Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Biol Chem ; 300(5): 107284, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38614208

RESUMO

Receptor-mediated cellular uptake of specific ligands constitutes an important step in the dynamic regulation of individual protein levels in extracellular fluids. With a focus on the inflammatory lung, we here performed a proteomics-based search for novel ligands regulated by the mannose receptor (MR), a macrophage-expressed endocytic receptor. WT and MR-deficient mice were exposed to lipopolysaccharide, after which the protein content in their lung epithelial lining fluid was compared by tandem mass tag-based mass spectrometry. More than 1200 proteins were identified in the epithelial lining fluid using this unbiased approach, but only six showed a statistically different abundance. Among these, an unexpected potential new ligand, thrombospondin-4 (TSP-4), displayed a striking 17-fold increased abundance in the MR-deficient mice. Experiments using exogenous addition of TSP-4 to MR-transfected CHO cells or MR-positive alveolar macrophages confirmed that TSP-4 is a ligand for MR-dependent endocytosis. Similar studies revealed that the molecular interaction with TSP-4 depends on both the lectin activity and the fibronectin type-II domain of MR and that a closely related member of the TSP family, TSP-5, is also efficiently internalized by the receptor. This was unlike the other members of this protein family, including TSPs -1 and -2, which are ligands for a close MR homologue known as urokinase plasminogen activator receptor-associated protein. Our study shows that MR takes part in the regulation of TSP-4, an important inflammatory component in the injured lung, and that two closely related endocytic receptors, expressed on different cell types, undertake the selective endocytosis of distinct members of the TSP family.


Assuntos
Lectinas Tipo C , Lesão Pulmonar , Receptor de Manose , Lectinas de Ligação a Manose , Proteômica , Receptores de Superfície Celular , Trombospondinas , Animais , Camundongos , Células CHO , Cricetulus , Endocitose , Lectinas Tipo C/metabolismo , Lectinas Tipo C/genética , Ligantes , Lipopolissacarídeos/toxicidade , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Lectinas de Ligação a Manose/metabolismo , Lectinas de Ligação a Manose/genética , Camundongos Knockout , Proteômica/métodos , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/genética , Trombospondinas/metabolismo , Trombospondinas/genética
2.
Matrix Biol ; 111: 307-328, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35878760

RESUMO

Thrombospondin-1 (TSP-1) is a matricellular protein with a multitude of functions in the pericellular and extracellular environment. We report a novel pathway for the regulation of extracellular TSP-1, governed by the endocytic collagen receptor, uPARAP (urokinase plasminogen activator receptor-associated protein; MRC2 gene product, also designated Endo180, CD280). First, using a novel proteomic approach for unbiased identification of ligands for endocytosis, we identify TSP-1 as a candidate ligand for specific uptake by uPARAP. We then show that uPARAP can efficiently internalize TSP-1 for lysosomal degradation, that this capability is not shared by other, closely related endocytic receptors and that uPARAP serves to regulate the extracellular levels of TSP-1 in vitro. Using wild type and uPARAP null mice, we also demonstrate uPARAP-mediated endocytosis of TSP-1 in dermal fibroblasts in vivo. Unlike other uPARAP ligands, the interaction with TSP-1 is sensitive to heparin and the responsible molecular motifs in uPARAP are overlapping, but not identical with those governing the interaction with collagens. Finally, we show that uPARAP can also mediate the endocytosis of TSP-2, a thrombospondin closely related to TSP-1, but not the more distantly related members of the same protein family, TSP-3, -4 and -5. These findings indicate that the role of uPARAP in ECM remodeling is not limited to the uptake of collagen for degradation but also includes an orchestrator function in the regulation of thrombospondins with numerous downstream effects. This is likely to be an important factor in the physiological and pathological roles of uPARAP in bone biology, fibrosis and cancer. The proteomic data has been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD031272.


Assuntos
Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Trombospondina 1/metabolismo , Animais , Colágeno/metabolismo , Endocitose , Ligantes , Camundongos , Camundongos Knockout , Proteômica , Trombospondina 1/genética
3.
Int J Mol Sci ; 22(21)2021 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-34768883

RESUMO

Malignant mesothelioma (MM) is a highly aggressive cancer with limited therapeutic options. We have previously shown that the endocytic collagen receptor, uPARAP, is upregulated in certain cancers and can be therapeutically targeted. Public RNA expression data display uPARAP overexpression in MM. Thus, to evaluate its potential use in diagnostics and therapy, we quantified uPARAP expression by immunohistochemical H-score in formalin-fixed paraffin-embedded bioptic/surgical human tissue samples and tissue microarrays. We detected pronounced upregulation of uPARAP in the three main MM subtypes compared to non-malignant reactive mesothelial proliferations, with higher expression in sarcomatoid and biphasic than in epithelioid MM. The upregulation appeared to be independent of patients' asbestos exposure and unaffected after chemotherapy. Using immunoblotting, we demonstrated high expression of uPARAP in MM cell lines and no expression in a non-malignant mesothelial cell line. Moreover, we showed the specific internalization of an anti-uPARAP monoclonal antibody by the MM cell lines using flow cytometry-based assays and confocal microscopy. Finally, we demonstrated the sensitivity of these cells towards sub-nanomolar concentrations of an antibody-drug conjugate formed with the uPARAP-directed antibody and a potent cytotoxin that led to efficient, uPARAP-specific eradication of the MM cells. Further studies on patient cohorts and functional preclinical models will fully reveal whether uPARAP could be exploited in diagnostics and therapeutic targeting of MM.


Assuntos
Lectinas de Ligação a Manose/metabolismo , Glicoproteínas de Membrana/metabolismo , Mesotelioma Maligno/metabolismo , Receptores de Superfície Celular/metabolismo , Adulto , Idoso , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Humanos , Imunoconjugados/metabolismo , Masculino , Lectinas de Ligação a Manose/fisiologia , Glicoproteínas de Membrana/fisiologia , Mesotelioma Maligno/diagnóstico , Mesotelioma Maligno/fisiopatologia , Pessoa de Meia-Idade , Receptores de Superfície Celular/fisiologia , Receptores de Colágeno/genética , Receptores de Colágeno/metabolismo , Receptores de Colágeno/fisiologia , Receptores Mitogênicos/genética , Transcriptoma , Regulação para Cima
4.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34202300

RESUMO

Cancer-induced bone degradation is part of the pathological process associated with both primary bone cancers, such as osteosarcoma, and bone metastases originating from, e.g., breast, prostate, and colon carcinomas. Typically, this includes a cancer-dependent hijacking of processes also occurring during physiological bone remodeling, including osteoclast-mediated disruption of the inorganic bone component and collagenolysis. Extensive research has revealed the significance of osteoclast-mediated bone resorption throughout the course of disease for both primary and secondary bone cancer. Nevertheless, cancer cells representing both primary bone cancer and bone metastasis have also been implicated directly in bone degradation. We will present and discuss observations on the contribution of osteoclasts and cancer cells in cancer-associated bone degradation and reciprocal modulatory actions between these cells. The focus of this review is osteosarcoma, but we will also include relevant observations from studies of bone metastasis. Additionally, we propose a model for cancer-associated bone degradation that involves a collaboration between osteoclasts and cancer cells and in which both cell types may directly participate in the degradation process.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias Ósseas/secundário , Reabsorção Óssea/etiologia , Reabsorção Óssea/metabolismo , Comunicação Celular , Osteoclastos/metabolismo , Osteossarcoma/complicações , Osteossarcoma/patologia , Animais , Neoplasias Ósseas/diagnóstico por imagem , Remodelação Óssea , Reabsorção Óssea/diagnóstico , Progressão da Doença , Suscetibilidade a Doenças , Humanos , Osteogênese
5.
Sci Rep ; 10(1): 19138, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33154487

RESUMO

The membrane-anchored matrix metalloprotease MT1-MMP is a potent collagenolytic enzyme with a well-established role in extracellular matrix turnover and cellular invasion into collagen-rich tissues. MT1-MMP is highly expressed in various types of cancer and has been demonstrated to be directly involved in several stages of tumor progression, including primary tumor growth, angiogenesis, invasion and metastasis. Osteosarcoma is the most common type of primary bone cancer. This disease is characterized by invasive tumor growth, leading to extensive bone destruction, and metastasis to the lungs. The tumor cells in human osteosarcoma display a strong expression of MT1-MMP, but the role of MT1-MMP in osteosarcoma progression is currently unknown. In this study, we investigated the role of MT1-MMP during various stages of osteosarcoma development. We utilized an optimized orthotopic murine osteosarcoma model and human osteosarcoma cells in which the MT1-MMP gene was knocked out using CRISPR/Cas9. We observed a strong expression of MT1-MMP in wildtype cells of both primary tumors and lung metastases, but, surprisingly, MT1-MMP deficiency did not affect primary tumor growth, bone degradation or the formation and growth of lung metastases. We therefore propose that, unlike findings reported in other cancers, tumor-expressed MT1-MMP is dispensable for all stages of osteosarcoma progression.


Assuntos
Neoplasias Ósseas/genética , Osso e Ossos/patologia , Proliferação de Células/genética , Neoplasias Pulmonares/genética , Metaloproteinase 14 da Matriz/genética , Osteossarcoma/genética , Animais , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Osso e Ossos/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Progressão da Doença , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Osteossarcoma/metabolismo , Osteossarcoma/secundário
6.
Acta Crystallogr F Struct Biol Commun ; 73(Pt 8): 486-490, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28777093

RESUMO

C4.4A is a glycosylphosphatidylinositol-anchored membrane protein comprised of two LU domains (Ly6/uPAR-like domains) and an extensively O-glycosylated C-terminal Ser/Thr/Pro-rich region. C4.4A is a novel biomarker for squamous epithelial differentiation. Its expression is dysregulated under various pathological conditions and it is a robust biomarker for poor prognosis in various malignant conditions such as pulmonary adenocarcinoma. To facilitate crystallization, the two LU domains were excised from intact C4.4A by limited proteolysis, purified and crystallized by the sitting-drop vapour-diffusion method. The crystals diffracted to 2.7 Šresolution and belonged to space group C2221, with unit-cell parameters a = 55.49, b = 119.63, c = 168.54 Å. The statistics indicated good quality of the data, which form a solid basis for the determination of the C4.4A structure.


Assuntos
Sequência de Aminoácidos , Biomarcadores Tumorais/química , Moléculas de Adesão Celular/química , Deleção de Sequência , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Clonagem Molecular , Cristalização , Cristalografia por Raios X , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Domínios Proteicos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Difração de Raios X
7.
Oncotarget ; 8(27): 44605-44624, 2017 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-28574834

RESUMO

A key task in developing the field of personalized cancer therapy is the identification of novel molecular targets that enable treatment of cancers not susceptible to other means of specific therapy. The collagen receptor uPARAP/Endo180 is overexpressed by malignant cells in several non-epithelial cancers, notably including sarcomas, glioblastomas and subsets of acute myeloid leukemia. In contrast, in healthy adult individuals, expression is restricted to minor subsets of mesenchymal cells. Functionally, uPARAP/Endo180 is a rapidly recycling endocytic receptor that delivers its cargo directly into the endosomal-lysosomal system, thus opening a potential route of entry into receptor-positive cells. This combination of specific expression and endocytic function appears well suited for targeting of uPARAP/Endo180-positive cancers by antibody-drug conjugate (ADC) mediated drug delivery. Therefore, we utilized a specific monoclonal antibody against uPARAP/Endo180, raised through immunization of a uPARAP/Endo180 knock-out mouse, which reacts with both the human and the murine receptor, to construct a uPARAP-directed ADC. This antibody was coupled to the highly toxic dolastatin derivative, monomethyl auristatin E, via a cathepsin-labile valine-citrulline linker. With this ADC, we show strong and receptor-dependent cytotoxicity in vitro in uPARAP/Endo180-positive cancer cell lines of sarcoma, glioblastoma and leukemic origin. Furthermore, we demonstrate the potency of the ADC in vivo in a xenograft mouse model with human uPARAP/Endo180-positive leukemic cells, obtaining a complete cure of all tested mice following intravenous ADC treatment with no sign of adverse effects. Our study identifies uPARAP/Endo180 as a promising target for novel therapy against several highly malignant cancer types.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Imunoconjugados/farmacologia , Lectinas de Ligação a Manose/antagonistas & inibidores , Glicoproteínas de Membrana/antagonistas & inibidores , Receptores de Superfície Celular/antagonistas & inibidores , Receptores Mitogênicos/antagonistas & inibidores , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Sobrevivência Celular , Modelos Animais de Doenças , Endocitose , Expressão Gênica , Humanos , Leucemia/tratamento farmacológico , Leucemia/metabolismo , Leucemia/mortalidade , Leucemia/patologia , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Terapia de Alvo Molecular , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Mitogênicos/genética , Receptores Mitogênicos/metabolismo , Sarcoma/tratamento farmacológico , Sarcoma/metabolismo , Sarcoma/mortalidade , Sarcoma/patologia , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Mol Carcinog ; 55(5): 717-31, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-25809119

RESUMO

The urokinase plasminogen activator system plays a key role in tissue degradation during cancer invasion. The linker region between domains I and II of the intact, three domain urokinase receptor uPAR(I-III) is highly susceptible to proteolytic cleavage and the resulting cleaved uPAR forms are strong prognostic biomarkers in several types of cancer, i.e., high levels of the cleaved uPAR forms indicate poor survival. To better understand the role of uPAR cleavage in cancer, we have designed immunoassays for specific quantification of intact mouse uPAR [muPAR(I-III)] and mouse uPAR domain I [muPAR(I)]. The level of muPAR(I) is significantly increased in mammary tumor-bearing mice compared to controls and, notably, there is a strong correlation to tumor volume. In contrast, the tumor volume is only weakly correlated to the level of intact muPAR(I-III), indicating that cleavage of muPAR is a more specific marker for cancer than increased expression of muPAR per se. The levels of the muPAR forms are dramatically affected by in vivo challenge with a urokinase -blocking antibody, demonstrating a functional role of uPA in uPAR cleavage. The levels of the muPAR forms are, however, unaffected by uPA-deficiency, suggesting that redundant proteases maintains the task of cleaving uPAR(I-III) when uPA is absent. Our findings emphasize the significance of the cleaved uPAR forms as cancer biomarkers. The strong correlation between muPAR(I) and the tumor volume in our experimental setup may motivate investigations of human uPAR(I) as biomarker for response to oncological treatment.


Assuntos
Neoplasias Mamárias Experimentais/patologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Feminino , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Transgênicos , Carga Tumoral , Ativador de Plasminogênio Tipo Uroquinase/química
9.
Data Brief ; 5: 107-13, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26504891

RESUMO

The urokinase-type plasminogen activator receptor (uPAR or CD87) is a glycolipid-anchored membrane protein often expressed in the microenvironment of invasive solid cancers and high levels are generally associated with poor patient prognosis (Kriegbaum et al., 2011 [1]). uPAR is organized as a dynamic modular protein structure composed of three homologous Ly6/uPAR domains (LU).This internally flexible protein structure of uPAR enables an allosteric regulation of the interactions with its two principal ligands: the serine protease urokinase-type plasminogen activator (uPA) and the provisional matrix protein vitronectin (Vn) (Mertens et al., 2012; Gårdsvoll et al., 2011; Madsen et al., 2007 [2-4]). The data presented here relates to the non-covalent trapping of one of these biologically relevant uPAR-conformations by a novel class of monoclonal antibodies (Zhao et al., 2015 [5]) and to the general mapping of the topographic epitope landscape on uPAR. The methods required to achieve these data include: (1) recombinant expression and purification of a uPAR-hybrid protein trapped in the desired conformation [patent; WO 2013/020898 A12013]; (2) developing monoclonal antibodies with unique specificities using this protein as antigen; (3) mapping the functional epitope on uPAR for these mAbs by surface plasmon resonance with a complete library of purified single-site uPAR mutants (Zhao et al., 2015; Gårdsvoll et al., 2006 [5,6]); and finally (4) solving the three-dimensional structures for one of these mAbs by X-ray crystallography alone and in complex with uPAR [deposited in the PDB database as 4QTH and 4QTI, respectively].

10.
Circ Res ; 116(4): 624-32, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25387803

RESUMO

RATIONALE: GPIHBP1, a GPI-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) in the subendothelial spaces and shuttles it to the capillary lumen. GPIHBP1 missense mutations that interfere with LPL binding cause familial chylomicronemia. OBJECTIVE: We sought to understand mechanisms by which GPIHBP1 mutations prevent LPL binding and lead to chylomicronemia. METHODS AND RESULTS: We expressed mutant forms of GPIHBP1 in Chinese hamster ovary cells, rat and human endothelial cells, and Drosophila S2 cells. In each expression system, mutation of cysteines in GPIHBP1's Ly6 domain (including mutants identified in patients with chylomicronemia) led to the formation of disulfide-linked dimers and multimers. GPIHBP1 dimerization/multimerization was not unique to cysteine mutations; mutations in other amino acid residues, including several associated with chylomicronemia, also led to protein dimerization/multimerization. The loss of GPIHBP1 monomers is relevant to the pathogenesis of chylomicronemia because only GPIHBP1 monomers-and not dimers or multimers-are capable of binding LPL. One GPIHBP1 mutant, GPIHBP1-W109S, had distinctive properties. GPIHBP1-W109S lacked the ability to bind LPL but had a reduced propensity for forming dimers or multimers, suggesting that W109 might play a more direct role in binding LPL. In support of that idea, replacing W109 with any of 8 other amino acids abolished LPL binding-and often did so without promoting the formation of dimers and multimers. CONCLUSIONS: Many amino acid substitutions in GPIHBP1's Ly6 domain that abolish LPL binding lead to protein dimerization/multimerization. Dimerization/multimerization is relevant to disease pathogenesis, given that only GPIHBP1 monomers are capable of binding LPL.


Assuntos
Hiperlipoproteinemia Tipo I/enzimologia , Lipase Lipoproteica/metabolismo , Mutação de Sentido Incorreto , Receptores de Lipoproteínas/metabolismo , Animais , Sítios de Ligação , Células CHO , Cricetulus , Cisteína , Drosophila/citologia , Drosophila/metabolismo , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Hiperlipoproteinemia Tipo I/genética , Modelos Moleculares , Fosfoinositídeo Fosfolipase C/metabolismo , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Ratos , Receptores de Lipoproteínas/química , Receptores de Lipoproteínas/genética , Transfecção
11.
J Biol Chem ; 289(28): 19491-9, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24847059

RESUMO

GPIHBP1, a glycosylphosphatidylinositol-anchored glycoprotein of microvascular endothelial cells, binds lipoprotein lipase (LPL) within the interstitial spaces and transports it across endothelial cells to the capillary lumen. The ability of GPIHBP1 to bind LPL depends on the Ly6 domain, a three-fingered structure containing 10 cysteines and a conserved pattern of disulfide bond formation. Here, we report a patient with severe hypertriglyceridemia who was homozygous for a GPIHBP1 point mutation that converted a serine in the GPIHBP1 Ly6 domain (Ser-107) to a cysteine. Two hypertriglyceridemic siblings were homozygous for the same mutation. All three homozygotes had very low levels of LPL in the preheparin plasma. We suspected that the extra cysteine in GPIHBP1-S107C might prevent the trafficking of the protein to the cell surface, but this was not the case. However, nearly all of the GPIHBP1-S107C on the cell surface was in the form of disulfide-linked dimers and multimers, whereas wild-type GPIHBP1 was predominantly monomeric. An insect cell GPIHBP1 expression system confirmed the propensity of GPIHBP1-S107C to form disulfide-linked dimers and to form multimers. Functional studies showed that only GPIHBP1 monomers bind LPL. In keeping with that finding, there was no binding of LPL to GPIHBP1-S107C in either cell-based or cell-free binding assays. We conclude that an extra cysteine in the GPIHBP1 Ly6 motif results in multimerization of GPIHBP1, defective LPL binding, and severe hypertriglyceridemia.


Assuntos
Homozigoto , Hiperlipoproteinemia Tipo I , Lipase Lipoproteica/metabolismo , Mutação de Sentido Incorreto , Multimerização Proteica/genética , Receptores de Lipoproteínas , Adulto , Substituição de Aminoácidos , Linhagem Celular , Humanos , Hiperlipoproteinemia Tipo I/genética , Hiperlipoproteinemia Tipo I/metabolismo , Hiperlipoproteinemia Tipo I/patologia , Lipase Lipoproteica/genética , Masculino , Ligação Proteica/genética , Estrutura Terciária de Proteína , Transporte Proteico/genética , Receptores de Lipoproteínas/genética , Receptores de Lipoproteínas/metabolismo
12.
J Histochem Cytochem ; 61(11): 802-13, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23896969

RESUMO

Several members of the Ly-6/uPAR (LU)-protein domain family are differentially expressed in human squamous epithelia. In some cases, they even play important roles in maintaining skin homeostasis, as exemplified by the secreted single domain member, SLURP-1, the deficiency of which is associated with the development of palmoplantar hyperkeratosis in the congenital skin disorder Mal de Meleda. In the present study, we have characterized a new member of the LU-protein domain family, which we find to be predominantly expressed in the stratum granulosum of human skin, thus resembling the expression of SLURP-1. In accordance with its expression pattern, we denote this protein product, which is encoded by the LYPD5 gene, as Haldisin (human antigen with LU-domains expressed in skin). Two of the five human glycolipid-anchored membrane proteins with multiple LU-domains characterized so far are predominantly confined to squamous epithelia (i.e., C4.4A), to stratum spinosum, and Haldisin to stratum granulosum under normal homeostatic conditions. Whether Haldisin is a prognostic biomarker for certain epithelial malignancies, like C4.4A and SLURP-1, remains to be explored.


Assuntos
Epiderme/metabolismo , Epiderme/patologia , Neoplasias de Células Escamosas/patologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/análise , Animais , Clonagem Molecular , Drosophila , Imunofluorescência , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Imuno-Histoquímica , Camundongos , Neoplasias de Células Escamosas/genética , Ratos , Ratos Sprague-Dawley , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética
13.
J Biol Chem ; 288(15): 10195-204, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23413031

RESUMO

The group of matrix metalloproteases (MMPs) is responsible for multiple processes of extracellular matrix remodeling in the healthy body but also for matrix and tissue destruction during cancer invasion and metastasis. The understanding of the contributions from each individual MMP, both in healthy and pathological events, has been complicated by the lack of specific inhibitors and the fact that some of the potent MMPs are multifunctional enzymes. These factors have also hampered the setup of therapeutic strategies targeting MMP activity. A tempting target is the membrane-associated MT1-MMP, which has well-documented importance in matrix degradation but which takes part in more than one pathway in this regard. In this report, we describe the selective targeting of a single function of this enzyme by means of a specific monoclonal antibody against MT1-MMP, raised in an MT1-MMP knock-out mouse. The antibody blocks the enzyme ability to activate proMMP-2 without interfering with the collagenolytic function or the general proteolytic activity of MT1-MMP. Using this antibody, we have shown that the MT1-MMP-catalyzed activation of proMMP-2 is involved in the outgrowth of cultured lymphatic endothelial cells in a collagen matrix in vitro, as well as in lymphatic vessel sprouting assayed ex vivo. This is the first example of the complete inactivation of a single function of a multifunctional MMP and the use of this strategy to pursue its role.


Assuntos
Matriz Extracelular/metabolismo , Linfangiogênese/fisiologia , Metaloproteinase 14 da Matriz/metabolismo , Animais , Anticorpos Monoclonais Murinos/química , Células CHO , Cricetinae , Ativação Enzimática/genética , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/patologia , Gelatinases/genética , Gelatinases/metabolismo , Humanos , Metaloproteinase 14 da Matriz/genética , Camundongos , Camundongos Knockout , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo
14.
Mol Cancer Res ; 10(12): 1532-43, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23038812

RESUMO

Data accumulated over the latest two decades have established that the serine protease urokinase-type plasminogen activator (uPA) is a potential therapeutic target in cancer. When designing inhibitors of the proteolytic activity of serine proteases, obtaining sufficient specificity is problematic, because the topology of the proteases' active sites are highly similar. In an effort to generate highly specific uPA inhibitors with new inhibitory modalities, we isolated uPA-binding RNA aptamers by screening a library of 35 nucleotides long 2'-fluoro-pyrimidine RNA molecules using a version of human pro-uPA lacking the epidermal growth factor-like and kringle domains as bait. One pro-uPA-binding aptamer sequence, referred to as upanap-126, proved to be highly specific for human uPA. Upanap-126 delayed the proteolytic conversion of human pro-uPA to active uPA, but did not inhibit plasminogen activation catalyzed by two-chain uPA. The aptamer also inhibited the binding of pro-uPA to uPAR and the binding of vitronectin to the preformed pro-uPA/uPAR complex, both in cell-free systems and on cell surfaces. Furthermore, upanap-126 inhibited human tumor cell invasion in vitro in the Matrigel assay and in vivo in the chick embryo assay of cell escape from microtumors. Finally, upanap-126 significantly reduced the levels of tumor cell intravasation and dissemination in the chick embryo model of spontaneous metastasis. Together, our findings show that usage of upanap-126 represents a novel multifunctional mechanistic modality for inhibition of uPA-dependent processes involved in tumor cell spread.


Assuntos
Aptâmeros de Nucleotídeos/genética , Aptâmeros de Nucleotídeos/farmacologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Linhagem Celular Tumoral , Sistema Livre de Células , Embrião de Galinha , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Células HEK293 , Humanos , Masculino , Terapia de Alvo Molecular , Invasividade Neoplásica , Plasminogênio/genética , Plasminogênio/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Proteases/genética , Serina Proteases/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
15.
J Biol Chem ; 287(41): 34304-15, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-22896701

RESUMO

The urokinase-type plasminogen activator receptor (uPAR) provides a rendezvous between proteolytic degradation of the extracellular matrix and integrin-mediated adhesion to vitronectin. These processes are, however, tightly linked because the high affinity binding of urokinase regulates the binding of uPAR to matrix-embedded vitronectin. Although crystal structures exist to define the corresponding static bi- and trimolecular receptor complexes, it is evident that the dynamic property of uPAR plays a decisive role in its function. In the present study, we combine small angle x-ray scattering, hydrogen-deuterium exchange, and surface plasmon resonance to develop a structural model describing the allosteric regulation of uPAR. We show that the flexibility of its N-terminal domain provides the key for understanding this allosteric mechanism. Importantly, our model has direct implications for understanding uPAR-assisted cell adhesion and migration as well as for translational research, including targeted intervention therapy and non-invasive tumor imaging in vivo.


Assuntos
Matriz Extracelular , Proteólise , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Vitronectina , Regulação Alostérica , Animais , Adesão Celular , Linhagem Celular , Movimento Celular , Medição da Troca de Deutério , Drosophila melanogaster , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Humanos , Invasividade Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Espalhamento de Radiação , Relação Estrutura-Atividade , Vitronectina/química , Vitronectina/metabolismo , Raios X
16.
J Biol Chem ; 286(38): 33544-56, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21799009

RESUMO

The urokinase-type plasminogen activator receptor (uPAR) is a glycolipid-anchored membrane protein with an established role in focalizing uPA-mediated plasminogen activation on cell surfaces. Distinct from this function, uPAR also modulates cell adhesion and migration on vitronectin-rich matrices. Although uPA and vitronectin engage structurally distinct binding sites on uPAR, they nonetheless cooperate functionally, as uPA binding potentiates uPAR-dependent induction of lamellipodia on vitronectin matrices. We now present data advancing the possibility that it is the burial of the ß-hairpin in uPA per se into the hydrophobic ligand binding cavity of uPAR that modulates the function of this receptor. Based on these data, we now propose a model in which the inherent interdomain mobility in uPAR plays a major role in modulating its function. Particularly one uPAR conformation, which is stabilized by engagement of the ß-hairpin in uPA, favors the proper assembly of an active, compact receptor structure that stimulates lamellipodia induction on vitronectin. This molecular model has wide implications for drug development targeting uPAR function.


Assuntos
Anticorpos Monoclonais/imunologia , Mapeamento de Epitopos , Pseudópodes/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Drosophila melanogaster , Humanos , Concentração Inibidora 50 , Cinética , Ligantes , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Peso Molecular , Peptídeos/química , Peptídeos/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Pseudópodes/efeitos dos fármacos , Relação Estrutura-Atividade , Transfecção , Ativador de Plasminogênio Tipo Uroquinase/química , Vitronectina/farmacologia
17.
J Biol Chem ; 286(37): 32736-48, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21768090

RESUMO

Collagens make up the most abundant component of interstitial extracellular matrices and basement membranes. Collagen remodeling is a crucial process in many normal physiological events and in several pathological conditions. Some collagen subtypes contain specific carbohydrate side chains, the function of which is poorly known. The endocytic collagen receptor urokinase plasminogen activator receptor-associated protein (uPARAP)/Endo180 plays an important role in matrix remodeling through its ability to internalize collagen for lysosomal degradation. uPARAP/Endo180 is a member of the mannose receptor protein family. These proteins all include a fibronectin type II domain and a series of C-type lectin-like domains, of which only a minor part possess carbohydrate recognition activity. At least two of the family members, uPARAP/Endo180 and the mannose receptor, interact with collagens. The molecular basis for this interaction is known to involve the fibronectin type II domain but nothing is known about the function of the lectin domains in this respect. In this study, we have investigated a possible role of the single active lectin domain of uPARAP/Endo180 in the interaction with collagens. By expressing truncated recombinant uPARAP/Endo180 proteins and analyzing their interaction with collagens with high and low levels of glycosylation we demonstrated that this lectin domain interacts directly with glycosylated collagens. This interaction is functionally important because it was found to modulate the endocytic efficiency of the receptor toward highly glycosylated collagens such as basement membrane collagen IV. Surprisingly, this property was not shared by the mannose receptor, which internalized glycosylated collagens independently of its lectin function. This role of modulating its uptake efficiency by a specific receptor is a previously unrecognized function of collagen glycosylation.


Assuntos
Colágeno Tipo IV/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Linhagem Celular Tumoral , Colágeno Tipo IV/química , Colágeno Tipo IV/genética , Matriz Extracelular/química , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Glicosilação , Humanos , Lectinas Tipo C/química , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/química , Lectinas de Ligação a Manose/genética , Lectinas de Ligação a Manose/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Mutantes , Estrutura Terciária de Proteína , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
Blood ; 116(9): 1593-603, 2010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20466854

RESUMO

The urokinase plasminogen activator receptor (uPAR) has emerged as a potential regulator of cell adhesion, cell migration, proliferation, differentiation, and cell survival in multiple physiologic and pathologic contexts. The urokinase plasminogen activator (uPA) was the first identified ligand for uPAR, but elucidation of the specific functions of the uPA-uPAR interaction in vivo has been difficult because uPA has important physiologic functions that are independent of binding to uPAR and because uPAR engages multiple ligands. Here, we developed a new mouse strain (Plau(GFDhu/GFDhu)) in which the interaction between endogenous uPA and uPAR is selectively abrogated, whereas other functions of both the protease and its receptor are retained. Specifically, we introduced 4 amino acid substitutions into the growth factor domain (GFD) of uPA that abrogate uPAR binding while preserving the overall structure of the domain. Analysis of Plau(GFDhu/GFDhu) mice revealed an unanticipated role of the uPA-uPAR interaction in suppressing inflammation secondary to fibrin deposition. In contrast, leukocyte recruitment and tissue regeneration were unaffected by the loss of uPA binding to uPAR. This study identifies a principal in vivo role of the uPA-uPAR interaction in cell-associated fibrinolysis critical for suppression of fibrin accumulation and fibrin-associated inflammation and provides a valuable model for further exploration of this multifunctional receptor.


Assuntos
Modelos Animais de Doenças , Fibrina/metabolismo , Inflamação/prevenção & controle , Pneumonia/prevenção & controle , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Apoptose , Western Blotting , Proliferação de Células , Feminino , Citometria de Fluxo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Inflamação/metabolismo , Inflamação/patologia , Fígado/imunologia , Fígado/lesões , Fígado/metabolismo , Lesão Pulmonar/imunologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/prevenção & controle , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/imunologia , Pneumonia/metabolismo , RNA Mensageiro/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Dermatopatias/imunologia , Dermatopatias/metabolismo , Dermatopatias/prevenção & controle , Taxa de Sobrevida , Ativador de Plasminogênio Tipo Uroquinase/genética , Cicatrização
19.
Protein Expr Purif ; 72(1): 87-94, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20214994

RESUMO

Matrix metalloproteinase-9 (MMP-9) is a 92-kDa soluble pro-enzyme implicated in pathological events including cancer invasion. It is therefore an attractive target for therapeutic intervention studies in mouse models. Development of inhibitors requires sufficient amounts of correctly folded murine MMP-9. Constructs encoding zymogens of full-length murine MMP-9 and a version lacking the O-glycosylated linker region and hemopexin domains were therefore generated and expressed in stably transfected Drosophila S2 insect cells. After 7 days of induction the expression levels of the full-length and truncated versions were 5 mg/l and 2 mg/l, respectively. The products were >95% pure after gelatin Sepharose chromatography and possessed proteolytic activity when analyzed by gelatin zymography. Using the purified full-length murine MMP-9 we raised polyclonal antibodies by immunizations of rabbits. These antibodies specifically identified pro-MMP-9 in incisional skin wound extracts from mice when used for Western blotting. Immunohistochemical analysis of paraffin embedded skin wounds from mice showed that MMP-9 protein was localized at the leading-edge keratinocytes in front of the migrating epidermal layer. No immunoreactivity was observed when the antibody was probed against skin wound material from MMP-9 deficient mice. In conclusion, we have generated and purified two proteolytically active recombinant murine MMP-9 protein constructs, which are critical reagents for future cancer drug discovery studies.


Assuntos
Drosophila/genética , Expressão Gênica , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/isolamento & purificação , Murinae/genética , Animais , Anticorpos/imunologia , Linhagem Celular , Cromatografia de Afinidade , Drosophila/citologia , Metaloproteinase 9 da Matriz/análise , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Estrutura Terciária de Proteína , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Pele/metabolismo
20.
J Biol Chem ; 285(14): 10982-92, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20133942

RESUMO

The high affinity interaction between the urokinase-type plasminogen activator (uPA) and its glycolipid-anchored receptor (uPAR) is decisive for cell surface-associated plasminogen activation. Because plasmin activity controls fibrinolysis in a variety of pathological conditions, including cancer and wound healing, several intervention studies have focused on targeting the uPA.uPAR interaction in vivo. Evaluations of such studies in xenotransplanted tumor models are, however, complicated by the pronounced species selectivity in this interaction. We now report the molecular basis underlying this difference by solving the crystal structure for the murine uPA.uPAR complex and demonstrate by extensive surface plasmon resonance studies that the kinetic rate constants for this interaction can be swapped completely between these orthologs by exchanging only two residues. This study not only discloses the structural basis required for a successful rational design of the species selectivity in the uPA.uPAR interaction, which is highly relevant for functional studies in mouse models, but it also suggests the possible development of general inhibitors that will target the uPA.uPAR interaction across species barriers.


Assuntos
Desenho de Fármacos , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Fator 1 Ativador da Transcrição/química , Fator 1 Ativador da Transcrição/genética , Fator 1 Ativador da Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Cristalização , Cristalografia por Raios X , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Neoplasias/tratamento farmacológico , Conformação Proteica , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Ressonância de Plasmônio de Superfície , Ativador de Plasminogênio Tipo Uroquinase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA