Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(6)2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38542421

RESUMEN

Extracellular vesicles produced by tumor cells (TEVs) influence all stages of cancer development and spread, including tumorigenesis, cancer progression, and metastasis. TEVs can trigger profound phenotypic and functional changes in target cells through three main general mechanisms: (i) docking of TEVs on target cells and triggering of intra-cellular signaling; (ii) fusion of TEVs and target cell membranes with release of TEVs molecular cargo in the cytoplasm of recipient cell; and (iii) uptake of TEVs by recipient cells. Though the overall tumor-promoting effects of TEVs as well as the general mechanisms involved in TEVs interactions with, and uptake by, recipient cells are relatively well established, current knowledge about the molecular determinants that mediate the docking and uptake of tumor-derived EVs by specific target cells is still rather deficient. These molecular determinants dictate the cell and organ tropism of TEVs and ultimately control the specificity of TEVs-promoted metastases. Here, we will review current knowledge on selected specific molecules that mediate the tropism of TEVs towards specific target cells and organs, including the integrins, ICAM-1 Inter-Cellular Adhesion Molecule), ALCAM (Activated Leukocyte Cell Adhesion Molecule), CD44, the metalloproteinases ADAM17 (A Disintegrin And Metalloproteinase member 17) and ADAM10 (A Disintegrin And Metalloproteinase member 10), and the tetraspanin CD9.


Asunto(s)
Desintegrinas , Vesículas Extracelulares , Humanos , Comunicación Celular , Tetraspaninas/metabolismo , Carcinogénesis/metabolismo , Vesículas Extracelulares/metabolismo
2.
Nature ; 530(7590): 349-53, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26863192

RESUMEN

In multiple sclerosis, brain-reactive T cells invade the central nervous system (CNS) and induce a self-destructive inflammatory process. T-cell infiltrates are not only found within the parenchyma and the meninges, but also in the cerebrospinal fluid (CSF) that bathes the entire CNS tissue. How the T cells reach the CSF, their functionality, and whether they traffic between the CSF and other CNS compartments remains hypothetical. Here we show that effector T cells enter the CSF from the leptomeninges during Lewis rat experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. While moving through the three-dimensional leptomeningeal network of collagen fibres in a random Brownian walk, T cells were flushed from the surface by the flow of the CSF. The detached cells displayed significantly lower activation levels compared to T cells from the leptomeninges and CNS parenchyma. However, they did not represent a specialized non-pathogenic cellular sub-fraction, as their gene expression profile strongly resembled that of tissue-derived T cells and they fully retained their encephalitogenic potential. T-cell detachment from the leptomeninges was counteracted by integrins VLA-4 and LFA-1 binding to their respective ligands produced by resident macrophages. Chemokine signalling via CCR5/CXCR3 and antigenic stimulation of T cells in contact with the leptomeningeal macrophages enforced their adhesiveness. T cells floating in the CSF were able to reattach to the leptomeninges through steps reminiscent of vascular adhesion in CNS blood vessels, and invade the parenchyma. The molecular/cellular conditions for T-cell reattachment were the same as the requirements for detachment from the leptomeningeal milieu. Our data indicate that the leptomeninges represent a checkpoint at which activated T cells are licensed to enter the CNS parenchyma and non-activated T cells are preferentially released into the CSF, from where they can reach areas of antigen availability and tissue damage.


Asunto(s)
Movimiento Celular , Líquido Cefalorraquídeo/citología , Encefalomielitis Autoinmune Experimental/patología , Meninges/patología , Esclerosis Múltiple/patología , Linfocitos T/patología , Traslado Adoptivo , Animales , Adhesión Celular , Líquido Cefalorraquídeo/inmunología , Quimiocinas/metabolismo , Plexo Coroideo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Integrina alfa4beta1/metabolismo , Activación de Linfocitos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Meninges/inmunología , Esclerosis Múltiple/inmunología , Ratas , Ratas Endogámicas Lew , Receptores CCR5/metabolismo , Receptores CXCR3/metabolismo , Linfocitos T/inmunología
3.
Int J Mol Sci ; 23(10)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35628559

RESUMEN

Colorectal cancer (CRC) and ovarian cancer (OvC) patients frequently develop peritoneal metastasis, a condition associated with a very poor prognosis. In these cancers, tumor-derived extracellular vesicles (EVs) cause immunosuppression, facilitate the direct attachment and invasion of cancer cells through the mesothelium, induce the conversion of peritoneal mesothelial cells (PMCs) into cancer-associated fibroblasts (CAFs) and transfer a more aggressive phenotype amongst cancer cells. Although the promoting role of EVs in CRC and OvC peritoneal metastasis is well established, the specific molecules that mediate the interactions between tumor-derived EVs and immune and non-immune target cells remain elusive. Here, we employed the SKOV-3 (ovarian adenocarcinoma) and Colo-320 (colorectal adenocarcinoma) human cell lines as model systems to study the interactions and uptake of EVs produced by ovarian carcinoma and colorectal carcinoma cells, respectively. We established that the adhesion molecule ALCAM/CD166 is involved in the interaction of cancer-derived EVs with recipient cancer cells (a process termed "EV binding" or "EV docking") and in their subsequent uptake by these cells. The identification of ALCAM/CD166 as a molecule mediating the docking and uptake of CRC and OvC-derived EVs may be potentially exploited to block the peritoneal metastasis cascade promoted by EVs in CRC and OvC patients.


Asunto(s)
Adenocarcinoma , Antígenos CD , Moléculas de Adhesión Celular Neuronal , Vesículas Extracelulares , Proteínas Fetales , Neoplasias Ováricas , Neoplasias Peritoneales , Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Adenocarcinoma/patología , Antígenos CD/metabolismo , Carcinoma Epitelial de Ovario/patología , Moléculas de Adhesión Celular Neuronal/metabolismo , Vesículas Extracelulares/metabolismo , Femenino , Proteínas Fetales/metabolismo , Humanos , Neoplasias Ováricas/patología , Neoplasias Peritoneales/metabolismo
4.
EMBO J ; 36(5): 629-645, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28122868

RESUMEN

We show that the three conformational states of integrin α5ß1 have discrete free energies and define activation by measuring intrinsic affinities for ligand of each state and the equilibria linking them. The 5,000-fold higher affinity of the extended-open state than the bent-closed and extended-closed states demonstrates profound regulation of affinity. Free energy requirements for activation are defined with protein fragments and intact α5ß1 On the surface of K562 cells, α5ß1 is 99.8% bent-closed. Stabilization of the bent conformation by integrin transmembrane and cytoplasmic domains must be overcome by cellular energy input to stabilize extension. Following extension, headpiece opening is energetically favored. N-glycans and leg domains in each subunit that connect the ligand-binding head to the membrane repel or crowd one another and regulate conformational equilibria in favor of headpiece opening. The results suggest new principles for regulating signaling in the large class of receptors built from extracellular domains in tandem with single-span transmembrane domains.


Asunto(s)
Integrina alfa5beta1/química , Integrina alfa5beta1/metabolismo , Línea Celular , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Termodinámica
5.
Nat Immunol ; 10(7): 753-60, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19503105

RESUMEN

The immunological synapse (IS) is a cell-cell junction formed between CD4(+) T cells and dendritic cells (DCs). Here we show in vitro and in vivo that IS formation inhibits apoptosis of DCs. Consistent with these results, IS formation induced antiapoptotic signaling events, including activation of the kinase Akt1 and localization of the prosurvival transcription factor NF-kappaB and the proapoptotic transcription factor FOXO1 to the nucleus and cytoplasm, respectively. Inhibition of phosphatidylinositol 3-OH kinase and Akt1 partially prevented the antiapoptotic effects of IS formation. Direct stimulation of the IS component CD40 on DCs leads to the activation of Akt1, suggesting the involvement of this receptor in the antiapoptotic effects observed upon IS formation.


Asunto(s)
Apoptosis/inmunología , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/metabolismo , Sinapsis Inmunológicas/inmunología , FN-kappa B/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD40/inmunología , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Humanos , Immunoblotting , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Transporte de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
6.
Int J Mol Sci ; 22(21)2021 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-34768926

RESUMEN

Most patients with ovarian cancer (OvCA) present peritoneal disseminated disease at the time of diagnosis. During peritoneal metastasis, cancer cells detach from the primary tumor and disseminate through the intraperitoneal fluid. The peritoneal mesothelial cell (PMC) monolayer that lines the abdominal cavity is the first barrier encountered by OvCA cells. Subsequent progression of tumors through the peritoneum leads to the accumulation into the peritoneal stroma of a sizeable population of carcinoma-associated fibroblasts (CAFs), which is mainly originated from a mesothelial-to-mesenchymal transition (MMT) process. A common characteristic of OvCA patients is the intraperitoneal accumulation of ascitic fluid, which is composed of cytokines, chemokines, growth factors, miRNAs, and proteins contained in exosomes, as well as tumor and mesothelial suspended cells, among other components that vary in proportion between patients. Exosomes are small extracellular vesicles that have been shown to mediate peritoneal metastasis by educating a pre-metastatic niche, promoting the accumulation of CAFs via MMT, and inducing tumor growth and chemoresistance. This review summarizes and discusses the pivotal role of exosomes and MMT as mediators of OvCA peritoneal colonization and as emerging diagnostic and therapeutic targets.


Asunto(s)
Carcinoma Epitelial de Ovario/patología , Transición Epitelial-Mesenquimal/fisiología , Exosomas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Peritoneales/secundario , Líquido Ascítico/química , Líquido Ascítico/citología , Línea Celular Tumoral , Citocinas/análisis , Epitelio/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/análisis , Peritoneo/patología
7.
Int J Mol Sci ; 22(18)2021 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-34576100

RESUMEN

Approximately 25% of colorectal cancer (CRC) patients develop peritoneal metastasis, a condition associated with a bleak prognosis. The CRC peritoneal dissemination cascade involves the shedding of cancer cells from the primary tumor, their transport through the peritoneal cavity, their adhesion to the peritoneal mesothelial cells (PMCs) that line all peritoneal organs, and invasion of cancer cells through this mesothelial cell barrier and underlying stroma to establish new metastatic foci. Exosomes produced by cancer cells have been shown to influence many processes related to cancer progression and metastasis. In epithelial ovarian cancer these extracellular vesicles (EVs) have been shown to favor different steps of the peritoneal dissemination cascade by changing the functional phenotype of cancer cells and PMCs. Little is currently known, however, about the roles played by exosomes in the pathogenesis and peritoneal metastasis cascade of CRC and especially about the molecules that mediate their interaction and uptake by target PMCs and tumor cells. We isolated exosomes by size-exclusion chromatography from CRC cells and performed cell-adhesion assays to immobilized exosomes in the presence of blocking antibodies against surface proteins and measured the uptake of fluorescently-labelled exosomes. We report here that the interaction between integrin α5ß1 on CRC cells (and PMCs) and its ligand ADAM17 on exosomes mediated the binding and uptake of CRC-derived exosomes. Furthermore, this process was negatively regulated by the expression of tetraspanin CD9 on exosomes.


Asunto(s)
Proteína ADAM17/metabolismo , Neoplasias Colorrectales/metabolismo , Exosomas/metabolismo , Integrina alfa5beta1/metabolismo , Adenocarcinoma/metabolismo , Adhesión Celular , Línea Celular Tumoral , Epitelio/patología , Exosomas/ultraestructura , Fibronectinas/metabolismo , Humanos , Peritoneo/patología , Tetraspanina 29/metabolismo
8.
Med Microbiol Immunol ; 209(4): 461-471, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32385608

RESUMEN

Human papillomaviruses (HPV) are causative agents of various tumours such as cervical cancer. HPV binding to the cell surface of keratinocytes leads to virus endocytosis at tetraspanin enriched microdomains. Complex interactions of the capsid proteins with host proteins as well as ADAM17-dependent ERK1/2 signal transduction enable the entry platform assembly of the oncogenic HPV type 16. Here, we studied the importance of tetraspanin CD9, also known as TSPAN29, in HPV16 infection of different epithelial cells. We found that both overexpression and loss of the tetraspanin decreased infection rates in cells with low endogenous CD9 levels, while reduction of CD9 expression in keratinocytes that exhibit high-CD9 protein amounts, led to an increase of infection. Therefore, we concluded that low-CD9 supports infection. Moreover, we found that changes in CD9 amounts affect the shedding of the ADAM17 substrate transforming growth factor alpha (TGFα) and the downstream phosphorylation of ERK. These effects correlate with those on infection rates suggesting that a specific CD9 optimum promotes ADAM17 activity, ERK signalling and virus infection. Together, our findings implicate that CD9 regulates HPV16 infection through the modulation of ADAM17 sheddase activity.


Asunto(s)
Proteína ADAM17/metabolismo , Sistema de Señalización de MAP Quinasas , Infecciones por Papillomavirus/metabolismo , Tetraspanina 29/metabolismo , Proteína ADAM17/genética , Endocitosis , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HaCaT , Células HeLa , Papillomavirus Humano 16 , Humanos , Queratinocitos/virología , Infecciones por Papillomavirus/virología , Tetraspanina 29/genética , Factor de Crecimiento Transformador alfa/metabolismo , Internalización del Virus
9.
Proc Natl Acad Sci U S A ; 113(27): E3872-81, 2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27317747

RESUMEN

Whether ß1 integrin ectodomains visit conformational states similarly to ß2 and ß3 integrins has not been characterized. Furthermore, despite a wealth of activating and inhibitory antibodies to ß1 integrins, the conformational states that these antibodies stabilize, and the relation of these conformations to function, remain incompletely characterized. Using negative-stain electron microscopy, we show that the integrin α5ß1 ectodomain adopts extended-closed and extended-open conformations as well as a bent conformation. Antibodies SNAKA51, 8E3, N29, and 9EG7 bind to different domains in the α5 or ß1 legs, activate, and stabilize extended ectodomain conformations. Antibodies 12G10 and HUTS-4 bind to the ß1 ßI domain and hybrid domains, respectively, activate, and stabilize the open headpiece conformation. Antibody TS2/16 binds a similar epitope as 12G10, activates, and appears to stabilize an open ßI domain conformation without requiring extension or hybrid domain swing-out. mAb13 and SG/19 bind to the ßI domain and ßI-hybrid domain interface, respectively, inhibit, and stabilize the closed conformation of the headpiece. The effects of the antibodies on cell adhesion to fibronectin substrates suggest that the extended-open conformation of α5ß1 is adhesive and that the extended-closed and bent-closed conformations are nonadhesive. The functional effects and binding sites of antibodies and fibronectin were consistent with their ability in binding to α5ß1 on cell surfaces to cross-enhance or inhibit one another by competitive or noncompetitive (allosteric) mechanisms.


Asunto(s)
Integrina alfa5beta1/química , Escherichia coli , Fibronectinas/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Conformación Proteica , Relación Estructura-Actividad
10.
Biochim Biophys Acta ; 1853(10 Pt A): 2464-80, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26003300

RESUMEN

The tetraspanin CD9 has been shown to interact with different members of the ß1 and ß3 subfamilies of integrins, regulating through these interactions cell adhesion, migration and signaling. Based on confocal microscopy co-localization and on co-immunoprecipitation results, we report here that CD9 associates with the ß2 integrin LFA-1 in different types of leukocytes including T, B and monocytic cells. This association is resistant to stringent solubilization conditions which, together with data from chemical crosslinking, in situ Proximity Ligation Assays and pull-down experiments, suggest a primary/direct type of interaction mediated by the Large Extracellular Loop of the tetraspanin. CD9 exerts inhibitory effects on the adhesive function of LFA-1 and on LFA-1-dependent leukocyte cytotoxic activity. The mechanism responsible for this negative regulation exerted by CD9 on LFA-1 adhesion does not involve changes in the affinity state of this integrin but seems to be related to alterations in its state of aggregation.


Asunto(s)
Leucocitos/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Tetraspanina 29/metabolismo , Adhesión Celular/fisiología , Femenino , Humanos , Leucocitos/citología , Masculino
11.
Blood ; 121(2): 403-15, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23074273

RESUMEN

Human endoglin is an RGD-containing transmembrane glycoprotein identified in vascular endothelial cells. Although endoglin is essential for angiogenesis and its expression is up-regulated in inflammation and at sites of leukocyte extravasation, its role in leukocyte trafficking is unknown. This function was tested in endoglin heterozygous mice (Eng(+/-)) and their wild-type siblings Eng(+/+) treated with carrageenan or LPS as inflammatory agents. Both stimuli showed that inflammation-induced leukocyte transendothelial migration to peritoneum or lungs was significantly lower in Eng(+/-) than in Eng(+/+) mice. Leukocyte transmigration through cell monolayers of endoglin transfectants was clearly enhanced in the presence of endoglin. Coating transwells with the RGD-containing extracellular domain of endoglin, enhanced leukocyte transmigration, and this increased motility was inhibited by soluble endoglin. Leukocytes stimulated with CXCL12, a chemokine involved in inflammation, strongly adhered to endoglin-coated plates and to endoglin-expressing endothelial cells. This endoglin-dependent adhesion was abolished by soluble endoglin, RGD peptides, the anti-integrin α5ß1 inhibitory antibody LIA1/2 and the chemokine receptor inhibitor AMD3100. These results demonstrate for the first time that endothelial endoglin interacts with leukocyte integrin α5ß1 via its RGD motif, and this adhesion process is stimulated by the inflammatory chemokine CXCL12, suggesting a regulatory role for endoglin in transendothelial leukocyte trafficking.


Asunto(s)
Antígenos CD/metabolismo , Quimiotaxis de Leucocito/fisiología , Células Endoteliales/metabolismo , Inflamación/metabolismo , Receptores de Superficie Celular/metabolismo , Migración Transendotelial y Transepitelial/fisiología , Animales , Adhesión Celular/fisiología , Ensayos de Migración de Leucocitos , Quimiocina CXCL12/metabolismo , Endoglina , Citometría de Flujo , Humanos , Integrina alfa5beta1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Fluorescente , Migración Transcelular de la Célula/fisiología
12.
Eur J Immunol ; 43(7): 1745-57, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23526587

RESUMEN

Janus kinases (JAKs) are central signaling molecules in cytokine receptor cascades. Although they have also been implicated in chemokine receptor signaling, this function continues to be debated. To address this issue, we established a nucleofection model in primary, nonactivated mouse T lymphocytes to silence JAK expression and to evaluate the ability of these cells to home to lymph nodes. Reduced JAK1 and JAK2 expression impaired naïve T-cell migration in response to gradients of the chemokines CXCL12 and CCL21. In vivo homing of JAK1/JAK2-deficient cells to lymph nodes decreased, whereas intranodal localization and motility were unaffected. JAK1 and JAK2 defects altered CXCL12- and CCL21-triggered ezrin/radixin/moesin (ERM) dephosphorylation and F-actin polymerization, as well as activation of lymphocyte function-associated Ag-1 and very late Ag-4 integrins. As a result, the cells did not adhere firmly to integrin substrates in response to these chemokines. The results demonstrate that JAK1/JAK2 participate in chemokine-induced integrin activation and might be considered a target for modulation of immune cell extravasation and therefore, control of inflammatory reactions.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , Integrinas/metabolismo , Janus Quinasa 1/metabolismo , Janus Quinasa 2/metabolismo , Linfocitos T/inmunología , Actinas/metabolismo , Animales , Western Blotting , Quimiocinas/inmunología , Quimiocinas/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Inmunohistoquímica , Integrinas/inmunología , Janus Quinasa 1/inmunología , Janus Quinasa 2/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Polimerizacion , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/metabolismo , Transfección
13.
J Pathol ; 231(4): 517-31, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24114721

RESUMEN

Peritoneal dissemination is a frequent metastatic route for cancers of the ovary and gastrointestinal tract. Tumour cells metastasize by attaching to and invading through the mesothelial cell (MC) monolayer that lines the peritoneal cavity. Metastases are influenced by carcinoma-associated fibroblasts (CAFs), a cell population that derives from different sources. Hence, we investigated whether MCs, through mesothelial-mesenchymal transition (MMT), were a source of CAFs during peritoneal carcinomatosis and whether MMT affected the adhesion and invasion of tumour cells. Biopsies from patients with peritoneal dissemination revealed the presence of myofibroblasts expressing mesothelial markers in the proximity of carcinoma implants. Prominent new vessel formation was observed in the peritoneal areas harbouring tumour cells when compared with tumour-free regions. The use of a mouse model of peritoneal dissemination confirmed the myofibroblast conversion of MCs and the increase in angiogenesis at places of tumour implants. Treatment of omentum MCs with conditioned media from carcinoma cell cultures resulted in phenotype changes reminiscent of MMT. Adhesion experiments demonstrated that MMT enhanced the binding of cancer cells to MCs in a ß1-integrin-dependent manner. Scanning electron microscopy imaging showed that the enhanced adhesion was mostly due to increased cell-cell interaction and not to a mere matrix exposure. Invasion assays suggested a reciprocal stimulation of the invasive capacity of tumour cells and MCs. Our results demonstrate that CAFs can derive from mesothelial cells during peritoneal metastasis. We suggest that MMT renders the peritoneum more receptive for tumour cell attachment/invasion and contributes to secondary tumour growth by promoting its vascularization.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Fibroblastos/patología , Neoplasias Peritoneales/secundario , Animales , Biopsia , Adhesión Celular , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Medios de Cultivo Condicionados/farmacología , Células Epiteliales/patología , Células Epiteliales/fisiología , Células Epiteliales/ultraestructura , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Fibroblastos/fisiología , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Microscopía Electrónica de Rastreo , Invasividad Neoplásica , Trasplante de Neoplasias , Neovascularización Patológica/patología , Neoplasias Ováricas/patología , Neoplasias Peritoneales/irrigación sanguínea , Neoplasias Peritoneales/patología , Neoplasias Peritoneales/ultraestructura
14.
Cell Mol Life Sci ; 70(13): 2395-410, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23420480

RESUMEN

Phagocytosis mediated by the complement receptor CR3 (also known as integrin αMß2 or Mac-1) is regulated by the recruitment of talin to the cytoplasmic tail of the ß2 integrin subunit. Talin recruitment to this integrin is dependent on Rap1 activation. However, the mechanism by which Rap1 regulates this event and CR3-dependent phagocytosis remains largely unknown. In the present work, we examined the role of the Rap1 effector RIAM, a talin-binding protein, in the regulation of complement-mediated phagocytosis. Using the human myeloid cell lines HL-60 and THP-1, we determined that knockdown of RIAM impaired αMß2 integrin affinity changes induced by stimuli fMLP and LPS. Phagocytosis of complement-opsonized RBC particles, but not of IgG-opsonized RBC particles, was impaired in RIAM knockdown cells. Rap1 activation via EPAC induced by 8-pCPT-2'-O-Me-cAMP resulted in an increase of complement-mediated phagocytosis that was abrogated by knockdown of RIAM in HL-60 and THP-1 cell lines and in macrophages derived from primary monocytes. Furthermore, recruitment of talin to ß2 integrin during complement-mediated phagocytosis was reduced in RIAM knockdown cells. These results indicate that RIAM is a critical component of the phagocytosis machinery downstream of Rap1 and mediates its function by recruiting talin to the phagocytic complement receptors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de la Membrana/fisiología , Fagocitosis/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos CD18/metabolismo , Antígenos CD18/fisiología , Células Cultivadas , Proteínas del Sistema Complemento/fisiología , Técnicas de Silenciamiento del Gen , Células HL-60 , Humanos , Antígeno de Macrófago-1/fisiología , Macrófagos/citología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Neutrófilos/citología , Neutrófilos/metabolismo , Talina/metabolismo , Talina/fisiología , Proteínas de Unión al GTP rap1/metabolismo , Proteínas de Unión al GTP rap1/fisiología
15.
Cell Mol Life Sci ; 70(3): 475-93, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23052204

RESUMEN

ALCAM/CD166 is a member of the immunoglobulin superfamily of cell adhesion molecules (Ig-CAMs) which mediates intercellular adhesion through either homophilic (ALCAM-ALCAM) or heterophilic (ALCAM-CD6) interactions. ALCAM-mediated adhesion is crucial in different physiological and pathological phenomena, with particular relevance in leukocyte extravasation, stabilization of the immunological synapse, T cell activation and proliferation and tumor growth and metastasis. Although the functional implications of ALCAM in these processes is well established, the mechanisms regulating its adhesive capacity remain obscure. Using confocal microscopy colocalization, and biochemical and functional analyses, we found that ALCAM directly associates with the tetraspanin CD9 on the leukocyte surface in protein complexes that also include the metalloproteinase ADAM17/TACE. The functional relevance of these interactions is evidenced by the CD9-induced upregulation of both homophilic and heterophilic ALCAM interactions, as reflected by increased ALCAM-mediated cell adhesion and T cell migration, activation and proliferation. The enhancement of ALCAM function induced by CD9 is mediated by a dual mechanism involving (1) augmented clustering of ALCAM molecules, and (2) upregulation of ALCAM surface expression due to inhibition of ADAM17 sheddase activity.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Tetraspanina 29/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Células CHO , Adhesión Celular , Línea Celular , Movimiento Celular , Cricetinae , Humanos , Células Jurkat , Células K562 , Leucocitos/metabolismo , Unión Proteica , Mapas de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Tetraspanina 29/antagonistas & inhibidores , Tetraspanina 29/genética , Regulación hacia Arriba
16.
J Exp Clin Cancer Res ; 43(1): 27, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38254102

RESUMEN

BACKGROUND: Peritoneal metastasis, which accounts for 85% of all epithelial ovarian carcinoma (EOC) metastases, is a multistep process that requires the establishment of adhesive interactions between cancer cells and the peritoneal membrane. Interrelations between EOC and the mesothelial stroma are critical to facilitate the metastatic process. No data is available so far on the impact of histone acetylation/deacetylation, a potentially relevant mechanism governing EOC metastasis, on mesothelial cells (MCs)-mediated adhesion. METHODS: Static adhesion and peritoneal clearance experiments were performed pretreating mesenchymal-like MCs and platinum-sensitive/resistant EOC cell lines with MS-275-a Histone deacetylase (HDAC)1-3 pharmacological inhibitor currently used in combination trials. Results were acquired by confocal microscopy and were analyzed with an automated Opera software. The role of HDAC1/2 was validated by genetic silencing. The role of α4-, α5-α1 Integrins and Fibronectin-1 was validated using specific monoclonal antibodies. Quantitative proteomic analysis was performed on primary MCs pretreated with MS-275. Decellularized matrices were generated from either MS-275-exposed or untreated cells to study Fibronectin-1 extracellular secretion. The effect of MS-275 on ß1 integrin activity was assessed using specific monoclonal antibodies. The role of Talin-1 in MCs/EOC adhesion was analyzed by genetic silencing. Talin-1 ectopic expression was validated as a rescue tool from MS-275-induced phenotype. The in vivo effect of MS-275-induced MC remodeling was validated in a mouse model of peritoneal EOC dissemination. RESULTS: Treatment of MCs with non-cytotoxic concentrations of MS-275 caused a consistent reduction of EOC adhesion. Proteomic analysis revealed several pathways altered upon MC treatment with MS-275, including ECM deposition/remodeling, adhesion receptors and actin cytoskeleton regulators. HDAC1/2 inhibition hampered actin cytoskeleton polymerization by downregulating actin regulators including Talin-1, impairing ß1 integrin activation, and leading to abnormal extracellular secretion and distribution of Fibronectin-1. Talin-1 ectopic expression rescued EOC adhesion to MS-275-treated MCs. In an experimental mouse model of metastatic EOC, MS-275 limited tumor invasion, Fibronectin-1 secretion and the sub-mesothelial accumulation of MC-derived carcinoma-associated fibroblasts. CONCLUSION: Our study unveils a direct impact of HDAC-1/2 in the regulation of MC/EOC adhesion and highlights the regulation of MC plasticity by epigenetic inhibition as a potential target for therapeutic intervention in EOC peritoneal metastasis.


Asunto(s)
Benzamidas , Carcinoma Epitelial de Ovario , Adhesión Celular , Histona Desacetilasa 1 , Histona Desacetilasa 2 , Neoplasias Ováricas , Neoplasias Peritoneales , Animales , Femenino , Humanos , Ratones , Citoesqueleto de Actina/metabolismo , Anticuerpos Monoclonales , Carcinoma Epitelial de Ovario/metabolismo , Epitelio , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas , Histona Desacetilasa 1/metabolismo , Integrina alfa5 , Integrina beta1/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/metabolismo , Proteómica , Piridinas , Talina/genética , Talina/metabolismo , Histona Desacetilasa 2/metabolismo , Adhesión Celular/genética
17.
J Biol Chem ; 286(21): 18492-504, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21454517

RESUMEN

The Mig-10/RIAM/lamellipodin (MRL) family member Rap1-GTP-interacting adaptor molecule (RIAM) interacts with active Rap1, a small GTPase that is frequently activated in tumors such as melanoma and prostate cancer. We show here that RIAM is expressed in metastatic human melanoma cells and that both RIAM and Rap1 are required for BLM melanoma cell invasion. RIAM silencing in melanoma cells led to inhibition of tumor growth and to delayed metastasis in a severe combined immunodeficiency xenograft model. Defective invasion of RIAM-silenced melanoma cells arose from impairment in persistent cell migration directionality, which was associated with deficient activation of a Vav2-RhoA-ROCK-myosin light chain pathway. Expression of constitutively active Vav2 and RhoA in cells depleted for RIAM partially rescued their invasion, indicating that Vav2 and RhoA mediate RIAM function. These results suggest that inhibition of cell invasion in RIAM-silenced melanoma cells is likely based on altered cell contractility and cell polarization. Furthermore, we show that RIAM depletion reduces ß1 integrin-dependent melanoma cell adhesion, which correlates with decreased activation of both Erk1/2 MAPK and phosphatidylinositol 3-kinase, two central molecules controlling cell growth and cell survival. In addition to causing inhibition of cell proliferation, RIAM silencing led to higher susceptibility to cell apoptosis. Together, these data suggest that defective activation of these kinases in RIAM-silenced cells could account for inhibition of melanoma cell growth and that RIAM might contribute to the dissemination of melanoma cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis/genética , Adhesión Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Silenciador del Gen , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Melanoma/genética , Melanoma/patología , Proteínas de la Membrana/genética , Ratones , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Trasplante Heterólogo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
18.
Cell Mol Life Sci ; 68(20): 3323-35, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21687991

RESUMEN

Several recent publications have described examples of physical and functional interations between tetraspanins and specific membrane proteases belonging to the TM-MMP and α-(ADAMs) and γ-secretases families. Collectively, these examples constitute an emerging body of evidence supporting the notion that tetraspanin-enriched microdomains (TEMs) represent functional platforms for the regulation of key cellular processes including the release of surface protein ectodomains ("shedding"), regulated intramembrane proteolysis ("RIPing") and matrix degradation and assembly. These cellular processes in turn play a crucial role in an array of physiological and pathological phenomena. Thus, TEMs may represent new therapeutical targets that may simultaneously affect the proteolytic activity of different enzymes and their substrates. Agonistic or antagonistic antibodies and blocking soluble peptides corresponding to tetraspanin functional regions may offer new opportunities in the treatment of pathologies such as chronic inflammation, cancer, or Alzheimer's disease. In this review article, we will discuss all these aspects of functional regulation of protease activities by tetraspanins.


Asunto(s)
Proteínas de la Membrana/metabolismo , Péptido Hidrolasas/metabolismo , Proteínas ADAM/metabolismo , Animales , Humanos , Metaloproteinasas de la Matriz/metabolismo , Tetraspaninas
19.
Cell Mol Life Sci ; 68(19): 3275-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21365281

RESUMEN

ADAM17/TACE is a metalloproteinase responsible for the shedding of the proinflammatory cytokine TNF-α and many other cell surface proteins involved in development, cell adhesion, migration, differentiation, and proliferation. Despite the important biological function of ADAM17, the mechanisms of regulation of its metalloproteinase activity remain largely unknown. We report here that the tetraspanin CD9 and ADAM17 partially co-localize on the surface of endothelial and monocytic cells. In situ proximity ligation, co-immunoprecipitation, crosslinking, and pull-down experiments collectively demonstrate a direct association between these molecules. Functional studies reveal that treatment with CD9-specific antibodies or neoexpression of CD9 exert negative regulatory effects on ADAM17 sheddase activity. Conversely, CD9 silencing increased the activity of ADAM17 against its substrates TNF-α and ICAM-1. Taken together, our results show that CD9 associates with ADAM17 and, through this interaction, negatively regulates the sheddase activity of ADAM17.


Asunto(s)
Proteínas ADAM/metabolismo , Antígenos CD/fisiología , Glicoproteínas de Membrana/fisiología , Proteínas ADAM/genética , Proteínas ADAM/fisiología , Proteína ADAM17 , Antígenos CD/genética , Antígenos CD/metabolismo , Línea Celular , Células Endoteliales/metabolismo , Silenciador del Gen , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Tetraspanina 29 , Factor de Necrosis Tumoral alfa/metabolismo
20.
Front Immunol ; 13: 951280, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36238292

RESUMEN

Activation of the integrin phagocytic receptors CR3 (αMß2, CD11b/CD18) and CR4 (αXß2, CD11c/CD18) requires Rap1 activation and RIAM function. RIAM controls integrin activation by recruiting Talin to ß2 subunits, enabling the Talin-Vinculin interaction, which in term bridges integrins to the actin-cytoskeleton. RIAM also recruits VASP to phagocytic cups and facilitates VASP phosphorylation and function promoting particle internalization. Using a CRISPR-Cas9 knockout approach, we have analyzed the requirement for RIAM, VASP and Vinculin expression in neutrophilic-HL-60 cells. All knockout cells displayed abolished phagocytosis that was accompanied by a significant and specific reduction in ITGAM (αM), ITGAX (αX) and ITGB2 (ß2) mRNA, as revealed by RT-qPCR. RIAM, VASP and Vinculin KOs presented reduced cellular F-actin content that correlated with αM expression, as treatment with the actin filament polymerizing and stabilizing drug jasplakinolide, partially restored αM expression. In general, the expression of αX was less responsive to jasplakinolide treatment than αM, indicating that regulatory mechanisms independent of F-actin content may be involved. The Serum Response Factor (SRF) was investigated as the potential transcription factor controlling αMß2 expression, since its coactivator MRTF-A requires actin polymerization to induce transcription. Immunofluorescent MRTF-A localization in parental cells was primarily nuclear, while in knockouts it exhibited a diffuse cytoplasmic pattern. Localization of FHL-2 (SRF corepressor) was mainly sub-membranous in parental HL-60 cells, but in knockouts the localization was disperse in the cytoplasm and the nucleus, suggesting RIAM, VASP and Vinculin are required to maintain FHL-2 close to cytoplasmic membranes, reducing its nuclear localization and inhibiting its corepressor activity. Finally, reexpression of VASP in the VASP knockout resulted in a complete reversion of the phenotype, as knock-ins restored αM expression. Taken together, our results suggest that RIAM, VASP and Vinculin, are necessary for the correct expression of αMß2 and αXß2 during neutrophilic differentiation in the human promyelocytic HL-60 cell line, and strongly point to an involvement of these proteins in the acquisition of a phagocytic phenotype.


Asunto(s)
Actinas , Talina , Proteínas Adaptadoras Transductoras de Señales , Moléculas de Adhesión Celular , Proteínas Co-Represoras , Células HL-60 , Humanos , Integrina alfaXbeta2 , Integrinas/metabolismo , Antígeno de Macrófago-1 , Proteínas de la Membrana , Proteínas de Microfilamentos , Neutrófilos/metabolismo , Fosfoproteínas , ARN Mensajero , Factor de Respuesta Sérica , Talina/genética , Talina/metabolismo , Vinculina/genética , Vinculina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA