Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 7290, 2024 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-38538704

RESUMEN

Bone destruction, a major source of morbidity, is mediated by heightened differentiation and activity of osteoclasts (OC), highly specialized multinucleated myeloid cells endowed with unique bone-resorptive capacity. The molecular mechanisms regulating OC differentiation in the bone marrow are still partly elusive. Here, we aimed to identify new regulatory circuits and actionable targets by comprehensive proteomic characterization of OCgenesis from mouse bone marrow monocytes, adopting two parallel unbiased comparative proteomic approaches. This work disclosed an unanticipated protein signature of OCgenesis, with most gene products currently unannotated in bone-related functions, revealing broad structural and functional cellular reorganization and divergence from macrophagic immune activity. Moreover, we identified the deubiquitinase UCHL1 as the most upregulated cytosolic protein in differentiating OCs. Functional studies proved it essential, as UCHL1 genetic and pharmacologic inhibition potently suppressed OCgenesis. Furthermore, proteomics and mechanistic dissection showed that UCHL1 supports OC differentiation by restricting the anti-OCgenic activity of NRF2, the transcriptional activator of the canonical antioxidant response, through redox-independent stabilization of the NRF2 inhibitor, KEAP1. Besides offering a valuable experimental framework to dissect OC differentiation, our study discloses the essential role of UCHL1, exerted through KEAP1-dependent containment of NRF2 anti-OCgenic activity, yielding a novel potential actionable pathway against bone loss.


Asunto(s)
Resorción Ósea , Osteólisis , Animales , Ratones , Resorción Ósea/metabolismo , Diferenciación Celular/genética , Enzimas Desubicuitinizantes/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Osteoclastos/metabolismo , Osteólisis/metabolismo , Proteómica , Ligando RANK/metabolismo
3.
Mol Imaging Biol ; 25(1): 122-132, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-34642899

RESUMEN

PURPOSE: Radical resection is paramount for curative oncological surgery. Fluorescence-guided surgery (FGS) aids in intraoperative identification of tumor-positive resection margins. This study aims to assess the feasibility of urokinase plasminogen activator receptor (uPAR) targeting antibody fragments for FGS in a direct comparison with their parent IgG in various relevant in vivo models. PROCEDURES: Humanized anti-uPAR monoclonal antibody MNPR-101 (uIgG) was proteolytically digested into F(ab')2 and Fab fragments named uFab2 and uFab. Surface plasmon resonance (SPR) and cell assays were used to determine in vitro binding before and after fluorescent labeling with IRDye800CW. Mice bearing subcutaneous HT-29 human colonic cancer cells were imaged serially for up to 120 h after fluorescent tracer administration. Imaging characteristics and ex vivo organ biodistribution were further compared in orthotopic pancreatic ductal adenocarcinoma (BxPc-3-luc2), head-and-neck squamous cell carcinoma (OSC-19-luc2-GFP), and peritoneal carcinomatosis (HT29-luc2) models using the clinical Artemis fluorescence imaging system. RESULTS: Unconjugated and conjugated uIgG, uFab2, and uFab specifically recognized uPAR in the nanomolar range as determined by SPR and cell assays. Subcutaneous tumors were clearly identifiable with tumor-to-background ratios (TBRs) > 2 after 72 h for uIgG-800F and 24 h for uFab2-800F and uFab-800F. For the latter two, mean fluorescence intensities (MFIs) dipped below predetermined threshold after 72 h and 36 h, respectively. Tumors were easily identified in the orthotopic models with uIgG-800F consistently having the highest MFIs and uFab2-800F and uFab-800F having similar values. In biodistribution studies, kidney and liver fluorescence approached tumor fluorescence after uIgG-800F administration and surpassed tumor fluorescence after uFab2-800F or uFab-800F administration, resulting in interference in the abdominal orthotopic mouse models. CONCLUSIONS: In a side-by-side comparison, FGS with uPAR-targeting antibody fragments compared with the parent IgG resulted in earlier tumor visualization at the expense of peak fluorescence intensity.


Asunto(s)
Neoplasias Pancreáticas , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Animales , Humanos , Ratones , Anticuerpos Monoclonales Humanizados , Línea Celular Tumoral , Colorantes Fluorescentes , Fragmentos Fab de Inmunoglobulinas , Inmunoglobulina G , Imagen Óptica/métodos , Neoplasias Pancreáticas/patología , Distribución Tisular
4.
Eur J Cancer ; 146: 11-20, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33561783

RESUMEN

With a 5-year recurrence rate of 30-78%, urothelial cell carcinoma (UCC) rates amongst the highest of all solid malignancies. Consequently, after transurethral resection, patients are subjugated to life-long endoscopic surveillance. A multimodal near-infrared (NIR) fluorescence-based imaging strategy can improve diagnosis, resection and surveillance, hence increasing quality of life. METHODS: Expression of urokinase plasminogen activator receptor (uPAR) and epithelial cell adhesion molecule (EpCAM) are determined on paraffin-embedded human UCC using immunohistochemistry and on UCC cell lines by flow cytometry. MNPR-101, a humanised monoclonal antibody targeting uPAR is conjugated to IRDye800CW and binding is validated in vitro using surface plasmon resonance and cell-based binding assays. In vivo NIR fluorescence and photoacoustic three-dimensional (3D) imaging are performed with subcutaneously growing human UM-UC-3luc2 cells in BALB/c-nude mice. The translational potential is confirmed in a metastasising UM-UC-3luc2 orthotopic mouse model. Infliximab-IRDye800CW and rituximab-IRDye800CW are used as controls. RESULTS: UCCs show prominent uPAR expression at the tumour-stroma interface and EpCAM on epithelial cells. uPAR and EpCAM are expressed by 6/7 and 4/7 UCC cell lines, respectively. In vitro, MNPR-101-IRDye800CW has a picomolar affinity for domain 2-3 of uPAR. In vivo fluorescence imaging with MNPR-101-IRDye800CW, specifically delineates both subcutaneous and orthotopic tumours with tumour-to-background ratios reaching as high as 6.8, differing significantly from controls (p < 0.0001). Photoacoustic 3D in depth imaging confirms the homogenous distribution of MNPR-101-IRDye800CW through the tumour. CONCLUSIONS: MNPR-101-IRDye800CW is suitable for multimodal imaging of UCC, awaiting clinical translation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Imagen Molecular/métodos , Imagen Óptica/métodos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Cirugía Asistida por Computador/métodos , Neoplasias de la Vejiga Urinaria/diagnóstico , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/cirugía , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cell Rep ; 32(12): 108162, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32966780

RESUMEN

FAM46C is a non-canonical poly(A) polymerase uniquely mutated in up to 20% of multiple myeloma (MM) patients, implying a tissue-specific tumor suppressor function. Here, we report that FAM46C selectively stabilizes mRNAs encoding endoplasmic reticulum (ER)-targeted proteins, thereby concertedly enhancing the expression of proteins that control ER protein import, folding, N-glycosylation, and trafficking and boosting protein secretion. This role requires the interaction with the ER membrane resident proteins FNDC3A and FNDC3B. In MM cells, FAM46C expression raises secretory capacity beyond sustainability, inducing ROS accumulation, ATP shortage, and cell death. FAM46C activity is regulated through rapid proteasomal degradation or the inhibitory interaction with the ZZ domain of the autophagic receptor p62 that hinders its association with FNDC3 proteins via sequestration in p62+ aggregates. Altogether, our data disclose a p62/FAM46C/FNDC3 circuit coordinating sustainable secretory activity and survival, providing an explanation for the MM-specific oncosuppressive role of FAM46C and uncovering potential therapeutic opportunities against cancer.


Asunto(s)
Fibronectinas/metabolismo , Homeostasis , Nucleotidiltransferasas/metabolismo , Proteostasis , Proteína Sequestosoma-1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Femenino , Silenciador del Gen/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Inmunoglobulinas/metabolismo , Membranas Intracelulares/metabolismo , Masculino , Ratones Endogámicos C57BL , Mieloma Múltiple/patología , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Inhibidores de Proteasoma/farmacología , Agregado de Proteínas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Proteostasis/efectos de los fármacos , Proteína Sequestosoma-1/química
6.
PLoS One ; 13(8): e0201621, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30067838

RESUMEN

To cope with intrinsic and environmental stress, cancer cells rely on adaptive pathways more than non-transformed counterparts. Such non-oncogene addiction offers new therapeutic targets and strategies to overcome chemoresistance. In an attempt to study the role of adaptive pathways in acquired drug resistance in carcinoma cells, we devised a model of in vitro conditioning to three standard chemotherapeutic agents, cisplatin, 5-fluorouracil, and docetaxel, from the epithelial cancer cell line, HEp-2, and investigated the mechanisms underlying reduced drug sensitivity. We found that triple-resistant cells suffered from higher levels of oxidative stress, and showed heightened anti-stress responses, including the antioxidant Nrf2 pathway and autophagy, a conserved pleiotropic homeostatic strategy, mediating the clearance of aggregates marked by the adapter p62/SQSTM1. As a result, re-administration of chemotherapeutic agents failed to induce further accumulation of reactive oxygen species and p62. Moreover, autophagy proved responsible for chemoresistance through the avoidance of p62 accumulation into toxic protein aggregates. Indeed, p62 ablation was sufficient to confer resistance in parental cells, and genetic and pharmacological autophagic inhibition restored drug sensitivity in resistant cells in a p62-dependent manner. Finally, exogenous expression of mutant p62 lacking the ubiquitin- and LC3-binding domains, required for autophagic engulfment, increased chemosensitivity in TDR HEp-2 cells. Altogether, these findings offer a cellular system to investigate the bases of acquired chemoresistance of epithelial cancers and encourage challenging the prognostic and antineoplastic therapeutic potential of p62 toxicity.


Asunto(s)
Antineoplásicos/farmacología , Regulación hacia Abajo , Resistencia a Antineoplásicos , Neoplasias Glandulares y Epiteliales/metabolismo , Proteína Sequestosoma-1/metabolismo , Autofagia , Línea Celular Tumoral , Cisplatino/farmacología , Docetaxel/farmacología , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mutación , Neoplasias Glandulares y Epiteliales/genética , Estrés Oxidativo , Dominios Proteicos , Proteína Sequestosoma-1/química , Proteína Sequestosoma-1/genética
7.
Blood ; 129(15): 2132-2142, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28130214

RESUMEN

Systemic light chain (AL) amyloidosis is caused by the clonal production of an unstable immunoglobulin light chain (LC), which affects organ function systemically. Although pathogenic LCs have been characterized biochemically, little is known about the biology of amyloidogenic plasma cells (PCs). Intrigued by the unique response rates of AL amyloidosis patients to the first-in-class proteasome inhibitor (PI) bortezomib, we purified and investigated patient-derived AL PCs, in comparison with primary multiple myeloma (MM) PCs, the prototypical PI-responsive cells. Functional, biochemical, and morphological characterization revealed an unprecedented intrinsic sensitivity of AL PCs to PIs, even higher than that of MM PCs, associated with distinctive organellar features and expression patterns indicative of cellular stress. These consisted of expanded endoplasmic reticulum (ER), perinuclear mitochondria, and a higher abundance of stress-related transcripts, and were consistent with reduced autophagic control of organelle homeostasis. To test whether PI sensitivity stems from AL LC production, we engineered PC lines that can be induced to express amyloidogenic and nonamyloidogenic LCs, and found that AL LC expression alters cell growth and proteostasis and confers PI sensitivity. Our study discloses amyloidogenic LC production as an intrinsic PC stressor, and identifies stress-responsive pathways as novel potential therapeutic targets. Moreover, we contribute a cellular disease model to dissect the biology of AL PCs.


Asunto(s)
Amiloidosis/tratamiento farmacológico , Amiloidosis/metabolismo , Bortezomib/farmacocinética , Cadenas Ligeras de Inmunoglobulina/biosíntesis , Células Plasmáticas/metabolismo , Inhibidores de Proteasoma/farmacocinética , Amiloidosis/patología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Femenino , Humanos , Masculino , Mitocondrias/metabolismo , Mitocondrias/patología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Plasmáticas/patología
8.
Int J Mol Sci ; 17(4): 443, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-27023526

RESUMEN

Autophagy favours metastatic growth through fuelling energy and nutrients and resistance to anoikis, typical of disseminated-tumour cells. The autophagic process, mediated by a unique organelle, the autophagosome, which fuses with lysosomes, is divided into three steps. Several stages, especially early omegasome formation and isolation-membrane initiation, remain controversial; molecular mechanisms involve the small-GTPase Rab5a, which regulates vesicle traffic for autophagosome formation. We examined Rab5a involvement in the function of key members of ubiquitin-conjugation systems, Atg7 and LC3-lipidated, interacting with the scaffold-protein p62. Immunohistochemistry of Rab5a was performed in human specimens of bone metastasis and pair-matched breast carcinoma; the autophagic-molecular mechanisms affected by Rab5a were evaluated in human 1833 bone metastatic cells, derived from breast-carcinoma MDA-MB231 cells. To clarify the role of Rab5a, 1833 cells were transfected transiently with Rab5a-dominant negative, and/or stably with the short-hairpin RNA Atg7, were exposed to two inhibitors of autolysosome function, and LC3II and p62 expression was measured. We showed basal autophagy in bone-metastatic cells and the pivotal role of Rab5a together with Beclin 1 between the early stages, elongation of isolation membrane/closed autophagosome mediated by Atg7, and the late-degradative stages. This regulatory network might occur in bone-metastasis and in high-grade dysplastic lesions, preceding invasive-breast carcinoma and conferring phenotypic characteristics for dissemination.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias de la Mama/patología , Proteínas de Unión al GTP rab5/metabolismo , Autofagia , Proteína 7 Relacionada con la Autofagia/antagonistas & inhibidores , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Beclina-1/genética , Beclina-1/metabolismo , Western Blotting , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/metabolismo , Estadificación de Neoplasias , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al GTP rab5/genética
9.
Autophagy ; 11(7): 1161-78, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26043024

RESUMEN

Multiple myeloma (MM) is the paradigmatic proteasome inhibitor (PI) responsive cancer, but many patients fail to respond. An attractive target to enhance sensitivity is (macro)autophagy, recently found essential to bone marrow plasma cells, the normal counterpart of MM. Here, integrating proteomics with hypothesis-driven strategies, we identified the autophagic cargo receptor and adapter protein, SQSTM1/p62 as an essential component of an autophagic reserve that not only synergizes with the proteasome to maintain proteostasis, but also mediates a plastic adaptive response to PIs, and faithfully reports on inherent PI sensitivity. Lentiviral engineering revealed that SQSTM1 is essential for MM cell survival and affords specific PI protection. Under basal conditions, SQSTM1-dependent autophagy alleviates the degradative burden on the proteasome by constitutively disposing of substantial amounts of ubiquitinated proteins. Indeed, its inhibition or stimulation greatly sensitized to, or protected from, PI-induced protein aggregation and cell death. Moreover, under proteasome stress, myeloma cells selectively enhanced SQSTM1 de novo expression and reset its vast endogenous interactome, diverting SQSTM1 from signaling partners to maximize its association with ubiquitinated proteins. Saturation of such autophagic reserve, as indicated by intracellular accumulation of undigested SQSTM1-positive aggregates, specifically discriminated patient-derived myelomas inherently susceptible to PIs from primarily resistant ones. These aggregates correlated with accumulation of the endoplasmic reticulum, which comparative proteomics identified as the main cell compartment targeted by autophagy in MM. Altogether, the data integrate autophagy into our previously established proteasome load-versus-capacity model, and reveal SQSTM1 aggregation as a faithful marker of defective proteostasis, defining a novel prognostic and therapeutic framework for MM.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia/efectos de los fármacos , Homeostasis/efectos de los fármacos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Inhibidores de Proteasoma/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citoprotección/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Agregado de Proteínas/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteína Sequestosoma-1 , Proteínas Ubiquitinadas/metabolismo
10.
PLoS One ; 8(7): e70606, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23923008

RESUMEN

Cell-associated receptor for urokinase plasminogen activator (uPAR) is released as both full-length soluble uPAR (suPAR) and cleaved (c-suPAR) form that maintain ability to bind to integrins and other receptors, thus triggering and modulating cell signaling responses. Concerning HIV-1 infection, plasma levels of suPAR have been correlated with the severity of disease, levels of immune activation and ineffective immune recovery also in individuals receiving combination anti-retroviral therapy (cART). However, it is unknown whether and which suPAR forms might contribute to HIV-1 induced pathogenesis and to the related state of immune activation. In this regard, lymphoid organs represent an import site of chronic immune activation and virus persistence even in individuals receiving cART. Lymphoid organs of HIV-1(+) individuals showed an enhanced number of follicular dendritic cells, macrophages and endothelial cells expressing the cell-associated uPAR in comparison to those of uninfected individuals. In order to investigate the potential role of suPAR forms in HIV-1 infection of secondary lymphoid organs, tonsil histocultures were established from HIV-1 seronegative individuals and infected ex vivo with CCR5- and CXCR4-dependent HIV-1 strains. The levels of suPAR and c-suPAR were significantly increased in HIV-infected tonsil histocultures supernatants in comparison to autologous uninfected histocultures. Supernatants from infected and uninfected cultures before and after immunodepletion of suPAR forms were incubated with the chronically infected promonocytic U1 cell line characterized by a state of proviral latency in unstimulated conditions. In the contest of HIV-conditioned supernatants we established that c-suPAR, but not suPAR, inhibited chemotaxis and induced virus expression in U1 cells. In conclusion, lymphoid organs are an important site of production and release of both suPAR and c-suPAR, this latter form being endowed with the capacity of inhibiting chemotaxis and inducing HIV-1 expression.


Asunto(s)
Quimiotaxis/genética , Regulación Viral de la Expresión Génica , Regulación de la Expresión Génica , Infecciones por VIH/genética , VIH-1/genética , Tejido Linfoide/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Adolescente , Adulto , Anciano , Terapia Antirretroviral Altamente Activa , Estudios de Casos y Controles , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiotaxis/inmunología , Femenino , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/virología , Tejido Linfoide/inmunología , Tejido Linfoide/virología , Masculino , Persona de Mediana Edad , Tonsila Palatina/metabolismo , Tonsila Palatina/virología , Proteolisis , Receptores CCR2/genética , Receptores CCR2/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Adulto Joven
11.
PLoS One ; 7(10): e39723, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23056166

RESUMEN

MYBBP1A is a predominantly nucleolar transcriptional regulator involved in rDNA synthesis and p53 activation via acetylation. However little further information is available as to its function. Here we report that MYBBP1A is developmentally essential in the mouse prior to blastocyst formation. In cell culture, down-regulation of MYBBP1A decreases the growth rate of wild type mouse embryonic stem cells, mouse embryo fibroblasts (MEFs) and of human HeLa cells, where it also promotes apoptosis. HeLa cells either arrest at G2/M or undergo delayed and anomalous mitosis. At mitosis, MYBBP1A is localized to a parachromosomal region and gene-expression profiling shows that its down-regulation affects genes controlling chromosomal segregation and cell cycle. However, MYBBP1A down-regulation increases the growth rate of the immortalized NIH3T3 cells. Such Mybbp1a down-regulated NIH3T3 cells are more susceptible to Ras-induced transformation and cause more potent Ras-driven tumors. We conclude that MYBBP1A is an essential gene with novel roles at the pre-mitotic level and potential tumor suppressor activity.


Asunto(s)
Proteínas Portadoras/genética , Ciclo Celular/genética , Embrión de Mamíferos/metabolismo , Mitosis/genética , Proteínas Nucleares/genética , Animales , Apoptosis/genética , Proteínas Portadoras/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Células Cultivadas , Proteínas de Unión al ADN , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Células 3T3 NIH , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Embarazo , Interferencia de ARN , Proteínas de Unión al ARN , Factores de Transcripción , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
Exp Cell Res ; 312(12): 2203-13, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16631161

RESUMEN

Colon cancer progression is associated with the activation of protein kinase C (PKC), the downregulation of functional E-cadherin and an increased expression of the serine protease urokinase (u-PA) and its receptor (u-PAR). HT29-M6 intestinal epithelial cells represent an in vitro model to study colon cancer progression. These cells are induced to scatter and to invade by phorbol esters. Using proteolytic and cell signaling inhibitors, we show that HT29-M6 cells require plasminogen for the acquisition of the scattering response to PMA. Our results indicate that, prior to inducing a state of competency for plasminogen-dependent scattering, PMA triggers an ordered succession of events where upregulation of the activity of u-PA precedes proteolysis of u-PAR and active degradation of the extracellular matrix (ECM). These events poise HT29-M6 cells to a scatter-competent state that allows the subsequent localized proteolytic activation of plasminogen to plasmin, required for the execution of scattering. Finally, we show that, in addition to its enzymatic activity directed at the degradation of ECM, plasmin generates an intracellular signal resulting in the phosphorylation of ERK 1/2. For a full motogenic activity, plasmin requires this signal since the use of a MEK inhibitor (PD98059) specifically blocks the plasmin-dependent phase of cell scattering. Our observations suggest that plasmin exerts a dual role in PMA-induced scattering of HT29-M6 cells, one directed extracellularly to promote proteolysis of the ECM and one directed to generate intracellular signaling.


Asunto(s)
Movimiento Celular/fisiología , Fibrinolisina/fisiología , Transducción de Señal/fisiología , Acetato de Tetradecanoilforbol/farmacología , Ácido Aminocaproico/farmacología , Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Medio de Cultivo Libre de Suero/farmacología , Inhibidores Enzimáticos/farmacología , Matriz Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrinolisina/metabolismo , Fibrinolisina/farmacología , Flavonoides/farmacología , Células HT29 , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Integrina beta1/metabolismo , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Plasminógeno/deficiencia , Plasminógeno/metabolismo , Plasminógeno/farmacología , Inhibidor 1 de Activador Plasminogénico/farmacología , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Transducción de Señal/efectos de los fármacos , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
13.
J Immunol Methods ; 308(1-2): 192-202, 2006 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-16386755

RESUMEN

The urokinase plasminogen activator receptor (uPAR) fragments D1 and D2D3 are often found in biological fluids from normal individuals and patients of cancer and other diseases. The D2D3 fragment may possess chemotactic activity depending on its N-terminal sequence. We have developed a sensitive and specific immunoassay for the chemotactic form of D2D3 and show that its level can be measured with high specificity and sensitivity in human serum and urine. Synthetic peptides (residues 84-92) derived from the linker region between domains 1 and 2 of uPAR were used as immunogens to generate mouse monoclonal antibodies. Recombinant soluble uPAR (D1D2D3(1-277)) was used to immunize rabbits to obtain polyclonal antibodies. A sandwich-type immunofluorimetric assay was developed with these antibodies. The assay specifically measures D2D3 containing the 84-88 residues, has a detection limit of 0.25 ng/ml and shows no cross-reactivity with D2D3(93-274). The assay is linear at 0-30 ng/ml, with an intra-assay CV of 10% (n=20), inter-assay CV of 15% (n=9) and a recovery of D2D3(84-274) added to urine samples of between 94% and 105%. A statistically significant difference level of D2D3(84-274) was found in two groups of tumor patients versus healthy volunteers (p

Asunto(s)
Fluoroinmunoensayo/métodos , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/biosíntesis , Western Blotting , Células CHO , Células COS , Línea Celular , Factores Quimiotácticos/análisis , Factores Quimiotácticos/genética , Factores Quimiotácticos/inmunología , Chlorocebus aethiops , Cricetinae , Ensayo de Inmunoadsorción Enzimática , Epítopos/análisis , Epítopos/genética , Citometría de Flujo , Fluoroinmunoensayo/estadística & datos numéricos , Humanos , Inmunoprecipitación , Ratones , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Conejos , Receptores de Superficie Celular/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Sensibilidad y Especificidad
14.
J Biol Chem ; 280(26): 24792-803, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15863511

RESUMEN

We investigated the interaction between the urokinase receptor (uPAR) and the integrin alphavbeta3. Vitronectin (VN) induces cell migration by binding to alphavbeta3, but expression of the uPAR boosts its efficacy. Thus, uPAR may regulate VN-induced cell migration by interacting laterally with alphavbeta3. In contrast, cells expressing a uPAR mutant lacking domain 2 do not migrate in response to VN. This effect is overcome by D2A, a synthetic peptide derived from the sequence of domain 2. In addition, D2A has chemotactic activity that requires alphavbeta3 and activates alphavbeta3-dependent signaling pathways such as the Janus kinase/Stat pathway. Moreover, D2A disrupts uPAR-alphavbeta3 and uPAR-alpha5beta1 co-immunoprecipitation, indicating that it can bind both of these integrins. We also identify the chemotactically active epitope harbored by peptide D2A. Mutating two glutamic acids into two alanines generates peptide D2A-Ala, which lacks chemotactic activity but inhibits VN-, FN-, and collagen-dependent cell migration. In fact, the GEEG peptide has potent chemotactic activity, and the GAAG sequence has inhibitory capacities. In summary, we have identified an integrin-interacting sequence located in domain 2 of uPAR, which is also a new chemotactic epitope that can activate alphavbeta3-dependent signaling pathways and stimulate cell migration. This sequence thus plays a pivotal role in the regulation of uPAR-integrin interactions. Moreover, we describe a novel, very potent inhibitor of integrin-dependent cell migration.


Asunto(s)
Integrinas/química , Receptores de Superficie Celular/química , Adenoviridae/genética , Alanina/química , Animales , Aorta/metabolismo , Adhesión Celular , Línea Celular , Movimiento Celular , Quimiotaxis , Citoesqueleto/metabolismo , Relación Dosis-Respuesta a Droga , Epítopos/química , Ácido Glutámico/química , Humanos , Inmunoprecipitación , Integrina alfaVbeta3/química , Ratones , Microscopía Fluorescente , Mutación , Miocitos del Músculo Liso/citología , Células 3T3 NIH , Péptidos/química , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Transducción de Señal , Temperatura , Factores de Tiempo , Transfección , Tirfostinos/farmacología
15.
J Cell Sci ; 117(Pt 14): 2909-16, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15173320

RESUMEN

We have previously shown that chymotrypsin-cleaved soluble uPAR (D2D3(88-274)) elicits migration of monocytic cells through interaction with FPRL-1, a G protein-coupled receptor that is homologous to the fMLP receptor. Here, we report that D2D3(88-274) also modulates the ability of monocytes to migrate in response to other chemokines. Pretreatment of monocytes with increasing amounts of D2D3(88-274) prevents cell migration in response to MCP-1, RANTES and fMLP. We demonstrate that D2D3(88-274) does not inhibit MCP-1 receptor binding, elicit CCR2 internalization and prevent MCP-1-induced intracellular Ca(2+) increase. Thus, CCR2 receptor desensitization cannot account for D2D3(88-274)-mediated inhibition of MCP-1-induced cell migration. Rather, we show that pretreatment of monocytes with D2D3(88-274) dramatically decreases chemokine-induced integrin-dependent rapid cell adhesion by interacting with FPRL-1. Together, our results indicate that chemokine-dependent cell migration can be regulated not only by homologous and heterologous receptor desensitization, but also by inhibition of integrin-dependent cell adhesion, an important step in cell transmigration.


Asunto(s)
Antígenos CD18/fisiología , Adhesión Celular/fisiología , Quimiotaxis de Leucocito/fisiología , Monocitos/fisiología , Receptores de Superficie Celular/fisiología , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Quimiocina CCL5/metabolismo , Quimiocina CCL5/farmacología , Quimiotaxis de Leucocito/efectos de los fármacos , Humanos , Monocitos/metabolismo , Receptores CCR2 , Receptores de Superficie Celular/metabolismo , Receptores de Quimiocina/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Proteínas Recombinantes/farmacología , Solubilidad
16.
Thromb Haemost ; 88(2): 298-306, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12195704

RESUMEN

Proteolytic cleavage of the urokinase plasminogen activator receptor (uPA(R)) prevents the binding of uPA and vitronectin while generating biologically active uPAR fragments. We have recently shown that matrix metalloproteinase-12 (MMP-12) releases cellular uPAR-antigen from stimulated human micro-vascular endothelial cells providing a novel feedback mechanism between the plasminogen activation and MMP systems. We now show that MMP-12 and other MMPs directly and efficiently cleave uPAR at the Thr86 paralal Tyr87 peptide bond located in the linker region connecting uPAR domains 1 and 2, releasing the major ligand binding domain 1 from the rest of the receptor. The possible biological importance of uPAR cleavage by MMPs is supported by the observation that also murine uPAR is cleaved by MMP-12 (at the Pro89 paralal Gln90 peptide bond), despite the limited sequence homology between the linker regions. Using an antibody raised against the human uPAR linker region we show that this region of uPAR, which contains the chemotactic SRSRY epitope, is exposed upon MMP cleavage.


Asunto(s)
Metaloendopeptidasas/metabolismo , Receptores de Superficie Celular/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Sitios de Unión , Epítopos , Humanos , Hidrólisis , Metaloproteinasa 12 de la Matriz , Ratones , Estructura Terciaria de Proteína , Receptores de Superficie Celular/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA