Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Biol Chem ; 295(27): 9157-9170, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32424040

RESUMEN

C-type lectins that contain collagen-like domains are known as collectins. These proteins are present both in the circulation and in extravascular compartments and are central players of the innate immune system, contributing to first-line defenses against viral, bacterial, and fungal pathogens. The collectins mannose-binding lectin (MBL) and surfactant protein D (SP-D) are regulated by tissue fibroblasts at extravascular sites via an endocytic mechanism governed by urokinase plasminogen activator receptor-associated protein (uPARAP or Endo180), which is also a collagen receptor. Here, we investigated the molecular mechanisms that drive the uPARAP-mediated cellular uptake of MBL and SP-D. We found that the uptake depends on residues within a protruding loop in the fibronectin type-II (FNII) domain of uPARAP that are also critical for collagen uptake. Importantly, however, we also identified FNII domain residues having an exclusive role in collectin uptake. We noted that these residues are absent in the related collagen receptor, the mannose receptor (MR or CD206), which consistently does not interact with collectins. We also show that the second C-type lectin-like domain (CTLD2) is critical for the uptake of SP-D, but not MBL, indicating an additional level of complexity in the interactions between collectins and uPARAP. Finally, we demonstrate that the same molecular mechanisms enable uPARAP to engage MBL immobilized on the surface of pathogens, thereby expanding the potential biological implications of this interaction. Our study reveals molecular details of the receptor-mediated cellular regulation of collectins and offers critical clues for future investigations into collectin biology and pathology.


Asunto(s)
Colectinas/metabolismo , Endocitosis/fisiología , Receptores Mitogénicos/genética , Animales , Células CHO , Proteínas Portadoras/metabolismo , Colágeno/metabolismo , Cricetulus , Fibroblastos/metabolismo , Células HEK293 , Humanos , Lectinas Tipo C , Receptor de Manosa , Lectina de Unión a Manosa/metabolismo , Lectinas de Unión a Manosa , Glicoproteínas de Membrana/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Receptores de Superficie Celular , Receptores de Colágeno/metabolismo , Receptores Mitogénicos/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo
2.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202300

RESUMEN

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.


Asunto(s)
Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Resorción Ósea/etiología , Resorción Ósea/metabolismo , Comunicación Celular , Osteoclastos/metabolismo , Osteosarcoma/complicaciones , Osteosarcoma/patología , Animales , Neoplasias Óseas/diagnóstico por imagen , Remodelación Ósea , Resorción Ósea/diagnóstico , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Humanos , Osteogénesis
3.
Int J Mol Sci ; 22(21)2021 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-34768883

RESUMEN

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.


Asunto(s)
Lectinas de Unión a Manosa/metabolismo , Glicoproteínas de Membrana/metabolismo , Mesotelioma Maligno/metabolismo , Receptores de Superficie Celular/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Femenino , Expresión Génica , Humanos , Inmunoconjugados/metabolismo , Masculino , Lectinas de Unión a Manosa/fisiología , Glicoproteínas de Membrana/fisiología , Mesotelioma Maligno/diagnóstico , Mesotelioma Maligno/fisiopatología , Persona de Mediana Edad , Receptores de Superficie Celular/fisiología , Receptores de Colágeno/genética , Receptores de Colágeno/metabolismo , Receptores de Colágeno/fisiología , Receptores Mitogénicos/genética , Transcriptoma , Regulación hacia Arriba
4.
BMC Biol ; 15(1): 46, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28571576

RESUMEN

BACKGROUND: Matriptase is a membrane serine protease essential for epithelial development, homeostasis, and regeneration, as well as a central orchestrator of pathogenic pericellular signaling in the context of inflammatory and proliferative diseases. Matriptase is an unusual protease in that its zymogen displays measurable enzymatic activity. RESULTS: Here, we used gain and loss of function genetics to investigate the possible biological functions of zymogen matriptase. Unexpectedly, transgenic mice mis-expressing a zymogen-locked version of matriptase in the epidermis displayed pathologies previously reported for transgenic mice mis-expressing wildtype epidermal matriptase. Equally surprising, mice engineered to express only zymogen-locked endogenous matriptase, unlike matriptase null mice, were viable, developed epithelial barrier function, and regenerated the injured epithelium. Compatible with these observations, wildtype and zymogen-locked matriptase were equipotent activators of PAR-2 inflammatory signaling. CONCLUSION: The study demonstrates that the matriptase zymogen is biologically active and is capable of executing developmental and homeostatic functions of the protease.


Asunto(s)
Precursores Enzimáticos/metabolismo , Epitelio/crecimiento & desarrollo , Homeostasis/genética , Regeneración/genética , Serina Endopeptidasas/metabolismo , Animales , Precursores Enzimáticos/genética , Epitelio/metabolismo , Femenino , Mutación con Ganancia de Función , Expresión Génica , Mutación con Pérdida de Función , Masculino , Ratones Transgénicos , Serina Endopeptidasas/genética
5.
J Immunol ; 191(3): 1324-33, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23817411

RESUMEN

The long pentraxin 3 (PTX3) has been shown to be important in maintaining internal tissue homeostasis and in protecting against fungal Aspergillus fumigatus infection. However, the molecular mechanisms of how these functions are elicited are poorly delineated. Ficolin-1 is a soluble pattern recognition molecule that interacts with PTX3. We hypothesized that heterocomplexes between ficolin-1 and PTX3 might mediate the signals necessary for sequestration of altered self-cells and A. fumigatus. We were able to show that ficolin-1 interacts with PTX3 via its fibrinogen-like domain. The interaction was affected in a pH- and divalent cation-sensitive manner. The primary binding site for ficolin-1 on PTX3 was located in the N-terminal domain portion of PTX3. Ficolin-1 and PTX3 heterocomplex formation occurred on dying host cells, but not on A. fumigatus. The heterocomplex formation was a prerequisite for enhancement of phagocytosis by human monocyte-derived macrophages and downregulation of IL-8 production during phagocytosis. On A. fumigatus, PTX3 exposed the C-terminal portion of the molecule, probably resulting in steric hindrance of ficolin-1 interaction with PTX3. These results demonstrate that ficolin-1 and PTX3 heterocomplex formation acts as a noninflammatory "find me and eat me" signal to sequester altered-host cells. The fact that the ficolin-1-PTX3 complex formation did not occur on A. fumigatus shows that PTX3 uses different molecular effector mechanisms, depending on which domains it exposes during ligand interaction.


Asunto(s)
Apoptosis , Proteína C-Reactiva/metabolismo , Interleucina-8/metabolismo , Lectinas/metabolismo , Leucocitos Mononucleares/inmunología , Componente Amiloide P Sérico/metabolismo , Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Sitios de Unión , Proteína C-Reactiva/química , Células Cultivadas , Humanos , Interleucina-8/biosíntesis , Macrófagos/inmunología , Fagocitosis , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Componente Amiloide P Sérico/química , Transducción de Señal , Resonancia por Plasmón de Superficie , Ficolinas
6.
Cell Rep ; 21(13): 3662-3671, 2017 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-29281816

RESUMEN

Physiologic turnover of interstitial collagen is mediated by a sequential pathway in which collagen is fragmented by pericellular collagenases, endocytosed by collagen receptors, and routed to lysosomes for degradation by cathepsins. Here, we use intravital microscopy to investigate if malignant tumors, which are characterized by high rates of extracellular matrix turnover, utilize a similar collagen degradation pathway. Tumors of epithelial, mesenchymal, or neural crest origin all display vigorous endocytic collagen degradation. The cells engaged in this process are identified as tumor-associated macrophage (TAM)-like cells that degrade collagen in a mannose receptor-dependent manner. Accordingly, mannose-receptor-deficient mice display increased intratumoral collagen. Whole-transcriptome profiling uncovers a distinct extracellular matrix-catabolic signature of these collagen-degrading TAMs. Lineage-ablation studies reveal that collagen-degrading TAMs originate from circulating CCR2+ monocytes. This study identifies a function of TAMs in altering the tumor microenvironment through endocytic collagen turnover and establishes macrophages as centrally engaged in tumor-associated collagen degradation.


Asunto(s)
Movimiento Celular , Colágeno/metabolismo , Endocitosis , Inflamación/patología , Macrófagos/patología , Monocitos/patología , Neoplasias/patología , Proteolisis , Animales , Polaridad Celular , Matriz Extracelular/metabolismo , Lectinas Tipo C , Macrófagos/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa , Ratones Endogámicos C57BL , Neoplasias/genética , Ratas , Receptores CCR2/metabolismo , Receptores de Superficie Celular , Transcriptoma/genética
7.
Oncotarget ; 8(27): 44605-44624, 2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28574834

RESUMEN

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.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Inmunoconjugados/farmacología , Lectinas de Unión a Manosa/antagonistas & inhibidores , Glicoproteínas de Membrana/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores Mitogénicos/antagonistas & inhibidores , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Supervivencia Celular , Modelos Animales de Enfermedad , Endocitosis , Expresión Génica , Humanos , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Leucemia/mortalidad , Leucemia/patología , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Terapia Molecular Dirigida , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Mitogénicos/genética , Receptores Mitogénicos/metabolismo , Sarcoma/tratamiento farmacológico , Sarcoma/metabolismo , Sarcoma/mortalidad , Sarcoma/patología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Cell Biol ; 202(6): 951-66, 2013 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-24019537

RESUMEN

Tissue remodeling processes critically depend on the timely removal and remodeling of preexisting collagen scaffolds. Nevertheless, many aspects related to the turnover of this abundant extracellular matrix component in vivo are still incompletely understood. We therefore took advantage of recent advances in optical imaging to develop an assay to visualize collagen turnover in situ and identify cell types and molecules involved in this process. Collagen introduced into the dermis of mice underwent cellular endocytosis in a partially matrix metalloproteinase-dependent manner and was subsequently routed to lysosomes for complete degradation. Collagen uptake was predominantly executed by a quantitatively minor population of M2-like macrophages, whereas more abundant Col1a1-expressing fibroblasts and Cx3cr1-expressing macrophages internalized collagen at lower levels. Genetic ablation of the collagen receptors mannose receptor (Mrc1) and urokinase plasminogen activator receptor-associated protein (Endo180 and Mrc2) impaired this intracellular collagen degradation pathway. This study demonstrates the importance of receptor-mediated cellular uptake to collagen turnover in vivo and identifies a key role of M2-like macrophages in this process.


Asunto(s)
Colágeno Tipo I/fisiología , Colágeno/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/fisiología , Receptores de Superficie Celular/fisiología , Receptores de Quimiocina/fisiología , Animales , Apoptosis , Western Blotting , Receptor 1 de Quimiocinas CX3C , Proliferación Celular , Células Cultivadas , Cadena alfa 1 del Colágeno Tipo I , Endocitosis/fisiología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Técnicas para Inmunoenzimas , Lisosomas/metabolismo , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Inmunológicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA