Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Oncol Rep ; 48(2)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35775375

RESUMEN

The clinical introduction of molecular imaging for the management of oropharyngeal squamous cell carcinoma (OPSCC) relies on the identification of relevant cancer­specific biomarkers. The application of three membrane­bound receptors, namely urokinase­type plasminogen activator receptor (uPAR), tissue factor (TF) and EGFR have been previously explored for targeted imaging and therapeutic strategies in a broad range of solid cancers. The present study aimed to investigate the expression patterns of uPAR, EGFR and TF by immunohistochemistry (IHC) to evaluate their potential for targeted imaging and prognostic value in OPSCC. In a retrospective cohort of 93 patients with primary OPSCC, who were balanced into the 45 human papillomavirus (HPV)­positive and 48 HPV­negative groups, the IHC­determined expression profiles of uPAR, TF and EGFR in large biopsy or tumor resection specimens were analyzed. Using the follow­up data, overall survival (OS) and recurrence­free survival were measured. Specifically, associations between survival outcome, biomarker expression and clinicopathological factors were examined using Cox proportional hazards model and log­rank test following Kaplan­Meier statistics. After comparing the expression pattern of biomarkers within the tumor compartment with that in the adjacent normal tissues, uPAR and TF exhibited a highly tumor­specific expression pattern, whereas EGFR showed a homogeneous expression within the tumor compartment as well as a consistent expression in the normal mucosal epithelium and salivary gland tissues. The positive expression rate of uPAR, TF and EGFR in the tumors was 98.9, 76.3 and 98.9%, respectively. No statistically significant association between biomarker expression and survival outcome could be detected. Higher uPAR expression levels had a trend towards reduced OS according to results from univariate analysis (P=0.07; hazard ratio=2.01; 95% CI=0.92­4.37). Taken together, these results suggest that uPAR, TF and EGFR may be suitable targets for molecular imaging and therapy in OPSCC. In particular, uPAR may be an attractive target owing to their high positive expression rates in tumors and a highly tumor­specific expression pattern.


Asunto(s)
Neoplasias Orofaríngeas , Infecciones por Papillomavirus , Carcinoma de Células Escamosas de Cabeza y Cuello , Biomarcadores de Tumor/biosíntesis , Receptores ErbB/biosíntesis , Humanos , Imagen Molecular , Terapia Molecular Dirigida , Neoplasias Orofaríngeas/diagnóstico por imagen , Neoplasias Orofaríngeas/tratamiento farmacológico , Neoplasias Orofaríngeas/patología , Neoplasias Orofaríngeas/virología , Papillomaviridae , Infecciones por Papillomavirus/diagnóstico por imagen , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología , Pronóstico , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Estudios Retrospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello/diagnóstico por imagen , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Tromboplastina/biosíntesis
2.
Curr Drug Targets ; 21(7): 647-656, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31755385

RESUMEN

The major proteases that constitute the fibrinolysis system are tightly regulated. Protease inhibitors target plasmin, the protease responsible for fibrin degradation, and the proteases that convert plasminogen into plasmin, including tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). A second mechanism by which fibrinolysis is regulated involves exosite interactions, which localize plasminogen and its activators to fibrin, extracellular matrix (ECM) proteins, and cell surfaces. Once plasmin is generated in association with cell surfaces, it may cleave transmembrane proteins, activate growth factors, release growth factors from ECM proteins, remodel ECM, activate metalloproteases, and trigger cell-signaling by cleaving receptors in the Proteaseactivated Receptor (PAR) family. These processes are all implicated in cancer. It is thus not surprising that a family of structurally diverse but functionally similar cell-surface proteins, called Plasminogen Receptors (PlgRs), which increase the catalytic efficiency of plasminogen activation, have received attention for their possible function in cancer and as targets for anticancer drug development. In this review, we consider four previously described PlgRs, including: α-enolase, annexin-A2, Plg-RKT, and cytokeratin-8, in human cancer. To compare the PlgRs, we mined transcriptome profiling data from The Cancer Genome Atlas (TCGA) and searched for correlations between PlgR expression and patient survival. In glioma, the expression of specific PlgRs correlates with tumor grade. In a number of malignancies, including glioblastoma and liver cancer, increased expression of α-enolase or annexin-A2 is associated with an unfavorable prognosis. Whether these correlations reflect the function of PlgRs as receptors for plasminogen or other activities is discussed.


Asunto(s)
Neoplasias/genética , Neoplasias/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Animales , Fibrinólisis/fisiología , Humanos , Terapia Molecular Dirigida , Clasificación del Tumor , Neoplasias/terapia , Pronóstico , Receptores Proteinasa-Activados/biosíntesis , Receptores Proteinasa-Activados/genética
3.
World J Urol ; 38(10): 2501-2511, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31797075

RESUMEN

PURPOSE: To investigate the prognostic role of expression of urokinase-type plasminogen activator system members, such as urokinase-type activator (uPA), uPA-receptor (uPAR), and plasminogen activator inhibitor-1 (PAI-1), in patients treated with radical prostatectomy (RP) for prostate cancer (PCa). METHODS: Immunohistochemical staining for uPA system was performed on a tissue microarray of specimens from 3121 patients who underwent RP. Cox regression analyses were performed to investigate the association of overexpression of these markers alone or in combination with biochemical recurrence (BCR). Decision curve analysis was used to assess the clinical impact of these markers. RESULTS: uPA, uPAR, and PAI-1 were overexpressed in 1012 (32.4%), 1271 (40.7%), and 1311 (42%) patients, respectively. uPA overexpression was associated with all clinicopathologic characteristics of biologically aggressive PCa. On multivariable analysis, uPA, uPAR, and PAI-1 overexpression were all three associated with BCR (HR: 1.75, p < 0.01, HR: 1.22, p = 0.01 and HR: 1.20, p = 0.03, respectively). Moreover, the probability of BCR increased incrementally with increasing cumulative number of overexpressed markers. Decision curve analysis showed that addition of uPA, uPAR, and PAI-1 resulted in a net benefit compared to a base model comparing standard clinicopathologic features across the entire threshold probability range. In subgroup analyses, overexpression of all three markers remained associated with BCR in patients with favorable pathologic characteristics. CONCLUSION: Overexpression of uPA, uPAR, and PAI-1 in PCa tissue were each associated with worse BCR. Additionally, overexpression of all three markers is informative even in patients with favorable pathologic characteristics potentially helping clinical decision-making regarding adjuvant therapy and/or intensified follow-up.


Asunto(s)
Biomarcadores de Tumor/fisiología , Recurrencia Local de Neoplasia/etiología , Inhibidor 1 de Activador Plasminogénico/fisiología , Prostatectomía , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/cirugía , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Anciano , Biomarcadores de Tumor/biosíntesis , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/epidemiología , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Pronóstico , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Estudios Retrospectivos , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
4.
Viruses ; 11(11)2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31739586

RESUMEN

The Ly6 (lymphocyte antigen-6)/uPAR (urokinase-type plasminogen activator receptor) superfamily protein is a group of molecules that share limited sequence homology but conserved three-fingered structures. Despite diverse cellular functions, such as in regulating host immunity, cell adhesion, and migration, the physiological roles of these factors in vivo remain poorly characterized. Notably, increasing research has focused on the interplays between Ly6/uPAR proteins and viral pathogens, the results of which have provided new insight into viral entry and virus-host interactions. While LY6E (lymphocyte antigen 6 family member E), one key member of the Ly6E/uPAR-family proteins, has been extensively studied, other members have not been well characterized. Here, we summarize current knowledge of Ly6/uPAR proteins related to viral infection, with a focus on uPAR and CD59. Our goal is to provide an up-to-date view of the Ly6/uPAR-family proteins and associated virus-host interaction and viral pathogenesis.


Asunto(s)
Antígenos CD59/metabolismo , Interacciones Huésped-Patógeno , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Virosis/metabolismo , Virosis/virología , Vías Biosintéticas , Antígenos CD59/biosíntesis , Antígenos CD59/química , Antígenos CD59/genética , Citocinas/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Familia de Multigenes , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/química , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Relación Estructura-Actividad
5.
Neuroscience ; 415: 184-200, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31362033

RESUMEN

Sushi repeat-containing protein X-linked 2 (SRPX2) is a novel hypothalamic protein and a ligand of the urokinase-type plasminogen activator receptor (uPAR), which is essential for proteolysis of extracellular matrix and tissue remodeling after an insult to the brain. However, little is known about regulation of SRPX2. Our objective was to investigate if SRPX2 expression is altered by (i) the deficiency of uPAR or uPA (urokinase-type plasminogen activator), and (ii) traumatic brain injury (TBI). SRPX2 expression was assessed in wild type (Wt), Plaur-/- (uPAR-deficient), and Plau-/- (uPA-deficient) mice, with and without controlled cortical impact injury (CCI). The number of SRPX2+ neurons in hypothalamus was comparable to that in Wt littermates in Plaur-/- (2985 ±â€¯138 vs. 2890 ±â€¯92, p > 0.05) and Plau-/- mice (2180 ±â€¯232 vs. 2027 ±â€¯77, p > 0.05). The number of hypothalamic SRPX2+ neurons in the Wt-CCI group was comparable to that in controls (3645 ±â€¯288 vs. 3385 ±â€¯192, p > 0.05). Hypothalamic, hippocampal and thalamic Srpx2 gene expression was unaltered after TBI. However, at 4 days post-TBI Srpx2 gene expression was upregulated in the perilesional cortex of Plau-/--CCI mice up to 123% of that in the sham group (p < 0.05). Unsupervised hierarchical clustering using SRPX2 expression did not identify genotype or injury-specific clusters. Our data demonstrate that SRPX2 expression in the hypothalamus is resistant to genetic deficiencies in the urokinase-system or to the hypothalamus-affecting TBI. The contribution of elevated Srpx2 gene expression in perilesional cortex to post-TBI recovery process, however, requires further exploration.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Animales , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/patología , Expresión Génica , Genotipo , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/deficiencia , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Transducción de Señal
6.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2342-2354, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28629718

RESUMEN

Primary forms of focal and segmental glomerulosclerosis (FSGS) are driven by circulating factors that cause dysfunction or loss podocytes. Rare genetic forms of FSGS can be caused by mutations in TRPC6, which encodes a Ca2+-permeable cationic channel expressed in mesangial cells and podocytes; and NPHS2, which encodes podocin, a TRPC6-binding protein expressed in podocyte slit diaphragm domains. Here we observed that exposing immortalized mouse podocytes to serum or plasma from recurrent FSGS patients for 24h increased the steady-state cell-surface abundance of TRPC6, accompanied by an increase in currents through endogenous TRPC6 channels evoked by a hypoosmotic stretch stimulus. These effects were mimicked by the soluble urokinase receptor (suPAR) and by tumor necrosis factor (TNF), circulating factors implicated in nephrotic syndromes. Most but not all of the recurrent FSGS plasma samples that we examined also caused a loss of podocin over a period of several hours. The loss of podocin was also seen following exposure to suPAR but not TNF. However, TNF increased the effects of suPAR on TRPC6 and podocin, and TNF and suPAR are required for the full effects of one of the recurrent FSGS plasma samples. The actions of FSGS plasma, suPAR and TNF on surface abundance of TRPC6 were blocked by cilengitide, an inhibitor of αvß3-integrin signaling. These data suggest that primary FSGS is a heterogeneous condition mediated by multiple circulating factors, and support TRPC6 and αvß3-integrin as potential therapeutic targets.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/metabolismo , Síndrome Nefrótico/metabolismo , Plasma , Podocitos/metabolismo , Suero , Canal Catiónico TRPC6/metabolismo , Adulto , Línea Celular Transformada , Femenino , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Integrina alfaVbeta3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Masculino , Proteínas de la Membrana/biosíntesis , Síndrome Nefrótico/patología , Permeabilidad , Podocitos/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo
7.
Int J Cardiol ; 227: 858-862, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27818020

RESUMEN

PURPOSE: Lipid lowering therapy constitutes the basis of cardiovascular disease therapy. The purpose of this study was to investigate effects of ezetimibe, a selective inhibitor of intestinal cholesterol absorption, on platelets and endothelial cells in an in vitro endothelial cell model. METHODS: After a 24h incubation period with ezetimibe (concentrations 1, 50, 100 and 1000ng/ml), human umbilical vein endothelial cells (HUVEC) were stimulated for 1h with lipopolysaccharide (LPS) and were then incubated in direct contact with activated platelets. Following this, the expression of CD40L and CD62P on platelets, and the expression of ICAM-1, VCAM-1, uPAR, and MT1-MMP on endothelial cells were measured by flow cytometry. Supernatants were analysed by enzyme linked immunosorbent assay for soluble MCP-1, IL-6 and MMP-1. RESULTS: The increased expression of uPAR on endothelial cells by proinflammatory stimulation with LPS and by direct endothelial contact with activated platelets was significantly reduced through pre-incubation with 100ng/ml and 1000ng/ml ezetimibe (p<0.05). Platelets directly incubated with ezetimibe but without endothelial cell contact showed significantly reduced CD62P and CD40L surface expression (p<0.05). Ezetimibe had no significant effects on HUVEC expression of MT1-MMP, ICAM-1 and VCAM-1 and on CD40L expression on platelets in direct contact with endothelial cells. Levels of soluble IL-6 in HUVEC supernatants were significantly lower after pre-incubation with ezetimibe. CONCLUSION: In this in vitro analysis, ezetimibe directly attenuates platelet activation and has significant endothelial cell mediated effects on selected markers of atherosclerosis.


Asunto(s)
Ezetimiba/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Anticolesterolemiantes/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Activación Plaquetaria/fisiología
8.
Int J Oncol ; 49(1): 336-42, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27176787

RESUMEN

A proton beam is a next generation tool to treat intractable cancer. Although the therapeutic effects of a proton beam are well known, the effect on tumor metastasis is not fully described. Here, we investigated the effects of a proton beam on metastasis in highly invasive 4T1 murine breast cancer cells and their orthotopic breast cancer model. Cells were irradiated with 2, 4, 8 or 16 Gy proton beam, and changes in cell proliferation, survival, and migration were observed by MTT, colony forming and wound healing assays. 4T1 breast cancer cell-implanted BALB/c mice were established and the animals were randomly divided into 4 groups when tumor size reached 200 mm3. Breast tumors were selectively irradiated with 10, 20 or 30 Gy proton beam. Breast tumor sizes were measured twice a week, and breast tumor and lung tissues were pathologically observed. Metastasis-regulating gene expression was assessed with quantitative RT-PCR. A proton beam dose-dependently decreased cell proliferation, survival and migration in 4T1 murine breast cancer cells. Also, growth of breast tumors in the 4T1 orthotopic breast cancer model was significantly suppressed by proton beam irradiation without significant change of body weight. Furthermore, fewer tumor nodules metastasized from breast tumor into lung in mice irradiated with 30 Gy proton beam, but not with 10 and 20 Gy, than in control. We observed correspondingly lower expression levels of urokinase plasminogen activator (uPA), uPA receptor, cyclooxygenase (COX)-2, and vascular endothelial growth factor (VEGF), which are important factors in cancer metastasis, in breast tumor irradiated with 30 Gy proton beam. Proton beam irradiation did not affect expressions of matrix metalloproteinase (MMP)-9 and MMP-2. Taken together, the data suggest that, although proton beam therapy is an effective tool for breast cancer treatment, a suitable dose is necessary to prevent metastasis-linked relapse and poor prognosis.


Asunto(s)
Neoplasias de la Mama/radioterapia , Movimiento Celular/efectos de la radiación , Neoplasias Pulmonares/radioterapia , Neoplasias Mamarias Experimentales/radioterapia , Animales , Peso Corporal/efectos de la radiación , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular/efectos de la radiación , Ciclooxigenasa 2/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/radioterapia , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis
9.
Invest Ophthalmol Vis Sci ; 57(6): 2600­2611, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27168367

RESUMEN

PURPOSE: A mouse model of age-related macular degeneration (AMD) was used to investigate the anti-angiogenic and anti-inflammatory role of UPARANT in laser-induced choroidal neovascularization (CNV). METHODS: Choroidal neovascularization was induced by laser photocoagulation, and UPARANT was intravitreally injected. Some experiments were also performed after either intravitreal injection of anti-VEGF drugs or systemic administration of UPARANT. Immunohistochemistry using CD31 antibodies was used to evaluate the area of CNV. Evans blue dye extravasation was quantitatively assessed. Transcripts of markers of outer blood retinal barrier were measured by quantitative RT-PCR, also used to evaluate angiogenesis and inflammation markers. Western blot was used to determine levels of transcription factors encoding genes involved in angiogenesis and inflammation. Levels of urokinase-type plasminogen activator (uPA), its receptor (uPAR), and formyl peptide receptors (FPRs) were determined at the transcript and the protein level. RESULTS: Intravitreal UPARANT reduced the CNV area and the leakage from the choroid. The uPA/uPAR/FPR system was upregulated in CNV, but was not influenced by UPARANT. UPARANT recovered laser-induced upregulation of transcription factors encoding angiogenic and inflammatory markers. Accordingly, angiogenic and inflammatory factors were also reduced. UPARANT as compared to anti-VEGF drugs displayed similar effects on CNV area. CONCLUSIONS: UPARANT mitigates laser-induced CNV by inhibiting angiogenesis and inflammation through an action on transcription factors encoding angiogenesis and inflammatory genes. The finding that UPARANT is effective against CNV may help to establish uPAR and its membrane partners as putative targets in the treatment of AMD.


Asunto(s)
Coroides/patología , Neovascularización Coroidal/tratamiento farmacológico , Oligopéptidos/administración & dosificación , Animales , Western Blotting , Neovascularización Coroidal/etiología , Neovascularización Coroidal/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Inmunohistoquímica , Inyecciones Intravítreas , Coagulación con Láser/efectos adversos , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/genética
10.
Oncol Rep ; 35(2): 992-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26718775

RESUMEN

Hypoxia occurs during development of cervical cancer and is considered to correlate with its invasion. Hypoxia mediates tumor cells to have more invasive property in a variety of cancers. Urokinase plasminogen activator receptor (uPAR) which mediates invasion is considered to be induced by hypoxia. We sought to determine the regulators of uPAR expression during hypoxia in cervical cancer. We showed that cervical cancer cell lines, CaSki and CA, were more invasive under hypoxic condition (1% O2) than under normoxic condition (20% O2) by invasion assays. Using western blot analysis, hypoxia enhanced the endogenous hypoxia-inducible factor (HIF)-1α and uPAR protein expression. uPAR mRNA level was also upregulated by hypoxia using real-time RT-PCR. Overexpression of HIF-1α which is induced by hypoxia activated the transcriptional activity of the uPAR promoter by luciferase assays. HIF-1 protein bound the putative HIF-1 response element on the uPAR promoter using electrophoretic mobility shift analysis, and additional luciferase assays show that this is essential for uPAR transactivation by HIF-1. HIF-1 overexpression enhanced the endogenous uPAR expression and introduction of siRNA for HIF-1α diminishes uPAR expression during hypoxia. These results indicate the upregulation of uPAR by hypoxia in cervical cancer cells is mediated through HIF-1. In cervical cancer tissues, we also demonstrated that uPAR protein expression was detected in cervical cancer but not in normal cervix or cervical intraepithelial neoplasia (CIN) by immunohistopathological staining. Our results provide evidence that regulation of uPAR expression by HIF-1 represents a mechanism for cervical cancer invasion during hypoxia.


Asunto(s)
Hipoxia de la Célula , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Neoplasias del Cuello Uterino/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Invasividad Neoplásica , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Proteínas Recombinantes de Fusión/genética , Elementos de Respuesta , Activación Transcripcional , Transfección , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Displasia del Cuello del Útero/genética , Displasia del Cuello del Útero/metabolismo
11.
J Comp Pathol ; 153(4): 278-82, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26286429

RESUMEN

Immunohistochemistry was used to assess the expression of urokinase plasminogen activator (uPA) and uPA receptor (uPAR) in 57 canine primary haemangiosarcomas (HSAs), 26 canine cutaneous haemangiomas (HAs) and in control sections of canine cutaneous granulation tissue. The correlation between uPA/uPAR expression and the Ki67 labelling index (LI) was estimated in the HSA and HA tissues. uPA was expressed by 73.2% and 75.0% of splenic HSAs and non-splenic HSAs, respectively. All HSA tissues tested expressed uPAR. Expression of both molecules was significantly higher in HSAs than in cutaneous HAs (3.8% for uPA and 30.7% for uPAR). The average Ki67 LI of the uPA(+)/uPAR(+) HSAs was significantly higher than that of uPA(-)/uPAR(+) HSAs and HA tissues (mean ± SDs 32.8 ± 15.3, 15.2 ± 7.2 and 2.1 ± 0.7, respectively; P <0.05). These results suggest that uPA and uPAR play a significant role in the malignant proliferation of canine HSA, regardless of the primary origin of the tumour.


Asunto(s)
Enfermedades de los Perros/metabolismo , Hemangioma/veterinaria , Hemangiosarcoma/veterinaria , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Animales , Enfermedades de los Perros/patología , Perros , Hemangioma/metabolismo , Hemangioma/patología , Hemangiosarcoma/metabolismo , Hemangiosarcoma/patología , Inmunohistoquímica
12.
Tumour Biol ; 36(11): 8715-25, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26050228

RESUMEN

Stromal macrophages of different phenotypes can contribute to the expression of proteins that affects metastasis such as urokinase-type plasminogen activator (uPA), its receptor uPAR, and plasminogen activator inhibitor-1 (PAI-1), but knowledge of how essential their contribution is in comparison to the cancer cells in small cell lung cancer (SCLC) and lung squamous cell carcinoma (SCC) is lacking. The expression of uPA, uPAR, and PAI-1 and of the matrix metalloproteinases (MMP)-2 and MMP-9 were studied in human macrophages of M1 and M2 phenotype and compared to a lung SCC (NCI-H520) and a SCLC (NCI-H69) cell line. Effects of treatment with conditioned media (CM) from M1 and M2 macrophages on the expression of these genes in H520 and H69 cells as well as effects on the cell growth were investigated. In addition, data on the stromal macrophages immunoreactivity of uPAR, MMP-2, and MMP-9 in a few SCC and SCLC biopsies was included. uPAR, MMP-2, and MMP-9 were confirmed in stromal cells including macrophages in the SCC and SCLC biopsies. In vitro, both macrophage phenotypes expressed considerably higher mRNA levels of uPA, uPAR, PAI-1, and MMP-9 compared to the cancer cell lines, and regarding uPAR, the highest level was found in the M1 macrophage phenotype. Furthermore, M1 CM treatment not only induced an upregulation of PAI-1 in both H520 and H69 cells but also inhibited cell growth in both cell lines, giving M1 macrophages both tumor-promoting and tumor-killing potential.


Asunto(s)
Carcinoma de Células Escamosas/genética , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Carcinoma Pulmonar de Células Pequeñas/genética , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Inhibidor 1 de Activador Plasminogénico/genética , ARN Mensajero/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Activador de Plasminógeno de Tipo Uroquinasa/genética
13.
Exp Mol Pathol ; 98(2): 136-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25661888

RESUMEN

The urokinase plasminogen activator receptor (uPAR) is closely associated with poor prognosis in various aggressive cancers including large-cell lung cancer (LCLC). Vasculogenic mimicry (VM) refers to the unique capability of aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks involving the blood supply in early tumor formation. We demonstrate the statistically positive correlation of uPAR expression with VM formation, metastasis, and poor prognosis of LCLC patients. uPAR(+) cells sorted from the LCLC H460 cell line show higher invasion, migration capacity, and tube structure formation capability on Matrigel compared with uPAR(-) cells. uPAR(+) tumor cells highly expressed vimentin and VE-cadherin; the epithelial marker E-cadherin was low expressed. Higher EMT-regulated protein twist and snail expressions were also observed in these cells. uPAR(+) cells injected subcutaneously into nude mice markedly increased tumor growth, induced VM formation and liver metastasis; by contrast, uPAR(-) cells did not. The data suggest that uPAR expression may predict VM formation, tumor metastasis and poorer prognosis of LCLC patients. The uPAR gene may be used as a novel therapeutic target for inhibiting angiogenesis and metastasis in LCLC.


Asunto(s)
Carcinoma de Células Grandes/patología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/patología , Neovascularización Patológica/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/mortalidad , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica/patología , Trasplante de Neoplasias , Proteínas Nucleares/biosíntesis , Pronóstico , Factores de Transcripción de la Familia Snail , Factores de Transcripción/biosíntesis , Trasplante Heterólogo , Proteína 1 Relacionada con Twist/biosíntesis , Vimentina/biosíntesis
14.
Urol Oncol ; 33(4): 165.e15-24, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25575713

RESUMEN

OBJECTIVES: To evaluate the expression-and localization pattern of the urokinase-type plasminogen activator receptor (uPAR), focusing on its clinical implications in patients with urothelial neoplasia of the bladder treated with radical cystectomy. uPAR is a central molecule in tissue remodeling during cancer invasion and metastasis and is an established prognostic marker in cancer. The expression and localization of uPAR and its prognostic significance is only limitedly investigated in urothelial bladder neoplasia. MATERIALS AND METHODS: The expression-and localization pattern of uPAR was investigated in formalin-fixed paraffin-embedded tumor tissue from 149 patients treated with radical cystectomy between 1988 and 2005. uPAR expression was determined by immunohistochemistry and scored as either negative or positive. Separate values were obtained for cancer cells, macrophages, and myofibroblasts at the invasive front and tumor core, respectively. Statistical analyses were performed to evaluate the association of uPAR localization and score with clinicopathologic covariates and survival. RESULTS: uPAR positivity was seen in 122/137 (89%) and 118/149 (74%) of the neoplasias at the invasive front and tumor core, respectively. uPAR was primarily expressed by myofibroblasts and macrophages in the surrounding stroma as well as some cancer cells. A significant association between uPAR positivity and T-stage as well as grade was found for all 3 cell types in tumor core (P ≤ 0.04 for all comparisons). In univariate analysis, the uPAR positive group had a shorter survival than the uPAR negative group (hazard ratio = 2.39; 95% CI: 1.15-5.01; P = 0.020). CONCLUSIONS: The expression of uPAR is a possible prognostic marker that could be useful in identification of patients with aggressive, highly invasive tumors that could benefit from additional chemotherapy or more intensive follow-up after cystectomy.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Transicionales/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Neoplasias de la Vejiga Urinaria/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Transicionales/metabolismo , Carcinoma de Células Transicionales/mortalidad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , Receptores del Activador de Plasminógeno Tipo Uroquinasa/análisis , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/mortalidad
15.
Tumour Biol ; 36(5): 3565-72, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25544710

RESUMEN

ETS gene fusions involving ERG, ETV1, ETV4, ETV5, and FLI1 define a distinct class of prostate cancer (PCa), and this might have a bearing on diagnosis, prognosis, and rational therapeutic targeting. In the current study, we focused on the clinicopathological significance of ETV4 in Chinese PCa patients and the mechanisms whereby ETV4 overexpression mediates tumor invasion in the prostate. Overall, ETV4 overexpression was identified in 30.4 % (45/148) of PCa cases by immunohistochemistry. Accordingly, ETV4 was rearranged in only 1.6 % (2/128) of PCa patients. Clinically, ETV4 overexpression was significantly correlated with Gleason score (P = 0.045) and pathological tumor stage (P = 0.041). Multivariate Cox regression analysis indicated that ETV4 is an unfavorable independent prognostic factor (P = 0.040). Functional studies further showed that small interfering RNA (siRNA) knockdown of ETV4 significantly decreases proliferation and invasion of PC-3 cell and partially reverses epithelial-mesenchymal transition in vitro. Notably, ETV4 knockdown significantly downregulated expression of urokinase plasminogen activator (uPA) and its receptor (uPAR) at messenger RNA (mRNA) and protein levels. Chromatin immunoprecipitation assay demonstrated that ETV4 regulates uPA expression through direct binding to its promoter region. Additionally, ETV4 knockdown was also observed to significantly inhibit expression of matrix metalloproteinase (MMP)-2 and MMP-9. In conclusion, for the first time, our study suggested that ETV4 is an independent poor prognostic factor in Chinese PCa patients. Silencing of ETV4 suppresses invasion of PCa cells by inhibiting the expression of uPA/uPAR as well as MMPs. Further studies will be needed to determine whether ETV4 could be regarded as a potential target for the management and prevention of PCa.


Asunto(s)
Proteínas E1A de Adenovirus/biosíntesis , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Neoplasias de la Próstata/genética , Proteínas Proto-Oncogénicas/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Proteínas E1A de Adenovirus/genética , Anciano , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/genética , Pronóstico , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-ets , ARN Interferente Pequeño
16.
Breast Cancer Res Treat ; 149(1): 99-108, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25519042

RESUMEN

The urokinase receptor (uPAR) plays a critical role in breast cancer (BC) progression and metastases and is a validated target for novel therapies. The current study investigates the effects of MV-uPA, an oncolytic measles virus fully retargeted against uPAR in syngeneic and xenograft BC metastases models. In vitro replication and cytotoxicity of MVs retargeted against human (MV-h-uPA) or mouse (MV-m-uPA) uPAR were assessed in human and murine cancer and non-cancer mammary epithelial cells. The in vivo effects of species-specific uPAR retargeted MVs were assessed in syngeneic and xenograft models of experimental metastases, established by intravenous administration of luciferase expressing 4T1 or MDA-MD-231 cells. Metastases progression was assessed by in vivo bioluminescence imaging. Tumor targeting was evaluated by qRT-PCR of MV-N, rescue of viable viral particles, and immunostaining of MV particles in lungs from tumor bearing mice. In vitro, MV-h-uPA and MV-m-uPA selectively infected, replicated, and induced cytotoxicity in cancer compared to non-cancer cells in a species-specific manner. In vivo, MV-m-uPA delayed 4T1 lung metastases progression and prolonged survival. These effects were associated with identification of viable viral particles, viral RNA, and detection of MV-N by immunostaining from lung tissues in treated mice. In the human MDA-MB-231 metastases model, intravenous administration of MV-h-uPA markedly inhibited metastases progression and significantly improved survival, compared to controls. No significant treatment-related toxicity was observed in treated mice. The above preclinical findings strongly suggest that uPAR retargeted measles virotherapy is a novel and feasible systemic therapy strategy against metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Mamarias Experimentales/genética , Virus del Sarampión/genética , Viroterapia Oncolítica , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Administración Intravenosa , Animales , Neoplasias de la Mama/terapia , Neoplasias de la Mama/virología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Mamarias Experimentales/terapia , Neoplasias Mamarias Experimentales/virología , Ratones , Metástasis de la Neoplasia , Virus Oncolíticos/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Minerva Anestesiol ; 81(2): 157-65, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24994498

RESUMEN

BACKGROUND: Osteopontin (OPN) and soluble urokinase plasminogen activator receptor (suPAR) have been proposed as markers of disease severity and risk-stratification in infection and inflammation. In breast cancer, OPN and the membrane bound form of urokinase plasminogen activator receptor (uPAR) are functionally related, as OPN-induced cell migration depends on uPAR triggering by urokinase plasminogen activator (uPA). The aim of this study was to prospectively evaluate the kinetic of OPN and suPAR blood levels in patients developing septic shock (SS) compared to those not developing SS, and to investigate the relationships between these two biomarkers in immune cells in vitro. METHODS: We measured the levels of OPN and suPAR for 15 days in forty-three patients, defined a priory as at risk to develop septic shock. Moreover, we investigated in vitro the effect of recombinant OPN on uPAR and suPAR expression in monocytes. RESULTS: We found that OPN and suPAR levels were directly correlated to each other both at intensive care unit admission and on the day patients met SIRS/sepsis or septic shock criteria. In patients developing septic shock, OPN increased prior to suPAR and was already detectable up to 4 days before the shock development. In vitro, OPN induced suPAR production in monocytes by increasing both uPAR gene expression, and suPAR release from the cell surface. CONCLUSION: These data suggest that OPN is partly responsible for the increased plasma levels of suPAR and might be a valuable tool to predict the occurrence of septic shock.


Asunto(s)
Osteopontina/farmacología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Adulto , Anciano , Biomarcadores , Calcitonina/biosíntesis , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Cinética , Masculino , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Estudios Prospectivos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Proteínas Recombinantes/farmacología , Choque Séptico/sangre
18.
PLoS One ; 9(12): e114446, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25517907

RESUMEN

AIMS: Cresols are present in antiseptics, coal tar, some resins, pesticides, and industrial solvents. Cresol intoxication leads to hepatic injury due to coagulopathy as well as disturbance of hepatic circulation in fatal cases. Patients with uremia suffer from cardiovascular complications, such as atherosclerosis, thrombosis, hemolysis, and bleeding, which may be partly due to p-cresol toxicity and its effects on vascular endothelial and mononuclear cells. Given the role of reactive oxygen species (ROS) and inflammation in vascular thrombosis, the objective of this study was to evaluate the effect of p-cresol on endothelial and mononuclear cells. METHODS: EA.hy926 (EAHY) endothelial cells and U937 cells were exposed to different concentrations of p-cresol. Cytotoxicity was evaluated by 3-(4,5-Dimethylthiazol-2-yl)-2,5 -diphenyltetrazolium bromide (MTT) assay and trypan blue dye exclusion technique, respectively. Cell cycle distribution was analyzed by propidium iodide flow cytometry. Endothelial cell migration was studied by wound closure assay. ROS level was measured by 2',7'-dichlorofluorescein diacetate (DCF) fluorescence flow cytometry. Prostaglandin F2α (PGF2α), plasminogen activator inhibitor-1 (PAI-1), soluble urokinase plasminogen activator receptor (suPAR), and uPA production were determined by Enzyme-linked immunosorbant assay (ELISA). RESULTS: Exposure to 100-500 µM p-cresol decreased EAHY cell number by 30-61%. P-cresol also decreased the viability of U937 mononuclear cells. The inhibition of EAHY and U937 cell growth by p-cresol was related to induction of S-phase cell cycle arrest. Closure of endothelial wounds was inhibited by p-cresol (>100 µM). P-cresol (>50 µM) also stimulated ROS production in U937 cells and EAHY cells but to a lesser extent. Moreover, p-cresol markedly stimulated PAI-1 and suPAR, but not PGF2α, and uPA production in EAHY cells. CONCLUSIONS: p-Cresol may contribute to atherosclerosis and thrombosis in patients with uremia and cresol intoxication possibly due to induction of ROS, endothelial/mononuclear cell damage and production of inflammation/atherosclerosis-related molecules.


Asunto(s)
Aterosclerosis/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Cresoles/toxicidad , Células Endoteliales/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dinoprost/biosíntesis , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Inflamación/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/química , Solubilidad , Células U937 , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis
19.
J Cell Mol Med ; 18(11): 2235-51, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25215932

RESUMEN

Pancreatic ductal adenocarcinoma is one of the lethal cancers with extensive local tumour invasion, metastasis, early systemic dissemination and poorest prognosis. Thus, understanding the mechanisms regulating invasion/metastasis and epithelial-mesenchymal transition (EMT), is the key for developing effective therapeutic strategies for pancreatic cancer (PaCa). Eukaryotic elongation factor-2 kinase (eEF-2K) is an atypical kinase that we found to be highly up-regulated in PaCa cells. However, its role in PaCa invasion/progression remains unknown. Here, we investigated the role of eEF-2K in cellular invasion, and we found that down-regulation of eEF-2K, by siRNA or rottlerin, displays impairment of PaCa cells invasion/migration, with significant decreases in the expression of tissue transglutaminase (TG2), the multifunctional enzyme implicated in regulation of cell attachment, motility and survival. These events were associated with reductions in ß1 integrin/uPAR/MMP-2 expressions as well as decrease in Src activity. Furthermore, inhibition of eEF-2K/TG2 axis suppresses the EMT, as demonstrated by the modulation of the zinc finger transcription factors, ZEB1/Snail, and the tight junction proteins, claudins. Importantly, while eEF-2K silencing recapitulates the rottlerin-induced inhibition of invasion and correlated events, eEF-2K overexpression, by lentivirus-based expression system, suppresses such rottlerin effects and potentiates PaCa cells invasion/migration capability. Collectively, our results show, for the first time, that eEF-2K is involved in regulation of the invasive phenotype of PaCa cells through promoting a new signalling pathway, which is mediated by TG2/ß1 integrin/Src/uPAR/MMP-2, and the induction of EMT biomarkers which enhance cancer cell motility and metastatic potential. Thus, eEF-2K could represent a novel potential therapeutic target in pancreatic cancer.


Asunto(s)
Quinasa del Factor 2 de Elongación/genética , Integrina beta1/biosíntesis , Neoplasias Pancreáticas/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Transglutaminasas/biosíntesis , Acetofenonas/administración & dosificación , Benzopiranos/administración & dosificación , Línea Celular Tumoral , Quinasa del Factor 2 de Elongación/metabolismo , Transición Epitelial-Mesenquimal/genética , Proteínas de Unión al GTP , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Integrina beta1/genética , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/patología , Proteína Glutamina Gamma Glutamiltransferasa 2 , ARN Interferente Pequeño , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Transducción de Señal/genética , Familia-src Quinasas/biosíntesis , Familia-src Quinasas/genética , Neoplasias Pancreáticas
20.
Int J Clin Exp Pathol ; 7(7): 3771-80, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25120753

RESUMEN

Urokinase-type plasminogen activator receptor (uPAR) and C-X-C-chemokine receptor-4 (CXCR4) are considered as key molecules in invasion and metastasis of several cancers via extracellular matrix degeneration and assist tumor metastasis to specific sites by chemotaxis. However, the combined effect of uPAR and CXCR4 on small cell lung cancer (SCLC), the most aggressive type of lung cancer, is not clear. In this study, we detected the expression of uPAR and CXCR4 in SCLC tissue samples (n = 50) by immunohistochemistry. The tumors with high expression of both uPAR and CXCR4 (12/50) had larger size, higher lymph node (LN) metastasis and worse prognosis of patients than those with low expression of uPAR and CXCR4 (38/50) (P < 0.05). We further identified and isolated the both uPAR and CXCR4 positive expression subpopulation cells (uPAR(+)CXCR4(+) cells) from the SCLC cell line H446 by flow cytometry. The uPAR(+)CXCR4(+) cancer cells showed a higher invasive and migrating capacity in the transwell and wound healing assays compared with other subpopulation cells (P < 0.05). uPAR(+)CXCR4(+) cells injected subcutaneously in nude mice markedly increased tumor growth and induced lung metastasis, while other subpopulation cells did not. In conclusion, these data suggest that uPAR and CXCR4 co-expression predicts worse prognosis of SCLC patients. uPAR(+)CXCR4(+) cells promote the tumor growth and play a potential role in metastasis of SCLC.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Pulmonares/patología , Receptores CXCR4/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Carcinoma Pulmonar de Células Pequeñas/patología , Adulto , Anciano , Animales , Movimiento Celular , Femenino , Citometría de Flujo , Xenoinjertos , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidad , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica/patología , Pronóstico , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/mortalidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA