Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
1.
J Am Soc Nephrol ; 28(7): 1998-2005, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28220032

RESUMEN

Activated fibroblasts are deemed the main executors of organ fibrosis. However, regulation of the pathologic functions of these cells in vivo is poorly understood. PDGF receptor ß (PDGFRß) is highly expressed in activated pericytes, a main source of fibroblasts. Studies using a PDGFRß promoter-driven Cre system to delete αv integrins in activated fibroblasts identified these integrins as core regulators of fibroblast activity across solid organs, including the kidneys. Here, we used the same PDGFRß-Cre line to isolate and study renal fibroblasts ex vivo We found that renal fibroblasts express three αv integrins, namely αvß1, αvß3, and αvß5. Blockade of αvß1 prevented direct binding of fibroblasts to the latency-associated peptide of TGF-ß1 and prevented activation of the latent TGF-ß complex. Continuous administration of a recently described potent small molecule inhibitor of αvß1, compound 8, starting the day of unilateral ureteral obstruction operation, inhibited collagen deposition in the kidneys of mice 14 days later. Compound 8 also effectively attenuated renal failure, as measured by BUN levels in mice fed an adenine diet known to cause renal injury followed by fibrosis. Inhibition of αvß1 integrin could thus hold promise as a therapeutic intervention in CKD characterized by renal fibrosis.


Asunto(s)
Guanidinas/farmacología , Guanidinas/uso terapéutico , Riñón/patología , Receptores de Vitronectina/antagonistas & inhibidores , Insuficiencia Renal/prevención & control , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Animales , Fibrosis/etiología , Fibrosis/prevención & control , Masculino , Ratones , Receptores de Vitronectina/fisiología , Insuficiencia Renal/etiología
2.
J Virol ; 91(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27974569

RESUMEN

Epidemic keratoconjunctivitis (EKC) is a severe, contagious ocular disease that affects 20 to 40 million individuals worldwide every year. EKC is mainly caused by six types of human adenovirus (HAdV): HAdV-8, -19, -37, -53, -54, and -56. Of these, HAdV-8, -19, and -37 use sialic acid-containing glycans as cellular receptors. αVß3, αVß5, and a few additional integrins facilitate entry and endosomal release of other HAdVs. With the exception of a few biochemical analyses indicating that HAdV-37 can interact physically with αVß5, little is known about the integrins used by EKC-causing HAdVs. Here, we investigated the overall integrin expression on human corneal cells and found expression of α2, α3, α6, αV, ß1, and ß4 subunits in human corneal in situ epithelium and/or in a human corneal epithelial (HCE) cell line but no or less accessible expression of α4, α5, ß3, or ß5. We also identified the integrins used by HAdV-37 through a series of binding and infection competition experiments and different biochemical approaches. Together, our data suggest that HAdV-37 uses αVß1 and α3ß1 integrins for infection of human corneal epithelial cells. Furthermore, to confirm the relevance of these integrins in the HAdV-37 life cycle, we developed a corneal multilayer tissue system and found that HAdV-37 infection correlated well with the patterns of αV, α3, and ß1 integrin expression. These results provide further insight into the tropism and pathogenesis of EKC-causing HAdVs and may be of importance for future development of new antiviral drugs.IMPORTANCE Keratitis is a hallmark of EKC, which is caused by six HAdV types (HAdV-8, -19, -37, -53, -54, and -56). HAdV-37 and some other HAdV types interact with integrin αVß5 in order to enter nonocular human cells. In this study, we found that αVß5 is not expressed on human corneal epithelial cells, thus proposing other host factors mediate corneal infection. Here, we first characterized integrin expression patterns on corneal tissue and corneal cells. Among the integrins identified, competition binding and infection experiments and biochemical assays pointed out αVß1 and α3ß1 to be of importance for HAdV-37 infection of corneal tissue. In the absence of a good animal model for EKC-causing HAdVs, we also developed an in vitro system with multilayer HCE cells and confirmed the relevance of the suggested integrins during HAdV-37 infection.


Asunto(s)
Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/fisiología , Integrina alfa3beta1/fisiología , Receptores de Vitronectina/fisiología , Células A549 , Córnea/patología , Córnea/virología , Humanos , Receptores Virales , Acoplamiento Viral , Internalización del Virus
3.
J Biol Chem ; 291(28): 14815-25, 2016 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-27226547

RESUMEN

Avian metapneumovirus (aMPV) fusion (F) protein mediates virus-cell membrane fusion to initiate viral infection, which requires F protein binding to its receptor(s) on the host cell surface. However, the receptor(s) for aMPV F protein is still not identified. All known subtype B aMPV (aMPV/B) F proteins contain a conserved Arg-Asp-Asp (RDD) motif, suggesting that the aMPV/B F protein may mediate membrane fusion via the binding of RDD to integrin. When blocked with integrin-specific peptides, aMPV/B F protein fusogenicity and viral replication were significantly reduced. Specifically we identified integrin αv and/or ß1-mediated F protein fusogenicity and viral replication using antibody blocking, small interfering RNAs (siRNAs) knockdown, and overexpression. Additionally, overexpression of integrin αv and ß1 in aMPV/B non-permissive cells conferred aMPV/B F protein binding and aMPV/B infection. When RDD was altered to RAE (Arg-Ala-Glu), aMPV/B F protein binding and fusogenic activity were profoundly impaired. These results suggest that integrin αvß1 is a functional receptor for aMPV/B F protein-mediated membrane fusion and virus infection, which will provide new insights on the fusogenic mechanism and pathogenesis of aMPV.


Asunto(s)
Fusión Celular , Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/fisiopatología , Receptores de Vitronectina/fisiología , Proteínas Virales de Fusión/fisiología , Animales , Línea Celular , Infecciones por Paramyxoviridae/virología , Replicación Viral
4.
PLoS One ; 11(4): e0154769, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27128974

RESUMEN

Human parechovirus 1 (HPeV-1) (family Picornaviridae) is a global cause of pediatric respiratory and CNS infections for which there is no treatment. Although biochemical and in vitro studies have suggested that HPeV-1 binds to αVß1, αVß3 and αVß6 integrin receptor(s), the actual cellular receptors required for infectious entry of HPeV-1 remain unknown. In this paper we analyzed the expression profiles of αVß1, αVß3, αVß6 and α5ß1 in susceptible cell lines (A549, HeLa and SW480) to identify which integrin receptors support HPeV-1 internalization and/or replication cycle. We demonstrate by antibody blocking assay, immunofluorescence microscopy and RT-qPCR that HPeV-1 internalizes and replicates in cell lines that express αVß1 integrin but not αVß3 or αVß6 integrins. To further study the role of ß1 integrin, we used a mouse cell line, GE11-KO, which is deficient in ß1 expression, and its derivate GE11-ß1 in which human integrin ß1 subunit is overexpressed. HPeV-1 (Harris strain) and three clinical HPeV-1 isolates did not internalize into GE11-KO whereas GE11-ß1 supported the internalization process. An integrin ß1-activating antibody, TS2/16, enhanced HPeV-1 infectivity, but infection occurred in the absence of visible receptor clustering. HPeV-1 also co-localized with ß1 integrin on the cell surface, and HPeV-1 and ß1 integrin co-endocytosed into the cells. In conclusion, our results demonstrate that in some cell lines the cellular entry of HPeV-1 is primarily mediated by the active form of αVß1 integrin without visible receptor clustering.


Asunto(s)
Parechovirus/patogenicidad , Infecciones por Picornaviridae/etiología , Receptores de Vitronectina/fisiología , Internalización del Virus , Animales , Antígenos de Neoplasias/fisiología , Línea Celular , Línea Celular Tumoral , Células HeLa , Humanos , Integrina alfaVbeta3/fisiología , Integrinas/fisiología , Ratones , Parechovirus/fisiología , Infecciones por Picornaviridae/fisiopatología , Infecciones por Picornaviridae/virología , Receptores Virales/fisiología
5.
J Dent Res ; 95(8): 939-45, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26912222

RESUMEN

Transforming growth factor ß (TGFß) regulates cell proliferation, differentiation, migration, apoptosis, and extracellular matrix production. It also plays a pivotal role in the pathogenesis of gingival overgrowth. Thrombin is a key player in tissue repair, remodeling, and fibrosis after an injury, and it exerts profibrotic effects by activating protease-activated receptors. Connective tissue growth factor (CTGF or CCN2) modulates cell adhesion, migration, proliferation, matrix production, and wound healing. It is overexpressed in many fibrotic disorders, including gingival overgrowth, and it is positively associated with the degree of fibrosis in gingival overgrowth. In human gingival fibroblasts, we previously found that TGFß1 induced CCN2 protein synthesis through c-jun N-terminal kinase and Smad3 activation. Thrombin stimulates CCN2 synthesis through protease-activated receptor 1 and c-jun N-terminal kinase signaling. Curcumin inhibited TGFß1- and thrombin-induced CCN2 synthesis. In this study, we demonstrated that thrombin and protease-activated receptor 1 agonist SFLLRN induced latent TGFß1 activation and Smad3 phosphorylation in human gingival fibroblasts. Pretreatment with a TGFß-neutralizing antibody, TGFß type I receptor inhibitor SB431542, and Smad3 inhibitor SIS3 inhibited approximately 86%, 94%, and 100% of thrombin-induced CCN2 synthesis, respectively. Furthermore, blocking integrin subunits αv and ß1 with antibodies effectively inhibited SFLLRN-induced Smad3 phosphorylation and CCN2 synthesis and increased activated TGFß1 levels; however, similar effects were not observed for integrins αvß3 and αvß5. These results suggest that protease-activated receptor 1-induced CCN2 synthesis in human gingival fibroblasts is mediated through integrin αvß1-induced latent TGFß1 activation and subsequent TGFß1 signaling. Moreover, curcumin dose dependently decreased thrombin-induced activated TGFß1 levels. Curcumin-inhibited thrombin-induced CCN2 synthesis in human gingival fibroblasts is caused by the suppression of latent TGFß1 activation.


Asunto(s)
Fibroblastos/fisiología , Encía/fisiología , Receptores de Vitronectina/fisiología , Trombina/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Western Blotting , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Curcumina/farmacología , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/efectos de los fármacos , Encía/citología , Encía/efectos de los fármacos , Humanos
6.
J Clin Invest ; 125(5): 1886-900, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25822023

RESUMEN

Liver cholestatic diseases, which stem from diverse etiologies, result in liver toxicity and fibrosis and may progress to cirrhosis and liver failure. We show that CCN1 (also known as CYR61), a matricellular protein that dampens and resolves liver fibrosis, also mediates cholangiocyte proliferation and ductular reaction, which are repair responses to cholestatic injury. In cholangiocytes, CCN1 activated NF-κB through integrin αvß5/αvß3, leading to Jag1 expression, JAG1/NOTCH signaling, and cholangiocyte proliferation. CCN1 also induced Jag1 expression in hepatic stellate cells, whereupon they interacted with hepatic progenitor cells to promote their differentiation into cholangiocytes. Administration of CCN1 protein or soluble JAG1 induced cholangiocyte proliferation in mice, which was blocked by inhibitors of NF-κB or NOTCH signaling. Knock-in mice expressing a CCN1 mutant that is unable to bind αvß5/αvß3 were impaired in ductular reaction, leading to massive hepatic necrosis and mortality after bile duct ligation (BDL), whereas treatment of these mice with soluble JAG1 rescued ductular reaction and reduced hepatic necrosis and mortality. Blockade of integrin αvß5/αvß3, NF-κB, or NOTCH signaling in WT mice also resulted in defective ductular reaction after BDL. These findings demonstrate that CCN1 induces cholangiocyte proliferation and ductular reaction and identify CCN1/αvß5/NF-κB/JAG1 as a critical axis for biliary injury repair.


Asunto(s)
Conductos Biliares/metabolismo , Proteína 61 Rica en Cisteína/fisiología , Hígado/metabolismo , FN-kappa B/metabolismo , Receptores de Vitronectina/fisiología , Animales , Conductos Biliares/fisiología , Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/farmacología , Proteínas de Unión al Calcio/uso terapéutico , División Celular , Células Cultivadas , Colestasis Extrahepática/genética , Colestasis Extrahepática/metabolismo , Colestasis Extrahepática/patología , Proteína 61 Rica en Cisteína/genética , Proteína 61 Rica en Cisteína/farmacología , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Humanos , Integrina alfaVbeta3 , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Proteína Jagged-1 , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas de la Membrana/farmacología , Proteínas de la Membrana/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , Receptores Notch/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Regeneración , Proteínas Serrate-Jagged
7.
Int J Cancer ; 136(6): E545-58, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25284767

RESUMEN

During melanoma progression, tumour cells show increased adhesiveness to the vascular wall, invade the extracellular matrix (ECM) and frequently form functional channels similar to vascular vessels (vasculogenic mimicry). These properties are mainly mediated by the interaction of integrins with ECM components. Since we had previously identified neuropilin 1 (NRP-1), a coreceptor of vascular endothelial growth factor A (VEGF-A), as an important determinant of melanoma aggressiveness, aims of this study were to identify the specific integrins involved in the highly invasive phenotype of NRP-1 expressing cells and to investigate their role as targets to counteract melanoma progression. Melanoma aggressiveness was evaluated in vitro as cell ability to migrate through an ECM layer and to form tubule-like structures using transfected cells. Integrins relevant to these processes were identified using specific blocking antibodies. The αvß5 integrin was found to be responsible for about 80% of the capability of NRP-1 expressing cells to adhere on vitronectin. In these cells αvß5 expression level was twice higher than in low-invasive control cells and contributed to the ability of melanoma cells to form tubule-like structures on matrigel. Cilengitide, a potent inhibitor of αν integrins activation, reduced ECM invasion, vasculogenic mimicry and secretion of VEGF-A and metalloproteinase 9 by melanoma cells. In conclusion, we demonstrated that ανß5 integrin is involved in the highly aggressive phenotype of melanoma cells expressing NRP-1. Moreover, we identified a novel mechanism that contributes to the antimelanoma activity of the αv integrin inhibitor cilengitide based on the inhibition of vasculogenic mimicry.


Asunto(s)
Melanoma/tratamiento farmacológico , Neuropilina-1/fisiología , Receptores de Vitronectina/antagonistas & inhibidores , Venenos de Serpiente/farmacología , Línea Celular Tumoral , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Melanoma/química , Melanoma/patología , Invasividad Neoplásica , Neuropilina-1/análisis , Receptores de Vitronectina/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis
8.
Lab Invest ; 94(12): 1419-30, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25310532

RESUMEN

Ulcerative colitis (UC) is characterized by increased epithelial cell death and subsequent breakdown of the intestinal epithelial barrier, which perpetuates chronic intestinal inflammation. Since fecal bile acid dysmetabolism is associated with UC and tauroursodeoxycholic acid (TUDCA) has been shown to improve murine colitis, we evaluated the effect of TUDCA on intestinal epithelial cell death in a mouse model of UC-like barrier dysfunction elicited by dextran sulfate sodium (DSS). We identified the prevention of colonic caspase-3 induction, a key proapoptotic marker which was also over-activated in UC, as the earliest event resulting in a clear clinical benefit. Whereas vehicle-treated mice showed a cumulative mortality of 40%, all TUDCA-treated mice survived the DSS experiment during a 14-day follow-up period. In line with a barrier protective effect, TUDCA decreased bacterial translocation to the spleen and stimulated mucin production. Similarly, TUDCA inhibited lipopolysaccharide-induced intestinal permeability and associated enterocyte apoptosis. The anti-apoptotic effect was confirmed in vitro by a dose-dependent inhibition of both receptor-dependent (using tumor necrosis factor and Fas ligand) and receptor-independent (staurosporine) caspase-3 induction in HT29 colonic epithelial cells. These data imply that caspase-3 activation is an early marker of colitis that is prevented by TUDCA treatment. These data, together with the previously reported beneficial effect in colitis, suggest that TUDCA could be an add-on strategy to current immunosuppressive treatment of UC patients.


Asunto(s)
Apoptosis/efectos de los fármacos , Colitis/prevención & control , Células Epiteliales/patología , Mucosa Intestinal/efectos de los fármacos , Ácido Tauroquenodesoxicólico/uso terapéutico , Adolescente , Adulto , Anciano , Animales , Caspasa 3/metabolismo , Niño , Colitis/inducido químicamente , Sulfato de Dextran , Células Epiteliales/metabolismo , Femenino , Células HT29 , Humanos , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/fisiología , Receptores de Vitronectina/fisiología
9.
PLoS One ; 9(9): e108464, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25247809

RESUMEN

Low survival rates of metastatic cancers emphasize the need for a drug that can prevent and/or treat metastatic cancer. αv integrins are involved in essential processes for tumor growth and metastasis and targeting of αv integrins has been shown to decrease angiogenesis, tumor growth and metastasis. In this study, the role of αv integrin and its potential as a drug target in bladder cancer was investigated. Treatment with an αv integrin antagonist as well as knockdown of αv integrin in the bladder carcinoma cell lines, resulted in reduced malignancy in vitro, as illustrated by decreased proliferative, migratory and clonogenic capacity. The CDH1/CDH2 ratio increased, indicating a shift towards a more epithelial phenotype. This shift appeared to be associated with downregulation of EMT-inducing transcription factors including SNAI2. The expression levels of the self-renewal genes NANOG and BMI1 decreased as well as the number of cells with high Aldehyde Dehydrogenase activity. In addition, self-renewal ability decreased as measured with the urosphere assay. In line with these observations, knockdown or treatment of αv integrins resulted in decreased metastatic growth in preclinical in vivo models as assessed by bioluminescence imaging. In conclusion, we show that αv integrins are involved in migration, EMT and maintenance of Aldehyde Dehydrogenase activity in bladder cancer cells. Targeting of αv integrins might be a promising approach for treatment and/or prevention of metastatic bladder cancer.


Asunto(s)
Carcinoma de Células Transicionales/patología , Vectores Genéticos/farmacología , Integrina alfaV/efectos de los fármacos , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Receptores de Vitronectina/antagonistas & inhibidores , Neoplasias de la Vejiga Urinaria/patología , Aldehído Deshidrogenasa/biosíntesis , Aldehído Deshidrogenasa/genética , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Corazón , Humanos , Integrina alfaV/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Trasplante de Neoplasias/métodos , Papiloma/patología , Receptores de Vitronectina/fisiología , Tibia , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transducción Genética , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cardiovasc Res ; 102(3): 407-17, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24639195

RESUMEN

AIMS: Pathological tissue remodelling by myofibroblast contraction is a hallmark of cardiac fibrosis. Myofibroblasts differentiate from cardiac fibroblasts under the action of transforming growth factor-ß1 (TGF-ß1), which is secreted into the extracellular matrix as a large latent complex. Integrin-mediated traction forces activate TGF-ß1 by inducing a conformational change in the latent complex. The mesenchymal integrins αvß5 and αvß3 are expressed in the heart, but their role in the activation of TGF-ß1 remains elusive. Here, we test whether targeting αvß5 and αvß3 integrins reduces latent TGF-ß1 activation by cardiac fibroblasts with the goal to prevent the formation of α-smooth muscle actin (α-SMA)-expressing cardiac myofibroblasts and their contribution to fibrosis. METHODS AND RESULTS: Using a porcine model of induced right ventricular fibrosis and pro-fibrotic culture conditions, we show that integrins αvß5 and αvß3 are up-regulated in myofibroblast-enriched fibrotic lesions and differentiated cultured human cardiac myofibroblasts. Both integrins autonomously contribute to latent TGF-ß1 activation and myofibroblast differentiation, as demonstrated by function-blocking peptides and antibodies. Acute blocking of both integrins leads to significantly reduced TGF-ß1 activation by cardiac fibroblast contraction and loss of α-SMA expression, which is restored by adding active TGF-ß1. Manipulating integrin protein levels in overexpression and shRNA experiments reveals that both integrins can compensate for each other with respect to TGF-ß1 activation and induction of α-SMA expression. CONCLUSIONS: Integrins αvß5 and αvß3 both control myofibroblast differentiation by activating latent TGF-ß1. Pharmacological targeting of mesenchymal integrins is a possible strategy to selectively block TGF-ß1 activation by cardiac myofibroblasts and progression of fibrosis in the heart.


Asunto(s)
Integrina alfaVbeta3/fisiología , Miofibroblastos/patología , Receptores de Vitronectina/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Actinas/análisis , Adulto , Animales , Diferenciación Celular , Células Cultivadas , Fibrosis , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Masculino , Miocardio/patología , Receptores de Vitronectina/antagonistas & inhibidores , Porcinos
11.
Front Biosci (Landmark Ed) ; 19(3): 408-15, 2014 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-24389192

RESUMEN

Interaction of pathogenic bacteria with human cells is usually an essential step in the infection process. The bacterial invasion is stimulated by microbial binding to mammalian extracellular matrix proteins such as vitronectin, fibronectin or integrins. We have recently shown that some strains isolated from a clinical environment are able to grow at/or above 37°C. In particular, we demonstrated that P. fluorescens AF181 binds specifically to the surface of A549 human respiratory epithelial cells and that adhesiveness modulates the inflammatory response. In this study, the involvement of Alpha(v)Beta5 integrins and its respective natural ligand vitronectin (VN) in P. fluorescens AF181 adherence and invasion was examined. The host cell cytoskeleton and cellular tyrosine kinases seem to be solicited during the P. fluorescens-respiratory cell interaction; consequently, cytochalasin D and genistein decreased the bacterial adherence and internalization. Gene silencing of α(v), ß5 integrins and vitronectin reduced P. fluorescens adherence and internalization to A549 cells. Taken together, these findings suggest that Alpha(v)Beta5 integrins and their natural ligand VN are involved in P. fluorescens adherence and invasion in human epithelial cells.


Asunto(s)
Adhesión Bacteriana/fisiología , Pseudomonas fluorescens/fisiología , Receptores de Vitronectina/fisiología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Pulmón/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Invest Ophthalmol Vis Sci ; 54(7): 5000-11, 2013 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-23821196

RESUMEN

PURPOSE: To investigate the roles of αvß3 and αvß5 integrins in phagocytosis in human trabecular meshwork (HTM) cells. METHODS: Immunofluorescence microscopy and FACS analysis were used to determine levels of αvß3 and αvß5 integrins in TM tissue and cultures of normal and immortalized TM cells. Phagocytosis was measured using pHrodo-labeled S. aureus bioparticles followed by FACS analysis. The role of αvß5 integrin in phagocytosis was evaluated by knocking down αvß5 integrin expression with siRNA against the human ß5 gene. Signaling from focal adhesion kinase (FAK) was blocked using FAK inhibitor 14. The role of αvß3 integrins in phagocytosis was determined by treating HTM cells with dexamethasone (DEX) or ethanol (EtOH) and by generating stable cell lines that overexpressed either wild type (WT) or constitutively active (CA) ß3 integrin subunit. RESULTS: Both TM tissue and cell lines expressed αvß3 and αvß5 integrins. Knockdown of αvß5 integrin reduced phagocytosis by ∼60% and FAK inhibition significantly reduced phagocytosis up to 84%, in a dose-dependent manner. DEX treatment increased αvß3 integrin expression in HTM cells but reduced phagocytosis by ∼50% compared with untreated and EtOH-treated cells. The CA ß3 integrin-expressing cell line showed increased αvß3 integrin levels and decreased phagocytosis by ∼50% compared with the control. CONCLUSIONS: The αvß5 integrin-FAK-mediated pathway regulates phagocytosis in TM cells and this pathway is inhibited by activation of αvß3 integrins. This suggests that changes in integrin expression and activity may be responsible for alterations in phagocytosis observed in steroid induced glaucoma.


Asunto(s)
Integrina alfaVbeta3/fisiología , Fagocitosis/fisiología , Receptores de Vitronectina/fisiología , Malla Trabecular/efectos de los fármacos , Adulto , Análisis de Varianza , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Humanos , Integrina alfaVbeta3/metabolismo , Fagocitosis/efectos de los fármacos , Receptores de Vitronectina/metabolismo , Transducción de Señal/fisiología , Malla Trabecular/metabolismo
13.
Cancer Res ; 73(10): 3155-67, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23536555

RESUMEN

The molecular mechanisms underlying metastatic dissemination are still not completely understood. We have recently shown that ß(1) integrin-dependent cell adhesion to fibronectin and signaling is affected by a transmembrane molecule, Trop-2, which is frequently upregulated in human carcinomas. Here, we report that Trop-2 promotes metastatic dissemination of prostate cancer cells in vivo and is abundantly expressed in metastasis from human prostate cancer. We also show here that Trop-2 promotes prostate cancer cell migration on fibronectin, a phenomenon dependent on ß(1) integrins. Mechanistically, we demonstrate that Trop-2 and the α(5)ß(1) integrin associate through their extracellular domains, causing relocalization of α(5)ß(1) and the ß(1)-associated molecule talin from focal adhesions to the leading edges. Trop-2 effect is specific as this molecule does not modulate migration on vitronectin, does not associate with the major vitronectin receptor, α(v)ß(3) integrin, and does not affect localization of α(v)ß(3) integrin as well as vinculin in focal adhesions. We show that Trop-2 enhances directional prostate cancer cell migration, through modulation of Rac1 GTPase activity. Finally, we show that Trop-2 induces activation of PAK4, a kinase that has been reported to mediate cancer cell migration. In conclusion, we provide the first evidence that ß(1) integrin-dependent migratory and metastatic competence of prostate cancer cells is enhanced by Trop-2.


Asunto(s)
Antígenos de Neoplasias/fisiología , Moléculas de Adhesión Celular/fisiología , Integrina beta1/fisiología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Movimiento Celular , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Multimerización de Proteína , Receptores de Vitronectina/fisiología
14.
J Immunol ; 188(11): 5776-84, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22547695

RESUMEN

Cysteine-rich protein 61 (Cyr61)/CCN1 is a product of an immediate early gene and functions in mediating cell adhesion and inducing cell migration. We previously showed that increased production of Cyr61 by fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA) promotes FLS proliferation and participates in RA pathogenesis with the IL-17-dependent pathway. However, whether Cyr61 in turn regulates Th17 cell differentiation and further enhances inflammation of RA remained unknown. In the current study, we explored the potential role of Cyr61 as a proinflammatory factor in RA pathogenesis. We found that Cyr61 treatment dramatically induced IL-6 production in FLS isolated from RA patients. Moreover, IL-6 production was attenuated by Cyr61 knockdown in FLS. Mechanistically, we showed that Cyr61 activated IL-6 production via the αvß5/Akt/NF-κB signaling pathway. Further, using a coculture system consisting of purified CD4(+) T cells and RA FLS, we found that RA FLS stimulated Th17 differentiation, and the pro-Th17 differentiation effect of RA FLS can be attenuated or stimulated by Cyr61 RNA interference or addition of exogenous Cyr61, respectively. Finally, using the collagen-induced arthritis animal model, we showed that treatment with the anti-Cyr61 mAb led to reduction of IL-6 levels, decrease of Th17 response, and attenuation of inflammation and disease progression in vivo. Taken together, our results reveal a novel role of Cyr61 in promoting Th17 development in RA via upregulation of IL-6 production by FLS, thus adding a new layer into the functional interplay between FLS and Th17 in RA pathogenesis. Our study also suggests that targeting of Cyr61 may represent a novel strategy in RA treatment.


Asunto(s)
Artritis Reumatoide/inmunología , Diferenciación Celular/inmunología , Proteína 61 Rica en Cisteína/fisiología , Fibroblastos/inmunología , Interleucina-6/biosíntesis , Membrana Sinovial/inmunología , Células Th17/inmunología , Adulto , Anciano , Animales , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Células Cultivadas , Técnicas de Cocultivo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Interleucina-6/fisiología , Masculino , Ratones , Ratones Endogámicos DBA , Persona de Mediana Edad , Receptores de Vitronectina/fisiología , Transducción de Señal/inmunología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Células Th17/patología
15.
Exp Eye Res ; 94(1): 63-70, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22138557

RESUMEN

Efficient phagocytosis of photoreceptor outer segments (POS) by retinal pigment epithelial cells (RPE) plays a key role in biological renewal of these highly peroxidizable structures and in maintenance of retina health. Here, we used an in vitro RPE cell phagocytosis assay to investigate how sub-lethal oxidative stress modifies the key components of the cell phagocytic machinery leading to severe impairment of phagocytosis. Sub-lethal oxidative treatment, induced by hydrogen peroxide (H(2)O(2)), significantly inhibited binding and uptake of POS by RPE cells. However, sub-lethal oxidative stress did not affect cell surface expression of αvß5 or RPE cell adhesion to αvß5. Similarly, the enzymatic activity of mature cathepsin D was not altered upon challenge by oxidative stress. In contrast, studies of signaling molecules in the RPE cell phagocytic machinery revealed that sub-lethal oxidative stress inhibits POS-induced activation of FAK and MerTK. Our data demonstrate that sub-lethal oxidative treatment with H(2)O(2) inhibits phagocytic activity of ARPE-19 cells, in part by inhibiting FAK and MerTK.


Asunto(s)
Quinasa 1 de Adhesión Focal/fisiología , Fagocitosis/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Vitronectina/fisiología , Epitelio Pigmentado de la Retina/metabolismo , Catepsina D/metabolismo , Adhesión Celular , Línea Celular , Supervivencia Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Peróxido de Hidrógeno/toxicidad , Immunoblotting , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Tirosina Quinasa c-Mer
16.
J Immunol ; 188(3): 1108-16, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22198947

RESUMEN

The intestinal immune system is constantly challenged by commensal bacteria; therefore, it must maintain quiescence via several regulatory mechanisms. Although intestinal macrophages (Ms) have been implicated in repression of excessive inflammation, it remains unclear how their functions are regulated during inflammation. In this study, we report that semaphorin 7A (Sema7A), a GPI-anchored semaphorin expressed in intestinal epithelial cells (IECs), induces IL-10 production by intestinal Mϕs to regulate intestinal inflammation. Sema7A-deficient mice showed severe signs of dextran sodium sulfate-induced colitis due to reduced intestinal IL-10 levels. We further identified CX3CR1(+)MHC class II(int)F4/80(hi)CD11b(hi) Mϕs as the main producers of IL-10 via αvß1 integrin in response to Sema7A. Notably, Sema7A was predominantly expressed on the basolateral side of IECs, and its expression pattern was responsible for protective effects against dextran sodium sulfate-induced colitis and IL-10 production by Mϕs during interactions between IECs and Mϕs. Furthermore, we determined that the administration of recombinant Sema7A proteins ameliorated the severity of colitis, and these effects were diminished by IL-10-blocking Abs. Therefore, our findings not only indicate that Sema7A plays crucial roles in suppressing intestinal inflammation through αvß1 integrin, but also provide a novel mode of IL-10 induction via interactions between IECs and Mϕs.


Asunto(s)
Antígenos CD/fisiología , Colitis/patología , Receptores de Vitronectina/fisiología , Semaforinas/fisiología , Animales , Comunicación Celular , Colitis/etiología , Células Epiteliales/metabolismo , Interleucina-10/biosíntesis , Interleucina-10/genética , Intestinos , Macrófagos , Ratones
17.
Bull Cancer ; 98(6): 47-57, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21636344

RESUMEN

Radiotherapy is successfully used to treat cancer. Emerging evidence, however, indicates that recurrences after radiotherapy are associated with increased local invasion, metastatic spreading and poor prognosis. Radiation-induced modifications of the tumor microenvironment have been proposed to contribute to increased aggressive tumor behavior, an effect also referred to as tumor bed effect, but the putative mechanisms involved have remained largely elusive. We have recently demonstrated that irradiation of the prospective tumor stroma impairs de novo angiogenesis through sustained inhibition of proliferation, migration and sprouting of endothelial cells. Experimental tumors growing within a pre-irradiated field have reduced tumor angiogenesis and tumor growth, increased hypoxia, necrosis, local invasion and distant metastasis. Mechanisms of progression involve adaptation of tumor cells to local hypoxic conditions as well as selection of cells with invasive and metastatic capacities. The matricellular protein CYR61 and integrin αVß5 emerged as molecules that cooperate to mediate lung metastasis. Cilengitide, a small molecular inhibitor of αV integrins prevented lung metastasis formation. These results represent a conceptual advance to the understanding of the tumor bed effect and indicate that αV integrin inhibition might be a potential therapeutic approach for preventing metastasis in patients at risk for post-radiation recurrences.


Asunto(s)
Neoplasias/radioterapia , Microambiente Tumoral/efectos de la radiación , Inhibidores de la Angiogénesis/uso terapéutico , Hipoxia de la Célula/fisiología , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Proteína 61 Rica en Cisteína/fisiología , Progresión de la Enfermedad , Células Endoteliales/efectos de la radiación , Humanos , Microvasos/efectos de la radiación , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Neoplasias/irrigación sanguínea , Neoplasias/fisiopatología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/radioterapia , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/fisiología , Venenos de Serpiente/uso terapéutico , Microambiente Tumoral/fisiología
18.
J Immunol ; 186(1): 464-70, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21098231

RESUMEN

Monocytes and macrophages are targets of HIV-1 infection and play critical roles in multiple aspects of viral pathogenesis. During the differentiation of monocytes to macrophages, adhesion molecules such as integrins are upregulated; therefore, they provide signals that control the process and subsequently may render macrophages more susceptible to HIV-1 infection. Previous work demonstrated that blocking α(v)-containing integrins triggered a signal transduction pathway leading to the inhibition of NF-κB-dependent HIV-1 transcription. In this paper, we show the influence of the different α(v)-coupled ß integrins in HIV-1 replication in macrophages. Inhibition of ß integrins, either by specific mAbs, small arginine-glycine-aspartic acid (RGD) mimetic compounds, or RNA interference, showed that integrin ß(5) was the major contributor to the integrin-mediated blockade of HIV-1 replication. Importantly, such inhibition did not induce changes in cell adhesion to the substrate. In conclusion, our results reveal a significant role of the integrin dimer α(v)ß(5) in HIV-1 infection of macrophages.


Asunto(s)
Antivirales/metabolismo , VIH-1/inmunología , Cadenas beta de Integrinas/fisiología , Multimerización de Proteína/inmunología , Receptores de Vitronectina/fisiología , Replicación Viral/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Predisposición Genética a la Enfermedad , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , VIH-1/patogenicidad , Humanos , Cadenas beta de Integrinas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/virología , Unión Proteica/genética , Unión Proteica/inmunología , Multimerización de Proteína/genética , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Replicación Viral/genética
19.
Biochem Pharmacol ; 81(5): 577-85, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21145881

RESUMEN

Osteosarcoma is characterized by a high malignant and metastatic potential. Cyclooxygenase (COX)-2, the inducible isoform of prostaglandin synthase, has been implicated in tumor metastasis. Nephroblastoma overexpressed gene (NOV), also called CCN3, was regulated proliferation and differentiation of cancer cells. However, the effect of NOV on migration activity and COX-2 expression in human osteosarcoma cells is mostly unknown. Here we found that NOV increased the migration and expression of COX-2 in human osteosarcoma cells. αvß5 monoclonal antibody (mAb), integrin-linked kinase (ILK) and Akt inhibitor reduced the NOV-enhanced the migration and COX-2 up-regulation of osteosarcoma cells. NOV stimulation increased the ILK kinase activity and phosphorylation of Akt. In addition, c-Jun siRNA also antagonized the NOV-mediated migration and COX-2 expression. Moreover, NOV enhanced the AP-1 binding activity and promoter activity. Taken together, these results suggest that the NOV acts through αvß5 integrin to activate ILK and Akt, which in turn activates c-Jun and AP-1, resulting in the activations of COX-2 and contributing the migration of human osteosarcoma cells.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Proteína Hiperexpresada del Nefroblastoma/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Vitronectina/fisiología , Factor de Transcripción AP-1/fisiología , Neoplasias Óseas , Línea Celular Tumoral , Movimiento Celular , Humanos , Osteosarcoma , Transducción de Señal , Regulación hacia Arriba
20.
J Gen Virol ; 91(Pt 1): 155-65, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19776235

RESUMEN

Previously published data suggest that the RGD-recognizing integrin, alphavbeta3, known as the vitronectin receptor, acts as a cellular receptor for RGD-containing enteroviruses, coxsackievirus A9 (CAV-9) and echovirus 9 (E-9), in several continuous cell lines as well as in primary human Langerhans' islets. As this receptor is also capable of binding the ligands by a non-RGD-dependent mechanism, we investigated whether vitronectin receptors, alpha v integrins, might act as receptors for other echoviruses that do not have the RGD motif. Blocking experiments with polyclonal anti-alphavbeta3 antibody showed that both primary human islets and a continuous laboratory cell line of green monkey kidney origin (GMK) are protected similarly from the adverse effects of several non-RGD-containing echovirus (E-7, -11, -25, -30, -32) infections. In contrast, corresponding studies on primary human endothelial cells showed that the receptor works only for E-25, E-30, E-32 and CAV-9. The inhibitory effect of the antibody was not restricted to prototype strains of echoviruses, as GMK cells infected with several field isolates of the corresponding serotypes were also protected from virus-induced cytopathic effects. Co-localization of virus particles with the receptor molecules in both GMK and primary human endothelial cells was demonstrated by live-cell stainings and confocal microscopy. Remarkably, in spite of similar virus-receptor co-localization and a comparable protective effect of the alphavbeta3 antibody, the entry pathways of the studied virus strains seemed to be divergent.


Asunto(s)
Células Endoteliales/virología , Enterovirus Humano B/fisiología , Integrina alfaV/fisiología , Islotes Pancreáticos/virología , Receptores Virales/fisiología , Receptores de Vitronectina/fisiología , Acoplamiento Viral , Animales , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Humanos , Microscopía Confocal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA