Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Eur Urol Open Sci ; 29: 19-29, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34337530

RESUMEN

BACKGROUND: Pelvic nodal metastasis in prostate cancer impacts patient outcome negatively. OBJECTIVE: To explore tumor-infiltrating immune cells as a potential predictive tool for regional lymph node (LN) metastasis. DESIGN SETTING AND PARTICIPANTS: We applied multiplex immunofluorescence and targeted transcriptomic analysis on 94 radical prostatectomy specimens in patients with (LN+) or without (LN-) pelvic nodal metastases. Both intraepithelial and stromal infiltrations of immune cells and differentially expressed genes (mRNA and protein levels) were correlated with the nodal status. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The identified CD4 effector cell signature of nodal metastasis was validated in a comparable independent patient cohort of 184 informative cases. Patient outcome analysis and decision curve analysis were performed with the CD4 effector cell density-based signature. RESULTS AND LIMITATIONS: In the discovery cohort, both tumor epithelium and stroma from patients with nodal metastasis had significantly lower infiltration of multiple immune cell types, with stromal CD4 effector cells highlighted as the top candidate marker. Targeted gene expression analysis and confirmatory protein analysis revealed key alteration of extracellular matrix components in tumors with nodal metastasis. Of note, stromal CD4 immune cell density was a significant independent predictor of LN metastasis (odds ratio [OR] = 0.15, p = 0.004), and was further validated as a significant predictor of nodal metastasis in the validation cohort (OR = 0.26, p < 0.001). CONCLUSIONS: Decreased T-cell infiltrates in the primary tumor (particularly CD4 effector cells) are associated with a higher risk of LN metastasis. Future evaluation of CD4-based assays on prostate cancer diagnostic biopsy materials may improve selection of at-risk patients for the treatment of LN metastasis. PATIENT SUMMARY: In this report, we found that cancer showing evidence of cancer metastasis to the lymph nodes tends to have less immune cells present within the tumor. We conclude that the extent of immune cells present within a prostate tumor can help doctors determine the most appropriate treatment plan for individual patients.

3.
Sci Immunol ; 4(36)2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-31175176

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are enriched in mucosal tissues (e.g., lung) and respond to epithelial cell-derived cytokines initiating type 2 inflammation. During inflammation, ILC2 numbers are increased in the lung. However, the mechanisms controlling ILC2 trafficking and motility within inflamed lungs remain unclear and are crucial for understanding ILC2 function in pulmonary immunity. Using several approaches, including lung intravital microscopy, we demonstrate that pulmonary ILC2s are highly dynamic, exhibit amoeboid-like movement, and aggregate in the lung peribronchial and perivascular spaces. They express distinct chemokine receptors, including CCR8, and actively home to CCL8 deposits located around the airway epithelium. Within lung tissue, ILC2s were particularly motile in extracellular matrix-enriched regions. We show that collagen-I drives ILC2 to markedly change their morphology by remodeling their actin cytoskeleton to promote environmental exploration critical for regulating eosinophilic inflammation. Our study provides previously unappreciated insights into ILC2 migratory patterns during inflammation and highlights the importance of environmental guidance cues in the lung in controlling ILC2 dynamics.


Asunto(s)
Pulmón/inmunología , Linfocitos/inmunología , Animales , Movimiento Celular/efectos de los fármacos , Colágeno/inmunología , Eosinófilos/inmunología , Matriz Extracelular/inmunología , Femenino , Fibronectinas/inmunología , Humanos , Inmunidad Innata , Inflamación/inmunología , Interleucina-33/farmacología , Linfocitos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteínas Recombinantes/farmacología
4.
Cell Rep ; 23(5): 1448-1460, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29719257

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is resistant to most therapies including single-agent immunotherapy and has a dense desmoplastic stroma, and most patients present with advanced metastatic disease. We reveal that macrophages are the dominant leukocyte population both in human PDAC stroma and autochthonous models, with an important functional contribution to the squamous subtype of human PDAC. We targeted macrophages in a genetic PDAC model using AZD7507, a potent selective inhibitor of CSF1R. AZD7507 caused shrinkage of established tumors and increased mouse survival in this difficult-to-treat model. Malignant cell proliferation diminished, with increased cell death and an enhanced T cell immune response. Loss of macrophages rewired other features of the TME, with global changes in gene expression akin to switching PDAC subtypes. These changes were markedly different to those elicited when neutrophils were targeted via CXCR2. These results suggest targeting the myeloid cell axis may be particularly efficacious in PDAC, especially with CSF1R inhibitors.


Asunto(s)
Carcinoma Ductal Pancreático/inmunología , Macrófagos/inmunología , Modelos Inmunológicos , Proteínas de Neoplasias/inmunología , Neoplasias Pancreáticas/inmunología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Linfocitos T/inmunología , Adulto , Compuestos de Anilina/farmacología , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Femenino , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Macrófagos/patología , Masculino , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Linfocitos T/patología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
PLoS One ; 13(1): e0183585, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29293509

RESUMEN

FRET biosensors have proven very useful tools for studying the activation of specific signalling pathways in living cells. Most biosensors designed to date have been predicated on fluorescent protein pairs that were identified by, and for use in, intensity based measurements, however fluorescence lifetime provides a more reliable measurement of FRET. Both the technology and fluorescent proteins available for FRET have moved on dramatically in the last decade. Lifetime imaging systems have become increasingly accessible and user-friendly, and there is an entire field of biology dedicated to refining and adapting different characteristics of existing and novel fluorescent proteins. This growing pool of fluorescent proteins includes the long-lifetime green and cyan fluorescent proteins Clover and mTurquoise2, the red variant mRuby2, and the dark acceptor sREACh. Here, we have tested these donors and acceptors in appropriate combinations against the standard or recommended norms (EGFP and mTFP as donors, mCherry and either Ypet or Venus as acceptors) to determine if they could provide more reliable, reproducible and quantifiable FLIM-FRET data to improve on the dynamic range compared to other donors and breadth of application of biosensor technologies. These tests were performed for comparison on both a wide-field, frequency domain system and a multiphoton, TCSPC time domain FLIM system. Clover proved to be an excellent donor with extended dynamic range in combination with mCherry on both platforms, while mRuby2 showed a high degree of variability and poor FRET efficiencies in all cases. mTFP-Venus was the most consistent cyan-yellow pair between the two FLIM methodologies, but mTurquoise2 has better dynamic range and transfers energy consistently over time to the dark acceptor sRCh. Combination of mTFP-sRCh with Clover-mCherry would allow the simultaneous use of two FLIM-FRET biosensors within one sample by eliminating the crosstalk between the yellow acceptor and green donor emissions.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Luminiscentes/metabolismo , Técnicas Biosensibles , Fluorescencia , Unión Proteica
6.
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
7.
EMBO J ; 36(16): 2373-2389, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28694244

RESUMEN

Tumor progression alters the composition and physical properties of the extracellular matrix. Particularly, increased matrix stiffness has profound effects on tumor growth and metastasis. While endothelial cells are key players in cancer progression, the influence of tumor stiffness on the endothelium and the impact on metastasis is unknown. Through quantitative mass spectrometry, we find that the matricellular protein CCN1/CYR61 is highly regulated by stiffness in endothelial cells. We show that stiffness-induced CCN1 activates ß-catenin nuclear translocation and signaling and that this contributes to upregulate N-cadherin levels on the surface of the endothelium, in vitro This facilitates N-cadherin-dependent cancer cell-endothelium interaction. Using intravital imaging, we show that knockout of Ccn1 in endothelial cells inhibits melanoma cancer cell binding to the blood vessels, a critical step in cancer cell transit through the vasculature to metastasize. Targeting stiffness-induced changes in the vasculature, such as CCN1, is therefore a potential yet unappreciated mechanism to impair metastasis.


Asunto(s)
Comunicación Celular , Células Endoteliales/fisiología , Melanocitos/fisiología , Cadherinas/análisis , Línea Celular , Proteína 61 Rica en Cisteína/análisis , Regulación de la Expresión Génica , Humanos , Espectrometría de Masas , beta Catenina/análisis
8.
Sci Transl Med ; 9(384)2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28381539

RESUMEN

The emerging standard of care for patients with inoperable pancreatic cancer is a combination of cytotoxic drugs gemcitabine and Abraxane, but patient response remains moderate. Pancreatic cancer development and metastasis occur in complex settings, with reciprocal feedback from microenvironmental cues influencing both disease progression and drug response. Little is known about how sequential dual targeting of tumor tissue tension and vasculature before chemotherapy can affect tumor response. We used intravital imaging to assess how transient manipulation of the tumor tissue, or "priming," using the pharmaceutical Rho kinase inhibitor Fasudil affects response to chemotherapy. Intravital Förster resonance energy transfer imaging of a cyclin-dependent kinase 1 biosensor to monitor the efficacy of cytotoxic drugs revealed that priming improves pancreatic cancer response to gemcitabine/Abraxane at both primary and secondary sites. Transient priming also sensitized cells to shear stress and impaired colonization efficiency and fibrotic niche remodeling within the liver, three important features of cancer spread. Last, we demonstrate a graded response to priming in stratified patient-derived tumors, indicating that fine-tuned tissue manipulation before chemotherapy may offer opportunities in both primary and metastatic targeting of pancreatic cancer.


Asunto(s)
Progresión de la Enfermedad , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/uso terapéutico , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Paclitaxel Unido a Albúmina/farmacología , Paclitaxel Unido a Albúmina/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Técnicas Biosensibles , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colágeno/metabolismo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Matriz Extracelular/metabolismo , Humanos , Hígado/patología , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Quinasas Asociadas a rho/metabolismo , Familia-src Quinasas/metabolismo , Gemcitabina
9.
EMBO Mol Med ; 9(2): 198-218, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28031255

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer death; identifying PDAC enablers may reveal potential therapeutic targets. Expression of the actomyosin regulatory ROCK1 and ROCK2 kinases increased with tumor progression in human and mouse pancreatic tumors, while elevated ROCK1/ROCK2 expression in human patients, or conditional ROCK2 activation in a KrasG12D/p53R172H mouse PDAC model, was associated with reduced survival. Conditional ROCK1 or ROCK2 activation promoted invasive growth of mouse PDAC cells into three-dimensional collagen matrices by increasing matrix remodeling activities. RNA sequencing revealed a coordinated program of ROCK-induced genes that facilitate extracellular matrix remodeling, with greatest fold-changes for matrix metalloproteinases (MMPs) Mmp10 and Mmp13 MMP inhibition not only decreased collagen degradation and invasion, but also reduced proliferation in three-dimensional contexts. Treatment of KrasG12D/p53R172H PDAC mice with a ROCK inhibitor prolonged survival, which was associated with increased tumor-associated collagen. These findings reveal an ancillary role for increased ROCK signaling in pancreatic cancer progression to promote extracellular matrix remodeling that facilitates proliferation and invasive tumor growth.


Asunto(s)
Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Colágeno/metabolismo , Transducción de Señal , Quinasas Asociadas a rho/metabolismo , Animales , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Ratones , Análisis de Supervivencia
10.
Cell Rep ; 14(1): 152-167, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26725115

RESUMEN

E-cadherin-mediated cell-cell junctions play a prominent role in maintaining the epithelial architecture. The disruption or deregulation of these adhesions in cancer can lead to the collapse of tumor epithelia that precedes invasion and subsequent metastasis. Here we generated an E-cadherin-GFP mouse that enables intravital photobleaching and quantification of E-cadherin mobility in live tissue without affecting normal biology. We demonstrate the broad applications of this mouse by examining E-cadherin regulation in multiple tissues, including mammary, brain, liver, and kidney tissue, while specifically monitoring E-cadherin mobility during disease progression in the pancreas. We assess E-cadherin stability in native pancreatic tissue upon genetic manipulation involving Kras and p53 or in response to anti-invasive drug treatment and gain insights into the dynamic remodeling of E-cadherin during in situ cancer progression. FRAP in the E-cadherin-GFP mouse, therefore, promises to be a valuable tool to fundamentally expand our understanding of E-cadherin-mediated events in native microenvironments.


Asunto(s)
Cadherinas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Imagen Óptica/métodos , Microambiente Tumoral , Animales , Cadherinas/genética , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Neoplasias Experimentales/genética , Especificidad de Órganos , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Curr Biol ; 25(11): 1520-5, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25981793

RESUMEN

Mesenchymal cell motility is driven by polarized actin polymerization [1]. Signals at the leading edge recruit actin polymerization machinery to promote membrane protrusion, while matrix adhesion generates tractive force to propel forward movement. To work effectively, cell motility is regulated by a complex network of signaling events that affect protein activity and localization. H2O2 has an important role as a diffusible second messenger [2], and mediates its effects through oxidation of cysteine thiols. One cell activity influenced by H2O2 is motility [3]. However, a lack of sensitive and H2O2-specific probes for measurements in live cells has not allowed for direct observation of H2O2 accumulation in migrating cells or protrusions. In addition, the identities of proteins oxidized by H2O2 that contribute to actin dynamics and cell motility have not been characterized. We now show, as determined by fluorescence lifetime imaging microscopy, that motile cells generate H2O2 at membranes and cell protrusions and that H2O2 inhibits cofilin activity through oxidation of cysteines 139 (C139) and 147 (C147). Molecular modeling suggests that C139 oxidation would sterically hinder actin association, while the increased negative charge of oxidized C147 would lead to electrostatic repulsion of the opposite negatively charged surface. Expression of oxidation-resistant cofilin impairs cell spreading, adhesion, and directional migration. These findings indicate that H2O2 production contributes to polarized cell motility through localized cofilin inhibition and that there are additional proteins oxidized during cell migration that might have similar roles.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Movimiento Celular , Peróxido de Hidrógeno/metabolismo , Adhesión Celular , Línea Celular Tumoral , Humanos , Oxidación-Reducción
12.
Cell Adh Migr ; 8(5): 478-86, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25482620

RESUMEN

Optimising response to tyrosine kinase inhibitors in cancer remains an extensive field of research. Intravital imaging is an emerging tool, which can be used in drug discovery to facilitate and fine-tune maximum drug response in live tumors. A greater understanding of intratumoural delivery and pharmacodynamics of a drug can be obtained by imaging drug target-specific fluorescence resonance energy transfer (FRET) biosensors in real time. Here, we outline our recent work using a Src-FRET biosensor as a readout of Src activity to gauge optimal tyrosine kinase inhibition in response to dasatinib treatment regimens in vivo. By simultaneously monitoring both the inhibition of Src using FRET imaging, and the modulation of the surrounding extracellular matrix using second harmonic generation (SHG) imaging, we were able to show enhanced drug penetrance and delivery to live pancreatic tumors. We discuss the implications of this dual intravital imaging approach in the context of altered tumor-stromal interactions, while summarising how this approach could be applied to assess other combination strategies or tyrosine kinase inhibitors in a preclinical setting.


Asunto(s)
Matriz Extracelular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Familia-src Quinasas/metabolismo , Técnicas Biosensibles , Línea Celular Tumoral , Dasatinib , Matriz Extracelular/enzimología , Transferencia Resonante de Energía de Fluorescencia , Humanos
13.
Cell Rep ; 6(6): 1153-1164, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24630994

RESUMEN

The small G protein family Rac has numerous regulators that integrate extracellular signals into tight spatiotemporal maps of its activity to promote specific cell morphologies and responses. Here, we have generated a mouse strain, Rac-FRET, which ubiquitously expresses the Raichu-Rac biosensor. It enables FRET imaging and quantification of Rac activity in live tissues and primary cells without affecting cell properties and responses. We assessed Rac activity in chemotaxing Rac-FRET neutrophils and found enrichment in leading-edge protrusions and unexpected longitudinal shifts and oscillations during protruding and stalling phases of migration. We monitored Rac activity in normal or disease states of intestinal, liver, mammary, pancreatic, and skin tissue, in response to stimulation or inhibition and upon genetic manipulation of upstream regulators, revealing unexpected insights into Rac signaling during disease development. The Rac-FRET strain is a resource that promises to fundamentally advance our understanding of Rac-dependent responses in primary cells and native environments.


Asunto(s)
Neutrófilos/enzimología , Proteínas de Unión al GTP rac/metabolismo , Animales , Activación Enzimática , Transferencia Resonante de Energía de Fluorescencia/métodos , Ratones , Neutrófilos/citología , Transducción de Señal , Análisis Espacio-Temporal , Proteínas de Unión al GTP rac/química
14.
J Biomed Opt ; 19(1): 16021, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24474507

RESUMEN

We have developed a nonlinear adaptive optics microscope utilizing a deformable membrane mirror (DMM) and demonstrated its use in compensating for system- and sample-induced aberrations. The optimum shape of the DMM was determined with a random search algorithm optimizing on either two photon fluorescence or second harmonic signals as merit factors. We present here several strategies to overcome photobleaching issues associated with lengthy optimization routines by adapting the search algorithm and the experimental methodology. Optimizations were performed on extrinsic fluorescent dyes, fluorescent beads loaded into organotypic tissue cultures and the intrinsic second harmonic signal of these cultures. We validate the approach of using these preoptimized mirror shapes to compile a robust look-up table that can be applied for imaging over several days and through a variety of tissues. In this way, the photon exposure to the fluorescent cells under investigation is limited to imaging. Using our look-up table approach, we show signal intensity improvement factors ranging from 1.7 to 4.1 in organotypic tissue cultures and freshly excised mouse tissue. Imaging zebrafish in vivo, we demonstrate signal improvement by a factor of 2. This methodology is easily reproducible and could be applied to many photon starved experiments, for example fluorescent life time imaging, or when photobleaching is a concern.


Asunto(s)
Diagnóstico por Imagen/métodos , Óptica y Fotónica , Fotoblanqueo , Algoritmos , Animales , Diferenciación Celular , Colágeno/química , Colorantes Fluorescentes/química , Humanos , Ratones , Microscopía Confocal , Microscopía Fluorescente , Neoplasias/metabolismo , Neoplasias/patología , Distribución Normal , Fotones , Refractometría , Piel/patología , Programas Informáticos , Pez Cebra
15.
Cancer Res ; 73(15): 4674-86, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23749641

RESUMEN

Cancer invasion and metastasis occur in a complex three-dimensional (3D) environment, with reciprocal feedback from the surrounding host tissue and vasculature-governing behavior. In this study, we used a novel intravital method that revealed spatiotemporal regulation of Src activity in response to the anti-invasive Src inhibitor dasatinib. A fluorescence lifetime imaging microscopy-fluorescence resonance energy transfer (FLIM-FRET) Src biosensor was used to monitor drug-targeting efficacy in a transgenic p53-mutant mouse model of pancreatic cancer. In contrast to conventional techniques, FLIM-FRET analysis allowed for accurate, time-dependent, live monitoring of drug efficacy and clearance in live tumors. In 3D organotypic cultures, we showed that a spatially distinct gradient of Src activity exists within invading tumor cells, governed by the depth of penetration into complex matrices. In parallel, this gradient was also found to exist within live tumors, where Src activity is enhanced at the invasive border relative to the tumor cortex. Upon treatment with dasatinib, we observed a switch in activity at the invasive borders, correlating with impaired metastatic capacity in vivo. Src regulation was governed by the proximity of cells to the host vasculature, as cells distal to the vasculature were regulated differentially in response to drug treatment compared with cells proximal to the vasculature. Overall, our results in live tumors revealed that a threshold of drug penetrance exists in vivo and that this can be used to map areas of poor drug-targeting efficiency within specific tumor microenvironments. We propose that using FLIM-FRET in this capacity could provide a useful preclinical tool in animal models before clinical translation.


Asunto(s)
Antineoplásicos/farmacología , Transferencia Resonante de Energía de Fluorescencia/métodos , Imagenología Tridimensional/métodos , Neoplasias Pancreáticas/metabolismo , Pirimidinas/farmacología , Tiazoles/farmacología , Familia-src Quinasas/metabolismo , Animales , Técnicas Biosensibles/métodos , Células Cultivadas , Dasatinib , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Microscopía Fluorescente
16.
Cell Stem Cell ; 12(6): 761-73, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23665120

RESUMEN

The Adenomatous Polyposis Coli (APC) gene is mutated in the majority of colorectal cancers (CRCs). Loss of APC leads to constitutively active WNT signaling, hyperproliferation, and tumorigenesis. Identification of pathways that facilitate tumorigenesis after APC loss is important for therapeutic development. Here, we show that RAC1 is a critical mediator of tumorigenesis after APC loss. We find that RAC1 is required for expansion of the LGR5 intestinal stem cell (ISC) signature, progenitor hyperproliferation, and transformation. Mechanistically, RAC1-driven ROS and NF-κB signaling mediate these processes. Together, these data highlight that ROS production and NF-κB activation triggered by RAC1 are critical events in CRC initiation.


Asunto(s)
Neoplasias Colorrectales/patología , Intestino Delgado/citología , FN-kappa B/metabolismo , Neuropéptidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células Madre/citología , Proteínas Wnt/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Células Madre/metabolismo
17.
FEBS J ; 280(21): 5177-97, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23678945

RESUMEN

The integration of signal transduction pathways plays a fundamental role in governing disease initiation, progression and outcome. It is therefore necessary to understand disease at the signalling level to enable effective treatment and to intervene in its progression. The recent extension of in vitro subcellular image-based analysis to live in vivo modelling of disease is providing a more complete picture of real-time, dynamic signalling processes or drug responses in live tissue. Intravital imaging offers alternative strategies for studying disease and embraces the biological complexities that govern disease progression. In the present review, we highlight how three-dimensional or live intravital imaging has uncovered novel insights into biological mechanisms or modes of drug action. Furthermore, we offer a prospective view of how imaging applications may be integrated further with the aim of understanding disease in a more physiological and functional manner within the framework of the drug discovery process.


Asunto(s)
Antineoplásicos/farmacología , Diagnóstico por Imagen , Descubrimiento de Drogas , Neoplasias/diagnóstico , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Animales , Humanos , Microscopía Fluorescente , Neoplasias/tratamiento farmacológico
18.
Small GTPases ; 2(4): 239-244, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22145098

RESUMEN

Many conceptual advances in biology have been achieved by experimental studies using planar two-dimensional cell culture systems. Recent adaptations of molecular techniques to three-dimensional model systems are bridging the gap in our understanding of biological events in vitro and in vivo in the study of disease progression. Recently, in vitro studies using Förster resonance energy transfer (FRET) have shown that the prototypical RhoGTPases Cdc42, Rac and RhoA are temporally and spatially synchronized during cell migration, with initial RhoA activity inducing protrusion prior to activation of Rac. This simultaneous FRET approach illustrates the tight control and dynamic regulation of RhoGTPase activity necessary for coordinated cell migration in vitro. Here, we discuss our recent work using FLIM-FRET analysis in a three-dimensional setting to reveal another layer of regulation in which RhoA activity is governed by the extracellular microenvironment. We demonstrate that RhoA is spatially regulated into discrete fractions of activity at the leading edge and rear of cells during invasion in vivo or within three-dimensional matrices. Significantly, this spatial regulation of RhoA was absent in two-dimensional in vitro settings. This distinct sub-cellular regulation of RhoA at the poles of invading cells in three-dimensions sets a precedent that other RhoGTPases or signaling proteins may also be differentially regulated in a con-text-dependent manner during key biological processes such as invasion.

19.
J Vis Exp ; (56): e3089, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-22025017

RESUMEN

Cell migration is fundamental to many aspects of biology, including development, wound healing, the cellular responses of the immune system, and metastasis of tumor cells. Migration has been studied on glass coverslips in order to make cellular dynamics amenable to investigation by light microscopy. However, it has become clear that many aspects of cell migration depend on features of the local environment including its elasticity, protein composition, and pore size, which are not faithfully represented by rigid two dimensional substrates such as glass and plastic. Furthermore, interaction with other cell types, including stromal fibroblasts and immune cells, has been shown to play a critical role in promoting the invasion of cancer cells. Investigation at the molecular level has increasingly shown that molecular dynamics, including response to drug treatment, of identical cells are significantly different when compared in vitro and in vivo. Ideally, it would be best to study cell migration in its naturally occurring context in living organisms, however this is not always possible. Intermediate tissue culture systems, such as cell derived matrix, matrigel, organotypic culture (described here) tissue explants, organoids, and xenografts, are therefore important experimental intermediates. These systems approximate certain aspects of an in vivo environment but are more amenable to experimental manipulation such as use of stably transfected cell lines, drug treatment regimes, long term and high-resolution imaging. Such intermediate systems are especially useful as proving grounds to validate probes and establish parameters required to image the dynamic response of cells and fluorescent reporters prior to undertaking imaging in vivo. As such, they can serve an important role in reducing the need for experiments on living animals.


Asunto(s)
Colágeno Tipo I , Técnicas de Cultivo de Órganos/métodos , Técnicas de Cultivo de Tejidos/métodos , Animales , Medios de Cultivo , Fibroblastos/citología , Ratas , Piel/citología
20.
J Cell Sci ; 124(Pt 17): 2877-90, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21878495

RESUMEN

Advances in fluorescence microscopy have enabled the study of membrane diffusion, cell adhesion and signal transduction at the molecular level in living cells grown in culture. By contrast, imaging in living organisms has primarily been restricted to the localization and dynamics of cells in tissues. Now, imaging of molecular dynamics is on the cusp of progressing from cell culture to living tissue. This transition has been driven by the understanding that the microenvironment critically determines many developmental and pathological processes. Here, we review recent progress in fluorescent protein imaging in vivo by drawing primarily on cancer-related studies in mice. We emphasize the need for techniques that can be easily combined with genetic models and complement fluorescent protein imaging by providing contextual information about the cellular environment. In this Commentary we will consider differences between in vitro and in vivo experimental design and argue for an approach to in vivo imaging that is built upon the use of intermediate systems, such as 3-D and explant culture models, which offer flexibility and control that is not always available in vivo. Collectively, these methods present a paradigm shift towards the molecular-level investigation of disease and therapy in animal models of disease.


Asunto(s)
Microscopía Fluorescente/métodos , Modelos Animales , Imagen Molecular/métodos , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...