Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(5)2021 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-33653008

RESUMEN

Gold nanoparticles (AuNPs) are foreseen as a promising tool in nanomedicine, both as drug carriers and radiosensitizers. They have been also proposed as a potential anticancer drug due to the anti-angiogenic effect in tumor tissue. In this work we investigated the effect of citrate-coated AuNPs of nominal diameter 20 nm on the growth and metastatic potential of 4T1 cells originated from a mouse mammary gland tumor inoculated into the mammary fat pad of Balb/ccmdb mice. To evaluate whether AuNPs can prevent the tumor growth, one group of inoculated mice was intragastrically (i.g.) administered with 1 mg/kg of AuNPs daily from day 1 to day 14 after cancer cell implantation. To evaluate whether AuNPs can attenuate the tumor growth, the second group was intravenously (i.v.) administered with 1 or 5 mg/kg of AuNPs, twice on day 5 and day 14 after inoculation. We did not observe any anticancer activity of i.v. nor i.g. administered AuNPs, as they did not affect neither the primary tumor growth rate nor the number of lung metastases. Unexpectedly, both AuNP treatment regimens caused a marked vasodilating effect in the tumor tissue. As no change of potential angiogenic genes (Fgf2, Vegfa) nor inducible nitric oxygenase (Nos2) was observed, we proposed that the vasodilation was caused by AuNP-dependent decomposition of nitrosothiols and direct release of nitric oxide in the tumor tissue.


Asunto(s)
Ácido Cítrico/uso terapéutico , Oro/uso terapéutico , Neoplasias Mamarias Animales/irrigación sanguínea , Nanopartículas del Metal/uso terapéutico , Animales , Línea Celular Tumoral , Ácido Cítrico/administración & dosificación , Femenino , Oro/administración & dosificación , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/terapia , Nanopartículas del Metal/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Nanomedicina , Tamaño de la Partícula , Vasodilatación
2.
Nanomedicine ; 21: 102074, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31376571

RESUMEN

The assessment of vascular permeability of malignant tumor plays an important role in the diagnosis and treatment of cancer. Dynamic contrast-enhanced magnetic resonance image (DCE-MRI) using Gd-encapsulated carbonaceous dots and Gd-DTPA-BMA as contrast agents was performed in 4T1 mouse breast cancer and HCC827 human non-small-cell lung cancer (NSNLC) xenograft models. Histopathological parameters of tumor vascularity microvessel density (MVD), microvessel area (MVA), endothelial area (EA) and α-SMA CD31 Co-expression (α-SMA/CD31%) were compared with the DCE-MRI parameters. Results demonstrated that DCE-MRI with the new nanoparticle Gd@C-dots can noninvasively evaluate vascular permeability. Ktrans measured by DCE-MRI with Gd@C-dots is an accurate parameter for the characterization of tumor permeability. EA is a reliable microvessel parameter to evaluate vessel permeability.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Medios de Contraste , Endotelio Vascular/diagnóstico por imagen , Gadolinio , Neoplasias Pulmonares/diagnóstico por imagen , Imagen por Resonancia Magnética , Neoplasias Mamarias Animales/diagnóstico por imagen , Nanopartículas/química , Neovascularización Patológica/diagnóstico por imagen , Animales , Carcinoma de Pulmón de Células no Pequeñas/irrigación sanguínea , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Medios de Contraste/química , Medios de Contraste/farmacocinética , Medios de Contraste/farmacología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Gadolinio/química , Gadolinio/farmacocinética , Gadolinio/farmacología , Xenoinjertos , Humanos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Trasplante de Neoplasias , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Trasplante Isogénico
3.
J Cell Mol Med ; 22(12): 5939-5954, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30291675

RESUMEN

The activity of a cell-surface ecto-adenosine deaminase (eADA) is markedly increased in the endothelial activation and vascular inflammation leading to decreased adenosine concentration and alterations in adenosine signalling. Depending on the specific pathway activated, extracellular purines mediate host cell response or regulate growth and cytotoxicity on tumour cells. The aim of this study was to test the effects of adenosine deaminase inhibition by 2'deoxycoformycin (dCF) on the breast cancer development. dCF treatment decreased a tumour growth and a final tumour mass in female BALB/c mice injected orthotopically with 4T1 cancer cells. dCF also counteracted cancer-induced endothelial dysfunction in orthotopic and intravenous 4T1 mouse breast cancer models. In turn, this low dCF dose had a minor effect on immune stimulation exerted by 4T1 cell implantation. In vitro studies revealed that dCF suppressed migration and invasion of 4T1 cells via A2a and A3 adenosine receptor activation as well as 4T1 cell adhesion and transmigration through the endothelial cell layer via A2a receptor stimulation. Similar effects of dCF were observed in human breast cancer cells. Moreover, dCF improved a barrier function of endothelial cells decreasing its permeability. This study highlights beneficial effects of adenosine deaminase inhibition on breast cancer development. The inhibition of adenosine deaminase activity by dCF reduced tumour size that was closely related to the decreased aggressiveness of tumour cells by adenosine receptor-dependent mechanisms and endothelial protection.


Asunto(s)
Inhibidores de la Adenosina Desaminasa/farmacología , Progresión de la Enfermedad , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Receptores Purinérgicos P1/metabolismo , Adenosina Desaminasa/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Espacio Extracelular/metabolismo , Femenino , Humanos , Neoplasias Mamarias Animales/sangre , Neoplasias Mamarias Animales/irrigación sanguínea , Ratones Endogámicos BALB C , Invasividad Neoplásica , Nucleótidos/sangre , Pentostatina/farmacología , Fenotipo , Migración Transendotelial y Transepitelial/efectos de los fármacos
4.
Breast Cancer Res ; 20(1): 20, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29566737

RESUMEN

BACKGROUND: Perfusion of breast cancer tissue limits oxygen availability and metabolism but angiogenesis inhibitors have hitherto been unsuccessful for breast cancer therapy. In order to identify abnormalities and possible therapeutic targets in mature cancer arteries, we here characterize the structure and function of cancer feed arteries and corresponding control arteries from female FVB/N mice with ErbB2-induced breast cancer. METHODS: We investigated the contractile function of breast cancer feed arteries and matched control arteries by isometric myography and evaluated membrane potentials and intracellular [Ca2+] using sharp electrodes and fluorescence microscopy, respectively. Arterial wall structure is assessed by transmission light microscopy of arteries mounted in wire myographs and by evaluation of histological sections using the unbiased stereological disector technique. We determined the expression of messenger RNA by reverse transcription and quantitative polymerase chain reaction and studied receptor expression by confocal microscopy of arteries labelled with the BODIPY-tagged α1-adrenoceptor antagonist prazosin. RESULTS: Breast cancer feed arteries are thin-walled and produce lower tension than control arteries of similar diameter in response to norepinephrine, thromboxane-analog U46619, endothelin-1, and depolarization with elevated [K+]. Fewer layers of similarly-sized vascular smooth muscle cells explain the reduced media thickness of breast cancer arteries. Evidenced by lower media stress, norepinephrine-induced and thromboxane-induced tension development of breast cancer arteries is reduced more than is explained by the thinner media. Conversely, media stress during stimulation with endothelin-1 and elevated [K+] is similar between breast cancer and control arteries. Correspondingly, vascular smooth muscle cell depolarizations and intracellular Ca2+ responses are attenuated in breast cancer feed arteries during norepinephrine but not during endothelin-1 stimulation. Protein expression of α1-adrenoceptors and messenger RNA levels for α1A-adrenoceptors are lower in breast cancer arteries than control arteries. Sympathetic vasocontraction elicited by electrical field stimulation is inhibited by α1-adrenoceptor blockade and reduced in breast cancer feed arteries compared to control arteries. CONCLUSION: Thinner media and lower α1-adrenoceptor expression weaken contractions of breast cancer feed arteries in response to sympathetic activity. We propose that abnormalities in breast cancer arteries can be exploited to modify tumor perfusion and thereby either starve cancer cells or facilitate drug and oxygen delivery during chemotherapy or radiotherapy.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Mamarias Animales/genética , Neovascularización Patológica/genética , Receptores Adrenérgicos alfa 1/genética , Antagonistas de Receptores Adrenérgicos alfa 1/administración & dosificación , Animales , Arterias/crecimiento & desarrollo , Arterias/patología , Arterias/ultraestructura , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Calcio/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/patología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Contracción Muscular/efectos de los fármacos , Miografía , Neovascularización Patológica/patología , Norepinefrina/administración & dosificación , Oxígeno/metabolismo , Prazosina/administración & dosificación , ARN Mensajero/genética , Receptor ErbB-2/genética , Receptores Adrenérgicos alfa 1/administración & dosificación
5.
Angiogenesis ; 21(3): 653-665, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29691683

RESUMEN

Anti-angiogenesis therapy is an established therapeutic strategy for cancer. The endogenous angiogenic inhibitor angiostatin contains the first 3-4 kringle domains of plasminogen and inhibits both angiogenesis and vascular permeability. We present here a 10-residue peptide, Angio-3, derived from plasminogen kringle 3, which retains the functions of angiostatin in inhibiting both angiogenesis and vascular permeability. NMR studies indicate that Angio-3 holds a solution structure similar to the corresponding region of kringle 3. Mechanistically, Angio-3 inhibited both VEGF- and bFGF-induced angiogenesis by inhibiting EC proliferation and migration while inducing apoptosis. Inhibition of VEGF-induced vascular permeability results from its ability to impede VEGF-induced dissociation of adherens junction and tight junction proteins as well as the formation of actin stress fibers. When administered intravenously, Angio-3 inhibited subcutaneous breast cancer and melanoma growth by suppressing both tumor angiogenesis and intra-tumor vascular permeability. Hence, Angio-3 is a novel dual inhibitor of angiogenesis and vascular permeability. It is valuable as a lead peptide that can be further developed as therapeutics for diseases involving excessive angiogenesis and/or vascular permeability.


Asunto(s)
Permeabilidad Capilar , Células Endoteliales de la Vena Umbilical Humana/patología , Neoplasias Mamarias Animales , Melanoma Experimental , Neovascularización Patológica/metabolismo , Péptidos/farmacología , Plasminógeno/farmacología , Animales , Apoptosis/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Imagen por Resonancia Magnética , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/patología , Péptidos/síntesis química , Péptidos/química , Plasminógeno/química , Fibras de Estrés/metabolismo , Fibras de Estrés/patología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Ultrasound Med ; 36(12): 2459-2466, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28646597

RESUMEN

OBJECTIVES: This study aimed to evaluate the impact of long-term exercise training on the vascularization of rat mammary tumors. METHODS: Female rats were divided into 4 groups: N-methyl-N-nitrosourea (MNU) treated sedentary, MNU treated exercised, control sedentary, and control exercised. Tumor development was induced in the MNU groups by MNU administration. Exercised groups were trained for 35 weeks. Tumor vascularization was evaluated by pulsed Doppler and contrast-enhanced ultrasonography. RESULTS: The pulsatility and resistive indices were slightly higher in the MNU sedentary group (P > .05). Mammary tumors mainly had centripetal and heterogeneous enhancement of the contrast, clear margins, and the presence of penetrating vessels. The MNU exercised group had a lower arrival time and time to peak and higher peak intensity, wash-in, and wash-out (P > .05). The area under the curve was similar between groups (P > .05). CONCLUSIONS: The contrast-enhanced ultrasonographic study did not detect differences in mammary tumor vascularization between MNU sedentary and MNU exercised groups previously detected by power Doppler imaging, B-flow imaging, and immunohistochemistry.


Asunto(s)
Medios de Contraste , Aumento de la Imagen/métodos , Neoplasias Mamarias Animales/irrigación sanguínea , Condicionamiento Físico Animal , Ultrasonografía/métodos , Animales , Modelos Animales de Enfermedad , Femenino , Neoplasias Mamarias Animales/diagnóstico por imagen , Neoplasias Mamarias Animales/patología , Neovascularización Patológica/diagnóstico por imagen , Ratas , Ratas Sprague-Dawley , Tiempo
7.
Microvasc Res ; 108: 69-74, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27569845

RESUMEN

PRIMARY OBJECTIVE: Utilizing the detailed vascular network obtained from micro-computed tomography (µCT) to establish a mathematical model of the temporal molecular distribution within a murine C3H mammary carcinoma. PROCEDURES: Female CDF1 mice with a C3H mammary carcinoma on the right rear foot were used in this study. Dynamic information for each tumour was achieved by Dynamic Contrast Enhanced-Magnetic Resonance Imaging (DCE-MRI) on a 16.4 T system. Detailed morphologic information on the tumour vasculature was obtained by ex vivo µCT and compared to CD34 immunohistochemical staining of tissue sections. The reconstructed vascular network served as origin for the diffusion (described by the apparent diffusion coefficient) within the tumour (the restricted volume described by the interstitial volume fraction derived from DCE-MRI). The resulting partial differential equation was solved using Finite-Element and a combined mathematical graph describing molecular distribution within the tumour was obtained. RESULTS: The established molecular distribution model predicted a heterogeneous distribution throughout the tumour related to the layout of the vascular network. Central tumour section concentration-time curves estimated from the established molecular distribution model were compared with physical measurements obtained by DCE-MRI of the same tumours and showed excellent correlation. CONCLUSIONS: A mathematical model describing temporal molecular distribution based on detailed vascular network structures was established and compared to DCE-MRI. The improved morphological insight will enhance future studies of heterogeneous tumours.


Asunto(s)
Antineoplásicos/metabolismo , Vasos Sanguíneos/diagnóstico por imagen , Angiografía por Tomografía Computarizada , Simulación por Computador , Imagen por Resonancia Magnética , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/diagnóstico por imagen , Modelos Biológicos , Técnicas de Diagnóstico Molecular/métodos , Microtomografía por Rayos X , Animales , Antígenos CD34/metabolismo , Antineoplásicos/administración & dosificación , Transporte Biológico , Vasos Sanguíneos/metabolismo , Difusión , Femenino , Análisis de Elementos Finitos , Inmunohistoquímica , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/metabolismo , Ratones , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
8.
Eur Radiol ; 26(6): 1843-51, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26334513

RESUMEN

OBJECTIVES: Intravital imaging within heterogenic solid tumours is important for understanding blood perfusion profiles responsible for establishment of multiple parameters within the tumour mass, such as hypoxic and nutrition gradients, cell viability, proliferation and drug response potentials. METHODS: Herein, we developed a method based on a volumetric multispectral optoacoustic tomography (vMSOT) for cancer imaging in preclinical models and explored its capacity for three-dimensional imaging of anatomic, vascular and functional tumour profiles in real time. RESULTS: In contrast to methods based on cross-sectional (2D) image acquisition as a basis for 3D rendering, vMSOT has attained concurrent observations from the entire tumour volume at 10 volumetric frames per second. This truly four dimensional imaging performance has enabled here the simultaneous assessment of blood oxygenation gradients and vascularization in solid breast tumours and revealed different types of blood perfusion profiles in-vivo. CONCLUSION: The newly introduced capacity for high-resolution three-dimensional tracking of fast tumour perfusion suggests vMSOT as a powerful method in preclinical cancer research and theranostics. As the imaging setup can be equally operated in both stationary and handheld mode, the solution is readily translatable for perfusion monitoring in a clinical setting. KEY POINTS: • vMSOT visualizes 3D anatomic, vascular and functional tumour profiles in real time. • Three types of blood perfusion profiles are revealed in breast tumour model. • The method is readily adaptable to operate in a handheld clinical mode.


Asunto(s)
Neoplasias Mamarias Animales/diagnóstico por imagen , Neovascularización Patológica/diagnóstico por imagen , Técnicas Fotoacústicas/métodos , Animales , Medios de Contraste/farmacocinética , Estudios Transversales , Femenino , Humanos , Imagenología Tridimensional/métodos , Verde de Indocianina/farmacocinética , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/patología , Ratones Desnudos , Trasplante de Neoplasias , Consumo de Oxígeno , Perfusión , Tomografía/métodos
9.
Proc Natl Acad Sci U S A ; 109(8): 2814-9, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-21576484

RESUMEN

Bone morphogenetic proteins (BMPs) are members of the TGF-ß superfamily of signaling molecules. BMPs can elicit a wide range of effects in many cell types and have previously been shown to induce growth inhibition in carcinoma cells as well as normal epithelia. Recently, it has been demonstrated that BMP4 and BMP7 are overexpressed in human breast cancers and may have tumor suppressive and promoting effects. We sought to determine whether disruption of the BMP receptor 2 (BMPR2) would alter mammary tumor progression in mice that express the Polyoma middle T antigen. Mice expressing Polyoma middle T antigen under the mouse mammary tumor virus promoter were combined with mice that have doxycycline-inducible expression of a dominant-negative (DN) BMPR2. We did not observe any differences in tumor latency. However, mice expressing the BMPR2-DN had a fivefold increase in lung metastases. We characterized several cell autonomous changes and found that BMPR2-DN-expressing tumor cells had higher rates of proliferation. We also identified unique changes in inflammatory cells and secreted chemokines/cytokines that accompanied BMPR2-DN-expressing tumors. By immunohistochemistry, it was found that BMPR2-DN primary tumors and metastases had an altered reactive stroma, indicating specific changes in the tumor microenvironment. Among the changes we discovered were increased myeloid derived suppressor cells and the chemokine CCL9. BMP was shown to directly regulate CCL9 expression. We conclude that BMPR2 has tumor-suppressive function in mammary epithelia and microenvironment and that disruption can accelerate mammary carcinoma metastases.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Comunicación Paracrina , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Movimiento Celular , Proliferación Celular , Quimiocinas/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Inflamación/patología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/irrigación sanguínea , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Células Mieloides/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Neovascularización Patológica , Transducción de Señal , Microambiente Tumoral
10.
Vopr Onkol ; 61(4): 642-6, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-26571837

RESUMEN

Age-dependent angiogenesis intensity changes have been studied in transgenic HER-2/neu (FBV/N) mice characteristic of breast tumors' high incidence with hyperexpression of HER-2/neu. Concentration of vascular endothelial growth factor, insulin-dependent growth factor 1, nitrogen monoxide, tissue plasminogen activator and type 1 plasminogen activator inhibitor were assessed by means of immune-enzyme assay. The results testify to angiogenesis processes activation side by side with aging and growth of the tumors. Maximum manifestation of these disturbances (growth factors' blood concentrations increase and endotheliocytes' functional activity inhibition) has been revealed in 6-month-old mice during neoplasma maximum intensive and aggressive growth period.


Asunto(s)
Adenocarcinoma/sangre , Envejecimiento/sangre , Biomarcadores de Tumor/sangre , Neoplasias Mamarias Animales/sangre , Neovascularización Patológica/sangre , Receptor ErbB-2/sangre , Adenocarcinoma/irrigación sanguínea , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Femenino , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Experimentales , Ratones , Ratones Endogámicos , Ratones Transgénicos , Óxido Nítrico/sangre , Inhibidor 1 de Activador Plasminogénico/sangre , Activador de Tejido Plasminógeno/sangre , Factor A de Crecimiento Endotelial Vascular/sangre
11.
Nanotechnology ; 25(44): 445101, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25302488

RESUMEN

Tumor vasculature is critically dependent on platelet mediated hemostasis and disruption of the same can augment delivery of nano-formulation based chemotherapeutic agents which depend on enhanced permeability and retention for tumor penetration. Here, we evaluated the role of Clopidogrel, a well-known inhibitor of platelet aggregation, in potentiating the tumor cytotoxicity of cisplatin nano-formulation in a murine breast cancer model. In vivo studies in murine syngeneic 4T1 breast cancer model showed a significant greater penetration of macromolecular fluorescent nanoparticles after clopidogrel pretreatment. Compared to self-assembling cisplatin nanoparticles (SACNs), combination therapy with clopidogrel and SACN was associated with a 4 fold greater delivery of cisplatin to tumor tissue and a greater reduction in tumor growth as well as higher survival rate. Clopidogrel enhances therapeutic efficiency of novel cisplatin based nano-formulations agents by increasing tumor drug delivery and can be used as a potential targeting agent for novel nano-formulation based chemotherapeutics.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cisplatino/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Mamarias Animales/tratamiento farmacológico , Nanosferas/uso terapéutico , Inhibidores de Agregación Plaquetaria/administración & dosificación , Ticlopidina/análogos & derivados , Animales , Línea Celular Tumoral , Cisplatino/química , Clopidogrel , Neoplasias Mamarias Animales/irrigación sanguínea , Ratones , Ratones Endogámicos BALB C , Nanosferas/química , Permeabilidad , Ticlopidina/administración & dosificación
12.
Am J Pathol ; 181(1): 278-93, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22698676

RESUMEN

Increasing chronological age is the most significant risk factor for human cancer development. To examine the effects of host aging on mammary tumor growth, we used caveolin (Cav)-1 knockout mice as a bona fide model of accelerated host aging. Mammary tumor cells were orthotopically implanted into these distinct microenvironments (Cav-1(+/+) versus Cav-1(-/-) age-matched young female mice). Mammary tumors grown in a Cav-1-deficient tumor microenvironment have an increased stromal content, with vimentin-positive myofibroblasts (a marker associated with oxidative stress) that are also positive for S6-kinase activation (a marker associated with aging). Mammary tumors grown in a Cav-1-deficient tumor microenvironment were more than fivefold larger than tumors grown in a wild-type microenvironment. Thus, a Cav-1-deficient tumor microenvironment provides a fertile soil for breast cancer tumor growth. Interestingly, the mammary tumor-promoting effects of a Cav-1-deficient microenvironment were estrogen and progesterone independent. In this context, chemoprevention was achieved by using the mammalian target of rapamycin (mTOR) inhibitor and anti-aging drug, rapamycin. Systemic rapamycin treatment of mammary tumors grown in a Cav-1-deficient microenvironment significantly inhibited their tumor growth, decreased their stromal content, and reduced the levels of both vimentin and phospho-S6 in Cav-1-deficient cancer-associated fibroblasts. Since stromal loss of Cav-1 is a marker of a lethal tumor microenvironment in breast tumors, these high-risk patients might benefit from treatment with mTOR inhibitors, such as rapamycin or other rapamycin-related compounds (rapalogues).


Asunto(s)
Envejecimiento/fisiología , Anticarcinógenos/uso terapéutico , Caveolina 1/fisiología , Neoplasias Mamarias Animales/prevención & control , Sirolimus/uso terapéutico , Animales , Caveolina 1/deficiencia , Femenino , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/fisiopatología , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Neovascularización Patológica/metabolismo , Ovariectomía , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Transducción de Señal/fisiología , Células del Estroma/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Microambiente Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancer Invest ; 31(8): 505-10, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24083814

RESUMEN

The L-methioninase-annexin V/selenomethionine enzyme prodrug system, designed to target the tumor vasculature and release the methylselenol anticancer drug in the tumor, was tested in mice with implanted MBA-MB-231 breast tumors. This therapy was able to cause a reduction in the size of the tumors during the treatment period. It was shown that L-methioninase-annexin V was uniformly bound at the blood vessel surface in the tumor and also that there was a substantial cutoff of blood flowing through the treated tumor, consistent with the therapy's design. This new approach for enzyme prodrug therapy of breast cancer appears promising.


Asunto(s)
Anexina A5/metabolismo , Antineoplásicos/uso terapéutico , Liasas de Carbono-Azufre/metabolismo , Neoplasias Mamarias Animales/tratamiento farmacológico , Metanol/análogos & derivados , Compuestos de Organoselenio/uso terapéutico , Selenometionina/metabolismo , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Terapia Enzimática , Femenino , Humanos , Neoplasias Mamarias Animales/irrigación sanguínea , Metanol/uso terapéutico , Ratones , Ratones SCID , Trasplante de Neoplasias , Profármacos/metabolismo , Profármacos/uso terapéutico
14.
FASEB J ; 25(8): 2626-37, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21518852

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are significantly increased in cancer patients and tumor bearing-animals. MDSCs infiltrate into tumors and promote tumor invasion and metastasis. To identify the mediator responsible for the prometastatic property of MDSCs, we used proteomics. We found neutrophilic granule protein (NGP) was decreased >2-fold in MDSCs from metastatic 4T1 tumor-bearing mice compared to nonmetastatic 67NR controls. NGP mRNA levels were decreased in bone marrow and in tumor-infiltrating MDSCs by 45 and 66%, respectively, in 4T1 tumor-bearing mice compared to 67NR controls. Interestingly, 4T1-conditioned medium reduced myeloid cell NGP expression by ∼ 40%, suggesting that a secreted factor mediates gene reduction. Sequence analysis shows a putative cystatin domain in NGP, and biochemical analysis confirms NGP a novel cathepsin inhibitor. It inhibited cathepsin B activity by nearly 40% in vitro. NGP expression in 4T1 tumor cells suppressed cell invasion, delayed primary tumor growth, and greatly reduced lung metastasis in vivo. A 2.8-fold reduction of cathepsin activity was found in tumors expressing NGP compared to controls. NGP significantly reduced tumor angiogenesis to 12.6 from 19.6 and lymphangiogenesis to 4.6 from 9.1 vessels/field. Necrosis was detectable only in NGP-expressing tumors, and the number of apoptotic cells increased to 22.4 from 8.3 in controls. Taken together, this study identifies a negative regulator of tumor metastasis in MDSCs, NGP, which is down-regulated in metastatic conditions. The finding suggests that malignant tumors promote invasion/metastasis not only through up-regulation of proteases but also down-regulation of protease inhibitors.


Asunto(s)
Cistatinas/fisiología , Células Mieloides/fisiología , Metástasis de la Neoplasia/prevención & control , Animales , Catepsina B/genética , Catepsina B/metabolismo , Línea Celular Tumoral , Proliferación Celular , Cistatinas/genética , Femenino , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/fisiopatología , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología , Neovascularización Patológica , Inhibidores de Proteasas/metabolismo , Proteómica
15.
Mol Oncol ; 16(2): 485-507, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34694686

RESUMEN

Alteration in glycosylation pattern of MUC1 mucin tandem repeats during carcinomas has been shown to negatively affect adhesive properties of malignant cells and enhance tumor invasiveness and metastasis. In addition, MUC1 overexpression is closely interrelated with angiogenesis, making it a great target for immunotherapy. Alongside, easier interaction of nanobodies (single-domain antibodies) with their antigens, compared to conventional antibodies, is usually associated with superior desirable results. Herein, we evaluated the preclinical efficacy of a recombinant nanobody against MUC1 tandem repeats in suppressing tumor growth, angiogenesis, invasion, and metastasis. Expressed nanobody demonstrated specificity only toward MUC1-overexpressing cancer cells and could internalize in cancer cell lines. The IC50 values (the concentration at which the nanobody exerted half of its maximal inhibitory effect) of the anti-MUC1 nanobody against MUC1-positive human cancer cell lines ranged from 1.2 to 14.3 nm. Similar concentrations could also effectively induce apoptosis in MUC1-positive cancer cells but not in normal cells or MUC1-negative human cancer cells. Immunohistochemical staining of spontaneously developed mouse breast tumors prior to in vivo studies confirmed cross-reactivity of nanobody with mouse MUC1 despite large structural dissimilarities between mouse and human MUC1 tandem repeats. In vivo, a dose of 3 µg nanobody per gram of body weight in tumor-bearing mice could attenuate tumor progression and suppress excessive circulating levels of IL-1a, IL-2, IL-10, IL-12, and IL-17A pro-inflammatory cytokines. Also, a significant decline in expression of Ki-67, MMP9, and VEGFR2 biomarkers, as well as vasculogenesis, was evident in immunohistochemically stained tumor sections of anti-MUC1 nanobody-treated mice. In conclusion, the anti-MUC1 tandem repeat nanobody of the present study could effectively overcome tumor growth, invasion, and metastasis.


Asunto(s)
Proliferación Celular/genética , Neoplasias Mamarias Animales/patología , Mucina-1/genética , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Neovascularización Patológica/genética , Anticuerpos de Dominio Único/genética , Secuencias Repetidas en Tándem , Animales , Apoptosis/genética , Línea Celular Tumoral , Quimiocinas/metabolismo , Reacciones Cruzadas , Citocinas/metabolismo , Femenino , Humanos , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos BALB C , Mucina-1/inmunología , Unión Proteica , Anticuerpos de Dominio Único/inmunología
16.
Am J Pathol ; 174(5): 1910-20, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19349372

RESUMEN

The (HER2/Neu) ErbB2 oncogene is commonly overexpressed in human breast cancer and is sufficient for mammary tumorigenesis in transgenic mice. Nuclear factor (NF)-kappaB activity is increased in both human and murine breast tumors. The immune response to mammary tumorigenesis may regulate tumor progression. The role of endogenous mammary epithelial cell NF-kappaB had not previously been determined in immune-competent animals. Furthermore, the role of the NF-kappaB components, p50 and p65, in tumor growth was not known. Herein, the expression of a stabilized form of the NF-kappaB-inhibiting IkappaBalpha protein (IkappaBalphaSR) in breast tumor cell lines that express oncogenic ErbB2 inhibited DNA synthesis and growth in both two- and three-dimensional cultures. Either NF-kappaB inhibition or selective silencing of p50 or p65 led to a loss of contact-independent tumor growth in vitro. IkappaBalphaSR reversed the features of the oncogene-induced phenotype under three-dimensional growth conditions. The NF-kappaB blockade inhibited ErbB2-induced mammary tumor growth in both immune-competent and immune-deficient mice. These findings were associated with both reduced tumor microvascular density and a reduction in the amount of vascular endothelial growth factor. The expression of IkappaBalphaSR in breast cancer tumors inhibited angiogenesis. Thus, mammary epithelial cell NF-kappaB activity enhances ErbB2-mediated mammary tumorigenesis in vivo by promoting both growth and survival signaling via the promotion of tumor vasculogenesis.


Asunto(s)
Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/patología , FN-kappa B/metabolismo , Receptor ErbB-2/metabolismo , Animales , Apoptosis/fisiología , Western Blotting , Adhesión Celular , Núcleo Celular/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Ensayo de Unidades Formadoras de Colonias , Citocinas/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Técnicas para Inmunoenzimas , Neoplasias Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Neovascularización Patológica , ARN Interferente Pequeño/farmacología , Receptor ErbB-2/genética , Venas Umbilicales/citología , Venas Umbilicales/metabolismo
17.
Bioconjug Chem ; 21(3): 556-62, 2010 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20170116

RESUMEN

The transition of a targeted ultrasound contrast agent from animal imaging to testing in clinical studies requires considerable chemical development. The nature of the construct changes from an agent that is chemically attached to microbubbles to one where the targeting group is coupled to a phospholipid, for direct incorporation to the bubble surface. We provide an efficient method to attach a heterodimeric peptide to a pegylated phospholipid and show that the resulting construct retains nanomolar affinity for its target, vascular endothelial growth factor receptor 2 (VEGFR2), for both the human (kinase insert domain-containing receptor - KDR) and the mouse (fetal liver kinase 1 - Flk-1) receptors. The purified phospholipid-PEG-peptide isolated from TFA-based eluents is not stable with respect to hydrolysis of the fatty ester moieties. This leads to the time-dependent formation of the lysophospholipid and the phosphoglycerylamide derived from the degradation of the product. Purification of the product using neutral eluent systems provides a stable product. Methods to prepare the lysophospholipid (hydrolysis product) are also included. Biacore binding data demonstrated the retention of binding of the lipopeptide to the KDR receptor. The phospholipid-PEG2000-peptide is smoothly incorporated into gas-filled microbubbles and provides imaging of angiogenesis in a rat tumor model.


Asunto(s)
Neoplasias Mamarias Animales/irrigación sanguínea , Neovascularización Patológica/diagnóstico por imagen , Péptidos , Fosfolípidos , Polietilenglicoles , Ultrasonografía , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Mamarias Animales/metabolismo , Ratones , Estructura Molecular , Neovascularización Patológica/patología , Péptidos/química , Fosfolípidos/química , Polietilenglicoles/química , Ratas , Ratas Endogámicas F344
18.
Can Vet J ; 51(10): 1109-14, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21197202

RESUMEN

The expression of 5 markers associated with angiogenesis, proliferation, and apoptosis was studied in 26 canine simple mammary gland adenocarcinomas (SMGAs). The adenocarcinomas were graded histologically, and tissue sections were immunohistochemically stained for the expression of vascular endothelial growth factor (VEGF), VEGF receptor-2 (VEGFR-2), intra-tumor microvessel density, and tumor proliferation (PI) using antibodies against VEGF, VEGFR-2, von Willebrand factor, and Ki-67 antigen, respectively. Apoptotic indices (AI) were determined by an apoptosis assay. Markers VEGF and VEGFR-2 were detected in 96% and 100% of SMGAs, respectively. A high correlation between histologic grade and PI (r = 0.73), a moderate correlation between VEGF and histologic grade (r = 0.33), and between VEGF and PI (r = 0.42) were found. There was a significant difference in median PI among the 3 histologic grade groups (r < 0.05). Vascular endothelial growth factor may stimulate tumor cell proliferation through an autocrine loop, since VEGF and VEGFR-2 were expressed in most tumors.


Asunto(s)
Adenocarcinoma/veterinaria , Enfermedades de los Perros/metabolismo , Neoplasias Mamarias Animales/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Enfermedades de los Perros/genética , Perros , Femenino , Regulación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Inmunohistoquímica/veterinaria , Antígeno Ki-67/metabolismo , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/veterinaria , Receptores de Factores de Crecimiento Endotelial Vascular/genética
19.
Mol Oncol ; 14(12): 3198-3210, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33080107

RESUMEN

Breast cancer metastasis is a complex process that depends not only on intrinsic characteristics of metastatic stem cells, but also on the particular microenvironment that supports their growth and modulates the plasticity of the system. In search for microenvironmental factors supporting cancer stem cell (CSC) growth and tumour progression to metastasis, we here investigated the role of the matricellular protein transforming growth factor beta induced (TGFBI) in breast cancer. We crossed the MMTV-PyMT model of mammary gland tumorigenesis with a TgfbiΔ/Δ mouse and studied the CSC content of the tumours. We performed RNAseq on wt and ko tumours, and analysed the tumour vasculature and the immune compartment by IHC and FACS. The source of TGFBI expression was determined by qPCR and by bone marrow transplantation experiments. Finally, we performed in silico analyses using the METABRIC cohort to assess the potential prognostic value of TGFBI. We observed that deletion of Tgfbi led to a dramatic decrease in CSC content and lung metastasis. Our results show that lack of TGFBI resulted in tumour vessel normalisation, with improved vessel perfusion and decreased hypoxia, a major factor controlling CSCs and metastasis. Furthermore, human data mining in a cohort of breast cancer patients showed that higher expression of TGFBI correlates with poor prognosis and is associated with the more aggressive subtypes of breast cancer. Overall, these data reveal a novel biological mechanism controlling metastasis that could potentially be exploited to improve the efficacy and delivery of chemotherapeutic agents in breast cancer.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Factor de Crecimiento Transformador beta/metabolismo , Hipoxia Tumoral , Animales , Femenino , Neoplasias Mamarias Animales/irrigación sanguínea , Ratones , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neovascularización Patológica/metabolismo , Pronóstico
20.
J Cell Mol Med ; 13(9B): 4002-13, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19426156

RESUMEN

The present study aims at investigating the mechanism by which membrane-type 4 matrix metalloproteinase (MT4-MMP), a membrane-anchored MMP expressed by human breast tumour cells promotes the metastatic dissemination into lung. We applied experimental (intravenous) and spontaneous (subcutaneous) models of lung metastasis using human breast adenocarcinoma MDA-MB-231 cells overexpressing or not MT4-MMP. We found that MT4-MMP does not affect lymph node colonization nor extravasation of cells from the bloodstream, but increases the intravasation step leading to metastasis. Ultrastructural and fluorescent microscopic observations coupled with automatic computer-assisted quantifications revealed that MT4-MMP expression induces blood vessel enlargement and promotes the detachment of mural cells from the vascular tree, thus causing an increased tumour vascular leak. On this basis, we propose that MT4-MMP promotes lung metastasis by disturbing the tumour vessel integrity and thereby facilitating tumour cell intravasation.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias Pulmonares/enzimología , Metaloproteinasa 17 de la Matriz/metabolismo , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/secundario , Metástasis Linfática , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/enzimología , Neoplasias Mamarias Animales/patología , Ratones , Ratones Desnudos , Microscopía Electrónica de Transmisión/métodos , Microscopía Fluorescente/métodos , Trasplante de Neoplasias , Neovascularización Patológica , Pericitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA