Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Bioeng Transl Med ; 9(4): e10636, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39036092

RESUMEN

Drug-carrying nanoparticles are a promising strategy to deliver therapeutics into the brain, but their translation requires better characterization of interactions between nanomaterials and endothelial cells of the blood-brain barrier (BBB). Here, we use a library of 18 layer-by-layer electrostatically assembled nanoparticles (NPs) to independently assess the impact of NP core and surface materials on in vitro uptake, transport, and intracellular trafficking in brain endothelial cells. We demonstrate that NP core stiffness determines the magnitude of transport, while surface chemistry directs intracellular trafficking. Finally, we demonstrate that these factors similarly dictate in vivo BBB transport using intravital imaging through cranial windows in mice. We identify that hyaluronic acid surface chemistry increases transport across the BBB in vivo, and flow conditions are necessary to replicate this finding in vitro. Taken together, these findings highlight the importance of assay geometry, cell biology, and fluid flow in developing nanocarriers for delivery to the brain.

2.
Proc Natl Acad Sci U S A ; 119(23): e2118697119, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35648828

RESUMEN

The blood­brain barrier represents a significant challenge for the treatment of high-grade gliomas, and our understanding of drug transport across this critical biointerface remains limited. To advance preclinical therapeutic development for gliomas, there is an urgent need for predictive in vitro models with realistic blood­brain-barrier vasculature. Here, we report a vascularized human glioblastoma multiforme (GBM) model in a microfluidic device that accurately recapitulates brain tumor vasculature with self-assembled endothelial cells, astrocytes, and pericytes to investigate the transport of targeted nanotherapeutics across the blood­brain barrier and into GBM cells. Using modular layer-by-layer assembly, we functionalized the surface of nanoparticles with GBM-targeting motifs to improve trafficking to tumors. We directly compared nanoparticle transport in our in vitro platform with transport across mouse brain capillaries using intravital imaging, validating the ability of the platform to model in vivo blood­brain-barrier transport. We investigated the therapeutic potential of functionalized nanoparticles by encapsulating cisplatin and showed improved efficacy of these GBM-targeted nanoparticles both in vitro and in an in vivo orthotopic xenograft model. Our vascularized GBM model represents a significant biomaterials advance, enabling in-depth investigation of brain tumor vasculature and accelerating the development of targeted nanotherapeutics.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Permeabilidad Capilar , Glioblastoma , Nanopartículas , Animales , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/metabolismo , Células Endoteliales/metabolismo , Glioblastoma/irrigación sanguínea , Glioblastoma/metabolismo , Humanos , Ratones , Microfluídica , Nanopartículas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Sci Adv ; 8(3): eabg6383, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35061540

RESUMEN

Access to electron acceptors supports oxidized biomass synthesis and can be limiting for cancer cell proliferation, but how cancer cells overcome this limitation in tumors is incompletely understood. Nontransformed cells in tumors can help cancer cells overcome metabolic limitations, particularly in pancreatic cancer, where pancreatic stellate cells (PSCs) promote cancer cell proliferation and tumor growth. However, whether PSCs affect the redox state of cancer cells is not known. By taking advantage of the endogenous fluorescence properties of reduced nicotinamide adenine dinucleotide and oxidized flavin adenine dinucleotide cofactors we use optical imaging to assess the redox state of pancreatic cancer cells and PSCs and find that direct interactions between PSCs and cancer cells promote a more oxidized state in cancer cells. This suggests that metabolic interaction between cancer cells and PSCs is a mechanism to overcome the redox limitations of cell proliferation in pancreatic cancer.


Asunto(s)
Neoplasias Pancreáticas , Células Estrelladas Pancreáticas , Línea Celular Tumoral , Humanos , Oxidación-Reducción , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/metabolismo , Células del Estroma , Neoplasias Pancreáticas
4.
Nat Commun ; 9(1): 1991, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29777137

RESUMEN

Effective treatment for glioblastoma (GBM) is limited by the presence of the blood-brain barrier (BBB) and rapid resistance to single agent therapies. To address these issues, we developed a transferrin-functionalized nanoparticle (Tf-NP) that can deliver dual combination therapies. Using intravital imaging, we show the ability of Tf-NPs to traverse intact BBB in mice as well as achieve direct tumor binding in two intracranial orthotopic models of GBM. Treatment of tumor-bearing mice with Tf-NPs loaded with temozolomide and the bromodomain inhibitor JQ1 leads to increased DNA damage and apoptosis that correlates with a 1.5- to 2-fold decrease in tumor burden and corresponding increase in survival compared to equivalent free-drug dosing. Immunocompetent mice treated with Tf-NP-loaded drugs also show protection from the effects of systemic drug toxicity, demonstrating the preclinical potential of this nanoscale platform to deliver novel combination therapies to gliomas and other central nervous system tumors.


Asunto(s)
Antineoplásicos Alquilantes/química , Antineoplásicos/administración & dosificación , Azepinas/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Glioma/tratamiento farmacológico , Nanopartículas/química , Temozolomida/administración & dosificación , Triazoles/administración & dosificación , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Azepinas/química , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/instrumentación , Glioma/metabolismo , Glioma/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Temozolomida/química , Triazoles/química , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Biomaterials ; 134: 202-215, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28482280

RESUMEN

Cell-intrinsic reporters such as luciferase (LUC) and red fluorescent protein (RFP) have been commonly utilized in preclinical studies to image tumor growth and to monitor therapeutic responses. While extrinsic reporters that emit near infrared I (NIR-I: 650-950 nm) or near-infrared II (NIR-II: 1000-1700 nm) optical signals have enabled minimization of tissue autofluorescence and light scattering, it has remained unclear as to whether their use has afforded more accurate tumor imaging in small animals. Here, we developed a novel optical imaging construct comprised of rare earth lanthanide nanoparticles coated with biodegradable diblock copolymers and doped with organic fluorophores, generating NIR-I and NIR-II emissive bands upon optical excitation. Simultaneous injection of multiple spectrally-unique nanoparticles into mice bearing tumor implants established via intraperitoneal dissemination of LUC+/RFP+ OVCAR-8 ovarian cancer cells enabled direct comparisons of imaging with extrinsic vs. intrinsic reporters, NIR-II vs. NIR-I signals, as well as targeted vs. untargeted exogenous contrast agents in the same animal and over time. We discovered that in vivo optical imaging at NIR-II wavelengths facilitates more accurate detection of smaller and earlier tumor deposits, offering enhanced sensitivity, improved spatial contrast, and increased depths of tissue penetration as compared to imaging with visible or NIR-I fluorescent agents. Our work further highlights the hitherto underappreciated enhancements in tumor accumulation that may be achieved with intraperitoneal as opposed to intravenous administration of nanoparticles. Lastly, we found discrepancies in the fidelity of tumor uptake that could be obtained by utilizing small molecules for in vivo as opposed to in vitro targeting of nanoparticles to disseminated tumors.


Asunto(s)
Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/diagnóstico , Espectroscopía Infrarroja Corta/métodos , Animales , Línea Celular Tumoral , Detección Precoz del Cáncer/métodos , Femenino , Humanos , Ratones , Ratones Desnudos , Nanopartículas/química , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Nat Med ; 23(2): 235-241, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28024083

RESUMEN

Mammalian tissues rely on a variety of nutrients to support their physiological functions. It is known that altered metabolism is involved in the pathogenesis of cancer, but which nutrients support the inappropriate growth of intact malignant tumors is incompletely understood. Amino acids are essential nutrients for many cancer cells that can be obtained through the scavenging and catabolism of extracellular protein via macropinocytosis. In particular, macropinocytosis can be a nutrient source for pancreatic cancer cells, but it is not fully understood how the tumor environment influences metabolic phenotypes and whether macropinocytosis supports the maintenance of amino acid levels within pancreatic tumors. Here we utilize miniaturized plasma exchange to deliver labeled albumin to tissues in live mice, and we demonstrate that breakdown of albumin contributes to the supply of free amino acids in pancreatic tumors. We also deliver albumin directly into tumors using an implantable microdevice, which was adapted and modified from ref. 9. Following implantation, we directly observe protein catabolism and macropinocytosis in situ by pancreatic cancer cells, but not by adjacent, non-cancerous pancreatic tissue. In addition, we find that intratumoral inhibition of macropinocytosis decreases amino acid levels. Taken together, these data suggest that pancreatic cancer cells consume extracellular protein, including albumin, and that this consumption serves as an important source of amino acids for pancreatic cancer cells in vivo.


Asunto(s)
Aminoácidos/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Pinocitosis , Proteolisis , Albúmina Sérica/metabolismo , Albúminas/metabolismo , Animales , Línea Celular Tumoral , Cromatografía de Gases , Modelos Animales de Enfermedad , Espacio Extracelular/metabolismo , Ratones , Microscopía de Fluorescencia por Excitación Multifotónica , Isótopos de Nitrógeno , Plasmaféresis , Proteínas/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
7.
Oncotarget ; 7(50): 82289-82304, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27793045

RESUMEN

The extracellular serine protease inhibitor serpinE2 is overexpressed in breast cancer and has been shown to foster metastatic spread. Here, we investigated the hypothesis that serpinE2 creates tumor-promoting conditions in the tumor microenvironment (TME) by affecting extracellular matrix remodeling. Using two different breast cancer models, we show that blocking serpinE2, either by knock-down (KD) in tumor cells or in response to a serpinE2 binding antibody, decreases metastatic dissemination from primary tumors to the lungs. We demonstrate that in response to serpinE2 KD or antibody treatment there are dramatic changes in the TME. Multiphoton intravital imaging revealed deposition of a dense extracellular collagen I matrix encapsulating serpinE2 KD or antibody-treated tumors. This is accompanied by a reduction in the population of tumor-promoting macrophages, as well as a decrease in chemokine ligand 2, which is known to affect macrophage abundance and polarization. In addition, TIMP-1 secretion is increased, which may directly inhibit matrix metalloproteases critical for collagen degradation in the tumor. In summary, our findings suggest that serpinE2 is required in the extracellular milieu of tumors where it acts in multiple ways to regulate tumor matrix deposition, thereby controlling tumor cell dissemination.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Matriz Extracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Serpina E2/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Colágeno Tipo I/metabolismo , Matriz Extracelular/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones Endogámicos BALB C , Ratones SCID , Invasividad Neoplásica , Fenotipo , Interferencia de ARN , Serpina E2/antagonistas & inhibidores , Serpina E2/genética , Transducción de Señal , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Transfección , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Res ; 75(16): 3255-67, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26122846

RESUMEN

The delivery of diagnostic and therapeutic agents to solid tumors is limited by physical transport barriers within tumors, and such restrictions directly contribute to decreased therapeutic efficacy and the emergence of drug resistance. Nanomaterials designed to perturb the local tumor environment with precise spatiotemporal control have demonstrated potential to enhance drug delivery in preclinical models. Here, we investigated the ability of one class of heat-generating nanomaterials called plasmonic nanoantennae to enhance tumor transport in a xenograft model of ovarian cancer. We observed a temperature-dependent increase in the transport of diagnostic nanoparticles into tumors. However, a transient, reversible reduction in this enhanced transport was seen upon reexposure to heating, consistent with the development of vascular thermotolerance. Harnessing these observations, we designed an improved treatment protocol combining plasmonic nanoantennae with diffusion-limited chemotherapies. Using a microfluidic endothelial model and genetic tools to inhibit the heat-shock response, we found that the ability of thermal preconditioning to limit heat-induced cytoskeletal disruption is an important component of vascular thermotolerance. This work, therefore, highlights the clinical relevance of cellular adaptations to nanomaterials and identifies molecular pathways whose modulation could improve the exposure of tumors to therapeutic agents.


Asunto(s)
Adaptación Fisiológica , Endotelio Vascular/metabolismo , Calor , Nanopartículas/metabolismo , Neoplasias Ováricas/metabolismo , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Células Cultivadas , Doxorrubicina/administración & dosificación , Doxorrubicina/metabolismo , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Femenino , Humanos , Hipertermia Inducida , Estimación de Kaplan-Meier , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Ratones Transgénicos , Nanopartículas/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/fisiopatología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Nat Mater ; 14(6): 643-51, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25985456

RESUMEN

The efficacy of implanted biomedical devices is often compromised by host recognition and subsequent foreign body responses. Here, we demonstrate the role of the geometry of implanted materials on their biocompatibility in vivo. In rodent and non-human primate animal models, implanted spheres 1.5 mm and above in diameter across a broad spectrum of materials, including hydrogels, ceramics, metals and plastics, significantly abrogated foreign body reactions and fibrosis when compared with smaller spheres. We also show that for encapsulated rat pancreatic islet cells transplanted into streptozotocin-treated diabetic C57BL/6 mice, islets prepared in 1.5-mm alginate capsules were able to restore blood-glucose control for up to 180 days, a period more than five times longer than for transplanted grafts encapsulated within conventionally sized 0.5-mm alginate capsules. Our findings suggest that the in vivo biocompatibility of biomedical devices can be significantly improved simply by tuning their spherical dimensions.


Asunto(s)
Reacción a Cuerpo Extraño/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Primates
10.
Nature ; 515(7525): 130-3, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25337873

RESUMEN

Secretion of C-C chemokine ligand 2 (CCL2) by mammary tumours recruits CCR2-expressing inflammatory monocytes to primary tumours and metastatic sites, and CCL2 neutralization in mice inhibits metastasis by retaining monocytes in the bone marrow. Here we report a paradoxical effect of CCL2 in four syngeneic mouse models of metastatic breast cancer. Surprisingly, interruption of CCL2 inhibition leads to an overshoot of metastases and accelerates death. This is the result of monocyte release from the bone marrow and enhancement of cancer cell mobilization from the primary tumour, as well as blood vessel formation and increased proliferation of metastatic cells in the lungs in an interleukin (IL)-6- and vascular endothelial growth factor (VEGF)-A-dependent manner. Notably, inhibition of CCL2 and IL-6 markedly reduced metastases and increased survival of the animals. CCL2 has been implicated in various neoplasias and adopted as a therapeutic target. However, our results call for caution when considering anti-CCL2 agents as monotherapy in metastatic disease and highlight the tumour microenvironment as a critical determinant of successful anti-metastatic therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/metabolismo , Metástasis de la Neoplasia , Neovascularización Patológica , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Monocitos/citología , Monocitos/metabolismo , Metástasis de la Neoplasia/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Análisis de Supervivencia , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Sci Signal ; 7(329): ra56, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24917593

RESUMEN

Memo is an evolutionarily conserved protein with a critical role in cell motility. We found that Memo was required for migration and invasion of breast cancer cells in vitro and spontaneous lung metastasis from breast cancer cell xenografts in vivo. Biochemical assays revealed that Memo is a copper-dependent redox enzyme that promoted a more oxidized intracellular milieu and stimulated the production of reactive oxygen species (ROS) in cellular structures involved in migration. Memo was also required for the sustained production of the ROS O2- by NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase 1 (NOX1) in breast cancer cells. Memo abundance was increased in >40% of the primary breast tumors tested, was correlated with clinical parameters of aggressive disease, and was an independent prognostic factor of early distant metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Cobre/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Hierro no Heme/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos NOD , Ratones SCID , NADP/genética , NADP/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Proteínas de Hierro no Heme/genética , Superóxidos/metabolismo
12.
Cell Rep ; 4(3): 429-36, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23911287

RESUMEN

A paracrine interaction between epidermal growth factor (EGF)-secreting tumor-associated macrophages (TAMs) and colony-stimulating factor 1 (CSF-1)-secreting breast carcinoma cells promotes invasion and metastasis. Here, we show that mice deficient in the hematopoietic-cell-specific Wiskott-Aldrich syndrome protein (WASp) are unable to support TAM-dependent carcinoma cell invasion and metastasis in both orthotopic and transgenic models of mammary tumorigenesis. Motility and invasion defects of tumor cells were recapitulated ex vivo upon coculture with WASp(-/-) macrophages. Mechanistically, WASp is required for macrophages to migrate toward CSF-1-producing carcinoma cells, as well as for the release of EGF through metalloprotease-dependent shedding of EGF from the cell surface of macrophages. Our findings suggest that WASp acts to support both the migration of TAMs and the production of EGF, which in concert promote breast tumor metastasis.


Asunto(s)
Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Progresión de la Enfermedad , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia , Ratas , Proteína del Síndrome de Wiskott-Aldrich/genética
13.
Curr Protoc Cell Biol ; Chapter 19: 19.7.1-19.7.19, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23456602

RESUMEN

This unit describes the methods that we have been developing for analyzing tumor cell motility in mouse and rat models of breast cancer metastasis. Rodents are commonly used both to provide a mammalian system for studying human tumor cells (as xenografts in immunocompromised mice) as well as for following the development of tumors from a specific tissue type in transgenic lines. The Basic Protocol in this unit describes the standard methods used for generation of mammary tumors and imaging them. Additional protocols for labeling macrophages, blood vessel imaging, and image analysis are also included.


Asunto(s)
Diagnóstico por Imagen/métodos , Neoplasias Mamarias Experimentales/patología , Animales , Vasos Sanguíneos/patología , Humanos , Macrófagos/patología , Ratones , Ratas
14.
Methods Mol Biol ; 872: 15-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22700401

RESUMEN

The analysis of cancer cell behavior in the primary tumor in living animals provides an opportunity to explore the process of invasion and intravasation in the complex microenvironment that is present in vivo. In this chapter, we describe the methods that we have developed for performing intravital imaging of mammary tumors. We provide procedures for generating tumors through injection of tumor cell lines, and multiphoton imaging using a skin-flap tumor dissection and a mammary imaging window.


Asunto(s)
Movimiento Celular/fisiología , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Animales , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones SCID , Ratas
15.
J Mammary Gland Biol Neoplasia ; 17(2): 125-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22581273

RESUMEN

A fundamental hallmark of cancer is progression to metastasis and the growth of breast cancer metastases in lung, bone, liver and/or brain causes fatal complications. Unfortunately, the cellular and biochemical mechanisms of the metastatic process remain ill-defined. Recent application of intravital multiphoton microscopy (MP-IVM) to image fluorescently labeled cells in mouse models of cancer has allowed dynamic observation of this multi-step process at the cellular and subcellular levels. In this article, we discuss the use of MP-IVM in studies of breast cancer metastasis, as well as surgical techniques for exposing tumors prior to imaging. We also describe a versatile multiphoton microscope for imaging tumor-stroma interactions.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Fluorescentes Verdes/metabolismo , Metástasis Linfática/patología , Glándulas Mamarias Animales/patología , Proteínas de Neoplasias/metabolismo , Animales , Neoplasias de la Médula Ósea/patología , Neoplasias de la Médula Ósea/secundario , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/genética , Huésped Inmunocomprometido , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica/instrumentación , Proteínas de Neoplasias/genética , Trasplante de Neoplasias/patología , Células del Estroma/metabolismo , Células del Estroma/patología
16.
J Cell Sci ; 125(Pt 3): 724-34, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22389406

RESUMEN

Invadopodia are proteolytic membrane protrusions formed by highly invasive cancer cells, commonly observed on substrate(s) mimicking extracellular matrix. Although invadopodia are proposed to have roles in cancer invasion and metastasis, direct evidence has not been available. We previously reported that neural Wiskott-Aldrich syndrome protein (N-WASP), a member of WASP family proteins that regulate reorganization of the actin cytoskeleton, is an essential component of invadopodia. Here, we report that N-WASP-mediated invadopodium formation is essential in breast cancer invasion, intravasation and lung metastasis. We established stable cell lines based on MTLn3 rat mammary adenocarcinoma cells that either overexpressed a dominant-negative (DN) N-WASP construct or in which N-WASP expression was silenced by a pSuper N-WASP shRNA. Both the N-WASP shRNA and DN N-WASP cells showed a markedly decreased ability to form invadopodia and degrade extracellular matrix. In addition, formation of invadopodia in primary tumors and collagen I degradation were reduced in the areas of invasion (collagen-rich areas in the invasive edge of the tumor) and in the areas of intravasation (blood-vessel-rich areas). Our results suggest that tumor cells in vivo that have a decreased activity of N-WASP also have a reduced ability to form invadopodia, migrate, invade, intravasate and disseminate to lung compared with tumor cells with parental N-WASP levels.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Invasividad Neoplásica/fisiopatología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/secundario , Animales , Secuencia de Bases , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Mamarias Experimentales/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones SCID , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , ARN Interferente Pequeño/genética , Ratas , Ratas Endogámicas F344 , Proteína Neuronal del Síndrome de Wiskott-Aldrich/antagonistas & inhibidores , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética
17.
Breast Cancer Res ; 14(1): R23, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22314082

RESUMEN

INTRODUCTION: Neu (HER2/ErbB2) is overexpressed in 25% to 30% of human breast cancer, correlating with a poor prognosis. Researchers in previous studies who used the mouse mammary tumor virus Neu-transgenic mouse model (MMTV-Neu) demonstrated that the Neu-YB line had increased production of CXCL12 and increased metastasis, whereas the Neu-YD line had decreased metastasis. In this study, we examined the role of increased production of CXCL12 in tumor cell invasion and malignancy. METHODS: We studied invasion in the tumor microenvironment using multiphoton intravital imaging, in vivo invasion and intravasation assays. CXCL12 signaling was altered by using the CXCR4 inhibitor AMD3100 or by increasing CXCL12 expression. The role of macrophage signaling in vivo was determined using a colony-stimulating factor 1 receptor (CSF-1R) blocking antibody. RESULTS: The Neu-YD strain was reduced in invasion, intravasation and metastasis compared to the Neu-YB and Neu deletion mutant (activated receptor) strains. Remarkably, in the Neu-YB strain, in vivo invasion to epidermal growth factor was dependent on both CXCL12-CXCR4 and CSF1-CSF-1R signaling. Neu-YB tumors had increased macrophage and microvessel density. Overexpression of CXCL12 in rat mammary adenocarcinoma cells increased in vivo invasion as well as microvessel and macrophage density. CONCLUSIONS: Expression of CXCL12 by tumor cells results in increased macrophage and microvessel density and in vivo invasiveness.


Asunto(s)
Adenocarcinoma/secundario , Quimiocina CXCL12/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/metabolismo , Animales , Movimiento Celular , Quimiocina CXCL12/fisiología , Femenino , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Macrófagos/fisiología , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Microscopía por Video , Invasividad Neoplásica , Trasplante de Neoplasias , Células Neoplásicas Circulantes/patología , Comunicación Paracrina , Ratas , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Carga Tumoral , Células Tumorales Cultivadas , Microambiente Tumoral
18.
Artículo en Inglés | MEDLINE | ID: mdl-25419258

RESUMEN

Cancer cells create a unique microenvironment in vivo that enables migration to distant organs. To better understand the tumor micro-environment, special tools and devices are required to monitor the interactions between different cell types and the effects of particular chemical gradients. Our study presents the design and optimization of a versatile chemotaxis device, the nano-intravital device (NANIVID), which consists of etched and bonded glass substrates that create a soluble factor reservoir. The device contains a customized hydrogel blend that is loaded with epidermal growth factor (EGF), which diffuses from the outlet to create a chemotactic gradient that can be sustained for many hours in order to attract specific cells to the device. A microelectrode array is under development for quantification of cell collection and will be incorporated into future device generations. Additionally, the NANIVID can be modified to generate gradients of other soluble factors in order to initiate controlled changes to the microenvironment including the induction of hypoxia, manipulation of extracellular matrix stiffness, etc. The focus of the article is to present the design and optimization of the device towards wide ranging applications of cancer cell dynamics in vitro and, ultimately, implantation for in vivo investigations.

19.
Nat Protoc ; 6(10): 1500-20, 2011 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-21959234

RESUMEN

Characterizing biological mechanisms dependent upon the interaction of many cell types in vivo requires both multiphoton microscope systems capable of expanding the number and types of fluorophores that can be imaged simultaneously while removing the wavelength and tunability restrictions of existing systems, and enhanced software for extracting critical cellular parameters from voluminous 4D data sets. We present a procedure for constructing a two-laser multiphoton microscope that extends the wavelength range of excitation light, expands the number of simultaneously usable fluorophores and markedly increases signal to noise via 'over-clocking' of detection. We also utilize a custom-written software plug-in that simplifies the quantitative tracking and analysis of 4D intravital image data. We begin by describing the optics, hardware, electronics and software required, and finally the use of the plug-in for analysis. We demonstrate the use of the setup and plug-in by presenting data collected via intravital imaging of a mouse model of breast cancer. The procedure may be completed in ∼24 h.


Asunto(s)
Neoplasias Mamarias Experimentales/patología , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Programas Informáticos , Animales , Femenino , Procesamiento de Imagen Asistido por Computador , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Microscopía de Fluorescencia por Excitación Multifotónica/instrumentación
20.
Cold Spring Harb Protoc ; 2011(10): 1167-84, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21969629

RESUMEN

Analysis of the individual steps in metastasis is crucial if insights at the molecular level are to be linked to the cell biology of cancer. A technical hurdle to achieving the analysis of the individual steps of metastasis is the fact that, at the gross level, tumors are heterogeneous in both animal models and patients. Human primary tumors show extensive variation in all properties ranging from growth and morphology of the tumor through tumor-cell density in the blood and formation and growth of metastases. Methods capable of the direct visualization and analysis of tumor-cell behavior at single-cell resolution in vivo have become crucial in advancing the understanding of mechanisms of metastasis, the definition of microenvironment, and the markers related to both. This article discusses the use of high-resolution multiphoton imaging of tumors (specifically breast tumors in mice) in vivo.


Asunto(s)
Neoplasias/patología , Animales , Modelos Animales de Enfermedad , Humanos , Microscopía Confocal , Fotones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...