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










Base de datos
Intervalo de año de publicación
1.
Ageing Res Rev ; 99: 102360, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38821417

RESUMEN

This article brings a new perspective on oral physiology by presenting the oral organ as an integrated entity within the entire organism and its surrounding environment. Rather than considering the mouth solely as a collection of discrete functions, this novel approach emphasizes its role as a dynamic interphase, supporting interactions between the body and external factors. As a resilient ecosystem, the equilibrium of mouth ecological niches is the result of a large number of interconnected factors including the heterogeneity of different oral structures, diversity of resources, external and internal pressures and biological actors. The manuscript seeks to deepen the understanding of age-related changes within the oral cavity and throughout the organism, aligning with the evolving field of gerophysiology. The strategic position and fundamental function of the mouth make it an invaluable target for early prevention, diagnosis, treatment, and even reversal of aging effects throughout the entire organism. Recognizing the oral cavity capacity for sensory perception, element capture and information processing underscores its vital role in continuous health monitoring. Overall, this integrated understanding of the oral physiology aims at advancing comprehensive approaches to the oral healthcare and promoting broader awareness of its implications on the overall well-being.

2.
R Soc Open Sci ; 11(1): 231456, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38298399

RESUMEN

The extracellular-matrix (ECM) is a complex interconnected three-dimensional network that provides structural support for the cells and tissues and defines organ architecture as key for their healthy functioning. However, the intimate mechanisms by which ECM acquire their three-dimensional architecture are still largely unknown. In this paper, we study this question by means of a simple three-dimensional individual based model of interacting fibres able to spontaneously crosslink or unlink to each other and align at the crosslinks. We show that such systems are able to spontaneously generate different types of architectures. We provide a thorough analysis of the emerging structures by an exhaustive parametric analysis and the use of appropriate visualization tools and quantifiers in three dimensions. The most striking result is that the emergence of ordered structures can be fully explained by a single emerging variable: the number of links per fibre in the network. If validated on real tissues, this simple variable could become an important putative target to control and predict the structuring of biological tissues, to suggest possible new therapeutic strategies to restore tissue functions after disruption, and to help in the development of collagen-based scaffolds for tissue engineering. Moreover, the model reveals that the emergence of architecture is a spatially homogeneous process following a unique evolutionary path, and highlights the essential role of dynamical crosslinking in tissue structuring.

3.
Nat Commun ; 9(1): 5178, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518756

RESUMEN

The development of new lymphatic vessels occurs in many cancerous and inflammatory diseases through the binding of VEGF-C to its receptors, VEGFR-2 and VEGFR-3. The regulation of VEGFR-2/VEGFR-3 heterodimerisation and its downstream signaling in lymphatic endothelial cells (LECs) remain poorly understood. Here, we identify the endocytic receptor, uPARAP, as a partner of VEGFR-2 and VEGFR-3 that regulates their heterodimerisation. Genetic ablation of uPARAP leads to hyperbranched lymphatic vasculatures in pathological conditions without affecting concomitant angiogenesis. In vitro, uPARAP controls LEC migration in response to VEGF-C but not VEGF-A or VEGF-CCys156Ser. uPARAP restricts VEGFR-2/VEGFR-3 heterodimerisation and subsequent VEGFR-2-mediated phosphorylation and inactivation of Crk-II adaptor. uPARAP promotes VEGFR-3 signaling through the Crk-II/JNK/paxillin/Rac1 pathway. Pharmacological Rac1 inhibition in uPARAP knockout mice restores the wild-type phenotype. In summary, our study identifies a molecular regulator of lymphangiogenesis, and uncovers novel molecular features of VEGFR-2/VEGFR-3 crosstalk and downstream signaling during VEGF-C-driven LEC sprouting in pathological conditions.


Asunto(s)
Linfangiogénesis , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Dimerización , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Masculino , Glicoproteínas de Membrana/genética , Ratones , Receptores de Superficie Celular/genética , Transducción de Señal , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/química , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
4.
J Immunol ; 201(12): 3815-3821, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30446570

RESUMEN

Mast cells (MC) are innate immune cells involved in many physiological and pathological processes. However, studies of MC function and biology are hampered by the difficulties to obtain human primary MC. To solve this problem, we established a new method to produce easily and rapidly high numbers of MC for in vitro studies using human adipose tissue, which is an abundant and easy access tissue. Stromal vascular fraction of adipose tissue, obtained from human abdominal dermolipectomy, was cultured as spheroids in serum free medium supplemented in stem cell factor. Using this method, we generated, within 3 wk, a highly pure population of connective tissue-type MC expressing MC typical peptidases (tryptase, chymase, and carboxypeptidase-A3) with a yield increasing over time. Stem cell factor was required for this culture, but unlike MC derived from CD34+ cells, this culture did not depend on IL-3 and -6. MC obtained with this method degranulated following FcεRI cross-linking or stimulation by C5a, compound 48/80, and substance P. Interestingly, activation by anti-IgE of both white adipose tissue-MC and MC obtained from peripheral blood-derived CD34+ pluripotent progenitor cells induced the production of PGs as well as proinflammatory cytokines (TNF-α, Il-6, and GM-CSF). In conclusion, we developed a new time saving and reproducible method to produce highly pure and functional human MC in 3 wk from human adipose tissue.


Asunto(s)
Abdomen/patología , Tejido Adiposo/citología , Técnicas de Cultivo de Célula , Endotelio Vascular/citología , Mastocitos/fisiología , Células del Estroma/fisiología , Abdomen/cirugía , Tejido Adiposo/cirugía , Degranulación de la Célula , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Quimasas/metabolismo , Humanos , Inmunidad Innata , Lipectomía , Esferoides Celulares/citología , Factor de Células Madre/metabolismo
5.
J Hematol Oncol ; 10(1): 122, 2017 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-28629427

RESUMEN

BACKGROUND: Lymphatic metastasis is one of the leading causes of death in patients with different types of cancer and is the main prognostic factor for the disease survival. The formation of new lymphatic vessels (lymphangiogenesis) in primary tumors facilitates tumor cell dissemination to regional lymph nodes and correlates with distant metastases. Lymphangiogenesis has thus emerged as a suitable therapeutic target to block metastases, but no anti-lymphangiogenic compounds have been approved for clinical use to date. Therefore, new or improved therapies blocking lymphatic metastases are urgently required. METHODS: We established murine breast tumors to assess the effect of AD0157 on tumor growth, lymphangiogenesis, and lymphatic dissemination. Then, a battery of in vivo (mouse corneal neovascularization and ear sponges), ex vivo (mouse lymphatic rings and rat mesentery explants), and in vitro (proliferation, tubulogenesis, wound-healing, Boyden chambers, and spheroids) assays was used to give insight into the lymphangiogenic steps affected by AD0157. Finally, we investigated the molecular pathways controlled by this drug. RESULTS: AD0157 was found to inhibit the growth of human breast cancer xenografts in mice, to strongly reduce tumor-associated lymphangiogenesis and to block metastatic dissemination to both lymph nodes and distant organs. The high anti-lymphangiogenic potency of AD0157 was further supported by its inhibitory activity at low micromolar range in two in vivo pathological models and in two ex vivo assays. In addition, AD0157 inhibited lymphatic endothelial cell proliferation, migration and invasion, cellular sprouting, and tube formation. Mechanistically, this compound induced apoptosis in lymphatic endothelial cells and decreased VEGFR-3/-2, ERK1/2, and Akt phosphorylations. CONCLUSIONS: These findings demonstrate the suitability of AD0157 to suppress tumor-associated lymphangiogenesis. Beyond discovering a new potent anti-lymphangiogenic drug that is worth considering in future clinical settings, our study supports the interest of designing anti-lymphangiogenic therapies to avoid distant metastatic processes.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Linfangiogénesis/efectos de los fármacos , Metástasis Linfática/prevención & control , Quinonas/uso terapéutico , Succinimidas/uso terapéutico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Metástasis Linfática/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Quinonas/farmacología , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Succinimidas/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
6.
Lab Invest ; 97(2): 194-206, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27918556

RESUMEN

Fatty acid synthase (FASN) is responsible for the endogenous production of fatty acids from acetyl-CoA and malonyl-CoA. Its overexpression is associated with poor prognosis in human cancers including melanomas. Our group has previously shown that the inhibition of FASN with orlistat reduces spontaneous lymphatic metastasis in experimental B16-F10 melanomas, which is a consequence, at least in part, of the reduction of proliferation and induction of apoptosis. Here, we sought to investigate the effects of pharmacological FASN inhibition on lymphatic vessels by using cell culture and mouse models. The effects of FASN inhibitors cerulenin and orlistat on the proliferation, apoptosis, and migration of human lymphatic endothelial cells (HDLEC) were evaluated with in vitro models. The lymphatic outgrowth was evaluated by using a murine ex vivo assay. B16-F10 melanomas and surgical wounds were produced in the ears of C57Bl/6 and Balb-C mice, respectively, and their peripheral lymphatic vessels evaluated by fluorescent microlymphangiography. The secretion of vascular endothelial growth factor C and D (VEGF-C and -D) by melanoma cells was evaluated by ELISA and conditioned media used to study in vitro lymphangiogenesis. Here, we show that cerulenin and orlistat decrease the viability, proliferation, and migration of HDLEC cells. The volume of lymph node metastases from B16-F10 experimental melanomas was reduced by 39% in orlistat-treated animals as well as the expression of VEGF-C in these tissues. In addition, lymphatic vessels from orlistat-treated mice drained more efficiently the injected FITC-dextran. Orlistat and cerulenin reduced VEGF-C secretion and, increase production of VEGF-D by B16-F10 and SK-Mel-25 melanoma cells. Finally, reduced lymphatic cell extensions, were observed following the treatment with conditioned medium from cerulenin- and orlistat-treated B16-F10 cells. Altogether, our results show that FASN inhibitors have anti-metastatic effects by acting on lymphatic endothelium and melanoma cells regardless the increase of lymphatic permeability promoted by orlistat.


Asunto(s)
Cerulenina/farmacología , Ácido Graso Sintasas/antagonistas & inhibidores , Lactonas/farmacología , Vasos Linfáticos/efectos de los fármacos , Melanoma Experimental/prevención & control , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Inhibidores de la Síntesis de Ácidos Grasos/farmacología , Humanos , Linfangiogénesis/efectos de los fármacos , Metástasis Linfática , Vasos Linfáticos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Orlistat , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor D de Crecimiento Endotelial Vascular/metabolismo
7.
Neoplasia ; 17(7): 552-63, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26297433

RESUMEN

Solid tumors comprise cancer cells and different supportive stromal cells, including mesenchymal stem cells (MSCs), which have recently been shown to enhance tumor growth and metastasis. We provide new mechanistic insights into how bone marrow (BM)-derived MSCs co-injected with Lewis lung carcinoma cells promote tumor growth and metastasis in mice. The proinvasive effect of BM-MSCs exerted on tumor cells relies on an unprecedented juxtacrine action of BM-MSC, leading to the trans-shedding of amphiregulin (AREG) from the tumor cell membrane by tumor necrosis factor-α-converting enzyme carried by the BM-MSC plasma membrane. The released soluble AREG activates cancer cells and promotes their invasiveness. This novel concept is supported by the exploitation of different 2D and 3D culture systems and by pharmacological approaches using a tumor necrosis factor-α-converting enzyme inhibitor and AREG-blocking antibodies. Altogether, we here assign a new function to BM-MSC in tumor progression and establish an uncovered link between AREG and BM-MSC.


Asunto(s)
Anfirregulina/metabolismo , Células de la Médula Ósea/metabolismo , Carcinoma Pulmonar de Lewis/patología , Comunicación Celular/fisiología , Células Madre Mesenquimatosas/metabolismo , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Membrana Celular/metabolismo , Proliferación Celular , Femenino , Transferencia Resonante de Energía de Fluorescencia , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Trasplante de Neoplasias , Esferoides Celulares , Células Tumorales Cultivadas
8.
PLoS One ; 9(9): e106976, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25222747

RESUMEN

It is now well accepted that multipotent Bone-Marrow Mesenchymal Stem Cells (BM-MSC) contribute to cancer progression through several mechanisms including angiogenesis. However, their involvement during the lymphangiogenic process is poorly described. Using BM-MSC isolated from mice of two different backgrounds, we demonstrate a paracrine lymphangiogenic action of BM-MSC both in vivo and in vitro. Co-injection of BM-MSC and tumor cells in mice increased the in vivo tumor growth and intratumoral lymphatic vessel density. In addition, BM-MSC or their conditioned medium stimulated the recruitment of lymphatic vessels in vivo in an ear sponge assay, and ex vivo in the lymphatic ring assay (LRA). In vitro, MSC conditioned medium also increased the proliferation rate and the migration of both primary lymphatic endothelial cells (LEC) and an immortalized lymphatic endothelial cell line. Mechanistically, these pro-lymphangiogenic effects relied on the secretion of Vascular Endothelial Growth Factor (VEGF)-A by BM-MSC that activates VEGF Receptor (VEGFR)-2 pathway on LEC. Indeed, the trapping of VEGF-A in MSC conditioned medium by soluble VEGF Receptors (sVEGFR)-1, -2 or the inhibition of VEGFR-2 activity by a specific inhibitor (ZM 323881) both decreased LEC proliferation, migration and the phosphorylation of their main downstream target ERK1/2. This study provides direct unprecedented evidence for a paracrine lymphangiogenic action of BM-MSC via the production of VEGF-A which acts on LEC VEGFR-2.


Asunto(s)
Linfangiogénesis , Células Madre Mesenquimatosas/fisiología , Animales , Movimiento Celular , Proliferación Celular , Medios de Cultivo Condicionados , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Femenino , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Fosforilación , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
PLoS One ; 9(5): e97019, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24806521

RESUMEN

The endothelial cell spheroid assay provides a suitable in vitro model to study (lymph) angiogenesis and test pro- and anti-(lymph) angiogenic factors or drugs. Usually, the extent of cell invasion, observed through optical microscopy, is measured. The present study proposes the spatial distribution of migrated cells as a new descriptor of the (lymph) angiogenic response. The utility of this novel method rests with its capacity to locally characterise spheroid structure, allowing not only the investigation of single and collective cell invasion but also the evolution of the spheroid core itself. Moreover, the proposed method can be applied to 2D-projected spheroid images obtained by optical microscopy, as well as to 3D images acquired by confocal microscopy. To validate the proposed methodology, endothelial cell invasion was evaluated under different experimental conditions. The results were compared with widely used global parameters. The comparison shows that our method prevents local spheroid modifications from being overlooked and leading to the possible misinterpretation of results.


Asunto(s)
Rastreo Celular/métodos , Células Endoteliales/ultraestructura , Neovascularización Patológica/genética , Esferoides Celulares/ultraestructura , Inhibidores de la Angiogénesis/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Células Endoteliales/efectos de los fármacos , Humanos , Imagenología Tridimensional , Microscopía Confocal , Neovascularización Patológica/tratamiento farmacológico , Esferoides Celulares/efectos de los fármacos , Telomerasa/química , Telomerasa/aislamiento & purificación , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Invest Ophthalmol Vis Sci ; 54(5): 3082-93, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23580490

RESUMEN

PURPOSE: To evaluate the antilymphangiogenic potential of multi-target tyrosine kinase inhibitor sunitinib in corneal neovascularization (NV). METHODS: Inflammatory corneal NV was induced by thermal cauterization applied in the central cornea of mice, to which sunitinib malate was daily administered by gavage or not. At days 6, 11, or 17 post cauterization, lymphatic and blood vessels, as well as inflammatory cells were immunostained and quantified in whole-mounted corneas. RT-PCRs were performed to evidence VEGF-A, VEGF-C, VEGF-D, placental growth factor (PlGF), and soluble vascular endothelial growth factor receptor (VEGFR)-1 and -2 (sVEGFR-1, sVEGFR-2) expressions. Macrophages were isolated from mice peritoneal cavity following thioglycollate injection to produce conditioned medium. The effects of sunitinib were evaluated in vitro in the aortic and lymphatic ring assays in the presence or not of macrophage conditioned medium. RESULTS: Sunitinib treatment drastically reduced pathologic corneal lymphangiogenesis and angiogenesis. Reduced F4/80+ cell infiltration was evidenced in sunitinib-treated mice and was associated to decreased VEGF-A (by 50%, P < 0.01) and VEGF-C (by 35%, P < 0.01) expressions, while VEGF-D and sVEGFR-2 expressions were not affected. In vitro, sunitinib dose-dependently inhibited aortic ring outgrowth, but failed to affect lymphangiogenesis in the lymphatic ring assay. However, macrophage conditioned medium-enhanced angiogenesis and lymphangiogenesis were both strongly counteracted by sunitinib treatment. Mechanistically, sunitinib blocked VEGFR-2 phosphorylation induced by VEGF-A released by macrophages. CONCLUSIONS: Sunitinib exerts antihemangiogenic and antilymphangiogenic effects in vivo by reducing F4/80+ cell recruitment and interacting with their released factors.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neovascularización de la Córnea/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Indoles/uso terapéutico , Linfangiogénesis/efectos de los fármacos , Pirroles/uso terapéutico , Animales , Proliferación Celular/efectos de los fármacos , Neovascularización de la Córnea/genética , Neovascularización de la Córnea/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente Indirecta , Glicoproteínas/metabolismo , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/metabolismo , Macrófagos Peritoneales , Masculino , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Placentario , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Gestacionales/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sunitinib , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
11.
Neoplasia ; 14(10): 943-51, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23097628

RESUMEN

Carcinoma-associated fibroblasts are key contributors of the tumor microenvironment that regulates carcinoma progression. They consist of a heterogeneous cell population with diverse origins, phenotypes, and functions. In the present report, we have explored the contribution of bone marrow (BM)-derived cells to generate different fibroblast subsets that putatively produce the matrix metalloproteinase 13 (MMP13) and affect cancer cell invasion. A murine model of skin carcinoma was applied to mice, irradiated, and engrafted with BM isolated from green fluorescent protein (GFP) transgenic mice. We provide evidence that one third of BM-derived GFP(+) cells infiltrating the tumor expressed the chondroitin sulfate proteoglycan NG2 (pericytic marker) or α-smooth muscle actin (α-SMA, myofibroblast marker), whereas almost 90% of Thy1(+) fibroblasts were originating from resident GFP-negative cells. MMP13producing cells were exclusively α-SMA(+) cells and derived from GFP(+) BM cells. To investigate their impact on tumor invasion, we isolated mesenchymal stem cells (MSCs) from the BM of wild-type and MMP13-deficient mice. Wild-type MSC promoted cancer cell invasion in a spheroid assay, whereas MSCs obtained from MMP13-deficient mice failed to. Our data support the concept of fibroblast subset specialization with BM-derived α-SMA(+) cells being the main source of MMP13, a stromal mediator of cancer cell invasion.


Asunto(s)
Médula Ósea/patología , Fibroblastos/patología , Metaloproteinasa 13 de la Matriz/fisiología , Células Madre Mesenquimatosas/patología , Miofibroblastos/patología , Neoplasias/patología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Médula Ósea/metabolismo , Femenino , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Hibridación in Situ , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miofibroblastos/metabolismo , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Microambiente Tumoral
12.
Blood ; 119(21): 5048-56, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22490679

RESUMEN

Lymphatic dysfunctions are associated with several human diseases, including lymphedema and metastatic spread of cancer. Although it is well recognized that lymphatic capillaries attach directly to interstitial matrix mainly composed of fibrillar type I collagen, the interactions occurring between lymphatics and their surrounding matrix have been overlooked. In this study, we demonstrate how matrix metalloproteinase (MMP)-2 drives lymphatic morphogenesis through Mmp2-gene ablation in mice, mmp2 knockdown in zebrafish and in 3D-culture systems, and through MMP2 inhibition. In all models used in vivo (3 murine models and thoracic duct development in zebrafish) and in vitro (lymphatic ring and spheroid assays), MMP2 blockage or down-regulation leads to reduced lymphangiogenesis or altered vessel branching. Our data show that lymphatic endothelial cell (LEC) migration through collagen fibers is affected by physical matrix constraints (matrix composition, density, and cross-linking). Transmission electron microscopy and confocal reflection microscopy using DQ-collagen highlight the contribution of MMP2 to mesenchymal-like migration of LECs associated with collagen fiber remodeling. Our findings provide new mechanistic insight into how LECs negotiate an interstitial type I collagen barrier and reveal an unexpected MMP2-driven collagenolytic pathway for lymphatic vessel formation and morphogenesis.


Asunto(s)
Linfangiogénesis/genética , Vasos Linfáticos/embriología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/fisiología , Animales , Animales Modificados Genéticamente , Células Cultivadas , Colágeno Tipo I/metabolismo , Colagenasas/genética , Colagenasas/metabolismo , Colagenasas/fisiología , Embrión no Mamífero , Líquido Extracelular/enzimología , Líquido Extracelular/metabolismo , Femenino , Humanos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/fisiología , Masculino , Metaloproteinasa 2 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pez Cebra
13.
BMC Cell Biol ; 12: 29, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21702933

RESUMEN

BACKGROUND: Abnormal lymphatic vessel formation (lymphangiogenesis) is associated with different pathologies such as cancer, lymphedema, psoriasis and graft rejection. Lymphatic vasculature displays distinctive features than blood vasculature, and mechanisms underlying the formation of new lymphatic vessels during physiological and pathological processes are still poorly documented. Most studies on lymphatic vessel formation are focused on organism development rather than lymphangiogenic events occurring in adults. We have here studied lymphatic vessel formation in two in vivo models of pathological lymphangiogenesis (corneal assay and lymphangioma). These data have been confronted to those generated in the recently set up in vitro model of lymphatic ring assay. Ultrastructural analyses through Transmission Electron Microscopy (TEM) were performed to investigate tube morphogenesis, an important differentiating process observed during endothelial cell organization into capillary structures. RESULTS: In both in vivo models (lymphangiogenic corneal assay and lymphangioma), migrating lymphatic endothelial cells extended long processes exploring the neighboring environment and organized into cord-like structures. Signs of intense extracellular matrix remodeling were observed extracellularly and inside cytoplasmic vacuoles. The formation of intercellular spaces between endothelial cells led to tube formation. Proliferating lymphatic endothelial cells were detected both at the tips of sprouting capillaries and inside extending sprouts. The different steps of lymphangiogenesis observed in vivo are fully recapitulated in vitro, in the lymphatic ring assay and include: (1) endothelial cell alignment in cord like structure, (2) intracellular vacuole formation and (3) matrix degradation. CONCLUSIONS: In this study, we are providing evidence for lymphatic vessel formation through tunneling relying on extensive matrix remodeling, migration and alignment of sprouting endothelial cells into tubular structures. In addition, our data emphasize the suitability of the lymphatic ring assay to unravel mechanisms underlying lymphangiogenesis.


Asunto(s)
Linfangiogénesis/fisiología , Vasos Linfáticos/fisiología , Animales , Movimiento Celular , Células Cultivadas , Córnea/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Linfático/metabolismo , Endotelio Linfático/patología , Femenino , Adyuvante de Freund , Hiperplasia , Técnicas In Vitro , Linfangioma/etiología , Linfangioma/patología , Vasos Linfáticos/patología , Vasos Linfáticos/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión
14.
Mol Aspects Med ; 32(2): 146-58, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21549745

RESUMEN

The main physiological function of the lymphatic vasculature is to maintain tissue fluid homeostasis. Lymphangiogenesis or de novo lymphatic formation is closely associated with tissue inflammation in adults (i.e. wound healing, allograft rejection, tumor metastasis). Until recently, research on lymphangiogenesis focused mainly on growth factor/growth factor-receptor pathways governing this process. One of the lymphatic vessel features is the incomplete or absence of basement membrane. This close association of endothelial cells with the underlying interstitial matrix suggests that cell-matrix interactions play an important role in lymphangiogenesis and lymphatic functions. However, the exploration of interaction between extracellular matrix (ECM) components and lymphatic endothelial cells is in its infancy. Herein, we describe ECM-cell and cell-cell interactions on lymphatic system function and their modification occurring in pathologies including cancer metastasis.


Asunto(s)
Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Linfangiogénesis/fisiología , Animales , Comunicación Celular/fisiología , Humanos , Leucocitos/metabolismo , Neoplasias/fisiopatología
15.
Cell Mol Life Sci ; 68(4): 677-86, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20700625

RESUMEN

In this study, we evaluate the potential involvement of collagenase-3 (MMP13), a matrix metalloproteinase (MMP) family member, in the exudative form of age-related macular degeneration characterized by a neovascularisation into the choroid. RT-PCR analysis revealed that human neovascular membranes issued from patients with AMD expressed high levels of Mmp13. The contribution of MMP13 in choroidal neovascularization (CNV) formation was explored by using a murine model of laser-induced CNV and applying it to wild-type mice (WT) and Mmp13-deficient mice (Mmp13 ( -/- ) mice). Angiogenic and inflammatory reactions were explored by immunohistochemistry. The implication of bone marrow (BM)-derived cells was determined by BM engraftment into irradiated mice and by injecting mesenchymal stem cells (MSC) isolated from WT BM. The deficiency of Mmp13 impaired CNV formation which was fully restored by WT BM engraftment and partially rescued by several injections of WT MSC. The present study sheds light on a novel function of MMP13 during BM-dependent choroidal vascularization and provides evidence for a role for MSC in the pathogenesis of CNV.


Asunto(s)
Neovascularización Coroidal/enzimología , Neovascularización Coroidal/etiología , Degeneración Macular/enzimología , Metaloproteinasa 13 de la Matriz/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Neovascularización Coroidal/genética , Neovascularización Coroidal/patología , Eliminación de Gen , Expresión Génica , Humanos , Degeneración Macular/genética , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL
16.
Cell Cycle ; 7(8): 1047-53, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18414048

RESUMEN

The metalloprotease 9 (MMP-9), a known mediator of tumour invasion, is secreted as a 92 kDa pro-form but a non-secreted variant of 85 Kda has been described. The importance of this variant pro-form in tumor progression remains poorly defined. We previously showed that the DNA repair protein Ku interacts at the cell surface of leukaemia cell lines with the 85 Kda pro-form of MMP-9 and these Ku/MMP-9 complexes regulates cell invasion, highlighting their importance in haematological malignancies. We demonstrate here that all samples of acute myeloid leukaemia (AML) blasts purified from bone marrow of 16 affected patients express the 85 Kda form of MMP-9. However, only AML that display monocytic lineage markers (AML4/5) express this form at the cell surface with co-expression of the membrane associated form of Ku. Blocking antibodies directed against Ku or MMP-9 specifically inhibited cell invasion of those expressing Ku/MMP-9 on the cell surface. The membrane form of Ku might represent an important factor in the exposition to the cell surface of this specific MMP-9 pro-form in AML with monocytic features. These results might have important functional significance in the occurrence of extra-medullar infiltrates of leukaemia cells that occurs frequently during the onset of monocyte-related AML sub-types.


Asunto(s)
ADN Helicasas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de la Membrana/metabolismo , Monocitos/metabolismo , Invasividad Neoplásica/fisiopatología , Adulto , Anciano , Western Blotting , Línea Celular Tumoral , Femenino , Humanos , Autoantígeno Ku , Masculino , Persona de Mediana Edad
17.
Int J Radiat Biol ; 83(11-12): 803-11, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18058368

RESUMEN

PURPOSE: The first reports that ionizing radiation (IR) induces rapid and persistent activation of transforming growth factor beta1 (TGFbeta) were nearly two decades ago. Subsequent studies have shown that TGFbeta is a major mediator of cellular and tissue responses to IR and have revealed novel facets of its complex biology. RESULTS: We and others have recently shown that inhibition of production or signaling of TGFbeta in epithelial cells modulates radiosensitivity and impedes activation of the DNA damage response program. The primary transducer of cellular response to DNA damage caused by ionizing radiation is the nuclear protein kinase ataxia telangiectasia mutated, whose activity is severely compromised when TGFbeta is inhibited. Thus, in conjunction, with its well-recognized contribution to normal tissue fibrosis, the role of TGFbeta in the genotoxic stress program provides a previously unsuspected avenue to modulate radiotherapy. CONCLUSIONS: We hypothesize that identification of the circumstances and tumors in which TGFbeta manipulation enhances tumor cell radiosensitivity, while protecting normal tissues, could significantly increase therapeutic index.


Asunto(s)
Neoplasias/radioterapia , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Humanos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/fisiología , Radiobiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transducción de Señal/efectos de la radiación , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo
18.
EMBO Rep ; 8(6): 583-8, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17496833

RESUMEN

Recent evidence shows that the DNA repair protein Ku is expressed on the surface of a subset of cells, where it contributes to adhesion and invasion processes, and also to viral or bacterial entry into target cells. Here, we show that Ku was expressed on the cell surface during activation of human monocytes and that its expression was independent of the conventional endoplasmic reticulum (ER)/Golgi secretory pathway. Ku inhibition, by blocking antibodies, decreases the migration of monocytes on fibronectin and laminin. On activation, nuclear Ku seems to move to the periphery of the cell and it shows a punctuate staining in the cytoplasm. The cytoplasmic distribution of Ku was shown to be unaltered by brefeldin A. Protease protection experiments show that Ku is contained within vesicles in activated monocytes. These data support a new role for Ku in the migration of monocytes into tissues, which depends on a tightly regulated pathway of intracellular redistribution.


Asunto(s)
Antígenos Nucleares/metabolismo , Movimiento Celular , Proteínas de Unión al ADN/metabolismo , Monocitos/citología , Monocitos/metabolismo , Señales de Clasificación de Proteína , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Humanos , Autoantígeno Ku , Factor Estimulante de Colonias de Macrófagos/farmacología , Monocitos/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo
19.
Cell Cycle ; 4(3): 438-41, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15738653

RESUMEN

The Ku heterodimer (Ku70/Ku80) plays a central role in DNA double strand break recognition and repair. It has been shown, more than ten years ago, that Ku is also expressed at the cell surface of different cells types along with its intracellular pool within the nucleus and the cytoplasm but involvement of Ku in cell-cell and cell-extracellular matrix adhesion has been only recently demonstrated. In addition, we have shown that Ku may have a second and unexpected activity in cell/microenvironment interaction. Indeed, Ku appears to be involved in extracellular proteolytic processes through its specific interaction, on the cell surface, with the matrix metalloprotease 9. Taken together, these results suggest that Ku function at the cell surface is likely to be important in tumour invasion. Various fundamental questions arise from these observations. How Ku is expressed on the cell surface, why a protein with completely unrelated functions also serve as an integrin-like molecule once expressed at the cell surface and is this functional moonlighting of Ku related to cell transformation remain open issues that will be discussed here.


Asunto(s)
Antígenos Nucleares/fisiología , Daño del ADN , Proteínas de Unión al ADN/fisiología , Animales , Adhesión Celular , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Reparación del ADN , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Autoantígeno Ku , Metaloproteinasa 9 de la Matriz/metabolismo , Modelos Biológicos , Neoplasias/metabolismo , Unión Proteica
20.
EMBO J ; 23(19): 3758-68, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15385961

RESUMEN

The Ku heterodimer (Ku70/Ku80) plays a central role in DNA double-strand breaks repair. Ku is also expressed on the cell surface of different types of cells where its function remains poorly understood. From a yeast two-hybrid screen, we have identified a specific interaction between the core region of Ku80 and the hemopexin domain of metalloproteinase 9 (MMP-9), a key enzyme involved in the degradation of extracellular matrix (ECM) components. Ku associates with MMP-9 on the surface of leukemic cells as demonstrated by co-immunoprecipitation experiments in membrane extracts and double-label immunofluorescence studies. In normal and tumoral migratory cells, Ku80 and MMP-9 colocalize at the periphery of leading edge of cells and cellular invasion of collagen IV matrices was blocked by antibodies directed against Ku70 or Ku80 subunits as well as by Ku80-specific antisense oligonucleotides. Our results indicate that Ku and MMP-9 interact at the cell membrane of highly invasive hematopoietic cells of normal and tumoral origin and document the unexpected importance of the membrane-associated form of Ku in the regulation of ECM remodelling.


Asunto(s)
Antígenos Nucleares/metabolismo , Membrana Celular/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Antígenos Nucleares/genética , Movimiento Celular , Colágeno Tipo IV/metabolismo , Proteínas de Unión al ADN/genética , Técnica del Anticuerpo Fluorescente , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunoprecipitación , Autoantígeno Ku , Leucemia/metabolismo , Leucemia/patología , Invasividad Neoplásica , Oligonucleótidos Antisentido/farmacología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...