Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Toxins (Basel) ; 13(4)2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807363

RESUMEN

Snake envenomation can result in hemorrhage, local necrosis, swelling, and if not treated properly can lead to adverse systemic effects such as coagulopathy, nephrotoxicity, neurotoxicity, and cardiotoxicity, which can result in death. As such, snake venom metalloproteinases (SVMPs) and disintegrins are two toxic components that contribute to hemorrhage and interfere with the hemostatic system. Administration of a commercial antivenom is the common antidote to treat snake envenomation, but the high-cost, lack of efficacy, side effects, and limited availability, necessitates the development of new strategies and approaches for therapeutic treatments. Herein, we describe the neutralization ability of anti-disintegrin polyclonal antibody on the activities of isolated disintegrins, P-II/P-III SVMPs, and crude venoms. Our results show disintegrin activity on platelet aggregation in whole blood and the migration of the SK-Mel-28 cells that can be neutralized with anti-disintegrin polyclonal antibody. We characterized a SVMP and found that anti-disintegrin was also able to inhibit its activity in an in vitro proteolytic assay. Moreover, we found that anti-disintegrin could neutralize the proteolytic and hemorrhagic activities from crude Crotalus atrox venom. Our results suggest that anti-disintegrin polyclonal antibodies have the potential for a targeted approach to neutralize SVMPs in the treatment of snakebite envenomations.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Antivenenos/farmacología , Venenos de Crotálidos/antagonistas & inhibidores , Crotalus , Desintegrinas/antagonistas & inhibidores , Metaloproteasas/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Mordeduras de Serpientes/tratamiento farmacológico , Regulación Alostérica , Animales , Especificidad de Anticuerpos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Reacciones Cruzadas , Venenos de Crotálidos/enzimología , Venenos de Crotálidos/inmunología , Modelos Animales de Enfermedad , Desintegrinas/inmunología , Desintegrinas/metabolismo , Hemorragia/enzimología , Hemorragia/etiología , Hemorragia/prevención & control , Humanos , Metaloproteasas/inmunología , Metaloproteasas/metabolismo , Ratones Endogámicos BALB C , Agregación Plaquetaria/efectos de los fármacos , Mordeduras de Serpientes/sangre , Mordeduras de Serpientes/enzimología , Mordeduras de Serpientes/inmunología
2.
Methods Mol Biol ; 2068: 205-223, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31576530

RESUMEN

Snake venoms could lead to the development of new drugs to treat a range of life-threatening conditions like cardiovascular diseases. Most snake venoms contain a large variety of lethal toxins as well as anti-adhesive proteins such as disintegrins, which have evolved from the harmless compounds ADAMs (proteins with a disintegrin and a metalloprotease domain) and C-type lectin proteins which disturb connective tissue and cell-matrix interaction. These anti-adhesive proteins target and block integrin receptors and disrupt normal biological processes in snakes' prey such as connective tissue physiology and blood clotting. This chapter provides the experimental details of a practical, cell-based adhesion protocol to help identify and isolate disintegrins and C-type lectin proteins from snake venoms, important tools in integrin research and lead compounds for drug discovery.


Asunto(s)
Adhesión Celular/fisiología , Venenos de Serpiente/análisis , Animales , Bioensayo , Coagulación Sanguínea/efectos de los fármacos , Desintegrinas/antagonistas & inhibidores , Humanos , Lectinas/química , Lectinas/farmacología
3.
Biochem Pharmacol ; 164: 188-204, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30905657

RESUMEN

A Disintegrin and Metalloproteinase (ADAM) is a family of proteolytic enzymes that possess sheddase function and regulate shedding of membrane-bound proteins, growth factors, cytokines, ligands and receptors. Typically, ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and a characteristic transmembrane domain. Most ADAMs are activated by proprotein convertases, but can also be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C activators. A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) is a family of secreted enzymes closely related to ADAMs. Like ADAMs, ADAMTS members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but they lack a transmembrane domain and instead have characteristic thrombospondin motifs. Activated ADAMs perform several functions and participate in multiple cardiovascular processes including vascular smooth muscle cell proliferation and migration, angiogenesis, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs may also be involved in pathological conditions and cardiovascular diseases such as atherosclerosis, hypertension, aneurysm, coronary artery disease, myocardial infarction and heart failure. Like ADAMs, ADAMTS have a wide-spectrum role in vascular biology and cardiovascular pathophysiology. ADAMs and ADAMTS activity is naturally controlled by endogenous inhibitors such as tissue inhibitors of metalloproteinases (TIMPs), and their activity can also be suppressed by synthetic small molecule inhibitors. ADAMs and ADAMTS can serve as important diagnostic biomarkers and potential therapeutic targets for cardiovascular disorders. Natural and synthetic inhibitors of ADAMs and ADAMTS could be potential therapeutic tools for the management of cardiovascular diseases.


Asunto(s)
Proteínas ADAM/metabolismo , Desintegrinas/metabolismo , Endotelio Vascular/metabolismo , Trombospondinas/metabolismo , Enfermedades Vasculares/metabolismo , Proteínas ADAM/antagonistas & inhibidores , Secuencias de Aminoácidos/efectos de los fármacos , Secuencias de Aminoácidos/fisiología , Animales , Desintegrinas/antagonistas & inhibidores , Endotelio Vascular/efectos de los fármacos , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/administración & dosificación , Trombospondinas/antagonistas & inhibidores , Enfermedades Vasculares/tratamiento farmacológico
4.
PLoS One ; 13(4): e0195116, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29617412

RESUMEN

Over expression of Tissue Inhibitor of Metalloproteinases-3 (TIMP-3) in vascular smooth muscle cells (VSMCs) induces apoptosis and reduces neointima formation occurring after saphenous vein interposition grafting or coronary stenting. In studies to address the mechanism of TIMP-3-driven apoptosis in human VSMCs we find that TIMP-3 increased activation of caspase-8 and apoptosis was inhibited by expression of Cytokine response modifier A (CrmA) and dominant negative FAS-Associated protein with Death Domain (FADD). TIMP-3 induced apoptosis did not cause mitochondrial depolarisation, increase activation of caspase-9 and was not inhibited by over-expression of B-cell Lymphoma 2 (Bcl2), indicating a mitochondrial independent/type-I death receptor pathway. TIMP-3 increased levels of the First Apoptosis Signal receptor (FAS) and depletion of FAS with shRNA showed TIMP-3-induced apoptosis was FAS dependent. TIMP-3 induced formation of the Death-Inducing Signalling Complex (DISC), as detected by immunoprecipitation and by immunofluorescence. Cellular-FADD-like IL-1 converting enzyme-Like Inhibitory Protein (c-FLIP) localised with FAS at the cell periphery in the absence of TIMP-3 and this localisation was lost on TIMP-3 expression with c-FLIP adopting a perinuclear localisation. Although TIMP-3 inhibited FAS shedding, this did not increase total surface levels of FAS but instead increased FAS levels within localised regions at the cell surface. A Disintegrin And Metalloproteinase 17 (ADAM17) is inhibited by TIMP-3 and depletion of ADAM17 with shRNA significantly decreased FAS shedding. However ADAM17 depletion did not induce apoptosis or replicate the effects of TIMP-3 by increasing localised clustering of cell surface FAS. ADAM17-depleted cells could activate caspase-3 when expressing levels of TIMP-3 that were otherwise sub-apoptotic, suggesting a partial role for ADAM17 mediated ectodomain shedding in TIMP-3 mediated apoptosis. We conclude that TIMP-3 induced apoptosis in VSMCs is highly dependent on FAS and is associated with changes in FAS and c-FLIP localisation, but is not solely dependent on shedding of the FAS ectodomain.


Asunto(s)
Apoptosis , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Receptor fas/metabolismo , Proteína ADAM17/antagonistas & inhibidores , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Apoptosis/efectos de los fármacos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasas/metabolismo , Células Cultivadas , Desintegrinas/antagonistas & inhibidores , Desintegrinas/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microscopía Confocal , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Estaurosporina/farmacología , Inhibidor Tisular de Metaloproteinasa-3/genética , Receptor fas/antagonistas & inhibidores , Receptor fas/genética
5.
Inflamm Res ; 64(3-4): 243-52, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25693597

RESUMEN

OBJECTIVE: To investigate the inhibitory effect of hyaluronan (HA) on mechanical stress- induced expression of a disintegrin and metalloproteinase with thrombospondin type 1 motifs (ADAMTS)-4, -5 and matrix metalloproteinase (MMP)-13 by human chondrocytes. MATERIALS AND METHODS: Normal human articular chondrocytes were pre-incubated with or without 1.0 mg/mL HA (2700 kDa) for 12 h at 37 °C in stretch chambers, then they were exposed to uni-axial cyclic tensile strain (CTS, 0.5 Hz, 10% elongation). The expression of ADAMTS-4, -5, and MMP-13 were analyzed by real-time polymerase chain reaction and Immunocytochemistry. The concentration of IL-1ß in the supernatant was measured using enzyme-linked immunosorbent assay (ELISA). The nuclear translocation of runt-related transcription factor 2 (RUNX-2) and nuclear factor-κB (NF-κB) was examined by ELISA and immunocytochemistry, and phosphorylation of NF-κB was examined by western blotting. RESULTS: HA inhibited mRNA expression of ADAMTS-4, -5, and MMP13 after 24 h CTS via inhibition of IL-1ß secretion and NF-κB activation. However, HA failed to inhibit CTS-induced RUNX-2 expression and subsequent expression of ADAMTS-5 and MMP-13 1 h after CTS. CONCLUSIONS: Our results demonstrated that HA significantly suppressed mechanical stress-induced expression of catabolic proteases by inhibition of the NF-κB-IL-1ß pathway, but did not suppress mechanical stress-induced RUNX-2 signaling.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Condrocitos/efectos de los fármacos , Desintegrinas/antagonistas & inhibidores , Ácido Hialurónico/farmacología , Procolágeno N-Endopeptidasa/antagonistas & inhibidores , Estrés Mecánico , Proteínas ADAM/metabolismo , Proteína ADAMTS4 , Proteína ADAMTS5 , Adolescente , Adulto , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Desintegrinas/metabolismo , Humanos , Técnicas In Vitro , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , FN-kappa B/metabolismo , Procolágeno N-Endopeptidasa/metabolismo , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Biochem Biophys Res Commun ; 421(1): 70-5, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22480688

RESUMEN

Angiotensin-I converting enzyme (ACE) is a zinc dependent peptidase with a major role in regulating vasoactive peptide metabolism. ACE, a transmembrane protein, undergoes proteolysis, or shedding, by an as yet unidentified proteinase to release a catalytically active soluble form of the enzyme. Physiologically, soluble ACE in plasma is derived primarily from endothelial cells. We demonstrate that ACE shedding from confluent endothelial cells is increased in response to bacterial lipopolysaccharide, but not phorbol esters. Characterisation of lipopolysaccharide stimulated shedding showed that there is a lag phase before soluble ACE can be detected which is sensitive to inhibitors of translation, NF-κB, TNFα and TNFR-I/II. The shedding phase is less sensitive to these inhibitors, but is ablated by BB-94, a Matrix Metalloproteinase (MMP)/A Disintegrin and Metalloproteinase (ADAM) inhibitor. Tissue Inhibitor of Metalloproteinase (TIMP) profiling suggested a requirement for ADAM9 in lipopolysaccharide induced ACE shedding, which was confirmed by depletion with siRNA. Transient transfection of ADAM9 and ACE cDNAs into HEK293 cells demonstrated that ADAM9 requires both membrane anchorage and its catalytic domain to shed ACE.


Asunto(s)
Proteínas ADAM/metabolismo , Células Endoteliales/enzimología , Lipopolisacáridos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas ADAM/genética , Dominio Catalítico , Membrana Celular/enzimología , ADN Complementario , Desintegrinas/antagonistas & inhibidores , Células Endoteliales/efectos de los fármacos , Células HEK293 , Humanos , Lipopolisacáridos/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Proteínas de la Membrana/genética , Metaloproteasas/antagonistas & inhibidores , FN-kappa B/metabolismo , Peptidil-Dipeptidasa A , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Ésteres del Forbol/metabolismo , Ésteres del Forbol/farmacología , Inhibidores de Proteasas/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Tiofenos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
7.
PLoS One ; 5(6): e10929, 2010 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-20532165

RESUMEN

Similar to other integrin-targeting strategies, disintegrins have previously shown good efficacy in animal cancer models with favorable pharmacological attributes and translational potential. Nonetheless, these polypeptides are notoriously difficult to produce recombinantly due to their particular structure requiring the correct pairing of multiple disulfide bonds for biological activity. Here, we show that a sequence-engineered disintegrin (called vicrostatin or VCN) can be reliably produced in large scale amounts directly in the oxidative cytoplasm of Origami B E. coli. Through multiple integrin ligation (i.e., alphavbeta3, alphavbeta5, and alpha5beta1), VCN targets both endothelial and cancer cells significantly inhibiting their motility through a reconstituted basement membrane. Interestingly, in a manner distinct from other integrin ligands but reminiscent of some ECM-derived endogenous anti-angiogenic fragments previously described in the literature, VCN profoundly disrupts the actin cytoskeleton of endothelial cells (EC) inducing a rapid disassembly of stress fibers and actin reorganization, ultimately interfering with EC's ability to invade and form tubes (tubulogenesis). Moreover, here we show for the first time that the addition of a disintegrin to tubulogenic EC sandwiched in vitro between two Matrigel layers negatively impacts their survival despite the presence of abundant haptotactic cues. A liposomal formulation of VCN (LVCN) was further evaluated in vivo in two animal cancer models with different growth characteristics. Our data demonstrate that LVCN is well tolerated while exerting a significant delay in tumor growth and an increase in the survival of treated animals. These results can be partially explained by potent tumor anti-angiogenic and pro-apoptotic effects induced by LVCN.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Desintegrinas/antagonistas & inhibidores , Integrinas/antagonistas & inhibidores , Neovascularización Patológica/prevención & control , Línea Celular Tumoral , Desintegrinas/farmacología , Humanos , Fosforilación
8.
J Biol Chem ; 285(16): 11793-9, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20164177

RESUMEN

Nerve growth factor (NGF) plays an important role in regulating mammalian neuronal/embryonic development, angiogenesis, and other physiological processes and has recently been investigated as a potential treatment for the neurodegenerative disorder, Alzheimer disease. In this study, we provide evidence that human NGF may also function as a metalloproteinase inhibitor, based on studies of NGF from snake venom. Originally, our aim was to isolate snake venom metalloproteinases targeting platelet receptors and/or ligands relevant to hemostasis and thrombosis, using Ni(2+)-agarose as a purification step based on the conserved metal ion-coordination motif in venom metalloproteinases. However, subsequent analysis of cobra (Naja kaouthia) venom led to the unexpected discovery that cobra venom NGF bound to Ni(2+)-agarose, eluting at approximately 15 mm imidazole, enabling a one-step purification. The identity of the purified protein was confirmed by mass spectrometry and N-terminal sequence analysis. Partial co-purification of NGF within metalloproteinase-enriched venom fractions led us to test whether NGF affected metalloproteinase activity. Venom NGF potently inhibited metalloproteinases isolated from the same or different venom and specifically bound to purified Nk metalloproteinase immobilized on agarose beads. Human NGF also interacted with human metalloproteinases because it blocked metalloproteinase-mediated shedding of the platelet collagen receptor, glycoprotein (GP)VI, and associated with recombinant ADAM10 by surface plasmon resonance. Together, these results suggest that NGF can function as a metalloproteinase inhibitor.


Asunto(s)
Desintegrinas/antagonistas & inhibidores , Metaloproteasas/antagonistas & inhibidores , Factor de Crecimiento Nervioso/farmacología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Secuencia de Aminoácidos , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Cromatografía en Agarosa , Venenos Elapídicos/química , Venenos Elapídicos/genética , Venenos Elapídicos/farmacología , Humanos , Técnicas In Vitro , Datos de Secuencia Molecular , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/aislamiento & purificación , Níquel , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
9.
Toxins (Basel) ; 2(11): 2606-21, 2010 11.
Artículo en Inglés | MEDLINE | ID: mdl-22069567

RESUMEN

Cell migration is a key process for the defense of pluricellular organisms against pathogens, and it involves a set of surface receptors acting in an ordered fashion to contribute directionality to the movement. Among these receptors are the integrins, which connect the cell cytoskeleton to the extracellular matrix components, thus playing a central role in cell migration. Integrin clustering at focal adhesions drives actin polymerization along the cell leading edge, resulting in polarity of cell movement. Therefore, small integrin-binding proteins such as the snake venom disintegrins that inhibit integrin-mediated cell adhesion are expected to inhibit cell migration. Here we review the current knowledge on disintegrin and disintegrin-like protein effects on cell migration and their potential use as pharmacological tools in anti-inflammatory therapy as well as in inhibition of metastatic invasion.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Desintegrinas/toxicidad , Neutrófilos/efectos de los fármacos , Venenos de Serpiente/química , Animales , Anoicis/efectos de los fármacos , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Desintegrinas/antagonistas & inhibidores , Humanos , Metástasis de la Neoplasia/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Venenos de Serpiente/farmacología
10.
Cancer Res ; 68(17): 7083-9, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18757423

RESUMEN

The ErbB family of receptors is overexpressed in numerous human tumors. Overexpression correlates with poor prognosis and resistance to therapy. Use of ErbB-specific antibodies to the receptors (Herceptin or Erbitux) or ErbB-specific small-molecule inhibitors of the receptor tyrosine kinase activity (Iressa or Tarceva) has shown clinical efficacy in several solid tumors. An alternative method of affecting ErbB-initiated tumor growth and survival is to block sheddase activity. Sheddase activity is responsible for cleavage of multiple ErbB ligands and receptors, a necessary step in availability of the soluble, active form of the ligand and a constitutively activated ligand-independent receptor. This sheddase activity is attributed to the ADAM (a disintegrin and metalloprotease) family of proteins. ADAM 10 is the main sheddase of epidermal growth factor (EGF) and HER-2/neu cleavage, whereas ADAM17 is required for cleavage of additional EGF receptor (EGFR) ligands (transforming growth factor-alpha, amphiregulin, heregulin, heparin binding EGF-like ligand). This study has shown that addition of INCB3619, a potent inhibitor of ADAM10 and ADAM17, reduces in vitro HER-2/neu and amphiregulin shedding, confirming that it interferes with both HER-2/neu and EGFR ligand cleavage. Combining INCB3619 with a lapatinib-like dual inhibitor of EGFR and HER-2/neu kinases resulted in synergistic growth inhibition in MCF-7 and HER-2/neu-transfected MCF-7 human breast cancer cells. Combining the INCB7839 second-generation sheddase inhibitor with lapatinib prevented the growth of HER-2/neu-positive BT474-SC1 human breast cancer xenografts in vivo. These results suggest that there may be an additional clinical benefit of combining agents that target the ErbB pathways at multiple points.


Asunto(s)
Desintegrinas/antagonistas & inhibidores , Metaloproteasas/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Ratones , Piperidinas/farmacología , Transducción de Señal , Compuestos de Espiro/farmacología
11.
J Biol Chem ; 282(49): 35712-21, 2007 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-17895248

RESUMEN

ADAM10 is a disintegrin metalloproteinase that processes amyloid precursor protein and ErbB ligands and is involved in the shedding of many type I and type II single membrane-spanning proteins. Like tumor necrosis factor-alpha-converting enzyme (TACE or ADAM17), ADAM10 is expressed as a zymogen, and removal of the prodomain results in its activation. Here we report that the recombinant mouse ADAM10 prodomain, purified from Escherichia coli, is a potent competitive inhibitor of the human ADAM10 catalytic/disintegrin domain, with a K(i) of 48 nM. Moreover, the mouse ADAM10 prodomain is a selective inhibitor as it only weakly inhibits other ADAM family proteinases in the micromolar range and does not inhibit members of the matrix metalloproteinase family under similar conditions. Mouse prodomains of TACE and ADAM8 do not inhibit their respective enzymes, indicating that ADAM10 inhibition by its prodomain is unique. In cell-based assays we show that the ADAM10 prodomain inhibits betacellulin shedding, demonstrating that it could be of potential use as a therapeutic agent to treat cancer.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursores Enzimáticos/antagonistas & inhibidores , Precursores Enzimáticos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Proteínas ADAM/uso terapéutico , Proteína ADAM10 , Proteína ADAM17 , Secretasas de la Proteína Precursora del Amiloide/uso terapéutico , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Antígenos CD/metabolismo , Betacelulina , Células COS , Chlorocebus aethiops , Desintegrinas/antagonistas & inhibidores , Desintegrinas/metabolismo , Desintegrinas/uso terapéutico , Precursores Enzimáticos/uso terapéutico , Receptores ErbB/metabolismo , Humanos , Proteínas de la Membrana/uso terapéutico , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Estructura Terciaria de Proteína/fisiología
12.
J Biol Chem ; 282(42): 30785-93, 2007 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17704059

RESUMEN

ADAM-9 belongs to a family of transmembrane, disintegrin-containing metalloproteinases involved in protein ectodomain shedding and cell-cell and cell-matrix interactions. The aim of this study was to analyze the expression of ADAM-9 in skin and to assess the role of this proteolytic/adhesive protein in skin physiology. In normal skin, ADAM-9 expression was detected in both the epidermis and dermis and in vitro in keratinocytes and fibroblasts. Here we report that ADAM-9 functions as a cell adhesion molecule via its disintegrin-cysteine-rich domain. Using solid phase binding assays and antibody inhibition experiments, we demonstrated that the recombinant disintegrin-cysteine-rich domain of ADAM-9 specifically interacts with the beta1 integrin subunit on keratinocytes. This was corroborated by co-immunoprecipitation. In addition, engagement of integrin receptors by the disintegrin-cysteine-rich domain resulted in ERK phosphorylation and increased MMP-9 synthesis. Treatment with the ERK inhibitor PD98059 inhibited MMP-9 induction. Furthermore, the presence of the soluble disintegrin-cysteine-rich domain did not interfere with cell migration on different substrates. However, keratinocytes adhering to the immobilized disintegrin-cysteine-rich domain showed increased motility, which was partially due to the induction of MMP-9 secretion. In summary, our results indicate that the ADAM-9 adhesive domain plays a role in regulating the motility of cells by interaction with beta1 integrins and modulates MMP synthesis.


Asunto(s)
Proteínas ADAM/biosíntesis , Moléculas de Adhesión Celular/biosíntesis , Movimiento Celular/fisiología , Desintegrinas/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Queratinocitos/enzimología , Proteínas de la Membrana/biosíntesis , Fenómenos Fisiológicos de la Piel , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/genética , Línea Celular , Movimiento Celular/efectos de los fármacos , Dermis/citología , Dermis/enzimología , Desintegrinas/antagonistas & inhibidores , Desintegrinas/genética , Células Epidérmicas , Epidermis/enzimología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavonoides/farmacología , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Queratinocitos/citología , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología
13.
J Med Chem ; 47(8): 1930-8, 2004 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-15055993

RESUMEN

As a part of synthetic studies on MMP (matrix metalloproteinase)/ADAM (a disintegrin and metalloproteinase) inhibitors, we have preliminarily communicated that azasugar-based compound 1a exhibited a potential inhibitory activity on some metalloprotease-catalyzed proteolytic reactions. To find promising candidates for the topical treatment of psoriasis, we investigated stability in aqueous solution of compound 1a and its derivative 1b and then optimized the P1' substuent (2-5). In the present study, we synthesized novel derivatives of compound 1a and evaluated their inhibitory activity toward MMP-1, -3, and -9, TACE, and HB-EGF shedding, from a viewpoint of versatility of azasugars as a functional scaffold. As a result, it was found that compound 1b demonstrated desirable inhibitory activity as an antipsoriatic agent, and some of the derivatives showed selective inhibitory activity. In addition, it was found that compound 1b exhibited a significant therapeutic effect on a mouse TPA-induced epidermal hyperplasia model. Therefore, compound 1b could become a promising candidate as a practical antipsoriatic agent.


Asunto(s)
Compuestos Aza/síntesis química , Carbohidratos/síntesis química , Desintegrinas/antagonistas & inhibidores , Ácidos Hidroxámicos/síntesis química , Inhibidores de la Metaloproteinasa de la Matriz , Inhibidores de Proteasas/síntesis química , Sulfonas/síntesis química , Proteínas ADAM , Proteína ADAM17 , Animales , Compuestos Aza/química , Compuestos Aza/farmacología , Carbohidratos/química , Carbohidratos/farmacología , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Similar a EGF de Unión a Heparina , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Hiperplasia , Péptidos y Proteínas de Señalización Intercelular , Metaloendopeptidasas/antagonistas & inhibidores , Ratones , Modelos Moleculares , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Psoriasis/tratamiento farmacológico , Piel/efectos de los fármacos , Piel/patología , Estereoisomerismo , Relación Estructura-Actividad , Sulfonas/química , Sulfonas/farmacología
14.
Matrix Biol ; 22(7): 557-60, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14996435

RESUMEN

Endothelial expression of matrix metalloproteinases has been implicated in angiogenesis and endothelial cell proliferation. Recently, it has been shown that high-density lipoproteins (HDLs) promote angiogenesis. In the present study, we investigated the effects of native HDLs on the expression of several proteases and their inhibitors in human umbilical vein endothelial cells. We show that ADAMTS-1 (a disintegrin and metalloproteinase with thrombospondin motif) was potently induced by incubation with lipopolysaccharide or tumor necrosis factor-alpha and that the expression was significantly reduced in the presence of HDL subfraction 3. Since ADAMTS-1 has recently been shown to inhibit endothelial cell proliferation, the result of the present work may represent a new mechanism by which HDL could have a positive effect on endothelial cell and vascular wall function.


Asunto(s)
Desintegrinas/antagonistas & inhibidores , Células Endoteliales/metabolismo , Lipopolisacáridos/farmacología , Lipoproteínas HDL/fisiología , Metaloendopeptidasas/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Proteínas ADAM , Proteína ADAMTS1 , Células Cultivadas , Desintegrinas/genética , Expresión Génica/efectos de los fármacos , Humanos , Lipoproteínas HDL/farmacología , Lipoproteínas HDL3 , Metaloendopeptidasas/genética , Metaloproteasas/metabolismo , ARN Mensajero/metabolismo
15.
Exp Cell Res ; 292(2): 371-84, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14697344

RESUMEN

The effects of jarastatin (JT), a monomeric RGD-disintegrin, were compared with those of the heterodimeric MLD-disintegrin, EC3, on human neutrophil activation and functions. Both disintegrins inhibited neutrophil chemotaxis induced by fMet-Leu-Phe and were also potent chemotactic agents. These effects were accompanied by an increase in actin polymerization, and both were inhibited by genistein, a tyrosine kinase inhibitor. While JT, but not other RGD-disintegrins, inhibited EC3-induced chemotaxis, EC3 was not able to inhibit JT effect. The chemotactic effect of JT was blocked by anti-alpha(M) antibody whereas anti-alpha(9)beta(1) inhibited EC3 effect. Both JT and EC3 induced focal adhesion kinase (FAK) and phosphoinositide 3-kinase (PI3K) activation. Accordingly, LY294002, a PI3K inhibitor, impaired their chemotactic effect on neutrophils. JT induced Erk-2 translocation to nucleus and a delay of the spontaneous apoptosis of neutrophils in vitro. In contrast, EC3 inhibited Erk-2 activation and had a proapoptotic effect. These effects were reverted by PD98059, an MEK 1/2 inhibitor and blocked by z-VAD-FMK, a caspase inhibitor. In addition, JT, but not EC3, increased the IL-8 mRNA levels in neutrophils. The data indicate that JT and EC3 directly activate an integrin-coupled signaling and modulate the MAPK pathway in different ways, leading the neutrophils to express different functional response.


Asunto(s)
Membrana Celular/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Desintegrinas/farmacología , Integrinas/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Venenos de Víboras/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Animales , Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Extractos Celulares , Membrana Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis de Leucocito/fisiología , Desintegrinas/antagonistas & inhibidores , Interacciones Farmacológicas/fisiología , Inhibidores Enzimáticos/farmacología , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Integrinas/metabolismo , Interleucina-8/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Neutrófilos/metabolismo , Fragmentos de Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo
16.
Eur J Biochem ; 270(11): 2394-403, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12755694

RESUMEN

Three mammalian ADAMTS enzymes, ADAMTS-1, -4 and -5, are known to cleave aggrecan at certain glutamyl bonds and are considered to be largely responsible for cartilage aggrecan catabolism observed during the development of arthritis. We have previously reported that certain catechins, polyphenolic compounds found in highest concentration in green tea (Camellia sinensis), are capable of inhibiting cartilage aggrecan breakdown in an in vitro model of cartilage degradation. We have now cloned and expressed recombinant human ADAMTS-1, -4 and -5 and report here that the catechin gallate esters found in green tea potently inhibit the aggrecan-degrading activity of these enzymes, with submicromolar IC50 values. Moreover, the concentration needed for total inhibition of these members of the ADAMTS group is approximately two orders of magnitude lower than that which is needed to partially inhibit collagenase or ADAM-10 activity. Catechin gallate esters therefore provide selective inhibition of certain members of the ADAMTS group of enzymes and could constitute an important nutritional aid in the prevention of arthritis as well as being part of an effective therapy in the treatment of joint disease and other pathologies involving the action of these enzymes.


Asunto(s)
Catequina/análogos & derivados , Catequina/metabolismo , Desintegrinas/antagonistas & inhibidores , Ésteres/metabolismo , Metaloendopeptidasas/antagonistas & inhibidores , Proteínas ADAM , Proteína ADAM10 , Proteína ADAMTS1 , Proteína ADAMTS4 , Proteína ADAMTS5 , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide , Animales , Western Blotting , Bovinos , Línea Celular , Medios de Cultivo Condicionados/farmacología , Desintegrinas/metabolismo , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Endopeptidasas/metabolismo , Inhibidores Enzimáticos/farmacología , Vectores Genéticos , Humanos , Concentración 50 Inhibidora , Insectos , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Datos de Secuencia Molecular , Procolágeno N-Endopeptidasa , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo ,
17.
Nat Med ; 8(1): 35-40, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11786904

RESUMEN

G-protein-coupled receptor (GPCR) agonists are well-known inducers of cardiac hypertrophy. We found that the shedding of heparin-binding epidermal growth factor (HB-EGF) resulting from metalloproteinase activation and subsequent transactivation of the epidermal growth factor receptor occurred when cardiomyocytes were stimulated by GPCR agonists, leading to cardiac hypertrophy. A new inhibitor of HB-EGF shedding, KB-R7785, blocked this signaling. We cloned a disintegrin and metalloprotease 12 (ADAM12) as a specific enzyme to shed HB-EGF in the heart and found that dominant-negative expression of ADAM12 abrogated this signaling. KB-R7785 bound directly to ADAM12, suggesting that inhibition of ADAM12 blocked the shedding of HB-EGF. In mice with cardiac hypertrophy, KB-R7785 inhibited the shedding of HB-EGF and attenuated hypertrophic changes. These data suggest that shedding of HB-EGF by ADAM12 plays an important role in cardiac hypertrophy, and that inhibition of HB-EGF shedding could be a potent therapeutic strategy for cardiac hypertrophy.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Desintegrinas/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/metabolismo , Glicina/análogos & derivados , Glicina/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Proteínas de la Membrana/antagonistas & inhibidores , Metaloendopeptidasas/antagonistas & inhibidores , Inhibidores de Proteasas/uso terapéutico , Proteínas ADAM , Proteína ADAM12 , Angiotensina II/farmacología , Animales , Aorta Torácica/cirugía , Modelos Animales de Enfermedad , Receptores ErbB/genética , Proteínas de Unión al GTP/metabolismo , Ventrículos Cardíacos , Factor de Crecimiento Similar a EGF de Unión a Heparina , Hipertensión , Péptidos y Proteínas de Señalización Intercelular , Masculino , Fenilefrina/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Transducción de Señal , Sístole , Activación Transcripcional
18.
Toxicon ; 39(2-3): 283-9, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-10978746

RESUMEN

The homodimeric disintegrin contortrostatin was compared directly to the monomeric disintegrins echistatin and flavoridin for the ability to affect protein tyrosine phosphorylation in tumor cells. It was observed that contortrostatin had a dramatic effect on the tyrosine phosphorylation status of several proteins in T24 human bladder cancer cells, including robust induction of phosphorylation of proteins in the range of 120-140 kDa. Echistatin alone had no effect on tyrosine phosphorylation in T24 cells, but dose-dependently inhibits the effects of contortrostatin when both are added simultaneously. Among the proteins that undergo tyrosine phosphorylation in response to contortrostatin treatment is CAS, a 130 kDa adapter protein involved in integrin signaling. Flavoridin alone was found to have no effect on CAS, but can completely block contortrostatin-induced phosphorylation of this protein in MDA-MB-435 cells. These observations strongly suggest that the homodimeric structure of contortrostatin functionally distinguishes it from other monomeric members of the disintegrin family.


Asunto(s)
Desintegrinas/farmacología , Péptidos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Tirosina/metabolismo , Venenos de Víboras/farmacología , Venenos de Crotálidos/farmacología , Desintegrinas/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Péptidos y Proteínas de Señalización Intercelular , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas
19.
Dev Biol ; 227(1): 197-210, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11076687

RESUMEN

ADAM13 is a cell surface metalloprotease expressed in cephalic neural crest cells during early Xenopus development. The cytoplasmic domain of ADAM13 contains three potential SH3 (Src homology type 3) binding sites, suggesting that this region may support interactions with intracellular proteins. In this report we describe the identification, by a new strategy, of three proteins that bind the ADAM13 cytoplasmic domain in vitro: X-Src1, X-An4, and X-PACSIN2. We focused our study on X-PACSIN2 protein because it colocalizes with ADAM13 in migrating neural crest cells during embryonic development. Using pull-down experiments we show that X-PACSIN2 binds to ADAM13 in vitro. Using Xenopus XTC cells, we demonstrate that ADAM13 and X-PACSIN2 colocalize to membrane ruffles and cytoplasmic vesicles. We also show that X-PACSIN2 overexpression can rescue developmental alterations induced by overexpression of ADAM13, suggesting that both proteins interact in vivo. Finally, our results suggest that X-PACSIN2 overexpression reduces endogenous ADAM13 function while a truncated X-PACSIN2 (DeltaSH3) increases this activity in cephalic neural crest cells. We propose that X-PACSIN2 may regulate ADAM13 activity by influencing either its subcellular localization or its catalytic activity. In agreement with this model, elimination of the ADAM13 cytoplasmic domain increased developmental alterations attributable to ADAM13 proteolytic activity.


Asunto(s)
Desintegrinas/metabolismo , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Proteínas/metabolismo , Proteínas de Xenopus , Proteínas ADAM , Secuencia de Aminoácidos , Animales , Desintegrinas/antagonistas & inhibidores , Desintegrinas/química , Desintegrinas/genética , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Metaloendopeptidasas/antagonistas & inhibidores , Metaloendopeptidasas/química , Metaloendopeptidasas/genética , Microinyecciones , Modelos Biológicos , Datos de Secuencia Molecular , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Fenotipo , Pruebas de Precipitina , Unión Proteica , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Xenopus laevis/embriología , Xenopus laevis/genética , Xenopus laevis/metabolismo , Dominios Homologos src
20.
Science ; 289(5483): 1308-10, 2000 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-10979856

RESUMEN

A series of molecular cues guide axons as they extend in the embryonic environment. In her Perspective, Pasquale discusses new findings that reveal how signals originating from adhesive contacts between axons and the cells they encounter can repel the axon, causing it to break the adhesive contacts and move away (Hattori et al. and Galko and Tessier-Lavigne).


Asunto(s)
Axones/fisiología , Desintegrinas/metabolismo , Proteínas de Drosophila , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor , Animales , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Membrana Celular/metabolismo , Receptor DCC , Desintegrinas/antagonistas & inhibidores , Efrina-A2 , Glicosilfosfatidilinositoles/metabolismo , Conos de Crecimiento/fisiología , Ligandos , Metaloendopeptidasas/antagonistas & inhibidores , Ratones , Receptores de Netrina , Netrina-1 , Inhibidores de Proteasas/farmacología , Receptores de Superficie Celular/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...