Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Sci Adv ; 8(21): eabn3774, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35613265

RESUMEN

The diffuse-type gastric cancer (DGC) is a subtype of gastric cancer (GC) associated with low HER2 positivity rate and insensitivity to chemotherapy and immune checkpoint inhibitors. Here, we identify urokinase-type plasminogen activator receptor (uPAR) as a potential therapeutic target for DGC. We have developed a novel anti-uPAR monoclonal antibody, which targets the domains II and III of uPAR and blocks the binding of urokinase-type plasminogen activator to uPAR. We show that the combination of anti-uPAR and anti-Programmed cell death protein 1 (PD-1) remarkably inhibits tumor growth and prolongs survival via multiple mechanisms, using cell line-derived xenograft and patient-derived xenograft mouse models. Furthermore, uPAR chimeric antigen receptor-expressing T cells based on the novel anti-uPAR effectively kill DGC patient-derived organoids and exhibit impressive survival benefit in the established mouse models, especially when combined with PD-1 blockade therapy. Our study provides a new possibility of DGC treatment by targeting uPAR in a unique manner.


Asunto(s)
Receptor de Muerte Celular Programada 1 , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Neoplasias Gástricas , Animales , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Humanos , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Transducción de Señal , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
2.
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
3.
Drug Discov Today ; 26(4): 1076-1085, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33486111

RESUMEN

Urokinase-type plasminogen activator receptor (uPAR) mediates a multitude of biological activities, has key roles in several clinical indications, including malignancies and inflammation, and, thus, has attracted intensive research over the past few decades. The pleiotropic functions of uPAR can be attributed to its interaction with an array of partners. Many inhibitors have been developed to intervene with the interaction of uPAR with these partners. Here, we review the development of these classes of uPAR inhibitor and their inhibitory mechanisms to promote the translation of these inhibitors to clinical applications.


Asunto(s)
Descubrimiento de Drogas/métodos , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Transducción de Señal/efectos de los fármacos , Humanos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo
4.
ChemMedChem ; 16(2): 377-387, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33107192

RESUMEN

The urokinase receptor (uPAR) is a cell surface receptor that binds to the serine protease urokinase-type plasminogen activator (uPA) with high affinity. This interaction is beneficial for extravascular fibrin clearance, but it has also been associated with a broad range of pathological conditions including cancer, atherosclerosis, and kidney disease. Here, starting with a small molecule that we previously discovered by virtual screening and cheminformatics analysis, we design and synthesize several derivatives that were tested for binding and inhibition of the uPAR ⋅ uPA interaction. To confirm the binding site and establish a binding mode of the compounds, we carried out biophysical studies using uPAR mutants, among them uPARH47C-N259C , a mutant previously developed to mimic the structure of uPA-bound uPAR. Remarkably, a substantial increase in potency is observed for inhibition of uPARH47C-N259C binding to uPA compared to wild-type uPAR, consistent with our use of the structure of uPAR in its uPA-bound state to design small-molecule uPAR ⋅ uPA antagonists. Combined with the biophysical studies, molecular docking followed by extensive explicit-solvent molecular dynamics simulations and MM-GBSA free energy calculations yielded the most favorable binding pose of the compound. Collectively, these results suggest that potent inhibition of uPAR binding to uPA with small molecules will likely only be achieved by developing small molecules that exhibit high-affinity to solution apo structures of uPAR, rather than uPA-bound structures of the receptor.


Asunto(s)
Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Sitios de Unión/efectos de los fármacos , Quimioinformática , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Conformación Molecular , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
5.
Cell Mol Life Sci ; 78(6): 3057-3072, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33237352

RESUMEN

Exosomes (Exos) have been reported to promote pre-metastatic niche formation, proliferation, angiogenesis and metastasis. We have investigated the role of uPAR in melanoma cell lines-derived Exos and their pro-angiogenic effects on human microvascular endothelial cells (HMVECs) and endothelial colony-forming cells (ECFCs). Melanoma Exos were isolated from conditioned media of A375 and M6 cells by differential centrifugation and filtration. Tunable Resistive Pulse Sensing (TRPS) and Nanoparticle tracking analysis were performed to analyze dimension and concentration of Exos. The CRISPR-Cas 9 technology was exploited to obtain a robust uPAR knockout. uPAR is expressed in melanoma Exos that are internalized by HMVECs and ECFCs, enhancing VE-Cadherin, EGFR and uPAR expression in endothelial cells that undergo a complete angiogenic program, including proliferation, migration and tube formation. uPAR loss reduced the pro-angiogenic effects of melanoma Exos in vitro and in vivo by inhibition of VE-Cadherin, EGFR and uPAR expression and of ERK1,2 signaling in endothelial cells. A similar effect was obtained with a peptide that inhibits uPAR-EGFR interaction and with the EGFR inhibitor Gefitinib, which also inhibited melanoma Exos-dependent EGFR phosphorylation. This study suggests that uPAR is required for the pro-angiogenic activity of melanoma Exos. We propose the identification of uPAR-expressing Exos as a potentially useful biomarker for assessing pro-angiogenic propensity and eventually monitoring the response to treatment in metastatic melanoma patients.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Exosomas/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal , Animales , Antígenos CD/genética , Cadherinas/genética , Línea Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gefitinib/farmacología , Edición Génica , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neovascularización Fisiológica , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética
6.
Biomolecules ; 10(6)2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32630411

RESUMEN

Ligand-targeted toxins (LTTs) are bioengineered molecules which are composed of a targeting component linked to a toxin that induces cell death once the LTT binds its target. Bispecific targeting allows for the simultaneous targeting of two receptors. In this review, we mostly focus on the epidermal growth factor receptor (EGFR) as a target. We discuss the development and testing of a bispecific LTT targeting EGFR and urokinase-type plasminogen activator receptor (uPAR) as two attractive targets implicated in tumor growth and in the regulation of the tumor microvasculature in solid tumors. In vitro and mouse xenograft studies have shown that EGFR-targeted bispecific angiotoxin (eBAT) is effective against human solid tumors. Canine studies have shown that eBAT is both safe and effective against canine hemangiosarcoma, which is physiologically similar to human angiosarcoma. Finding the appropriate dosing strategy and sequencing of eBAT administration, in combination with other therapeutics, are among important factors for future directions. Together, the data indicate that eBAT targets cancer stem cells, it may have a role in inhibiting human tumor vasculature, and its bispecific conformation may have a role in reducing toxicity in comparative oncologic trials in dogs.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Hemangiosarcoma/tratamiento farmacológico , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Animales , Receptores ErbB/metabolismo , Hemangiosarcoma/metabolismo , Humanos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo
7.
Biomater Sci ; 8(2): 720-738, 2020 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-31777866

RESUMEN

In the present work, an iridium (Ir) complex loaded theranostic nanoplatform was designed for high-efficiency triple-negative breast cancer (TNBC) therapy. For this purpose, the Ir complex was firstly loaded on a photothermal agent of gold nanostars (GNS) by simply mixing followed by functionalization using a urokinase-type plasminogen activator receptor (uPAR) targeted polyetherimide-AE105 peptide conjugate (P-AE105) with the formation of GNS@Ir@P-AE105. It was demonstrated that the resultant GNS@Ir@P-AE105 was a multifunctional nanoplatform with advantages of (1) NIR laser controlled release of the Ir complex; (2) precise delivery of the Ir complex to TNBC cells; (3) excellent photothermal (PT)/photoacoustic (PA)/X-ray computed tomography (CT) tri-modal imaging ability; and (4) a synergistic photothermal-chemotherapeutic effect. An in-depth investigation of the mechanism revealed that binding forces of the Ir complex-GNS and P-AE105-GNS were significantly diminished upon NIR laser irradiation, which conferred an NIR laser-responsive Ir complex release property to the nanoplatform. Moreover, the nanoplatform exerted high efficiency anti-TNBC effects via a ROS-induced p53 apoptotic pathway. Specifically, combinational photothermal-chemotherapeutic treatments stimulated intracellular ROS generation, which significantly up-regulated apoptotic-relative p53 gene expression either by causing severe DNA damage or inducing an arrest effect on the sub-G1 phase of the cell cycle. Taken together, our work provides a novel theranostic nanoplatform for efficient and simultaneous diagnosis and therapy of TNBC.


Asunto(s)
Antineoplásicos/farmacología , Rayos Láser , Imagen Multimodal , Fototerapia , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/terapia , Animales , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Liberación de Fármacos , Humanos , Rayos Infrarrojos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Imagen Óptica , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas
8.
World Neurosurg ; 131: 252-263.e2, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31376551

RESUMEN

BACKGROUND: Glioblastoma (GBM) is the most common and deadly form of brain tumor. After standard treatment of resection, radiotherapy, and chemotherapy, the 5-year survival is <5%. In recent years, research has uncovered several potential targets within the Notch signaling pathway, which may lead to improved patient outcomes. METHODS: A literature search was performed for articles containing the terms "Glioblastoma" and "Receptors, Notch" between 2003 and July 2015. Of the 62 articles retrieved, 46 met our criteria and were included in our review. Nine articles were identified from other sources and were subsequently included, leaving 55 articles reviewed. RESULTS: Of the 55 articles reviewed, 47 used established human GBM cell lines. Seventeen articles used human GBM surgical samples. Forty-five of 48 articles that assessed Notch activity showed increased expression in GBM cell lines. Targeting the Notch pathway was carried out through Notch knockdown and overexpression and targeting δ-like ligand, Jagged, γ-secretase, ADAM10, ADAM17, and Mastermindlike protein 1. Arsenic trioxide, microRNAs, and several other compounds were shown to have an effect on the Notch pathway in GBM. Notch activity in GBM was also shown to be associated with hypoxia and certain cancer-related molecular pathways such as PI3K/AKT/mTOR and ERK/MAPK. Most articles concluded that Notch activity amplifies malignant characteristics in GBM and targeting this pathway can bring about amelioration of these effects. CONCLUSIONS: Recent literature suggests targeting the Notch pathway has great potential for future therapies for GBM.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Proteínas de Neoplasias/antagonistas & inhibidores , Receptores Notch/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Proteínas ADAM/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Antineoplásicos/farmacología , Trióxido de Arsénico/farmacología , Neoplasias Encefálicas/irrigación sanguínea , Hipoxia de la Célula , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Glioblastoma/irrigación sanguínea , Humanos , Proteínas Inhibidoras de la Diferenciación/antagonistas & inhibidores , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , MicroARNs/farmacología , Microvasos , Terapia Molecular Dirigida/métodos , Netrina-1/antagonistas & inhibidores , Niclosamida/farmacología , Receptores Notch/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Resveratrol/farmacología , Transducción de Señal/genética , Tretinoina/farmacología
9.
Sci Rep ; 9(1): 12179, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434965

RESUMEN

Drug resistance to anti-cancer agents is a major concern regarding the successful treatment of malignant tumors. Recent studies have suggested that acquired resistance to anti-epidermal growth factor receptor (EGFR) therapies such as cetuximab are in part caused by genetic alterations in patients with oral squamous cell carcinoma (OSCC). However, the molecular mechanisms employed by other complementary pathways that govern resistance remain unclear. In the current study, we performed gene expression profiling combined with extensive molecular validation to explore alternative mechanisms driving cetuximab-resistance in OSCC cells. Among the genes identified, we discovered that a urokinase-type plasminogen activator receptor (uPAR)/integrin ß1/Src/FAK signal circuit converges to regulate ERK1/2 phosphorylation and this pathway drives cetuximab-resistance in the absence of EGFR overexpression or acquired EGFR activating mutations. Notably, the polyphenolic phytoalexin resveratrol, inhibited uPAR expression and consequently the signaling molecules ERK1/2 downstream of EGFR thus revealing additive effects on promoting OSCC cetuximab-sensitivity in vitro and in vivo. The current findings indicate that uPAR expression plays a critical role in acquired cetuximab resistance of OSCC and that combination therapy with resveratrol may provide an attractive means for treating these patients.


Asunto(s)
Carcinoma de Células Escamosas/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Boca/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Resveratrol/farmacología , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Cetuximab/uso terapéutico , Resistencia a Antineoplásicos/genética , Humanos , Ratones , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/metabolismo , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Resveratrol/uso terapéutico , Transducción de Señal , Trasplante Heterólogo
10.
Cells ; 8(8)2019 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-31426601

RESUMEN

Dysregulation of vascular networks is characteristic of eye diseases associated with retinal cell degeneration and visual loss. Visual impairment is also the consequence of photoreceptor degeneration in inherited eye diseases with a major inflammatory component, but without angiogenic profile. Among the pathways with high impact on vascular/degenerative diseases of the eye, a central role is played by a system formed by the ligand urokinase-type plasminogen activator (uPA) and its receptor uPAR. The uPAR system, although extensively investigated in tumors, still remains a key issue in vascular diseases of the eye and even less studied in inherited retinal pathologies such as retinitis pigmantosa (RP). Its spectrum of action has been extended far beyond a classical pro-angiogenic function and has emerged as a central actor in inflammation. Preclinical studies in more prevalent eye diseases characterized by neovascular formation, as in retinopathy of prematurity, wet macular degeneration and rubeosis iridis or vasopermeability excess as in diabetic retinopathy, suggest a critical role of increased uPAR signaling indicating the potentiality of its modulation to counteract neovessel formation and microvascular dysfunction. The additional observation that the uPAR system plays a major role in RP by limiting the inflammatory cascade triggered by rod degeneration rises further questions about its role in the diseased eye.


Asunto(s)
Inflamación/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Enfermedades de la Retina , Activador de Plasminógeno de Tipo Uroquinasa , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Sprague-Dawley , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/fisiología
11.
Bull Exp Biol Med ; 167(3): 315-319, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31346863

RESUMEN

Vitronectin, extracellular matrix protein, plays an important role in embryonic development and in organ and tissue reparation. A unique characteristic of vitronectin is specific binding of various biological molecules, including urokinase receptor (uPAR), extracellular matrix components, adhesion receptors, growth factors, thus supporting the modulation of cell behavior. Vitronectin is in fact not found in intact myocardium, while after infarction its level increases significantly, which correlates with accumulation of uPAR+ progenitor cardiac cells in the focus. The cells isolated from the heart of wild type mice are characterized by higher adhesion to vitronectin than progenitor cardiac cells from the myocardium of uPAR knockout mice. In addition, inhibition of urokinase receptor with specific antibodies on the surface of the progenitor cardiac cells of wild type mice leads to attenuation of their adhesive activity and flattening on vitronectin matrix, which can be important for their distribution in the postinfarction myocardium and realization of the reparative functions.


Asunto(s)
Adhesión Celular/fisiología , Miocardio/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Células Madre/fisiología , Vitronectina/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/patología , Miocardio/citología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética
12.
Cell Death Dis ; 10(6): 401, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31127093

RESUMEN

Urokinase plasminogen activator receptor (uPAR) is upregulated in podocytes of glomerular diseases and crucially mediates podocyte injury through integrin ß3 (ITGB3). We previously showed that the miR-30 family maintains podocyte structure and function by inhibiting injurious calcineurin signaling through nuclear factor of activated T cells C (NFATC). Here, we tested whether the miR-30-calcineurin-NFATC and uPAR-ITGB3 pathways, two of the major pathways leading to podocyte injury, could interact. We found that podocyte-specific miR-30 knockdown in mice induced uPAR upregulation and ITGB3 activation, accompanied by proteinuria and podocyte injury. These effects of miR-30 knockdown were reduced using inhibitors of ITGB3, calcineurin, and NFATC, respectively, which are known to be antiproteinuric. These results indicate that miR-30 deficiency leads to calcineurin-NFATC signaling activation, which in turn activates the uPAR-ITGB3 pathway. In cultured podocytes, miR-30 knockdown also activated uPAR-ITGB3 signaling, leading to Rho GTPase activation, synaptopodin downregulation and podocyte injury. To explore uPAR-ITGB3 signaling regulation by miR-30 in podocytopathy development, we treated mice with lipopolysaccharide (LPS) and found that miR-30 was downregulated in podocytes, accompanied by uPAR upregulation and ITGB3 activation. We obtained the same results in cultured podocytes treated with LPS. Podocyte-specific transgenic miR-30 abolished uPAR-ITGB3 signaling and ameliorated podocyte injury and proteinuria in mice. Taken together, these experiments show that uPAR-ITGB3 signaling is negatively regulated by miR-30 through calcineurin-NFATC pathway, a novel mechanism underlying podocyte injury in glomerular diseases. Our study has elucidated the relationship among the crucial players governing podocyte pathophysiology and the antiproteinuric actions of drugs commonly used for podocytopathies.


Asunto(s)
Calcineurina/metabolismo , MicroARNs/metabolismo , Factores de Transcripción NFATC/metabolismo , Transducción de Señal , Animales , Antagomirs/metabolismo , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Humanos , Integrina beta3/química , Integrina beta3/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Transgénicos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Proteínas de Microfilamentos/metabolismo , Podocitos/citología , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal/efectos de los fármacos , Tacrolimus/farmacología , Regulación hacia Arriba/efectos de los fármacos , Proteínas de Unión al GTP rho/metabolismo
13.
ChemMedChem ; 14(1): 119-131, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30548204

RESUMEN

There is growing interest in the use of structure-based virtual screening to identify small molecules that inhibit challenging protein-protein interactions (PPIs). In this study, we investigated how effectively chemical library members docked at the PPI interface mimic the position of critical side-chain residues known as "hot spots". Three compound collections were considered, a commercially available screening collection (ChemDiv), a collection of diversity-oriented synthesis (DOS) compounds that contains natural-product-like small molecules, and a library constructed using established reactions (the "screenable chemical universe based on intuitive data organization", SCUBIDOO). Three different tight PPIs for which hot-spot residues have been identified were selected for analysis: uPAR⋅uPA, TEAD4⋅Yap1, and CaV α⋅CaV ß. Analysis of library physicochemical properties was followed by docking to the PPI receptors. A pharmacophore method was used to measure overlap between small-molecule substituents and hot-spot side chains. Fragment-like conformationally restricted small molecules showed better hot-spot overlap for interfaces with well-defined pockets such as uPAR⋅uPA, whereas better overlap was observed for more complex DOS compounds in interfaces lacking a well-defined binding site such as TEAD4⋅Yap1. Virtual screening of conformationally restricted compounds targeting uPAR⋅uPA and TEAD4⋅Yap1 followed by experimental validation reinforce these findings, as the best hits were fragment-like and had few rotatable bonds for the former, while no hits were identified for the latter. Overall, such studies provide a framework for understanding PPIs in the context of additional chemical matter and new PPI definitions.


Asunto(s)
Productos Biológicos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Productos Biológicos/síntesis química , Productos Biológicos/química , Canales de Calcio/química , Canales de Calcio/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/química , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Factores de Transcripción de Dominio TEA , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Proteínas Señalizadoras YAP
14.
J Cancer Res Clin Oncol ; 144(3): 415-430, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29288363

RESUMEN

Autophagy is a catabolic process for degrading dysfunctional proteins and organelles, and closely associated with cancer cell survival under therapeutic, metabolic stress, hypoxia, starvation and lack of growth factors, contributing to resistance to therapies. However, the role of autophagy in breast cancer cells is not well understood. In the present study, we investigated the role of autophagy in highly aggressive and metastatic triple negative breast cancer (TNBC) and non-metastatic breast cancer cells and demonstrated that the knockdown of autophagy-related genes (LC3 and Beclin-1) inhibited autophagy and significantly suppressed cell proliferation, colony formation, migration/invasion and induced apoptosis in MDA-MB-231 and BT-549 TNBC cells. Knockdown of LC3 and Beclin-1 led to inhibition of multiple proto-oncogenic signaling pathways, including cyclin D1, uPAR/integrin-ß1/Src, and PARP1. In conclusion, our study suggests that LC3 and Beclin-1 are required for cell proliferation, survival, migration and invasion, and may contribute to tumor growth and progression of highly aggressive and metastatic TNBC cells and therapeutic targeting of autophagy genes may be a potential therapeutic strategy for TNBC in breast cancer.


Asunto(s)
Beclina-1/antagonistas & inhibidores , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Terapia Molecular Dirigida/métodos , ARN Interferente Pequeño/farmacología , Neoplasias de la Mama Triple Negativas/patología , Autofagia/genética , Beclina-1/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Ciclina D1/antagonistas & inhibidores , Ciclina D1/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Integrina beta1/metabolismo , Células MCF-7 , Proteínas Asociadas a Microtúbulos/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias de la Mama Triple Negativas/genética , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
15.
ChemMedChem ; 12(21): 1794-1809, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-28960868

RESUMEN

Tight protein-protein interactions (Kd <100 nm) that occur over a large binding interface (>1000 Å2 ) are highly challenging to disrupt with small molecules. Historically, the design of small molecules to inhibit protein-protein interactions has focused on mimicking the position of interface protein ligand side chains. Here, we explore mimicry of the pairwise intermolecular interactions of the native protein ligand with residues of the protein receptor to enrich commercial libraries for small-molecule inhibitors of tight protein-protein interactions. We use the high-affinity interaction (Kd =1 nm) between the urokinase receptor (uPAR) and its ligand urokinase (uPA) to test our methods. We introduce three methods for rank-ordering small molecules docked to uPAR: 1) a new fingerprint approach that represents uPA's pairwise interaction energies with uPAR residues; 2) a pharmacophore approach to identify small molecules that mimic the position of uPA interface residues; and 3) a combined fingerprint and pharmacophore approach. Our work led to small molecules with novel chemotypes that inhibited a tight uPAR⋅uPA protein-protein interaction with single-digit micromolar IC50 values. We also report the extensive work that identified several of the hits as either lacking stability, thiol reactive, or redox active. This work suggests that mimicking the binding profile of the native ligand and the position of interface residues can be an effective strategy to enrich commercial libraries for small-molecule inhibitors of tight protein-protein interactions.


Asunto(s)
Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Sitios de Unión , Ensayo de Inmunoadsorción Enzimática , Ensayos Analíticos de Alto Rendimiento , Humanos , Concentración 50 Inhibidora , Cinética , Simulación del Acoplamiento Molecular , Dominios y Motivos de Interacción de Proteínas/efectos de los fármacos , Estructura Terciaria de Proteína , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad , Termodinámica , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores
16.
Sci Rep ; 7(1): 9388, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28839232

RESUMEN

The urokinase-type plasminogen activator receptor (uPAR) is a GPI-anchored cell membrane receptor that focuses urokinase (uPA) proteolytic activity on the cell surface. Its expression is increased in many human cancers, including non-small cell lung cancer (NSCLC) and colorectal cancer (CRC), and correlates with a poor prognosis and early invasion and metastasis. uPAR is able to control, through a cross-talk with tyrosine kinase receptors, the shift between tumor dormancy and proliferation, that usually precedes metastasis formation. Therefore, we investigated the role of uPAR expression in RAS mutated NSCLC and CRC cells. In this study we provided evidence, for the first time, that RAS mutational condition is functionally correlated to uPAR overexpression in NSCLC and CRC cancer cell lines and patient-derived tissue samples. Moreover, oncogenic features related to uPAR overexpression in RAS mutated NSCLC and CRC, such as adhesion, migration and metastatic process may be targeted, in vitro and in vivo, by new anti-uPAR small molecules, specific inhibitors of uPAR-vitronectin interaction. Therefore, anti-uPAR drugs could represent an effective pharmacological strategy for NSCLC and CRC patients carrying RAS mutations.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias/genética , Neoplasias/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Proteínas ras/genética , Animales , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/metabolismo
17.
Elife ; 62017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28849762

RESUMEN

The urokinase receptor (uPAR) is a glycosylphosphatidylinositol (GPI)-anchored protein that promotes tissue remodeling, tumor cell adhesion, migration and invasion. uPAR mediates degradation of the extracellular matrix through protease recruitment and enhances cell adhesion, migration and signaling through vitronectin binding and interactions with integrins. Full-length uPAR is released from the cell surface, but the mechanism and significance of uPAR shedding remain obscure. Here we identify transmembrane glycerophosphodiesterase GDE3 as a GPI-specific phospholipase C that cleaves and releases uPAR with consequent loss of function, whereas its homologue GDE2 fails to attack uPAR. GDE3 overexpression depletes uPAR from distinct basolateral membrane domains in breast cancer cells, resulting in a less transformed phenotype, it slows tumor growth in a xenograft model and correlates with prolonged survival in patients. Our results establish GDE3 as a negative regulator of the uPAR signaling network and, furthermore, highlight GPI-anchor hydrolysis as a cell-intrinsic mechanism to alter cell behavior.


Asunto(s)
Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Hidrolasas Diéster Fosfóricas/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Femenino , Técnicas de Inactivación de Genes/métodos , Células HEK293 , Humanos , Hidrólisis , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Desnudos , Modelos Moleculares , Trasplante de Neoplasias , Hidrolasas Diéster Fosfóricas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal , Carga Tumoral , Vitronectina/genética , Vitronectina/metabolismo
18.
Oncol Rep ; 38(4): 2325-2334, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28849196

RESUMEN

Epidermal growth factor (EGF) signaling has been shown to induce epithelial to mesenchymal transition (EMT) in many types of cancer cells. However, the molecular mechanism of EGF-induced EMT in gastric cancer remains largely unknown. In the present study, we found that human gastric cancer cell lines SGC-7901 and BGC-823 underwent EMT phenotypic changes upon exposure to EGF. The induction of EMT was consistent with aggressive characteristics such as increased cell migration, invasion and clonogenic growth. Additionally, EGF stimulation also led to the upregulation of urokinase plasminogen activator receptor (uPAR) both at mRNA and protein levels. Knockdown of uPAR by siRNA significantly attenuated EMT induction by EGF in SGC-7901 and BGC-823 cells. Furthermore, EGF increased ERK1/2 activity and blocking ERK1/2 signaling with its inhibitor, U0126, markedly inhibited EGF-induced uPAR expression and consequently EMT. Collectively, the present study demonstrated that EGF induced aggressiveness of gastric cancer cells by activating EMT, which involved the activation of the ERK1/2 pathway and, subsequently, uPAR expression.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Transición Epitelial-Mesenquimal/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Neoplasias Gástricas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , ARN Interferente Pequeño/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Neoplasias Gástricas/patología
19.
Sci Rep ; 7(1): 1312, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28465589

RESUMEN

The development of metastases is a multistep process that requires the activation of physiological and biochemical processes that govern migration, invasion and entry of metastatic cells into blood vessels. The urokinase receptor (uPAR) promotes cell migration by interacting with the Formyl Peptide Receptors (FPRs). Since both uPAR and FPR1 are involved in tumor progression, the uPAR-FPR1 interaction is an attractive therapeutic target. We previously described peptide antagonists of the uPAR-FPR1 interaction that inhibited cell migration and angiogenesis. To develop enzyme-resistant analogues, we applied here the Retro-Inverso (RI) approach, whereby the topology of the side chains is maintained by inverting the sequence of the peptide and the chirality of all residues. Molecular dynamics suggests that peptide RI-3 adopts the turn structure typical of uPAR-FPR1 antagonists. Accordingly, RI-3 is a nanomolar competitor of N-formyl-Met-Leu-Phe for binding to FPR1 and inhibits migration, invasion, trans-endothelial migration of sarcoma cells and VEGF-triggered endothelial tube formation. When sarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density, circulating tumor cells and pulmonary metastases were significantly reduced in animals treated daily with 6 mg/Kg RI-3 as compared to animals treated with vehicle only. Thus, RI-3 represents a promising lead for anti-metastatic drugs.


Asunto(s)
Neovascularización Patológica/tratamiento farmacológico , Péptidos/administración & dosificación , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Sarcoma/tratamiento farmacológico , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Simulación de Dinámica Molecular , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Formil Péptido/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Sarcoma/genética , Sarcoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Mol Cancer Ther ; 16(5): 956-965, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28193671

RESUMEN

Sarcomas differ from carcinomas in their mesenchymal origin. Therapeutic advancements have come slowly, so alternative drugs and models are urgently needed. These studies report a new drug for sarcomas that simultaneously targets both tumor and tumor neovasculature. eBAT is a bispecific angiotoxin consisting of truncated, deimmunized Pseudomonas exotoxin fused to EGF and the amino terminal fragment of urokinase. Here, we study the drug in an in vivo "ontarget" companion dog trial as eBAT effectively kills canine hemangiosarcoma and human sarcoma cells in vitro We reasoned the model has value due to the common occurrence of spontaneous sarcomas in dogs and a limited lifespan allowing for rapid accrual and data collection. Splenectomized dogs with minimal residual disease were given one cycle of eBAT followed by adjuvant doxorubicin in an adaptive dose-finding, phase I-II study of 23 dogs with spontaneous, stage I-II, splenic hemangiosarcoma. eBAT improved 6-month survival from <40% in a comparison population to approximately 70% in dogs treated at a biologically active dose (50 µg/kg). Six dogs were long-term survivors, living >450 days. eBAT abated expected toxicity associated with EGFR targeting, a finding supported by mouse studies. Urokinase plasminogen activator receptor and EGFR are targets for human sarcomas, so thorough evaluation is crucial for validation of the dog model. Thus, we validated these markers for human sarcoma targeting in the study of 212 human and 97 canine sarcoma samples. Our results support further translation of eBAT for human patients with sarcomas and perhaps other EGFR-expressing malignancies. Mol Cancer Ther; 16(5); 956-65. ©2017 AACR.


Asunto(s)
Receptores ErbB/genética , Hemangiosarcoma/tratamiento farmacológico , Terapia Molecular Dirigida , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , ADP Ribosa Transferasas/administración & dosificación , ADP Ribosa Transferasas/química , ADP Ribosa Transferasas/genética , Animales , Toxinas Bacterianas/administración & dosificación , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perros , Doxorrubicina/administración & dosificación , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/genética , Receptores ErbB/antagonistas & inhibidores , Exotoxinas/administración & dosificación , Exotoxinas/química , Exotoxinas/genética , Hemangiosarcoma/genética , Hemangiosarcoma/patología , Humanos , Ratones , Estadificación de Neoplasias , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/genética , Factores de Virulencia/administración & dosificación , Factores de Virulencia/química , Factores de Virulencia/genética , Exotoxina A de Pseudomonas aeruginosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA