Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Ann Neurol ; 87(3): 405-418, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31900946

RESUMEN

OBJECTIVE: Leucine-rich glioma-inactivated 1 (LGI1) encephalitis is the second most common antibody-mediated encephalopathy, but insight into the intrathecal B-cell autoimmune response, including clonal relationships, isotype distribution, frequency, and pathogenic effects of single LGI1 antibodies, has remained limited. METHODS: We cloned, expressed, and tested antibodies from 90 antibody-secreting cells (ASCs) and B cells from the cerebrospinal fluid (CSF) of several patients with LGI1 encephalitis. RESULTS: Eighty-four percent of the ASCs and 21% of the memory B cells encoded LGI1-reactive antibodies, whereas reactivities to other brain epitopes were rare. All LGI1 antibodies were of IgG1, IgG2, or IgG4 isotype and had undergone affinity maturation. Seven of the overall 26 LGI1 antibodies efficiently blocked the interaction of LGI1 with its receptor ADAM22 in vitro, and their mean LGI1 signal on mouse brain sections was weak compared to the remaining, non-ADAM22-competing antibodies. Nevertheless, both types of LGI1 antibodies increased the intrinsic cellular excitability and glutamatergic synaptic transmission of hippocampal CA3 neurons in slice cultures. INTERPRETATION: Our data show that the patients' intrathecal B-cell autoimmune response is dominated by LGI1 antibodies and that LGI1 antibodies alone are sufficient to promote neuronal excitability, a basis of seizure generation. Fundamental differences in target specificity and antibody hypermutations compared to the CSF autoantibody repertoire in N-methyl-D-aspartate receptor encephalitis underline the clinical concept that autoimmune encephalitides are very distinct entities. Ann Neurol 2020;87:405-418.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Autoanticuerpos/farmacología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Neuronas/fisiología , Proteínas ADAM/efectos de los fármacos , Anciano , Animales , Anticuerpos Monoclonales/líquido cefalorraquídeo , Autoanticuerpos/líquido cefalorraquídeo , Región CA3 Hipocampal/fisiología , Células Cultivadas , Encefalitis/líquido cefalorraquídeo , Encefalitis/inmunología , Femenino , Enfermedad de Hashimoto/líquido cefalorraquídeo , Enfermedad de Hashimoto/inmunología , Humanos , Isotipos de Inmunoglobulinas , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas del Tejido Nervioso/efectos de los fármacos , Ratas , Transmisión Sináptica/efectos de los fármacos
2.
Int J Biochem Cell Biol ; 105: 123-133, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30343150

RESUMEN

The Eph-ephrin signaling pathway mediates developmental processes and the proper functioning of the adult human body. This distinctive bidirectional signaling pathway includes a canonical downstream signal cascade inside the Eph-bearing cells, as well as a reverse signaling in the ephrin-bearing cells. The signaling is terminated by ADAM metalloproteinase cleavage, internalization, and degradation of the Eph/ephrin complexes. Consequently, the Eph-ephrin-ADAM signaling cascade has emerged as a key target with immense therapeutic potential particularly in the context of cancer. An interesting twist was brought forth by the emergence of ephrins as the entry receptors for the pathological Henipaviruses, which has spurred new studies to target the viral entry. The availability of high-resolution structures of the multi-modular Eph receptors in complexes with ephrins and other binding partners, such as peptides, small molecule inhibitors and antibodies, offers a wealth of information for the structure-guided development of therapeutic intervention. Furthermore, genomic data mining of Eph mutants involved in cancer provides information for targeted drug development. In this review we summarize the distinct avenues for targeting the Eph-ephrin signaling pathway, including its termination by ADAM proteinases. We highlight the latest developments in Eph-related pharmacology in the context of Eph-ephrin-ADAM-based antibodies and small molecules. Finally, the future prospects of genomics- and proteomics-based medicine are discussed.


Asunto(s)
Efrinas/efectos de los fármacos , Efrinas/metabolismo , Receptores de la Familia Eph/efectos de los fármacos , Receptores de la Familia Eph/metabolismo , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/metabolismo , Anticuerpos/química , Anticuerpos/farmacología , Antineoplásicos/farmacología , Sitios de Unión , Desarrollo de Medicamentos , Efrinas/química , Humanos , Modelos Biológicos , Modelos Moleculares , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Receptores de la Familia Eph/genética , Transducción de Señal/efectos de los fármacos
3.
ACS Chem Neurosci ; 7(1): 40-5, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26559179

RESUMEN

The ADAM family of metalloproteases cleave a diverse range of transmembrane substrates, resulting in the release of their soluble ectodomains. This process of protein shedding, termed α-secretase processing, is involved in many facets of both normal and disease related cellular function. While the processing of substrates has been well documented, the regulation and trafficking of the ADAMs are less well understood. Tools that allow for the study of ADAMs under their native environment will allow for a better understanding of their regulation and activity. Here we describe the design and evaluation of a novel fluorescent analogue of a well-characterized ADAM inhibitor, Batimastat. This probe exhibited similar activity for inhibiting α-secretase processing in cells as did Batimastat. Importantly, this probe specifically labeled ADAMs fluorescently in both fixed and living cells, enabling the possibility to study the trafficking of α-secretase proteins in a dynamic environment.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Fenilalanina/análogos & derivados , Inhibidores de Proteasas/farmacología , Tiofenos/farmacología , Proteínas ADAM/efectos de los fármacos , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Glicoproteínas de Membrana/metabolismo , Microscopía Fluorescente , Fenilalanina/química , Fenilalanina/farmacología , Inhibidores de Proteasas/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Tiofenos/química , Transfección
4.
Osteoarthritis Cartilage ; 24(1): 178-187, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26256766

RESUMEN

OBJECTIVE: To determine whether mandibular condylar cartilage degradation induced by experimentally abnormal occlusion could be ameliorated via systemic administration of strontium or NBD peptide. METHODS: Six-week-old female C57BL/6J mice were used. From the seventh day after mock operation or unilateral anterior crossbite (UAC) treatment, the control and UAC mice were further respectively pharmacologically treated for 2 weeks or 4 weeks of saline (CON + Saline and UAC + Saline groups), SrCl2 (CON + SrCl2 and UAC + SrCl2 groups) or NBD peptide (CON + NBD peptide and UAC + NBD peptide groups). Changes in condylar cartilage and subchondral bone were assessed 21 and 35 days after mock operation or UAC procedure by histology and micro-CT. Real-time PCR and/or immunohistochemistry (IHC) were performed to evaluate changes in expression levels of col2a1, aggrecan, ADAMTS-5, tnf-α, il-1ß, nfkbia, nuclear factor-kappaB phospho-p65 in condylar cartilage, and rankl/rank/opg in both condylar cartilage and subchondral bone. RESULTS: Cartilage degradation with decreased col2a1 and aggrecan expression, and increased ADAMTS-5, tnf-α/il1-ß, nfkbia and NF-κB phospho-p65 was observed in UAC + Saline groups. Subchondral bone loss with increased osteoclast numbers and decreased opg/rankl ratio was found in UAC + Saline groups compared to age-match CON + Saline groups. Cartilage degradation and subchondral bone loss were reversed by treatment of SrCl2 or NBD peptide while the same dosage in control mice induced few changes in condylar cartilage and subchondral bone. CONCLUSIONS: The results demonstrate reverse effect of systemic administration of strontium or NBD peptide on UAC-induced condylar cartilage degradation and subchondral bone loss.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Maloclusión , Cóndilo Mandibular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Péptidos/farmacología , ARN Mensajero/efectos de los fármacos , Estroncio/farmacología , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS5 , Agrecanos/efectos de los fármacos , Agrecanos/genética , Agrecanos/metabolismo , Animales , Cartílago Articular/metabolismo , Colágeno Tipo II/efectos de los fármacos , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Oclusión Dental , Femenino , Proteínas I-kappa B/efectos de los fármacos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inmunohistoquímica , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Cóndilo Mandibular/metabolismo , Cóndilo Mandibular/patología , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , Osteoclastos/metabolismo , Osteoprotegerina/efectos de los fármacos , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Proteoglicanos/efectos de los fármacos , Proteoglicanos/metabolismo , Ligando RANK/efectos de los fármacos , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Activador del Factor Nuclear kappa-B/efectos de los fármacos , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Factor de Transcripción ReIA/efectos de los fármacos , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
Int Immunopharmacol ; 29(2): 863-868, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26371861

RESUMEN

Hypothemycin, a resorcylic acid lactone polyketide, has been shown to inhibit oncogenic ras-transformation and T cell activation. In the present study, we investigated the effect of hypothemycin on tumor necrosis factor-α (TNF-α) production in macrophages and the molecular mechanisms involved in this effect. Hypothemycin potently suppressed the TNF-α production without affecting nitric oxide production in lipopolysaccharide (LPS)-stimulated macrophages. However, hypothemycin had no effect on the activity of TNF-α-converting enzyme, a key enzyme for converting membrane-bound pro-TNF-α into soluble TNF-α. Further study demonstrated that the stability of TNF-α mRNA was decreased by hypothemycin treatment. In addition, hypothemycin suppressed LPS-induced phosphorylation of p38 MAPK and ERK. Moreover, knockdown of tristetraprolin (TTP), which is an important trans-acting regulator of TNF-α mRNA stability and downstream target of p38 MAPK and ERK, reversed hypothemycin-mediated inhibition of TNF-α mRNA expression. Collectively, our results suggest that hypothemycin suppresses TNF-α production by TTP-dependent destabilization of TNF-α mRNA and this is mediated, at least in part, by blocking the activation of p38 MAPK and ERK.


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Estabilidad del ARN/efectos de los fármacos , Tristetraprolina/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis , Zearalenona/análogos & derivados , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Regulación hacia Abajo/efectos de los fármacos , Humanos , Macrófagos/efectos de los fármacos , Ratones , Óxido Nítrico/biosíntesis , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Zearalenona/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
6.
Invest Ophthalmol Vis Sci ; 56(9): 5162-73, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26244291

RESUMEN

PURPOSE: The a-disintegrin-and-metalloprotease (ADAM) family proteins are widely expressed in the different layers of the retina throughout development. The effect of ADAM proteins on the epithelial-to-mesenchymal transition (EMT) in proliferative vitreoretinopathy (PVR) or AMD is yet to be elucidated. In this study we used Epstein-Barr virus (EBV)-transformed adult retinal pigment epithelial (ARPE) cells to investigate how sorafenib, a multikinase inhibitor, modulates ADAM proteins to control EMT. METHODS: Epithelial to mesenchymal transition and related mechanisms in EBV-infected ARPE cells were determined by RT-PCR, Western blot, invasion assay, ELISA assay, and gene silencing with siRNA. RESULTS: Mesenchymal-like ARPE/EBV cells exhibited considerably increased cellular migration and invasion compared with ARPE cells and produced EMT-related cytokines. Sorafenib significantly inhibited production of TGF-ß1, VEGF, IL-6, IL-8, MCP-1, and TNF-α and blocked the activation of migration-related signaling molecules, such as HIF-1α, p-STAT3, MMP2, and Ang-1. The expression of mature ADAM10, ADAM17, and cleaved Notch 1 proteins in ARPE/EBV cells was downregulated after treatment with sorafenib through the regulatory activity of nardilysin (NRD-1). Gene silencing of NRD-1 in ARPE/EBV cells attenuated secretion of EMT-related cytokines and expression of ADAM10 and 17 and upregulated epithelial markers. CONCLUSIONS: Sorafenib controls the mesenchymal characteristics of EBV-infected ARPE cells. Nardilysin and ADAM family proteins might be new targets for the prevention or control of EMT in retinal diseases.


Asunto(s)
Proteínas ADAM/genética , Secretasas de la Proteína Precursora del Amiloide/genética , ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Herpesvirus Humano 4 , Proteínas de la Membrana/genética , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Epitelio Pigmentado de la Retina/metabolismo , Proteínas ADAM/biosíntesis , Proteínas ADAM/efectos de los fármacos , Proteína ADAM10 , Proteína ADAM17 , Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Secretasas de la Proteína Precursora del Amiloide/efectos de los fármacos , Movimiento Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Humanos , Immunoblotting , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/efectos de los fármacos , Niacinamida/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Sorafenib , Factor de Necrosis Tumoral alfa , Vitreorretinopatía Proliferativa/genética , Vitreorretinopatía Proliferativa/patología , Vitreorretinopatía Proliferativa/virología
8.
Am J Physiol Renal Physiol ; 307(5): F551-9, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899059

RESUMEN

Polycystic kidney disease (PKD) is a common genetic disorder leading to cyst formation in the kidneys and other organs that ultimately results in kidney failure and death. Currently, there is no therapy for slowing down or stopping the progression of PKD. In this study, we identified the disintegrin metalloenzyme 17 (ADAM17) as a key regulator of cell proliferation in kidney tissues of conditional knockout Ift88(-/-) mice and collecting duct epithelial cells from Ift88°(rpk) mice, animal models of autosomal recessive polycystic kidney disease (ARPKD). Using Western blotting, an enzyme activity assay, and a growth factor-shedding assay in the presence or absence of the specific ADAM17 inhibitor TMI-005, we show that increased expression and activation of ADAM17 in the cystic kidney and in collecting duct epithelial cells originating from the Ift88°(rpk) mice (designated as PKD cells) lead to constitutive shedding of several growth factors, including heparin-binding EGF-like growth factor (HB-EGF), amphiregulin, and transforming growth factor-α (TGF-α). Increased growth factor shedding induces activation of the EGFR/MAPK/ERK pathway and maintains higher cell proliferation rate in PKD cells compared with control cells. PKD cells also displayed increased lactate formation and extracellular acidification indicative of aerobic glycolysis (Warburg effect), which was blocked by ADAM17 inhibition. We propose that ADAM17 is a key promoter of cellular proliferation in PKD cells by activating the EGFR/ERK axis and a proproliferative glycolytic phenotype.


Asunto(s)
Proteínas ADAM/fisiología , Proliferación Celular/fisiología , Células Epiteliales/patología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Glucólisis/fisiología , Túbulos Renales Colectores/patología , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/efectos de los fármacos , Proteína ADAM17 , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Receptores ErbB/fisiología , Femenino , Factor de Crecimiento Similar a EGF de Unión a Heparina/fisiología , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/fisiopatología , Masculino , Ratones , Ratones Noqueados , Morfolinas/farmacología , Fenotipo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Factor de Crecimiento Transformador alfa/fisiología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
9.
BMC Gastroenterol ; 13: 155, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24172289

RESUMEN

BACKGROUND: Ursodeoxycholic acid (UDCA) is used to treat primary biliary cirrhosis, intrahepatic cholestasis, and other cholestatic conditions. Although much has been learned about the molecular basis of the disease pathophysiology, our understanding of the effects of UDCA remains unclear. Possibly underlying its cytoprotective, anti-apoptotic, anti-oxidative effects, UDCA was reported to regulate the expression of TNFα and other inflammatory cytokines. However, it is not known if this effect involves also modulation of ADAM family of metalloproteinases, which are responsible for release of ectodomains of inflammatory cytokines from the cell surface. We hypothesized that UDCA modulates ADAM17 activity, resulting in amelioration of cholestasis in a murine model of bile duct ligation (BDL). METHODS: The effect of UDCA on ADAM17 activity was studied using the human liver hepatocellular carcinoma cell line HepG2. Untransfected cells or cells ectopically expressing human ADAM17 were cultured with or without UDCA and further activated using phorbol-12-myristate-13-acetate (PMA). The expression and release of ADAM17 substrates, TNFα, TGFα, and c-Met receptor (or its soluble form, sMet) were evaluated using ELISA and quantitative real-time (qRT) PCR. Immunoblotting analyses were conducted to evaluate expression and activation of ADAM17 as well as the level of ERK1/2 phosphorylation after UDCA treatment. The regulation of tissue inhibitor of metalloproteinases-1 (TIMP-1) by UDCA was studied using zymography and qRT-PCR. A mouse model of acute cholestasis was induced by common BDL technique, during which mice received daily orogastric gavage with either UDCA or vehicle only. Liver injury was quantified using alkaline phosphatase (ALP), relative liver weight, and confirmed by histological analysis. ADAM17 substrates in sera were assessed using a bead multiplex assay. RESULTS: UDCA decreases amount of shed TNFα, TGFα, and sMet in cell culture media and the phosphorylation of ERK1/2. These effects are mediated by the reduction of ADAM17 activity in PMA stimulated cells although the expression ADAM17 is not affected. UDCA reduced the level of the mature form of ADAM17. Moreover, UDCA regulates the expression of TIMP-1 and gelatinases activity in PMA stimulated cells. A BDL-induced acute cholangitis model was characterized by increased relative liver weight, serum levels of ALP, sMet, and loss of intracellular glycogen. UDCA administration significantly decreased ALP and sMet levels, and reduced relative liver weight. Furthermore, hepatocytes of UDCA-treated animals retained their metabolic activity as evidenced by the amount of glycogen storage. CONCLUSIONS: The beneficial effect of UDCA appears to be mediated in part by the inhibition of ADAM17 activation and, thus, the release of TNFα, a strong pro-inflammatory factor. The release of other ADAM17 substrates, TGFα and sMet, are also regulated this way, pointing to a general impact on the release of ADAM17 substrates, which are pivotal for liver regeneration and function. In parallel, UDCA upregulates TIMP-1 that in turn inhibits matrix metalloproteinases, which destroy the hepatic ECM in diseased liver. This control of extracellular matrix turnover represents an additional beneficial path of UDCA treatment.


Asunto(s)
Proteínas ADAM/efectos de los fármacos , Colagogos y Coleréticos/farmacología , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Ácido Ursodesoxicólico/farmacología , Proteína ADAM17 , Animales , Conductos Biliares/cirugía , Colestasis , Células Hep G2 , Humanos , Ligadura , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proteínas Proto-Oncogénicas c-met/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/metabolismo , Factor de Crecimiento Transformador alfa/efectos de los fármacos , Factor de Crecimiento Transformador alfa/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 32(10): 2444-51, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22904271

RESUMEN

OBJECTIVE: Endothelium dysfunction is an initiating factor in atherosclerosis. A disintegrin and metalloproteinase 15 (ADAM 15) is a multidomain metalloprotease recently identified as a regulator of endothelial permeability. However, whether and how ADAM15 contributes to atherosclerosis remains unknown. METHODS AND RESULTS: Genetic ablation of ADAM15 in apolipoprotein E-deficient mice led to a significant reduction in aortic atherosclerotic lesion size (by 52%), plaque macrophage infiltration (by 69%), and smooth muscle cell deposition (by 82%). In vitro studies implicated endothelial-derived ADAM15 in barrier dysfunction and monocyte transmigration across mouse aortic and human umbilical vein endothelial cell monolayers. This role of ADAM15 depended on intact functioning of the cytoplasmic domain, as evidenced in experiments with site-directed mutagenesis targeting the metalloprotease active site (E349A), the disintegrin domain (Arginine-Glycine-Aspartic acid→Threonine-Aspartic acid-Aspartic acid), or the cytoplasmic tail. Further investigations revealed that ADAM15-induced barrier dysfunction was concomitant with dissociation of endothelial adherens junctions (vascular endothelial [VE]-cadherin/γ-catenin), an effect that was sensitive to Src family kinase inhibition. Through small interfering RNA-mediated knockdown of distinct Src family kinase members, c-Src and c-Yes were identified as important mediators of these junctional effects of ADAM15. CONCLUSIONS: These results suggest that endothelial cell-derived ADAM15, signaling through c-Src and c-Yes, contributes to atherosclerotic lesion development by disrupting adherens junction integrity and promoting monocyte transmigration.


Asunto(s)
Proteínas ADAM/fisiología , Aterosclerosis/fisiopatología , Endotelio Vascular/fisiopatología , Proteínas de la Membrana/fisiología , Transducción de Señal/fisiología , Familia-src Quinasas/fisiología , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/genética , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Proteína Tirosina Quinasa CSK , Movimiento Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Humanos , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/patología , Monocitos/fisiología , Proteínas Proto-Oncogénicas c-yes/efectos de los fármacos , Proteínas Proto-Oncogénicas c-yes/genética , Proteínas Proto-Oncogénicas c-yes/fisiología , ARN Interferente Pequeño/farmacología , Familia-src Quinasas/efectos de los fármacos , Familia-src Quinasas/genética
11.
Toxicol Appl Pharmacol ; 263(1): 61-72, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22687607

RESUMEN

Mer receptor tyrosine kinase (Mer) regulates macrophage activation and promotes apoptotic cell clearance. Mer activation is regulated through proteolytic cleavage of the extracellular domain. To determine if membrane-bound Mer is cleaved during bleomycin-induced lung injury, and, if so, how preventing the cleavage of Mer enhances apoptotic cell uptake and down-regulates pulmonary immune responses. During bleomycin-induced acute lung injury in mice, membrane-bound Mer expression decreased, but production of soluble Mer and activity as well as expression of disintegrin and metalloproteinase 17 (ADAM17) were enhanced . Treatment with the ADAM inhibitor TAPI-0 restored Mer expression and diminished soluble Mer production. Furthermore, TAPI-0 increased Mer activation in alveolar macrophages and lung tissue resulting in enhanced apoptotic cell clearance in vivo and ex vivo by alveolar macrophages. Suppression of bleomycin-induced pro-inflammatory mediators, but enhancement of hepatocyte growth factor induction were seen after TAPI-0 treatment. Additional bleomycin-induced inflammatory responses reduced by TAPI-0 treatment included inflammatory cell recruitment into the lungs, levels of total protein and lactate dehydrogenase activity in bronchoalveolar lavage fluid, as well as caspase-3 and caspase-9 activity and alveolar epithelial cell apoptosis in lung tissue. Importantly, the effects of TAPI-0 on bleomycin-induced inflammation and apoptosis were reversed by coadministration of specific Mer-neutralizing antibodies. These findings suggest that restored membrane-bound Mer expression by TAPI-0 treatment may help resolve lung inflammation and apoptosis after bleomycin treatment.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Fagocitosis/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/fisiología , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/metabolismo , Proteína ADAM17 , Lesión Pulmonar Aguda/inducido químicamente , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bleomicina/farmacología , Western Blotting , Caspasa 3/metabolismo , Caspasa 9/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Dipéptidos/farmacología , Ensayo de Inmunoadsorción Enzimática , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas Receptoras/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Arch Toxicol ; 86(10): 1537-46, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22581348

RESUMEN

Human exposure to PM(2.5) (particulate matter with an aerodynamic diameter below 2.5 µm) is known to be responsible for airway inflammation and may also induce airway remodelling. In respiratory epithelial cells exposed to PM(2.5), releases of pro-inflammatory cytokines such as granulocyte macrophage-colony stimulating factor (GM-CSF) and growth factor ligands of the epidermal growth factor receptor (EGFR) are increased. The present study aimed at determining the involvement of EGFR ligands by autocrine effects in PM(2.5)-induced GM-CSF release. PM(2.5) exposure triggers GM-CSF release by human bronchial epithelial (HBE) cells. This release is dependent on EGFR activation by ligand binding as it is inhibited by AG1478, an inhibitor of EGFR tyrosine kinase activity as well as by a neutralizing anti-EGFR antibody. The use of conditioned medium from cells previously exposed to PM(2.5) demonstrates that PM(2.5)-exposed cells release soluble EGFR ligands able to induce GM-CSF release by an autocrine manner. It was further demonstrated by inhibiting tumour-necrosis factor-alpha converting enzyme (TACE) that is involved in some EGFR ligand shedding. TAPI-2 and GM-6001, two TACE inhibitors, prevented the PM(2.5)-induced GM-CSF release as well as the silencing of TACE by siRNA. We provide evidence that the pro-inflammatory response induced by PM(2.5) exposure on HBE cells, results from an autocrine effect of EGFR ligands released by TACE activity. This autocrine loop by eliciting and sustaining inflammation could contribute to exacerbation of airway remodelling in respiratory-compromised individuals.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Receptores ErbB/efectos de los fármacos , Inflamación/inducido químicamente , Material Particulado/toxicidad , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Comunicación Autocrina/efectos de los fármacos , Bronquios/citología , Bronquios/efectos de los fármacos , Línea Celular , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Silenciador del Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inflamación/patología , Tamaño de la Partícula , Quinazolinas/farmacología , ARN Interferente Pequeño/metabolismo , Tirfostinos/farmacología
13.
Eur Cell Mater ; 23: 41-57, 2012 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-22287113

RESUMEN

This study investigated whether treatment of articular cartilage chondrocytes with a combination of oncostatin M (OSM) and interleukin-1 (IL-1) could induce a degradative phenotype that was mediated through the protein kinase R (PKR) signalling pathway. High-density monolayer cultures of full depth, bovine chondrocytes were treated with a combination of OSM and IL-1 (OSM+IL-1) for 7 days. To inhibit the activation of PKR, a pharmacological inhibitor of PKR was added to duplicate cultures. Pro- and active matrix metalloproteinase-9 (MMP9) and MMP9 mRNA were significantly upregulated by OSM+IL-1 through a PKR dependent mechanism. ADAMTS4 and ADAMTS5 mRNA were also upregulated by OSM+IL-1. The upregulation of ADAMTS4 and ADAMTS5 were, in part, mediated through PKR. OSM+IL-1 resulted in a loss of type II collagen, which could not be rescued by PKR inhibition. OSM+IL-1 reduced the expression of COL2A1 (type II collagen), COL9A1 (type IX collagen), COL11A1 (type XI collagen), and ACAN (aggrecan) mRNAs. Expression of type II and XI collagen and aggrecan was reduced further when PKR was inhibited. OSM+IL-1 resulted in an 11-fold increase in TNFa mRNA which was, in part, mediated through the PKR pathway. This study demonstrates, for the first time, that a number of catabolic and pro-inflammatory effects known to be important in human arthritis and induced by OSM and IL-1, are mediated by the PKR signalling pathway.


Asunto(s)
Cartílago Articular/citología , Cartílago Articular/metabolismo , Condrocitos/metabolismo , Interleucina-1/administración & dosificación , Oncostatina M/administración & dosificación , Oncostatina M/metabolismo , eIF-2 Quinasa/metabolismo , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/metabolismo , Animales , Bovinos , Condrocitos/efectos de los fármacos , Colágeno Tipo II/efectos de los fármacos , Colágeno Tipo II/metabolismo , Combinación de Medicamentos , Inhibidores Enzimáticos , Metaloproteinasa 13 de la Matriz/efectos de los fármacos , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Cultivo Primario de Células , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , eIF-2 Quinasa/antagonistas & inhibidores
14.
Nephrol Dial Transplant ; 27(5): 2114-22, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22015440

RESUMEN

BACKGROUND: Interstitial fibrosis and tubular atrophy (IF/TA) is an important cause of renal function loss and ischaemia-reperfusion (I/R) injury is considered to play an important role in its pathophysiology. The aim of the present study was to investigate the role of a disintegrin and metalloproteinase 17 (ADAM17) in human renal allograft disease and in experimental I/R injury of the kidney. METHODS: We studied the expression of ADAM17 messenger RNA (mRNA) in IF/TA and control kidneys by reverse transcription-polymerase chain reaction and in situ hybridization. Moreover, we assessed ADAM17-mediated heparin-binding epidermal growth factor (HB-EGF) shedding in immortalized human cells. Finally, we studied the effect of pharmacological ADAM17 inhibition in a model of renal I/R injury in rats. RESULTS: ADAM17 mRNA was up-regulated in IF/TA when compared to control kidneys. In normal kidneys, ADAM17 mRNA was weakly expressed in proximal tubules, peritubular capillaries, glomerular endothelium and parietal epithelium. In IF/TA, tubular, capillary and glomerular ADAM17 expression was strongly enhanced with de novo expression in the mesangium. In interstitial fibrotic lesions, we observed co-localization of ADAM17 with HB-EGF protein. In vitro, inhibition of ADAM17 with TNF484 resulted in a dose-dependent reduction of HB-EGF shedding in phorbol 12-myrisate 13-acetate-stimulated cells and non-stimulated cells. In vivo, ADAM17 inhibition significantly reduced the number of glomerular and interstitial macrophages at Day 4 of reperfusion. CONCLUSIONS: In conclusion, HB-EGF co-expresses with ADAM17 in renal interstitial fibrosis, suggesting a potential interaction in IF/TA. Targeting ADAM17 to reduce epidermal growth factor receptor phosphorylation could be a promising way of intervention in human renal disease.


Asunto(s)
Proteínas ADAM/metabolismo , Trasplante de Riñón , Riñón/metabolismo , Riñón/patología , Daño por Reperfusión/metabolismo , Regulación hacia Arriba , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/efectos de los fármacos , Proteína ADAM17 , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Atrofia , Células Cultivadas , Niño , Preescolar , Relación Dosis-Respuesta a Droga , Femenino , Fibrosis , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Ácidos Hidroxámicos/farmacología , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/efectos de los fármacos , Masculino , Persona de Mediana Edad , Modelos Animales , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Daño por Reperfusión/patología , Adulto Joven
15.
Int J Pharm ; 416(2): 486-92, 2011 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-21382458

RESUMEN

UNLABELLED: Encapsulation of glucocorticoids into long-circulating liposomes provides targeting of these drugs to the inflamed synovium in experimental arthritis and thereby strongly improves their therapeutic index. The goal of this study was to evaluate the effect and mechanisms of intravenous liposomal delivery of prednisolone phosphate (Lip-PLP) on protease mediated cartilage destruction during murine antigen-induced arthritis (AIA). Mice treated with a single injection of Lip-PLP showed a pronounced suppression of synovial immune cell infiltration compared to control, PBS-treated mice. Liposomal PLP also significantly suppressed interleukin 1ß (3.6 fold) in the synovium, but not in the blood serum. Furthermore, expression of the proteases MMP-3, -9, -13 and -14 and ADAMTS-4 and -5 was suppressed by Lip-PLP in the synovium, but not within the articular cartilage of the femur and tibia, demonstrating the specific action of Lip-PLP on the synovium. Lip-PLP is phagocytosed by macrophages in vitro and suppresses their expression of IL-1ß and MMPs after LPS activation. Moreover, Lip-PLP suppresses destruction of the cartilage matrix during AIA mediated by active MMPs and ADAMTS, as assessed by immunostaining of their respective neoepitopes VDIPEN and NITEGE in various cartilage layers of the knee joint. CONCLUSION: liposomal delivery of glucocorticoids protects against cartilage matrix destruction during experimental arthritis by inhibiting protease expression and activity in the inflamed synovium.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Glucocorticoides/farmacología , Prednisolona/análogos & derivados , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/metabolismo , Animales , Antígenos/toxicidad , Artritis Experimental/patología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Preparaciones de Acción Retardada , Glucocorticoides/administración & dosificación , Interleucina-1beta/metabolismo , Liposomas , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Prednisolona/administración & dosificación , Prednisolona/farmacología , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología
16.
Cell Tissue Res ; 340(1): 179-88, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20217136

RESUMEN

Pro-inflammatory cytokines induce meniscal matrix degradation and inhibition of endogenous repair mechanisms, but the pathogenic mechanisms behind this are mostly unknown. Therefore, we investigated details of interleukin-1 (IL-1alpha)-induced aggrecan turnover in mature meniscal tissue explants. Fibro-cartilagenous disks (3 mm diameter x 1 mm thickness) were isolated from the central, weight-bearing region of menisci from 2-year-old cattle. After 3 or 6 days of IL-1alpha-treatment, GAG loss (DMMB assay), biosynthetic activity ([(35)SO(4)]-sulfate and [(3)H]-proline incorporation), gene expression (quantitative RT-PCR) and the abundance (zymography, Western blot) of matrix-degrading enzymes and specific aggrecan products were determined. Meniscal fibrocartilage had a 4-fold lower GAG content (per wet weight) than adjacent articular cartilage, and expressed MMPs-1, -2, -3 and ADAMTS4 constitutively, whereas ADAMTS5 m-RNA was essentially undetectable. Significant IL-1 effects were a decrease in biosynthetic activity, an increase in GAG release and in the expression/abundance of MMP-2, MMP-3 and ADAMTS4. Fresh tissue contained aggrecan core protein products similar to those previously described for bovine articular cartilage of this age. IL-1 induced the release of aggrecanase-generated CS-substituted products including both high (>250 kDa) and low molecular weight (about 75 kDa) species. TIMP-3 (but not TIMP-1 and -2 or a broad spectrum MMP inhibitor) inhibited IL-1-dependent GAG loss. In addition, IL-1 induced the release of preformed pools of three known G1-bearing products. We conclude that aggrecanases are responsible for IL-1-stimulated GAG release from meniscal explants, and that IL-1 also stimulates release of G1-bearing products, by a process possibly involving hyaluronan fragmentation.


Asunto(s)
Agrecanos/metabolismo , Artritis/inmunología , Glicosaminoglicanos/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-1alfa/metabolismo , Meniscos Tibiales/inmunología , Proteínas ADAM/efectos de los fármacos , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS4 , Agrecanos/efectos de los fármacos , Animales , Artritis/metabolismo , Artritis/fisiopatología , Calpaína/efectos de los fármacos , Calpaína/genética , Calpaína/metabolismo , Bovinos , Endopeptidasas/efectos de los fármacos , Endopeptidasas/genética , Endopeptidasas/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Ácido Hialurónico/metabolismo , Mediadores de Inflamación/farmacología , Interleucina-1alfa/farmacología , Metaloproteinasas de la Matriz/efectos de los fármacos , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Meniscos Tibiales/efectos de los fármacos , Meniscos Tibiales/metabolismo , Modelos Biológicos , Procolágeno N-Endopeptidasa/efectos de los fármacos , Procolágeno N-Endopeptidasa/genética , Procolágeno N-Endopeptidasa/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/efectos de los fármacos , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo
17.
Osteoarthritis Cartilage ; 18(3): 455-63, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19909832

RESUMEN

OBJECTIVE: Glucosamine has been previously shown to suppress cartilage aggrecan catabolism in explant cultures. We determined the effect of glucosamine on ADAMTS5 (a disintegrin-like and metalloprotease domain (reprolysin type) with thrombospondin type-1 motifs 5), a major aggrecanase in osteoarthritis, and investigated a potential mechanism underlying the observed effects. DESIGN: HEK293F and CHO-K1 cells transiently transfected with ADAMTS5 cDNA were treated with glucosamine or the related hexosamine mannosamine. Glucosamine effects on FURIN transcription were determined by quantitative RT-PCR. Effects on furin-mediated processing of ADAMTS5 zymogen, and aggrecan processing by glucosamine-treated cells, were determined by western blotting. Post-translational modification of furin and N-glycan deficient furin mutants generated by site-directed mutagenesis was analyzed by western blotting, and the mutants were evaluated for their ADAMTS5 processing ability in furin-deficient CHO-RPE.40 cells. RESULTS: Ten mM glucosamine and 5-10mM mannosamine reduced excision of the ADAMTS5 propeptide, indicating interference with the propeptide excision mechanism, although mannosamine compromised cell viability at these doses. Although glucosamine had no effect on furin mRNA levels, western blot of furin from glucosamine-treated cells suggested altered post-translational modification. Glucosamine treatment led to decreased glycosylation of cellular furin, with reduced furin autoactivation as the consequence. Recombinant furin treated with peptide N-glycanase F had reduced activity against a synthetic peptide substrate. Indeed, site-directed mutagenesis of two furin N-glycosylation sites, Asn(387) and Asn(440), abrogated furin activation and this mutant was unable to rescue ADAMTS5 processing in furin-deficient cells. CONCLUSIONS: Ten mM glucosamine reduces excision of the ADAMTS5 propeptide via interference with post-translational modification of furin and leads to reduced aggrecanase activity of ADAMTS5.


Asunto(s)
Proteínas ADAM/efectos de los fármacos , Furina/efectos de los fármacos , Glucosamina/metabolismo , Proteína ADAMTS5 , Western Blotting , Células Cultivadas , Humanos , Procesamiento Proteico-Postraduccional , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadística como Asunto
18.
Osteoarthritis Cartilage ; 18(1): 106-16, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19786147

RESUMEN

OBJECTIVE: Although most studies have focused on the cholesterol-lowering activity of stigmasterol, other bioactivities have been ascribed to this plant sterol compound, one of which is a potential anti-inflammatory effect. To investigate the effects of stigmasterol, a plant sterol, on the inflammatory mediators and metalloproteinases produced by chondrocytes. METHOD: We used a model of newborn mouse chondrocytes and human osteoarthritis (OA) chondrocytes in primary culture stimulated with or without IL-1beta (10 ng/ml), for 18 h. Cells were pre-incubated for 48 h with stigmasterol (20 microg/ml) compared to untreated cells. We initially investigated the presence of stigmasterol in chondrocyte, compared to other phytosterols. We then assessed the role of stigmasterol on the expression of various genes involved in inflammation (IL-6) and cartilage turn-over (MMP-3, -13, ADAMTS-4, -5, type II collagen, aggrecan) by quantitative Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR). Additional experiments were carried out to monitor the production of MMP-3 and prostaglandin E2 (PGE(2)) by specific immuno-enzymatic assays. We eventually looked at the role of stigmasterol on NF-kappaB activation by western blot, using an anti-IkappaBalpha antibody. RESULTS: After 18 h of IL-1beta treatment, MMP-3, MMP-13, ADAMTS-4, but not ADAMTS-5 RNA expression were elevated, as well as MMP-3 and PGE(2) protein levels in mouse and human chondrocytes. Type II collagen and aggrecan mRNA levels were significatively reduced. Pre-incubation of stigmasterol to IL-1beta-treated cells significantly decreased these effects described above (significant reduction of MMP-3 mRNA in human and mouse, MMP-3 protein in mouse, MMP-13 mRNA in mouse and human, ADAMTS-4 mRNA in human, PGE(2) protein in human and mouse) Finally, stigmasterol was capable of counteracting the IL-1beta-induced NF-kappaB pathway. CONCLUSION: This study shows that stigmasterol inhibits several pro-inflammatory and matrix degradation mediators typically involved in OA-induced cartilage degradation, at least in part through the inhibition of the NF-kappaB pathway. These promising results justify further ex vivo and in vivo investigations with stigmasterol.


Asunto(s)
Proteínas ADAM/metabolismo , Condrocitos/efectos de los fármacos , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Osteoartritis de la Rodilla/metabolismo , Estigmasterol/farmacología , Proteínas ADAM/efectos de los fármacos , Proteína ADAMTS5 , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Muerte Celular , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Condrocitos/inmunología , Condrocitos/metabolismo , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Interleucina-1beta/farmacología , L-Lactato Deshidrogenasa/análisis , Metaloproteinasa 13 de la Matriz/efectos de los fármacos , Metaloproteinasa 3 de la Matriz/efectos de los fármacos , Ratones , Persona de Mediana Edad , Modelos Animales , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Blood Coagul Fibrinolysis ; 20(8): 619-26, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19809308

RESUMEN

The von Willebrand factor (VWF) is elevated in patients with diabetes mellitus and degraded by a metalloprotease, ADAMTS13. We hypothesized that this elevation is due to a decreased function of ADAMTS13. Thus, we investigated ADAMTS13 in patients with diabetes mellitus without and with peripheral artery occlusive disease (PAOD). When treating the latter group with dalteparin, VWF is reported to increase significantly, and we therefore measured ADAMTS13 also in these patients. VWF antigen and ADAMTS13 antigen and activity concentrations were measured in patients with diabetes mellitus but without PAOD (diabetes mellitus; n = 23) and with diabetes mellitus and PAOD (diabetes mellitus + PAOD; n = 65) before and after treatment with dalteparin or placebo. In the diabetes mellitus group, concentration of VWF antigen was significantly higher, whereas that of ADAMTS13 activity was significantly lower than in the healthy controls. In the diabetes mellitus along with PAOD group, VWF antigen was significantly higher, but ADAMTS13 antigen or activity did not differ significantly from those of healthy controls. The ADAMTS13 activity/antigen ratio was lower than in controls only in the diabetes mellitus patient group. VWF antigen increased significantly during dalteraprin treatment, whereas ADAMTS13 activity and antigen remained unchanged. Only patients with diabetes mellitus had significantly lower concentrations of ADAMTS13 activity in plasma than controls, although the diabetes mellitus along with PAOD had a more pronounced VWF antigen elevation than diabetes mellitus patients, illustrating a possible link between ADAMTS13 and microangiopathy in diabetes mellitus patients. The increase in VWF antigen concentration during treatment with dalteparin does not seem to be due to changes in ADAMTS13.


Asunto(s)
Proteínas ADAM/sangre , Arteriopatías Oclusivas/etiología , Diabetes Mellitus/sangre , Factor de von Willebrand/análisis , Proteínas ADAM/efectos de los fármacos , Proteína ADAMTS13 , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Dalteparina , Complicaciones de la Diabetes/etiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factor de von Willebrand/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...