated that MECs could significantly suppress the viability of cancer cells at different time points (P<0.05). At the end of the fourth and fifth weeks, treated mice had smaller tumor volume compared with control animals. Average tumor volume was significantly less in treated groups than control group at days 21 (0.38±0.19 vs. 1.99±0.13 cm3), 28 (0.57±0.3 vs. 2.5±0.37 cm3) and 35 (0.7±0.35 vs. 2.65±0.4 cm3) after tumor cell injection (P<0.05). No hematological, hepatocellular, and renal toxicities were seen in MECs treated groups. Ultrastructural features revealed severe relationship between adjacent tumoral cells and loose interconnections of neoplastic cells in treated group. Immunohistochemical examinations of breast tumors showed high p63 and low alpha-smooth muscle actin protein expression in treated mice compared to control (P<0.05). MRNA expressions of TNF-α, smooth muscle-myosin heavy chain, connexin 43, and maspin were significantly up-regulated in breast tumor tissues in treated group compared to control (P<0.05). VEGF and alpha-smooth muscle actin mRNA expression were reduced in treated animals (P<0.05). The present study highlighted the potential tumor suppression effects of MECs on breast cancer in a typical animal model.


Asunto(s)
Adenocarcinoma/terapia , Trasplante de Células/métodos , Células Epiteliales/trasplante , Glándulas Mamarias Animales/citología , Neoplasias Mamarias Experimentales/terapia , Adenocarcinoma/metabolismo , Adenocarcinoma/ultraestructura , Animales , Línea Celular Tumoral , Supervivencia Celular , Técnicas de Cocultivo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Citometría de Flujo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , ARN/metabolismo
5.
Nat Commun ; 4: 2153, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23842578

RESUMEN

The ability to modulate the fluorescence of optical probes can be used to enhance signal-to-noise ratios for imaging within highly autofluorescent environments, such as intact tissues and living organisms. Here, we report two bacteriophytochrome-based photoactivatable near-infrared fluorescent proteins, named PAiRFP1 and PAiRFP2. PAiRFPs utilize haem-derived biliverdin, ubiquitous in mammalian tissues, as the chromophore. Initially weakly fluorescent PAiRFPs undergo photoconversion into a highly fluorescent state with excitation/emission at 690/717 nm following a brief irradiation with far-red light. After photoactivation, PAiRFPs slowly revert back to initial state, enabling multiple photoactivation-relaxation cycles. Low-temperature optical spectroscopy reveals several intermediates involved in PAiRFP photocycles, which all differ from that of the bacteriophytochrome precursor. PAiRFPs can be photoactivated in a spatially selective manner in mouse tissues, and optical modulation of their fluorescence allows for substantial contrast enhancement, making PAiRFPs advantageous over permanently fluorescent probes for in vivo imaging conditions of high autofluorescence and low signal levels.


Asunto(s)
Adenocarcinoma/ultraestructura , Proteínas Bacterianas/genética , Colorantes Fluorescentes/metabolismo , Neoplasias Mamarias Experimentales/ultraestructura , Imagen Molecular/métodos , Fitocromo/genética , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Agrobacterium tumefaciens/química , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/metabolismo , Biliverdina/metabolismo , Femenino , Células HeLa , Humanos , Rayos Infrarrojos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Procesos Fotoquímicos , Fitocromo/metabolismo , Ingeniería de Proteínas , Relación Señal-Ruido , Espectrometría de Fluorescencia , Transfección
6.
BMC Cancer ; 11: 22, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21244707

RESUMEN

BACKGROUND: ARHI is a Ras-related imprinted gene that inhibits cancer cell growth and motility. ARHI is downregulated in the majority of breast cancers, and loss of its expression is associated with its progression from ductal carcinoma in situ (DCIS) to invasive disease. In ovarian cancer, re-expression of ARHI induces autophagy and leads to autophagic death in cell culture; however, ARHI re-expression enables ovarian cancer cells to remain dormant when they are grown in mice as xenografts. The purpose of this study is to examine whether ARHI induces autophagy in breast cancer cells and to evaluate the effects of ARHI gene re-expression in combination with paclitaxel. METHODS: Re-expression of ARHI was achieved by transfection, by treatment with trichostatin A (TSA) or by a combination of TSA and 5-aza-2'-deoxycytidine (DAC) in breast cancer cell cultures and by liposomal delivery of ARHI in breast tumor xenografts. RESULTS: ARHI re-expression induces autophagy in breast cancer cells, and ARHI is essential for the induction of autophagy. When ARHI was re-expressed in breast cancer cells treated with paclitaxel, the growth inhibitory effect of paclitaxel was enhanced in both the cell culture and the xenografts. Although paclitaxel alone did not induce autophagy in breast cancer cells, it enhanced ARHI-induced autophagy. Conversely, ARHI re-expression promoted paclitaxel-induced apoptosis and G2/M cell cycle arrest. CONCLUSIONS: ARHI re-expression induces autophagic cell death in breast cancer cells and enhances the inhibitory effects of paclitaxel by promoting autophagy, apoptosis, and G2/M cell cycle arrest.


Asunto(s)
Autofagia/genética , Regulación Neoplásica de la Expresión Génica/genética , Paclitaxel/farmacología , Proteínas de Unión al GTP rho/genética , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Autofagia/efectos de los fármacos , Azacitidina/análogos & derivados , Azacitidina/farmacología , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Decitabina , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Proteínas de Unión al GTP rho/metabolismo
7.
Am J Pathol ; 173(4): 1173-85, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18787105

RESUMEN

Inhibitors of angiogenesis and radiation induce compensatory changes in the tumor vasculature both during and after treatment cessation. To assess the responses to irradiation and vascular endothelial growth factor-receptor tyrosine kinase inhibition (by the vascular endothelial growth factor tyrosine kinase inhibitor PTK787/ZK222854), mammary carcinoma allografts were investigated by vascular casting; electron, light, and confocal microscopy; and immunoblotting. Irradiation and anti-angiogenic therapy had similar effects on the tumor vasculature. Both treatments reduced tumor vascularization, particularly in the tumor medulla. After cessation of therapy, the tumor vasculature expanded predominantly by intussusception with a plexus composed of enlarged sinusoidal-like vessels containing multiple transluminal tissue pillars. Tumor revascularization originated from preserved alpha-smooth muscle actin-positive vessels in the tumor cortex. Quantification revealed that recovery was characterized by an angiogenic switch from sprouting to intussusception. Up-regulated alpha-smooth muscle actin-expression during recovery reflected the recruitment of alpha-smooth muscle actin-positive cells for intussusception as part of the angio-adaptive mechanism. Tumor recovery was associated with a dramatic decrease (by 30% to 40%) in the intratumoral microvascular density, probably as a result of intussusceptive pruning and, surprisingly, with only a minimal reduction of the total microvascular (exchange) area. Therefore, the vascular supply to the tumor was not severely compromised, as demonstrated by hypoxia-inducible factor-1alpha expression. Both irradiation and anti-angiogenic therapy cause a switch from sprouting to intussusceptive angiogenesis, representing an escape mechanism and accounting for the development of resistance, as well as rapid recovery, after cessation of therapy.


Asunto(s)
Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/terapia , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/radioterapia , Ftalazinas/uso terapéutico , Piridinas/uso terapéutico , Radiación Ionizante , Actinas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Inmunohistoquímica , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Ratones Desnudos , Necrosis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta , Factores de Tiempo , Supervivencia Tisular/efectos de los fármacos , Supervivencia Tisular/efectos de la radiación , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Biomed Res ; 28(3): 127-37, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17625345

RESUMEN

The purpose of this study was to examine the localization of macrophages, B-lymphocytes and osteoclasts in tumoral lesions of mammary carcinoma metastasized to bone of non-immunocompromised mice. Mouse mammary carcinoma cells (BALB/c-MC) were injected through the left cardiac ventricle into 5-week-old female wild-type Balb/c mice. The femora and tibiae of mice with metastasized cancer were extracted, and thereafter processed for histochemical analyses. The foci of metastasized tumor cells occupied the metaphyseal area, and the cell death zones could be identified within the tumor mass. Abundant tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts were found among the alkaline phosphatase (ALP)-reactive osteoblastic cell layer that covered the bone surface neighboring the metastatic lesion. In contrast, F4/80-positive macrophages/monocytes were localized adjacent to, or invading the metastatic tissue. In addition, some F4/80-positive cells were found in the aforementioned cell death zones. Unlike F4/80-positive cells, CD45R-positive B-lymphocytes did not accumulate at the surfaces of the tumor lesions, nor infiltrate into them, but were found scattered over bone marrow. Interestingly, some CD45R-positive cells were observed close to TRAP-positive osteoclasts in the stromal tissue surrounding the tumor lesion. Our findings suggest that, in the bone metastatic lesions of non-immunocompromised mice, F4/80-positive macrophages/monocytes accumulated on and/or infiltrated into the tumor nests, while CD45R-positive B-lymphocytes were associated with osteoclasts, rather than attacking metastatic tumor cells.


Asunto(s)
Linfocitos B/patología , Neoplasias Óseas/secundario , Carcinoma/inmunología , Carcinoma/patología , Macrófagos/patología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Osteoclastos/patología , Animales , Linfocitos B/ultraestructura , Neoplasias Óseas/patología , Neoplasias Óseas/ultraestructura , Carcinoma/ultraestructura , Linaje de la Célula/inmunología , Femenino , Macrófagos/ultraestructura , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Osteoclastos/ultraestructura
9.
BMC Med ; 4(1): 38, 2006 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-17190588

RESUMEN

BACKGROUND: Stromal-epithelial interactions are of particular significance in breast tissue as misregulation of these interactions can promote tumorigenesis and invasion. Moreover, collagen-dense breast tissue increases the risk of breast carcinoma, although the relationship between collagen density and tumorigenesis is not well understood. As little is known about epithelial-stromal interactions in vivo, it is necessary to visualize the stroma surrounding normal epithelium and mammary tumors in intact tissues to better understand how matrix organization, density, and composition affect tumor formation and progression. METHODS: Epithelial-stromal interactions in normal mammary glands, mammary tumors, and tumor explants in three-dimensional culture were studied with histology, electron microscopy, and nonlinear optical imaging methodologies. Imaging of the tumor-stromal interface in live tumor tissue ex vivo was performed with multiphoton laser-scanning microscopy (MPLSM) to generate multiphoton excitation (MPE) of endogenous fluorophores and second harmonic generation (SHG) to image stromal collagen. RESULTS: We used both laser-scanning multiphoton and second harmonic generation microscopy to determine the organization of specific collagen structures around ducts and tumors in intact, unfixed and unsectioned mammary glands. Local alterations in collagen density were clearly seen, allowing us to obtain three-dimensional information regarding the organization of the mammary stroma, such as radiating collagen fibers that could not have been obtained using classical histological techniques. Moreover, we observed and defined three tumor-associated collagen signatures (TACS) that provide novel markers to locate and characterize tumors. In particular, local cell invasion was found predominantly to be oriented along certain aligned collagen fibers, suggesting that radial alignment of collagen fibers relative to tumors facilitates invasion. Consistent with this observation, primary tumor explants cultured in a randomly organized collagen matrix realigned the collagen fibers, allowing individual tumor cells to migrate out along radially aligned fibers. CONCLUSION: The presentation of these tumor-associated collagen signatures allowed us to identify pre-palpable tumors and see cells at the tumor-stromal boundary invading into the stroma along radially aligned collagen fibers. As such, TACS should provide indications that a tumor is, or could become, invasive, and may serve as part of a strategy to help identify and characterize breast tumors in animal and human tissues.


Asunto(s)
Colágeno/ultraestructura , Glándulas Mamarias Animales/ultraestructura , Neoplasias Mamarias Experimentales/ultraestructura , Invasividad Neoplásica , Animales , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Femenino , Glándulas Mamarias Animales/citología , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Microscopía Confocal , Microscopía Electrónica de Transmisión , Células del Estroma/metabolismo , Células del Estroma/ultraestructura
10.
Mol Cell Biochem ; 290(1-2): 169-76, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16941229

RESUMEN

This study was to demonstrate by histological grading whether soy protein protects against dimethylbenz[a]anthracene (DMBA) -induced breast tumors in female rats. At 25 days of age, rats were fed diets containing either casein or soy protein. After 25 days on diets, a single dose of DMBA in sesame oil (80 mg/kg) was administered by gavage. All tumors were detected by palpation. The number of tumors per rat was less in soy group than that in casein group at any time point up to 122 days after DMBA administration. Incidence of tumors was less in soy protein group than that in casein group. Casein group had 20% grade I, 60% grade II, and 20% grade III adenocarcinoma. However, the soy group had 100% grade I adenocarcinoma and no aggressive grade II or grade III tumor. There was a delay in the development of tumor in the soy protein group in comparison to the casein group. Again, unlike casein, the soy group had cessation of angiogenesis at several sites of tumor, and reduced levels of angiogenic markers, VEGF and bFGF. Immunohistochemical analysis of the breast tissues did not show any CD-31 positive stain in soy protein group, whereas some CD-31 positive stain was revealed in casein group, which further suggests that soy protein controls angiogenesis. Furthermore, proliferative index as assessed by Ki-67 staining was less in soy protein group than that in casein group. These findings suggest that the soy protein may protect against the development of a more aggressive breast carcinoma.


Asunto(s)
Adenocarcinoma/prevención & control , Antracenos , Proteínas en la Dieta/administración & dosificación , Neoplasias Mamarias Experimentales/patología , Neovascularización Patológica/terapia , Proteínas de Soja/administración & dosificación , Adenocarcinoma/patología , Adenocarcinoma/ultraestructura , Animales , Western Blotting , Mama/efectos de los fármacos , Mama/ultraestructura , Femenino , Neoplasias Mamarias Experimentales/prevención & control , Neoplasias Mamarias Experimentales/ultraestructura , Estadificación de Neoplasias , Ratas , Ratas Sprague-Dawley
11.
J Cell Biol ; 169(6): 977-85, 2005 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-15967816

RESUMEN

We recently reported that uPARAP/Endo180 can mediate the cellular uptake and lysosomal degradation of collagen by cultured fibroblasts. Here, we show that uPARAP/Endo180 has a key role in the degradation of collagen during mammary carcinoma progression. In the normal murine mammary gland, uPARAP/Endo180 is widely expressed in periductal fibroblast-like mesenchymal cells that line mammary epithelial cells. This pattern of uPARAP/Endo180 expression is preserved during polyomavirus middle T-induced mammary carcinogenesis, with strong uPARAP/Endo180 expression by mesenchymal cells embedded within the collagenous stroma surrounding nests of uPARAP/Endo180-negative tumor cells. Genetic ablation of uPARAP/Endo180 impaired collagen turnover that is critical to tumor expansion, as evidenced by the abrogation of cellular collagen uptake, tumor fibrosis, and blunted tumor growth. These studies identify uPARAP/Endo180 as a key mediator of collagen turnover in a pathophysiological context.


Asunto(s)
Carcinoma/metabolismo , Transformación Celular Neoplásica/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Glicoproteínas de Membrana/metabolismo , Mesodermo/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Carcinoma/genética , Carcinoma/ultraestructura , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/ultraestructura , Células Cultivadas , Modelos Animales de Enfermedad , Matriz Extracelular/ultraestructura , Femenino , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/ultraestructura , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/ultraestructura , Glicoproteínas de Membrana/genética , Mesodermo/patología , Mesodermo/ultraestructura , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Invasividad Neoplásica , Poliomavirus , Receptores de Superficie Celular/genética , Células del Estroma/metabolismo , Células del Estroma/patología , Células del Estroma/ultraestructura
12.
Stem Cell Rev ; 1(3): 215-23, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-17142858

RESUMEN

I have used the paradigm of mammary cancer induction by the mouse mammary tumor virus (MMTV) to illustrate the body of evidence that supports the hypothesis that mammary epithelial stem/progenitor cells represent targets for oncogenic transformation. Further, it is argued that this is not a special case applicable only to MMTV-induced mammary cancer, because MMTV acts as an environmental mutagen producing random interruptions in the somatic DNA of infected cells by insertion of proviral DNA copies. In addition to disrupting the host genome, the proviral DNA also influences gene expression through its associated enhancer sequences over significant inter-genome distances. Genes commonly affected by MMTV insertion in multiple individual tumors include the Wnt genes, the fibroblast growth factor (FGF) gene family, and the Notch gene family. All of these gene families are known to play essential roles in stem cell maintenance and behavior in a variety of organs. The MMTV-induced mutations accumulate in cells that are long-lived and possess the properties of stem cells, namely, self-renewal and the capacity to produce divergent epithelial progeny through asymmetric division. The evidence shows that epithelial cells with these properties are present in normal mammary glands, may be infected with MMTV, and become transformed to produce epithelial hyperplasia through MMTV-induced mutagenesis and progress to frank mammary malignancy. Retroviral marking via MMTV proviral insertion demonstrates that this process progresses from a single mammary epithelial cell that possesses all the features ascribed to tissue-specific stem cells.


Asunto(s)
Transformación Celular Viral , Neoplasias Mamarias Animales/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Mutagénesis Insercional , Células Madre Neoplásicas/metabolismo , Integración Viral , Animales , Transformación Celular Viral/genética , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Células Epiteliales/virología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/ultraestructura , Neoplasias Mamarias Animales/virología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/ultraestructura , Neoplasias Mamarias Experimentales/virología , Virus del Tumor Mamario del Ratón/genética , Ratones , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/virología , Integración Viral/genética
13.
Cancer Epidemiol Biomarkers Prev ; 13(7): 1173-84, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15247128

RESUMEN

An experimental model developed to investigate premalignant stages of breast cancer was used to establish a rationale for designing experiments that target angiogenesis for cancer prevention. Blood vessels were identified via CD31 immunostaining, and all vessels that occurred in a 50 microm wide region circumscribing each pathology were counted using a digital imaging technique. The blood vessel density associated with terminal end buds was unaffected by carcinogen treatment, whereas vessel density was higher in intraductal proliferations and ductal carcinoma in situ than in terminal end buds (P < 0.001) and total vascularity increased with morphologic progression. In comparison with intraductal proliferation or ductal carcinoma in situ, mammary carcinomas had higher vascular density in the tissue surrounding the cancer with a marked increase in the number of blood vessels <25 microm(2). These data suggest that antiangiogenic chemopreventive agents would inhibit cancer occurrence if initiated at any premalignant stage of the carcinogenic process. Because increased vascular density observed during premalignancy could be explained by the size expansion of the lesion and its encroachment on a preexisting blood supply, by pathology-associated vessel expansion, and/or by angiogenesis, it remains to be determined if antiangiogenic agents will reduce the prevalence of premalignant lesions or cause their accumulation by blocking conversion to carcinomas. Failure to recognize the patterns of vascularization that accompany morphologic progression could limit the success of efforts to target angiogenesis for cancer prevention and lead to misinformation about how agents that affect blood vessel formation or growth inhibit the carcinogenic process.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Carcinoma in Situ/irrigación sanguínea , Glándulas Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Experimentales/irrigación sanguínea , Metilnitrosourea/toxicidad , Neovascularización Patológica/sangre , Animales , Carcinógenos/toxicidad , Carcinoma in Situ/inducido químicamente , Carcinoma in Situ/ultraestructura , Femenino , Glándulas Mamarias Animales/ultraestructura , Neoplasias Mamarias Experimentales/prevención & control , Neoplasias Mamarias Experimentales/ultraestructura , Ratas , Ratas Sprague-Dawley , Proyectos de Investigación
14.
Eur J Pharm Biopharm ; 57(1): 53-63, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14729080

RESUMEN

A novel injectable thermosensitive in situ gelling hydrogel has been developed. The system, which falls under the BST-Gel platform technology developed at Biosyntech Inc. (Laval, QC, Canada), consists of a chitosan solution (C) neutralized with beta-glycerophosphate (GP) that is liquid at room temperature but gels when heated to body temperature. We propose to use this thermosensitive hydrogel for the sustained release of paclitaxel at tumor resection sites in order to prevent local tumor recurrence. The in vitro release profiles demonstrated controlled delivery over 1 month. The initial drug loading substantially affected the release. Local delivery of paclitaxel from the formulation injected intratumorally was investigated using EMT-6 tumors implanted subcutaneously on Balb/c mice. These experiments showed that one intratumoral injection of the thermosensitive hydrogel containing paclitaxel was as efficacious as four intravenous injections of Taxol in inhibiting the growth of EMT-6 cancer cells in mice, but in a less toxic manner. Further histological analysis revealed that while the proportion of necrotic areas was similar for the C/GP/paclitaxel and the Taxol-treated tumors, a disparity between tumor-associated inflammatory cell populations may suggest differing anti-tumor mechanisms.


Asunto(s)
Temperatura Corporal/fisiología , Quitosano/farmacología , Hidrogeles/farmacología , Inyecciones Intralesiones/métodos , Paclitaxel/administración & dosificación , Animales , Línea Celular Tumoral , Quitosano/química , Quitosano/metabolismo , Sistemas de Liberación de Medicamentos/instrumentación , Sistemas de Liberación de Medicamentos/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Estabilidad de Medicamentos , Estudios de Evaluación como Asunto , Femenino , Rayos gamma , Glicerofosfatos/química , Glicerofosfatos/metabolismo , Glicerofosfatos/farmacología , Hidrogeles/química , Hidrogeles/metabolismo , Inyecciones Intravenosas , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/ultraestructura , Ensayo de Materiales/métodos , Ratones , Ratones Endogámicos BALB C , Paclitaxel/metabolismo , Paclitaxel/uso terapéutico , Soluciones/efectos de la radiación , Esterilización/métodos
15.
Cancer Res ; 62(5): 1410-9, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11888913

RESUMEN

FTY720 is a unique immunosuppressive agent that exerts its activity by inducing apoptosis in lymphocytes. We conducted the present study to investigate the effects of FTY720 on cancer growth and metastasis, as well as its mechanism of action. In vitro treatment with FTY720 induced dramatic cancer cell apoptosis in a mouse breast cancer cell line, JygMC(A). Electron microscopy revealed distinct changes on the cell surface with decreased filopodias and microvilli in cancer cells treated with FTY720 at 2 microM and clear evidence of apoptosis at 10 microM. Interestingly, the effect of FTY720 was significantly less in the normal fibroblasts than in the cancer cells, indicating greater susceptibility of cancer cells to the agent. We then tested the in vivo effect of FTY720 in a mouse breast cancer model created by inoculating JygMC(A) cells (s.c.) in the flank region of BALB/c-nu/nu mice at three different dosages (2, 5, and 10 mg/kg/day; n = 30/group). Tumor growth was markedly suppressed at a dosage of 5 mg/kg or more without notable side effects. In addition, tumor metastasis, which was dramatically evident in control mice, was significantly prevented even at a low dose (2 mg/kg/day), resulting in a significant prolongation of animal survival. These data led us to additionally investigate the mechanism of action, especially the prevention of metastasis at a low dose. FTY720 treatment at 2 microM caused a remarkable cytoskeletal change with deformed and decreased filopodias in cancer cells. In addition, it significantly decreased the ability of cancer cells to adhere and migrate to extracellular matrix components, and markedly reduced the expression of integrins on the cancer cell surface. These results indicate that FTY720 is a potent anticancer agent that induces cancer cell apoptosis and is markedly effective for prevention of metastasis. The changes of cellular structure with reduction of integrin expression may be one of its underlying mechanisms of action.


Asunto(s)
Inmunosupresores/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Metástasis de la Neoplasia/prevención & control , Glicoles de Propileno/farmacología , Actinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Femenino , Clorhidrato de Fingolimod , Integrinas/análisis , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Glicoles de Propileno/efectos adversos , Esfingosina/análogos & derivados , Células Tumorales Cultivadas
16.
Nat Med ; 7(3): 297-303, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11231627

RESUMEN

The initiation of T-cell-mediated antitumor immune responses requires the uptake and processing of tumor antigens by dendritic cells and their presentation on MHC-I molecules. Here we show in a human in vitro model system that exosomes, a population of small membrane vesicles secreted by living tumor cells, contain and transfer tumor antigens to dendritic cells. After mouse tumor exosome uptake, dendritic cells induce potent CD8+ T-cell-dependent antitumor effects on syngeneic and allogeneic established mouse tumors. Therefore, exosomes represent a novel source of tumor-rejection antigens for T-cell cross priming, relevant for immunointerventions.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Mamarias Experimentales/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Células Dendríticas/inmunología , Humanos , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Microscopía Inmunoelectrónica , Células Tumorales Cultivadas
17.
Virchows Arch ; 435(5): 509-19, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10592055

RESUMEN

Previous studies showed that the hormone relaxin acts on human breast cancer MCF-7 cells in vitro by modulating cell proliferation and promoting cell differentiation toward a duct epithelial phenotype. The present study was designed to investigate whether relaxin retains these properties when acting in vivo on MCF-7 cell tumors developed in athymic nude mice. Mice bearing MCF-7 cell tumors transplanted under the mammary fat pad and estrogenized to sustain tumor growth were treated systemically with relaxin (10 microg/day) for 19 days. Vehicle-treated mice were used as controls. Thirty days later, the mice were sacrificed and tumor fragments were analyzed by light and electron microscopy and immunocytochemistry. Measurements of tumor volume were recorded weekly for the overall experimental period. The results obtained indicate that relaxin treatment promotes differentiation of tumor cells towards both myoepithelial-like and epithelial-like cells, as judged by the ultrastructural features of the cells and by the increased expression of smooth muscle actin and cadherins. Measurements of tumor size and of the number of cycling cells show that relaxin, at the doses and times of exposure used in this study, does not significantly influence tumor growth and cell proliferation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/ultraestructura , Relaxina/farmacología , Actinas/biosíntesis , Animales , Cadherinas/biosíntesis , División Celular/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Desnudos , Microscopía Electrónica , Trasplante de Neoplasias , Fenotipo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Células Tumorales Cultivadas
18.
J Cell Biol ; 145(2): 331-45, 1999 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-10209028

RESUMEN

Using both light and high resolution electron microscopy, we analyzed the spatial and temporal relationships between the Arp2/3 complex and the nucleation activity that is required for lamellipod extension in mammary carcinoma cells after epidermal growth factor stimulation. A rapid two- to fourfold increase in filament barbed end number occurs transiently after stimulation and remains confined almost exclusively to the extreme outer edge of the extending lamellipod (within 100-200 nm of the plasma membrane). This is accompanied by an increase in filament density at the leading edge and a general decrease in filament length, with a specific loss of long filaments. Concomitantly, the Arp2/3 complex is recruited with a 1.5-fold increase throughout the entire cortical filament network extending 1-1.5 microm in depth from the membrane at the leading edge. The recruitment of the Arp2/3 complex at the membrane of the extending lamellipod indicates that Arp2/3 may be involved in initial generation of growing filaments. However, only a small subset of the complex present in the cortical network colocalizes near free barbed ends. This suggests that the 100-200-nm submembraneous compartment at the leading edge of the extending lamellipod constitutes a special biochemical microenvironment that favors the generation and maintenance of free barbed ends, possibly through the locally active Arp2/3 complex, severing or decreasing the on-rate of capping protein. Our results are inconsistent with the hypothesis suggesting uncapping is the dominant mechanism responsible for the generation of nucleation activity. However, they support the hypothesis of an Arp2/3-mediated capture of actin oligomers that formed close to the membrane by other mechanisms such as severing. They also support pointed-end capping by the Arp2/3 complex, accounting for its wide distribution at the leading edge.


Asunto(s)
Actinas/metabolismo , Membrana Celular/ultraestructura , Proteínas del Citoesqueleto , Citoesqueleto/ultraestructura , Factor de Crecimiento Epidérmico/farmacología , Neoplasias Mamarias Experimentales/ultraestructura , Proteína 2 Relacionada con la Actina , Proteína 3 Relacionada con la Actina , Animales , Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular , Citoesqueleto/efectos de los fármacos , Femenino , Neoplasias Mamarias Experimentales/patología , Microscopía Fluorescente , Modelos Biológicos , Ratas
19.
Arch Pharm (Weinheim) ; 332(2): 59-69, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10191716

RESUMEN

[meso-1,2-Bis(2,6-dichloro-4-hydroxyphenyl)ethylenediamine]- dichloroplatinum(II) (meso-1-PtCl2), an estrogenic and cytotoxic platinum complex, shows activity against ER+ but not against ER- breast cancers in vivo (ER: estrogen receptor; ER+ and ER- indicate the presence or absence of the ER). To clarify whether its estrogenic or its cytotoxic potency or both properties are the cause of this specific inhibitory effect, we tested meso-1-PtCl2 comparatively in vivo on an ER+ and an ER- murine breast cancer (MXT-M-3.2 MC and MXT-M-3.2(ovex) MC, respectively), and in vitro on two cell lines derived from the former in vivo models (MXT+ and MXT-, respectively). The estrogens diethylstilbestrol (DES) and the ligand of meso-1-PtCl2 (meso-1), responsible for the hormonal effect of meso-1-PtCl2, and the cytotoxic drug cisplatin (cDDP) were used as comparative substances. Meso-1-PtCl2. DES and cDDP showed a strong and comparable activity on the ER+ MXT-M-3.2 MC in vivo, meso-1 being somewhat less inhibitory. In experiments on the murine, ER- MXT-M-3.2(ovex) MC only cDDP caused a marked inhibitory effect. The other compounds were inactive or only marginally active. In accordance with the in vivo results cDDP was also very active on the MXT+ and MXT- breast cancer cell line. In contrast to this meso-1-PtCl2, meso-1, and DES proved to be only weakly active or inactive on both cell lines. From these results it can be concluded that there is only little if any contribution of the cytotoxic PtCl2 moiety of meso-1-PtCl2 to the anti-breast cancer activity in vivo. On the ER+ MXT-M-3.2 MC transplanted into ovariectomized mice meso-1-PtCl2 yielded a biphasic dose activity curve, i.e. an increase of the tumor growth at low doses followed by a decrease at high doses, identical with those of the estrogens DES and meso-1. These results indicate that meso-1-PtCl2 inhibits ER+ breast cancers by its estrogenicity in the same manner as meso-1 and DES. The complex mechanism of anti-breast cancer active estrogens involves presumably the endocrine and/or the immune system. Its investigation is the subject of further studies.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Compuestos Organoplatinos/farmacología , Animales , Dietilestilbestrol/farmacología , Femenino , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Ratones Endogámicos , Neoplasias Hormono-Dependientes/ultraestructura , Ovariectomía , Receptores de Estrógenos/fisiología
20.
Int J Mol Med ; 2(4): 437-44, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9857231

RESUMEN

To investigate the roles of telomere length (mean length of the terminal restriction fragments; TRFs), telomerase activity (TA) and telomerase RNA (mTR) expression in relation to mouse mammary tumor progression, we examined a pregnancy-dependent mouse mammary tumor line (TPDMT-4) and its four autonomous sublines (T4-OI320: non-metastatic; and T4-OI165, -OI96, and -OI145: artificial metastatic) of DDD/1 mouse origin, and an autonomous growing mammary tumor (JYG-MC) showing spontaneous lung metastasis developed in BALB/c mice infected with a Chinese feral mice (Sub-Jyg)-derived mouse mammary tumor virus (JYG-MTV). Compared with normal (pregnant) mammary tissue, the TA was elevated in the TPDMT-4 tumor and in the non-metastatic subline tumor (T4-OI320) (x10 fold, respectively), and was further increased (x13-15 fold) in parallel with the acquisition of metastatic potential (T4-OI165, -OI96, and -OI145). The mTR level was upregulated (x2.7-2.8 fold) in all autonomous growing tumors compared to the normal counter-part, but not in TPDMT-4. The TRF was shorter in accord with tumor progression (normal mammary tissue, 48 kb; TPDMT-4, 45 kb; T4-OI320, 37 kb; T4-OI165, -OI96 and -OI145, mean 37.7 kb; and JYG-MC, 21 kb). These results suggest that the activation of TA occurs as an early event at the stage of hormone-dependent tumorigenesis, followed by the up-regulation of mTR expression in accordance with the acquisition of autonomous growth, and then further activation of TA occurs when the tumor acquires metastatic potential. The TRF shortening was in parallel with the tumor progression.


Asunto(s)
Neoplasias Mamarias Experimentales/enzimología , Neoplasias Hormono-Dependientes/enzimología , ARN Mensajero/biosíntesis , Telomerasa/metabolismo , Telómero/ultraestructura , Animales , Femenino , Neoplasias Pulmonares/secundario , Glándulas Mamarias Animales/enzimología , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Neoplasias Hormono-Dependientes/patología , Neoplasias Hormono-Dependientes/ultraestructura , Embarazo , Complicaciones Neoplásicas del Embarazo/enzimología , Complicaciones Neoplásicas del Embarazo/patología , Telomerasa/biosíntesis , Telomerasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...