Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Immunol ; 200(3): 957-965, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29282305

RESUMEN

Genetic absence of the urokinase-type plasminogen activator (uPA) reduces arthritis progression in the collagen-induced arthritis (CIA) mouse model to an extent just shy of disease abrogation, but this remarkable observation has not been translated into therapeutic intervention. Our aim was to test the potential in mice of an Ab that blocks the proteolytic capacity of uPA in the CIA model and the delayed-type hypersensitivity arthritis model. A second aim was to determine the cellular origins of uPA and the uPA receptor (uPAR) in joint tissue from patients with rheumatoid arthritis. A mAb that neutralizes mouse uPA significantly reduced arthritis progression in the CIA and delayed-type hypersensitivity arthritis models. In the CIA model, the impact of anti-uPA treatment was on par with the effect of blocking TNF-α by etanercept. A pharmacokinetics evaluation of the therapeutic Ab revealed target-mediated drug disposition consistent with a high turnover of endogenous uPA. The cellular expression patterns of uPA and uPAR were characterized by double immunofluorescence in the inflamed synovium from patients with rheumatoid arthritis and compared with synovium from healthy donors. The arthritic synovium showed expression of uPA and uPAR in neutrophils, macrophages, and a fraction of endothelial cells, whereas there was little or no expression in synovium from healthy donors. The data from animal models and human material provide preclinical proof-of-principle that validates uPA as a novel therapeutic target in rheumatic diseases.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Artritis Experimental/patología , Artritis Reumatoide/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Membrana Sinovial/patología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/inmunología , Etanercept/farmacología , Femenino , Humanos , Hipersensibilidad Tardía/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Neutrófilos/inmunología , Membrana Sinovial/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
2.
Front Immunol ; 15: 1404121, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38720900

RESUMEN

Pharmacodynamic assessment of T-cell-based cancer immunotherapies often focus on detecting rare circulating T-cell populations. The therapy-induced immune cells in blood-derived clinical samples are often present in very low frequencies and with the currently available T-cell analytical assays, amplification of the cells of interest prior to analysis is often required. Current approaches aiming to enrich antigen-specific T cells from human Peripheral Blood Mononuclear Cells (PBMCs) depend on in vitro culturing in presence of their cognate peptides and cytokines. In the present work, we improved a standard, publicly available protocol for T-cell immune analyses based on the in vitro expansion of T cells. We used PBMCs from healthy subjects and well-described viral antigens as a model system for optimizing the experimental procedures and conditions. Using the standard protocol, we first demonstrated significant enrichment of antigen-specific T cells, even when their starting frequency ex vivo was low. Importantly, this amplification occurred with high specificity, with no or neglectable enrichment of irrelevant T-cell clones being observed in the cultures. Testing of modified culturing timelines suggested that the protocol can be adjusted accordingly to allow for greater cell yield with strong preservation of the functionality of antigen-specific T cells. Overall, our work has led to the refinement of a standard protocol for in vitro stimulation of antigen-specific T cells and highlighted its reliability and reproducibility. We envision that the optimized protocol could be applied for longitudinal monitoring of rare blood-circulating T cells in scenarios with limited sample material.


Asunto(s)
Linfocitos T , Humanos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Antígenos Virales/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Células Cultivadas , Vacunas contra el Cáncer/inmunología
3.
FASEB J ; 26(11): 4445-57, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22815383

RESUMEN

Rearrangement of the skin during wound healing depends on plasmin and plasminogen, which serve to degrade fibrin depositions in the provisional matrix and thereby facilitate keratinocyte migration. In the current study, we investigated whether plasmin and plasminogen likewise played a role during the development of skin cancer. To test this, we set up a chemically induced skin tumor model in a cohort of mice and found that skin tumor growth in Plg(-/-) male mice was reduced by 52% compared with wild-type controls. Histological analyses suggested that the growth-restricting effect of plasminogen deficiency was due to thrombosis and lost patency of the tumor vasculature, resulting in tumor necrosis. The connection between plasmin-dependent fibrinolysis, vascular patency, and tumor growth was further substantiated as the effect of plasminogen deficiency on tumor growth could be reverted by superimposing heterozygous fibrinogen deficiency on Plg(-/-) mice. Tumors derived from these Fib(-/+);Plg(-/-) mice displayed a significantly decreased level of tumor thrombosis compared with Plg(-/-) mice. In summary, these data indicate that plasmin-driven fibrinolysis facilitates tumor growth by maintaining patency of the tumor vasculature.


Asunto(s)
Fibrinolisina/metabolismo , Fibrinólisis/fisiología , Neoplasias Cutáneas/patología , Animales , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Fibrina/genética , Fibrina/metabolismo , Fibrinógeno/genética , Fibrinógeno/metabolismo , Fibrinolisina/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Laminina/genética , Laminina/metabolismo , Masculino , Ratones , Ratones Noqueados , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/metabolismo , Ovariectomía , Factores Sexuales , Neoplasias Cutáneas/metabolismo , Trombosis/metabolismo
4.
J Pathol ; 227(1): 94-105, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22294280

RESUMEN

Fibrosis of the liver and its end-stage, cirrhosis, represent major health problems worldwide. In these fibrotic conditions, activated fibroblasts and hepatic stellate cells display a net deposition of collagen. This collagen deposition is a major factor leading to liver dysfunction, thus making it crucially important to understand both the collagen synthesis and turnover mechanisms in this condition. Here we show that the endocytic collagen receptor, uPARAP/Endo180, is a major determinant in governing the balance between collagen deposition and degradation. Cirrhotic human livers displayed a marked up-regulation of uPARAP/Endo180 in activated fibroblasts and hepatic stellate cells located close to the collagen deposits. In a hepatic stellate cell line, uPARAP/Endo180 was shown to be active in, and required for, the uptake and intracellular degradation of collagen. To evaluate the functional importance of this collagen receptor in vivo, liver fibrosis was induced in uPARAP/Endo180-deficient mice and littermate wild-type mice by chronic CCl(4) administration. A strong up-regulation of uPARAP/Endo180 was observed in wild-type mice, and a quantitative comparison of collagen deposits in the two groups of mice clearly revealed a fibrosis protective role of uPARAP/Endo180. This effect appeared to directly reflect the activity of the collagen receptor, since no compensatory events were noted when comparing the mRNA expression profiles of the two groups of mice in an array system focused on matrix-degrading components. This function of uPARAP/Endo180 defines a novel role of intracellular collagen turnover in fibrosis protection.


Asunto(s)
Colágeno/metabolismo , Endocitosis/fisiología , Cirrosis Hepática Experimental/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Línea Celular , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática Experimental/genética , Cirrosis Hepática Experimental/prevención & control , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos , Ratones Noqueados , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Regulación hacia Arriba
5.
Front Immunol ; 14: 1166546, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37114047

RESUMEN

The global SARS-CoV-2 pandemic caused significant social and economic disruption worldwide, despite highly effective vaccines being developed at an unprecedented speed. Because the first licensed vaccines target only single B-cell antigens, antigenic drift could lead to loss of efficacy against emerging SARS-CoV-2 variants. Improving B-cell vaccines by including multiple T-cell epitopes could solve this problem. Here, we show that in silico predicted MHC class I/II ligands induce robust T-cell responses and protect against severe disease in genetically modified K18-hACE2/BL6 mice susceptible to SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Vacunas de ADN , Animales , Ratones , COVID-19/prevención & control , ADN , Epítopos de Linfocito T , Inmunización , SARS-CoV-2
6.
NPJ Vaccines ; 8(1): 77, 2023 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-37244905

RESUMEN

Recent findings have positioned tumor mutation-derived neoepitopes as attractive targets for cancer immunotherapy. Cancer vaccines that deliver neoepitopes via various vaccine formulations have demonstrated promising preliminary results in patients and animal models. In the presented work, we assessed the ability of plasmid DNA to confer neoepitope immunogenicity and anti-tumor effect in two murine syngeneic cancer models. We demonstrated that neoepitope DNA vaccination led to anti-tumor immunity in the CT26 and B16F10 tumor models, with the long-lasting presence of neoepitope-specific T-cell responses in blood, spleen, and tumors after immunization. We further observed that engagement of both the CD4+ and CD8+ T cell compartments was essential to hamper tumor growth. Additionally, combination therapy with immune checkpoint inhibition provided an additive effect, superior to either monotherapy. DNA vaccination offers a versatile platform that allows the encoding of multiple neoepitopes in a single formulation and is thus a feasible strategy for personalized immunotherapy via neoepitope vaccination.

8.
Front Immunol ; 14: 1234912, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37720215

RESUMEN

Introduction: Tumor-specific mutations generate neoepitopes unique to the cancer that can be recognized by the immune system, making them appealing targets for therapeutic cancer vaccines. Since the vast majority of tumor mutations are patient-specific, it is crucial for cancer vaccine designs to be compatible with individualized treatment strategies. Plasmid DNA vaccines have substantiated the immunogenicity and tumor eradication capacity of cancer neoepitopes in preclinical models. Moreover, early clinical trials evaluating personalized neoepitope vaccines have indicated favorable safety profiles and demonstrated their ability to elicit specific immune responses toward the vaccine neoepitopes. Methods: By fusing in silico predicted neoepitopes to molecules with affinity for receptors on the surface of APCs, such as chemokine (C-C motif) ligand 19 (CCL19), we designed an APC-targeting cancer vaccine and evaluated their ability to induce T-cell responses and anti-tumor efficacy in the BALB/c syngeneic preclinical tumor model. Results: In this study, we demonstrate how the addition of an antigen-presenting cell (APC) binding molecule to DNA-encoded cancer neoepitopes improves neoepitope-specific T-cell responses and the anti-tumor efficacy of plasmid DNA vaccines. Dose-response evaluation and longitudinal analysis of neoepitope-specific T-cell responses indicate that combining APC-binding molecules with the delivery of personalized tumor antigens holds the potential to improve the clinical efficacy of therapeutic DNA cancer vaccines. Discussion: Our findings indicate the potential of the APC-targeting strategy to enhance personalized DNA cancer vaccines while acknowledging the need for further research to investigate its molecular mechanism of action and to translate the preclinical results into effective treatments for cancer patients.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Vacunas de ADN , Humanos , Neoplasias/genética , Neoplasias/terapia , Células Presentadoras de Antígenos , Mutación
9.
Dev Biol ; 358(1): 56-67, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21802414

RESUMEN

Urokinase-type plasminogen activator (uPA) and matrix metalloproteinase-9 (MMP9, gelatinase B) have separately been recognized to play important roles in various tissue remodeling processes. In this study, we demonstrate that deficiency for MMP9 in combination with ablation of either uPA- or tissue-type plasminogen activator (tPA)-catalyzed plasminogen activation is critical to accomplish normal gestation in mice. Gestation was also affected by simultaneous lack of MMP9 and the uPA receptor (uPAR). Interestingly, uPA-deficiency additionally exacerbated the effect of MMP9-deficiency on bone growth and an additive effect caused by combined lack in MMP9 and uPA was observed during healing of cutaneous wounds. By comparison, MMP9-deficiency combined with absence of either tPA or uPAR resulted in no significant effect on wound healing, indicating that the role of uPA during wound healing is independent of uPAR, when MMP9 is absent. Notably, compensatory upregulation of uPA activity was seen in wounds from MMP9-deficient mice. Taken together, these studies reveal essential functional dependency between MMP9 and uPA during gestation and tissue repair.


Asunto(s)
Metaloproteinasa 9 de la Matriz/deficiencia , Embarazo/fisiología , Fenómenos Fisiológicos de la Piel , Activador de Plasminógeno de Tipo Uroquinasa/deficiencia , Cicatrización de Heridas/fisiología , Animales , Western Blotting , Pesos y Medidas Corporales , Cartilla de ADN/genética , Electroforesis en Gel de Poliacrilamida , Femenino , Técnicas Histológicas , Hibridación in Situ , Ratones , Cicatrización de Heridas/genética
10.
Mediators Inflamm ; 2012: 157894, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22529519

RESUMEN

Besides their evident importance in host defense, macrophages have been shown to play a detrimental role in different pathological conditions, including chronic inflammation, atherosclerosis, and cancer. Regardless of the exact situation, macrophage activation and migration are intimately connected to extracellular matrix degradation. This process is accomplished by multiple proteolytic enzymes, including serine proteases and members of the matrix metalloproteinase family. In this study, we have utilized qPCR arrays to simultaneously analyze the temporal expression pattern of a range of genes involved in extracellular matrix metabolism in the mouse derived-macrophage cell line RAW 264.7 following stimulation with LPS. Our results revealed that LPS induces the expression of matrix metalloproteinases while at the same time decreased the expression of matrix metalloproteinase inhibitors. The opposite scenario was found for the genes encoding serine proteases, which were downregulated while their inhibitors were upregulated. In addition, intergenic comparison of the expression levels of related proteases revealed large differences in their basal expression level. These data highlight the complexity of the gene expression regulation implicated in macrophage-dependent matrix degradation and furthermore emphasize the value of qPCR array techniques for the investigation of the complex regulation of the matrix degradome.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Lipopolisacáridos/metabolismo , Macrófagos/enzimología , Péptido Hidrolasas/biosíntesis , Inhibidores de Proteasas/metabolismo , ARN/biosíntesis , Algoritmos , Animales , Línea Celular , Dosificación de Gen , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Reconocimiento de Normas Patrones Automatizadas , Péptido Hidrolasas/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Transducción de Señal , Factores de Tiempo , Transcripción Genética
11.
Exp Dermatol ; 19(8): e234-40, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20163454

RESUMEN

During healing of incisional skin wounds, migrating keratinocytes dissect their way under the crust to re-epithelialize the wounded area. The efficiency of this tissue remodelling process depends on the concomitant activity of several extracellular proteases, including members of the plasminogen activation (PA) system and the matrix metalloproteinase (MMP) family. Treatment with the broad spectrum MMP inhibitor, galardin, delays wound healing in wildtype mice and completely arrest wound healing in plasminogen (Plg)-deficient mice, indicating a functional overlap between plasmin- and galardin-sensitive MMPs during wound healing. To address whether MMP2 is accountable for the galardin-induced healing deficiency in wildtype and Plg-deficient mice, incisional skin wounds were generated in MMP2 single-deficient mice and in MMP2/Plg double-deficient mice and followed until healed. Alternatively, tissue was isolated 7 days post wounding for histological and biochemical analyses. No difference was found in the time from wounding to overt gross restoration of the epidermal surface between MMP2-deficient and wildtype control littermate mice. MMP2/Plg double-deficient mice were viable and fertile, and displayed an unchallenged general phenotype resembling that of Plg-deficient mice, including development of rectal prolapses. MMP2/Plg double-deficient mice displayed a slight increase in the wound length throughout the healing period compared with Plg-deficient mice. However, the overall time to complete healing was not significantly different between Plg-deficient and MMP2/Plg double-deficient mice. These results show that MMP2 activity is not essential for wound healing and indicate that lack of MMP2 only marginally potentiates the effect of Plg deficiency.


Asunto(s)
Metaloproteinasa 2 de la Matriz/deficiencia , Plasminógeno/deficiencia , Piel/lesiones , Cicatrización de Heridas/fisiología , Animales , Dipéptidos/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Modelos Animales , Plasminógeno/genética , Plasminógeno/metabolismo , Inhibidores de Proteasas/farmacología , ARN Mensajero/metabolismo , Piel/metabolismo
12.
Thromb Haemost ; 97(6): 1013-22, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17549305

RESUMEN

Binding of urokinase plasminogen activator (uPA) to its cellular receptor, uPAR, potentiates plasminogen activation and localizes it to the cell surface. Focal plasminogen activation is involved in both normal and pathological tissue remodeling processes including cancer invasion. The interaction between uPA and uPAR therefore represents a potential target for anti-invasive cancer therapy. Inhibitors of the human uPA-uPAR interaction have no effect in the murine system. To enable in-vivo studies in murine cancer models we have now generated murine monoclonal antibodies (mAbs) against murine uPAR (muPAR) by immunizing uPAR-deficient mice with recombinant muPAR and screened for antibodies, which inhibit the muPA-muPAR interaction. Two of the twelve mAbs obtained, mR1 and mR2, interfered with the interaction between muPAR and the amino-terminal fragment of muPA (mATF) when analyzed by surface plasmon resonance. The epitope for mR1 is located on domain I of muPAR, while that of mR2 is on domains (II-III). In cell binding experiments using radiolabelled mATF, the maximal inhibition obtained with mR1 was 85% while that obtained with mR2 was 50%. The IC(50) value for mR1 was 0.67 nM compared to 0.14 nM for mATF. In an assay based on modified anthrax toxins, requiring cell-bound muPA activity for its cytotoxity, an approximately 50% rescue of the cells could be obtained by addition of mR1. Importantly, in-vivo efficacy of mR1 was demonstrated by the ability of mR1 to rescue mice treated with a lethal dose of uPA-activatable anthrax toxins.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Macrófagos/efectos de los fármacos , Plasminógeno/metabolismo , Receptores de Superficie Celular/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Reacciones Antígeno-Anticuerpo , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/toxicidad , Sitios de Unión de Anticuerpos , Unión Competitiva , Línea Celular , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Mapeo Epitopo , Femenino , Humanos , Hibridomas/metabolismo , Inmunización , Radioisótopos de Yodo/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Fragmentos de Péptidos/metabolismo , Estructura Terciaria de Proteína , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Factores de Tiempo
13.
Mol Cancer Ther ; 5(1): 89-96, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16432166

RESUMEN

Previously, we have generated a potent prodrug consisting of modified anthrax toxins that is activated by urokinase plasminogen activator (uPA). The cytotoxicity of the drug, PrAg-U2 + FP59, is dependent on the presence of receptor-associated uPA activity. Local intradermal administration of PrAg-U2 + FP59 adjacent to the tumor nodules in mice with transplanted solid tumors had a potent antitumor effect. In succession of these experiments, we have now investigated the systemic antitumor efficacy of PrAg-U2 + FP59. C57Bl/6J mice bearing syngenic tumors derived from B16 melanoma, T241 fibrosarcoma, or Lewis lung carcinoma cells were treated with different mass ratios and doses of PrAg-U2 + FP59. Tumor volumes were recorded daily by caliper measurements. In some experiments, dexamethasone was coadministered. Our data show a significant antitumor effect of systemic administration of PrAg-U2 + FP59 in three syngenic tumor models. Optimal antitumor effect and low toxicity was obtained with a 25:1 mass ratio between the two components (PrAg-U2 and FP59). The experiments show that PrAg-U2 + FP59 displays a clear dose-response relationship with regard to both antitumor efficacy and systemic toxicity. Dose-limiting toxicity seemed to be due to activation of the prodrug by uPA and its receptor in the intestinal mucosa. Concurrent treatment with dexamethasone was found to prevent dose-limiting toxicity. Taken together, these data indicate that uPA-activated toxins may be promising candidates for targeted therapy of human cancers that overexpress uPA and its receptor.


Asunto(s)
Antígenos Bacterianos/farmacología , Antineoplásicos/farmacología , Toxinas Bacterianas/farmacología , Profármacos/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Antígenos Bacterianos/toxicidad , Antineoplásicos/toxicidad , Toxinas Bacterianas/toxicidad , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Profármacos/química , Proteínas Recombinantes de Fusión/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/efectos de los fármacos
14.
Sci Rep ; 6: 30739, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27491826

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancer, representing approximately 75% of all renal neoplasms. ccRCC is known to be strongly associated with silencing of the von Hippel Lindau (VHL) tumor suppressor gene, yet VHL deficiency alone does not seem to be sufficient to drive the oncogenic transformation of normal renal epithelium and induce renal tumorigenesis. We, and others, have previously suggested that constitutive activation of the Notch signaling pathway, alongside with VHL loss, contribute to the oncogenic features of ccRCC. Here we report a prevailing hyperactivation of the Notch1 receptor in human ccRCC relative to the healthy counterpart. To explore the consequences of the elevated Notch1 signaling observed in ccRCC patient material, we made use of a conditional mouse model based on concurrent ectopic expression of constitutively active Notch1 (NICD1) and deletion of the Vhl gene. Histological examination of the kidneys of the conditional mice demonstrate the existence of nests of dysplastic cells with a clear cytoplasm as a consequence of lipid accumulation, thus displaying a one important hallmark of human ccRCC.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Receptor Notch1/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Humanos , Riñón/metabolismo , Riñón/patología , Neoplasias Renales/genética , Metabolismo de los Lípidos , Ratones , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Receptor Notch1/química , Transducción de Señal , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
15.
PLoS One ; 8(3): e59942, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527289

RESUMEN

The fibrinolytic activity of plasmin plays a fundamental role in resolution of blood clots and clearance of extravascular deposited fibrin in damaged tissues. These vital functions of plasmin are exploited by malignant cells to accelerate tumor growth and facilitate metastases. Mice lacking functional plasmin thus display decreased tumor growth in a variety of cancer models. Interestingly, this role of plasmin has, in regard to skin cancer, been shown to be restricted to male mice. It remains to be clarified whether gender also affects other phenotypic characteristics of plasmin deficiency or if this gender effect is restricted to skin cancer. To investigate this, we tested the effect of gender on plasmin dependent immune cell migration, accumulation of hepatic fibrin depositions, skin composition, and skin wound healing. Gender did not affect immune cell migration or hepatic fibrin accumulation in neither wildtype nor plasmin deficient mice, and the existing differences in skin composition between males and females were unaffected by plasmin deficiency. In contrast, gender had a marked effect on the ability of plasmin deficient mice to heal skin wounds, which was seen as an accelerated wound closure in female versus male plasmin deficient mice. Further studies showed that this gender effect could not be reversed by ovariectomy, suggesting that female sex-hormones did not mediate the accelerated skin wound healing in plasmin deficient female mice. Histological examination of healed wounds revealed larger amounts of fibrotic scars in the provisional matrix of plasmin deficient male mice compared to female mice. These fibrotic scars correlated to an obstruction of cell infiltration of the granulation tissue, which is a prerequisite for wound healing. In conclusion, the presented data show that the gender dependent effect of plasmin deficiency is tissue specific and may be secondary to already established differences between genders, such as skin thickness and composition.


Asunto(s)
Plasminógeno/deficiencia , Caracteres Sexuales , Cicatrización de Heridas/fisiología , Análisis de Varianza , Animales , Movimiento Celular/fisiología , Femenino , Fibrina/metabolismo , Tejido de Granulación/citología , Tejido de Granulación/metabolismo , Técnicas Histológicas , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ovariectomía
16.
Dis Model Mech ; 4(2): 212-27, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21123624

RESUMEN

The family of matrix metalloproteinases (MMPs) is responsible for extracellular matrix degradation during physiological and pathophysiological tissue remodeling processes such as embryogenesis, tissue repair and cancer progression. Despite these important roles of MMPs, inhibition or ablation of individual members of the MMP family in animal models have been shown to have little effect. It has been speculated that this results from a functional overlap between individual MMPs and (as-yet-unclassified) functional overlaps between MMPs and other protease systems. We here present genetic data showing that concomitant ablation of MMP9 (gelatinase B) and the serine protease plasmin results in lethal inflammatory mass lesions in the colon. These lesions possessed several histological attributes that are characteristic of mucosal prolapse seen in humans, and they were found to be associated with splenomegaly, enlarged mesenteric lymph nodes, decreased thymus size and altered populations of circulating immune cells. A time-course study provided evidence that the massive lymphoid hyperplasia and reactive changes were secondary to discrete fibrinous lesions also observed in mice only deficient for plasminogen (Plg), the zymogen for plasmin. These data demonstrate a non-appreciated vital protective role for MMP9 in the absence of Plg.


Asunto(s)
Colon/enzimología , Colon/patología , Inflamación/enzimología , Inflamación/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Sustancias Protectoras/metabolismo , Alelos , Animales , Linfocitos B/metabolismo , Células Epiteliales/patología , Fibrina/metabolismo , Granulocitos , Recuento de Leucocitos , Activación de Linfocitos/inmunología , Tejido Linfoide/patología , Metaloproteinasa 9 de la Matriz/deficiencia , Ratones , Plasminógeno/deficiencia , Plasminógeno/metabolismo , Bazo/patología , Linfocitos T/inmunología , Timo/patología , Cicatrización de Heridas
17.
PLoS One ; 5(9): e12746, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20856796

RESUMEN

BACKGROUND: Proteolytic degradation by plasmin and metalloproteinases is essential for epidermal regeneration in skin wound healing. Plasminogen deficient mice have severely delayed wound closure as have mice simultaneously lacking the two plasminogen activators, urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA). In contrast, individual genetic deficiencies in either uPA or tPA lead to wound healing kinetics with no or only slightly delayed closure of skin wounds. METHODOLOGY/PRINCIPAL FINDINGS: To evaluate the therapeutic potential in vivo of a murine neutralizing antibody directed against mouse uPA we investigated the efficacy in skin wound healing of tPA-deficient mice. Systemic administration of the anti-mouse uPA monoclonal antibody, mU1, to tPA-deficient mice caused a dose-dependent delay of skin wound closure almost similar to the delayed kinetics observed in uPA;tPA double-deficient mice. Analysis of wound extracts showed diminished levels of plasmin in the mU1-treated tPA-deficient mice. Immunohistochemistry revealed that fibrin accumulated in the wounds of such mU1-treated tPA-deficient mice and that keratinocyte tongues were aberrant. Together these abnormalities lead to compromised epidermal closure. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that inhibition of uPA activity with a monoclonal antibody in adult tPA-deficient mice mimics the effect of simultaneous genetic ablation of uPA and tPA. Thus, application of the murine inhibitory mU1 antibody provides a new and highly versatile tool to interfere with uPA-activity in vivo in mouse models of disease.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Fibrinolisina/metabolismo , Piel/fisiopatología , Activador de Tejido Plasminógeno/deficiencia , Activador de Plasminógeno de Tipo Uroquinasa/inmunología , Cicatrización de Heridas/efectos de los fármacos , Heridas y Lesiones/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Piel/efectos de los fármacos , Piel/lesiones , Activador de Tejido Plasminógeno/genética , Heridas y Lesiones/metabolismo , Heridas y Lesiones/fisiopatología
18.
J Invest Dermatol ; 128(8): 2092-101, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18337830

RESUMEN

Genetic ablation of plasminogen (Plg) and pharmacological inhibition of metalloproteinase activity by galardin delay skin wound healing in mice, whereas the combined inhibition of these two enzyme systems completely prevents healing. In this study, the impact of plasmin and metalloproteinases as profibrinolytic enzymes has been investigated by comparing skin wound healing in the absence and presence of fibrin. Plg deficiency impairs skin wound healing kinetics, but this delay is only partially restored in the absence of fibrin. This suggests that plasmin-mediated fibrinolysis is the primary, but not the exclusive, requirement for healing of wounds in these mice. In addition, we observe that lack of fibrin reduces Plg activation significantly during wound healing. The profibrinolytic role of metalloproteinases is revealed by the finding that lack of fibrin partially restores the otherwise arrested healing of Plg-deficient wounds after metalloproteinase inhibition. In conclusion, the residual impairment of skin wound healing in the absence of fibrin suggests the existence of a fibrin-independent substrate(s) for plasmin and metalloproteinases. Furthermore, these in vivo data reveal that galardin-sensitive metalloproteinases mediate compensatory fibrinolysis to facilitate wound healing in the absence of plasmin.


Asunto(s)
Fibrinólisis/fisiología , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Plasminógeno/metabolismo , Piel/metabolismo , Cicatrización de Heridas/fisiología , Animales , Dipéptidos/farmacología , Fibrina/metabolismo , Fibrinógeno/genética , Fibrinógeno/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasminógeno/genética , Inhibidores de Proteasas/farmacología , Piel/patología
19.
J Biol Chem ; 283(47): 32506-15, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18799467

RESUMEN

Urokinase-type plasminogen activator (uPA) plays a central role in tissue remodeling processes. Most of our understanding of the role of uPA in vivo is derived from studies using gene-targeted uPA-deficient mice. To enable in vivo studies on the specific interference with uPA functionality in mouse models, we have now developed murine monoclonal antibodies (mAbs) directed against murine uPA by immunization of uPA-deficient mice with the recombinant protein. Guided by enzyme-linked immunosorbent assay, Western blotting, surface plasmon resonance, and enzyme kinetic analyses, we have selected two highly potent and inhibitory anti-uPA mAbs (mU1 and mU3). Both mAbs recognize epitopes located on the B-chain of uPA that encompasses the catalytic site. In enzyme activity assays in vitro, mU1 blocked uPA-catalyzed plasminogen activation as well as plasmin-mediated pro-uPA activation, whereas mU3 only was directed against the first of these reactions. We additionally provide evidence that mU1, but not mU3, successfully targets uPA-dependent processes in vivo. Hence, systemic administration of mU1 (i) rescued mice treated with a uPA-activable anthrax protoxin and (ii) impaired uPA-mediated hepatic fibrinolysis in tissue-type plasminogen activator (tPA)-deficient mice, resulting in a phenotype mimicking that of uPA;tPA double deficient mice. Importantly, this is the first report demonstrating specific antagonist-directed targeting of mouse uPA at the enzyme activity level in a normal physiological process in vivo.


Asunto(s)
Fibrinólisis , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Anticuerpos/química , Anticuerpos Monoclonales/química , Ensayo de Inmunoadsorción Enzimática , Femenino , Cinética , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Unión Proteica , Proteínas Recombinantes/química , Resonancia por Plasmón de Superficie
20.
Nat Methods ; 3(4): 259-61, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16554829

RESUMEN

We describe a simple, sensitive and noninvasive assay that uses nontoxic, reengineered anthrax toxin-beta-lactamase fusion proteins with altered protease cleavage specificity to visualize specific cell-surface proteolytic activity in single living cells. The assay could be used to specifically image endogenous cell-surface furin, urokinase plasminogen activator and metalloprotease activity. We have adapted the assay for fluorescence microscopy, flow cytometry and fluorescent plate reader formats, and it is amenable for automation and high-throughput analysis.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Bioensayo/métodos , Membrana Celular/metabolismo , Células Cultivadas/metabolismo , Automatización , Membrana Celular/ultraestructura , Células Cultivadas/ultraestructura , Citometría de Flujo , Furina/metabolismo , Metaloproteasas/metabolismo , Microscopía Fluorescente , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato , beta-Lactamasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA