Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Org Biomol Chem ; 17(46): 9913-9923, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31720670

RESUMEN

Nucleotide pyrophosphatase/phosphodiesterase-1 (NPP1) inhibitors have been suggested as a potential treatment for calcium pyrophosphate dihydrate (CPPD) deposition disease. Here, we targeted the development of improved NPP1 inhibitors based on acyclic mimics of Pα,α-phosphorodithioate-substituted adenine nucleotides, 7-10. The latter were obtained in a facile two-step synthesis from adenine-(methoxy)ethanol. Among analogs 7-10, adenine-(methoxy)ethoxy-Pα,α-dithio-triphosphate, 8, was the most potent NPP1 inhibitor both with purified enzyme (IC50 0.645 µM) and in osteoarthritic human chondrocytes (IC50 0.033 µM). Furthermore, it efficaciously (10-fold vs. control) inhibited ATP-induced CPPD in human articular chondrocytes. Importantly, 8 was a highly selective NPP1 inhibitor which showed only minor inhibition of NPP3, CD39 and CD73, and did not inhibit TNAP (tissue nonspecific alkaline phosphatase) activity in human chondrocytes. Furthermore, 8 did not activate P2Y1,2,6 receptors. Analog 8 was not toxic to cultured chondrocytes at 100 µM. Therefore, 8 may be suitable for further development as a drug candidate for the treatment of CPPD arthritis and other NPP1-related diseases.


Asunto(s)
Adenina/farmacología , Pirofosfato de Calcio/antagonistas & inhibidores , Condrocitos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Osteoartritis de la Rodilla/tratamiento farmacológico , Polifosfatos/farmacología , Pirofosfatasas/antagonistas & inhibidores , Compuestos de Sulfhidrilo/farmacología , Adenina/síntesis química , Adenina/química , Pirofosfato de Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Hidrolasas Diéster Fosfóricas/metabolismo , Polifosfatos/química , Pirofosfatasas/metabolismo , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/química
2.
PLoS One ; 7(7): e40272, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792258

RESUMEN

INTRODUCTION: Treatment for osteoporosis commonly includes the use of bisphosphonates. Serious side effects of these drugs are caused by the inhibition of bone resorption as a result of osteoclast apoptosis. Treatment using calcitonin along with bisphosphonates overcomes these side-effects in some patients. Calcitonin is known to inhibit bone resorption without reducing the number of osteoclasts and is thought to prolong osteoclast survival through the inhibition of apoptosis. Further understanding of how calcitonin inhibits apoptosis could prove useful to the development of alternative treatment regimens for osteoporosis. This study aimed to analyze the mechanism by which calcitonin influences osteoclast apoptosis induced by a bisphosphate analog, sintered dicalcium pyrophosphate (SDCP), and to determine the effects of co-treatment with calcitonin and SDCP on apoptotic signaling in osteoclasts. METHODS: Isolated osteoclasts were treated with CT, SDCP or both for 48 h. Osteoclast apoptosis assays, pit formation assays, and tartrate-resistant acid phosphatase (TRAP) staining were performed. Using an osteoporosis rat model, ovariectomized (OVX) rats received calcitonin, SDCP, or calcitonin + SDCP. The microarchitecture of the fifth lumbar trabecular bone was investigated, and histomorphometric and biochemical analyses were performed. RESULTS: Calcitonin inhibited SDCP-induced apoptosis in primary osteoclast cultures, increased Bcl-2 and Erk activity, and decreased Mcl-1 activity. Calcitonin prevented decreased osteoclast survival but not resorption induced by SDCP. Histomorphometric analysis of the tibia revealed increased bone formation, and microcomputed tomography of the fifth lumbar vertebrate showed an additive effect of calcitonin and SDCP on bone volume. Finally, analysis of the serum bone markers CTX-I and P1NP suggests that the increased bone volume induced by co-treatment with calcitonin and SDCP may be due to decreased bone resorption and increased bone formation. CONCLUSIONS: Calcitonin reduces SDCP-induced osteoclast apoptosis and increases its efficacy in an in vivo model of osteoporosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Calcitonina/farmacología , Pirofosfato de Calcio/farmacología , Osteoclastos/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Animales , Conservadores de la Densidad Ósea/antagonistas & inhibidores , Pirofosfato de Calcio/antagonistas & inhibidores , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Sistema de Señalización de MAP Quinasas , Osteoclastos/fisiología , Osteoporosis/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley
3.
Curr Med Chem ; 18(14): 2196-203, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21517761

RESUMEN

In this review we consider diseases associated with pathological mineralization/ossification, namely, ankylosing spondylitis (AS), osteoarthritis (OA), generalized artery calcification of infancy (GACI), vascular calcification as well as chondrocalcinosis (CC) and pseudo gout. Deciphering the key enzymes implicated in the calcification process is an objective of prime importance and the ultimate goal is to synthesize inhibitors of these enzymes in order to provide efficient alternate therapeutic strategies that will slow down the pathologic mineralization and complement the arsenal of anti-inflammatory drugs. One of the difficulties in the definition of diseases associated with pathologic mineralization/ossification lies in the controversial relationship between the type of calcification and the nature of the disease. Here, we propose to clarify this relationship by making a distinction between diseases associated with hydroxyapatite (HA) and calcium pyrophosphate dihydrate (CPPD) deposits. AS, OA, GACI and vascular calcification are usually characterized by mineralization/ossification associated with HA deposits, while CC and pseudo gout are mostly characterized by CPPD deposits. Although both HA and CPPD deposits may occur concomitantly, as in chronic pyrophosphate arthritis or in OA with CPPD, they are formed as a result of two antagonistic processes indicating that treatment of distinct diseases can be only achieved by disease-specific drug therapies. The hydrolysis of PPi, an inhibitor of HA formation, is mostly controlled by tissue non-specific alkaline phosphatase TNAP, while PPi production in the extracellular medium is controlled by ANK, a PPi transporter, and/or NPP1 which generates PPi from nucleotide triphosphates. Low PPi concentration may lead to a preferential deposition of HA while high PPi concentration will favor the formation of CPPD deposits. Thus, HA and CCPD deposition cannot occur concomitantly because they are determined by the Pi/PPi ratio which, in turn, depends on the relative activities of antagonistic enzymes, TNAP hydrolyzing PPi or ANK and NPP1 producing PPi. TNAP inhibitors could prevent HA formation in AS, in late OA, in GACI, as well as in vascular calcifications, while ANK or NPP1 inhibitors could slow down CCPD deposition in CC and pseudo gout.


Asunto(s)
Calcinosis/metabolismo , Pirofosfato de Calcio/metabolismo , Condrocalcinosis/metabolismo , Durapatita/metabolismo , Osteoartritis/metabolismo , Espondilitis Anquilosante/metabolismo , Enfermedades Vasculares/metabolismo , Fosfatasa Alcalina/antagonistas & inhibidores , Fosfatasa Alcalina/metabolismo , Animales , Calcinosis/tratamiento farmacológico , Calcinosis/enzimología , Pirofosfato de Calcio/antagonistas & inhibidores , Condrocalcinosis/tratamiento farmacológico , Condrocalcinosis/enzimología , Durapatita/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Osteoartritis/tratamiento farmacológico , Osteoartritis/enzimología , Espondilitis Anquilosante/tratamiento farmacológico , Espondilitis Anquilosante/enzimología , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/enzimología
4.
J Rheumatol ; 36(12): 2758-65, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19884277

RESUMEN

OBJECTIVE: The enzymatic activities of tissue-nonspecific alkaline phosphatase (TNAP) including capacity to inhibit calcium pyrophosphate dihydrate (CPPD) crystal dissolution are known to be inhibited by endogenous amino acids, notably cysteine. As cysteine is recognized as a strong TNAP inhibitor, we investigated whether cysteine-related metabolites such as mercaptopyruvate (MPA) could show similar enzyme inhibition effects and, if so, whether these effects might be synergistic with cysteine at approximate physiologic concentrations of the amino acids. METHODS: We studied the inhibitory effects of MPA as well as MPA and cysteine combined in equimolar concentrations on TNAP's phosphatase, inorganic pyrophosphatase, and CPPD crystal dissolution activities. Kinetic parameters V(max), K(M), concentration for 50% inhibition (I(50)), inhibitor constant (K(I)), and specific activities calculated from initial velocity, Eadie-Hofstee, Simple, Dixon, and secondary plots were used to assess enzyme inhibition. RESULTS: MPA significantly inhibited TNAP's phosphatase and pyrophosphatase activities at 10x and 100x physiological concentrations. In the presence of calcium [Ca(2+)] and [Mg(2+)] = 1 mM, MPA inhibited uncompetitively TNAP's phosphatase activity and inhibited noncompetitively its pyrophosphatase activity. CPPD crystal dissolution activity was also inhibited. Cysteine and MPA together in equimolar concentrations inhibited TNAP enzyme activities and CPPD crystal dissolution much more effectively than MPA or cysteine alone, reducing CPPD dissolution to 38% of controls at approximate physiologic inhibitor concentrations. CONCLUSION: Endogenous amino acids like cysteine and its derivative MPA have the capacity to inhibit TNAP activities at physiologic concentrations. Downregulation of their inhibiting concentration in the cartilage interstitial fluid environment may provide a therapeutic avenue to controlled dissolution of CPPD crystal deposition in tissues.


Asunto(s)
Fosfatasa Alcalina/antagonistas & inhibidores , Pirofosfato de Calcio , Condrocalcinosis , Cisteína/análogos & derivados , Isoenzimas/antagonistas & inhibidores , Fosfatasa Alcalina/metabolismo , Pirofosfato de Calcio/antagonistas & inhibidores , Pirofosfato de Calcio/química , Pirofosfato de Calcio/metabolismo , Condrocalcinosis/patología , Condrocalcinosis/fisiopatología , Cisteína/química , Cisteína/metabolismo , Humanos , Isoenzimas/metabolismo , Estructura Molecular
5.
J Biol Chem ; 272(30): 18920-5, 1997 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-9228071

RESUMEN

Calcium deposition diseases caused by calcium pyrophosphate dihydrate (CPPD) and basic calcium phosphate (BCP) crystals are a significant source of morbidity in the elderly. We have shown previously that both types of crystals can induce mitogenesis, as well as metalloproteinase synthesis and secretion by fibroblasts and chondrocytes. These responses may promote degradation of articular tissues. We have also shown previously that both CPPD and BCP crystals activate expression of the c-fos and c-jun proto-oncogenes. Phosphocitrate (PC) can specifically block mitogenesis and proto-oncogene expression induced by either BCP or CPPD crystals in 3T3 cells and human fibroblasts, suggesting that PC may be an effective therapy for calcium deposition diseases. To understand how PC inhibits BCP and CPPD-mediated cellular effects, we have investigated the mechanism by which BCP and CPPD transduce signals to the nucleus. Here we demonstrate that BCP and CPPD crystals activate a protein kinase signal transduction pathway involving p42 and p44 mitogen-activated protein (MAP) kinases (ERK 2 and ERK 1). BCP and CPPD also cause phosphorylation of a nuclear transcription factor, cyclic AMP response element-binding protein (CREB), on serine 133, a residue essential for CREB's ability to transactivate. Treatment of cells with PC at concentrations of 10(-3) to 10(-5) M blocked both the activation of p42/p44 MAP kinases, and CREB serine 133 phosphorylation, in a dose-dependent fashion. At 10(-3) M, a PC analogue, n-sulfo-2-aminotricarballylate and citrate also modulate this signal transduction pathway. Inhibition by PC is specific for BCP- and CPPD-mediated signaling, since all three compounds had no effect on serum-induced p42/P44 or interleukin-1beta induced p38 MAP kinase activities. Treatment of cells with an inhibitor of MEK1, an upstream activator of MAPKs, significantly inhibited crystal-induced cell proliferation, suggesting that the MAPK pathway is a significant mediator of crystal-induced signals.


Asunto(s)
Fosfatos de Calcio/farmacología , Pirofosfato de Calcio/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Citratos/farmacología , Proteínas Quinasas Activadas por Mitógenos , Transducción de Señal , Fosfatos de Calcio/antagonistas & inhibidores , Pirofosfato de Calcio/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , División Celular , Células Cultivadas , Cristalización , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Humanos , Interleucina-1/farmacología , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Fosforilación , Proto-Oncogenes Mas , Transducción de Señal/efectos de los fármacos , Ácidos Tricarboxílicos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos
6.
Immunology ; 90(4): 502-10, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9176102

RESUMEN

The activation of MAP kinase in human neutrophils stimulated by both uncoated and plasma-opsonized crystals of triclinic calcium pyrophosphate dihydrate (CPPD) was investigated. The effect of taxol on MAP kinase activation and on the responses of neutrophils stimulated by plasma-opsonized crystals was determined. MAP kinase activation was identified and quantified in Mono Q chromatography separated fractions of neutrophils that had been incubated with CPPD crystals by measuring [gamma-32P]adenosine triphosphate (ATP) phosphorylation of myelin basic protein and using immunoblotting techniques. Human neutrophils were incubated with taxol (0-50 microM), added to plasma-opsonized CPPD (50 mg/ml) and MAP kinase activation, chemiluminescence, superoxide anion generation, lysozyme and myeloperoxidase release were monitored. Both uncoated and plasma coated CPPD crystals induced a large increase in MAP kinase activity in neutrophils over control levels within 1 min of incubation. Pretreatment of neutrophils with taxol was able to suppress this activation of MAP kinase. Taxol produced a concentration-dependent inhibition of opsonized CPPD-induced neutrophil chemiluminescence, superoxide anion production and myeloperoxide release. Taxol at 28 microM also significantly inhibited chemiluminescence, superoxide anion production and myeloperoxidase release from neutrophils stimulated by opsonized zymosan. This is the first report of crystal-induced activation of MAP kinase in neutrophils. Microtubule-associated processes, such as signal transduction, secretion and phagocytosis are involved in particulate-induced neutrophil responses. We have suggested that the inhibitory effect of taxol observed in this work is due to its stabilizing effect on microtubules and disruption of MAP kinase activation associated with microtubules.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Pirofosfato de Calcio/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Activación Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Paclitaxel/farmacología , Superóxidos/sangre , Western Blotting , Pirofosfato de Calcio/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/sangre , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Técnicas de Cultivo de Célula , Degranulación de la Célula/efectos de los fármacos , Cristalización , Humanos , Mediciones Luminiscentes , Activación Neutrófila/fisiología , Neutrófilos/enzimología
7.
Biochem Med Metab Biol ; 41(1): 56-63, 1989 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2540780

RESUMEN

The protection afforded by phosphocitrate, a phosphorylated polycarboxylic acid, against crystal-induced membrane damage to polymorphonuclear leukocytes was studied in vitro. Membranolysis was assessed by nitro blue tetrazolium salt reduction, lactate dehydrogenase release, and scanning electron microscopy. Phosphocitrate protected strongly against hydroxyapatite crystal-induced damage, an action attributable to crystal surface binding of phosphocitrate rather than to the membrane. The ability of phosphocitrate to prevent hydroxyapatite crystallization, together with its membrane protective effect against preformed crystals, would suggest that the compound might have a useful future role against crystal-induced arthropathies.


Asunto(s)
Apatitas/toxicidad , Pirofosfato de Calcio/toxicidad , Citratos/farmacología , Difosfatos/toxicidad , Neutrófilos/efectos de los fármacos , Apatitas/antagonistas & inhibidores , Pirofosfato de Calcio/antagonistas & inhibidores , Membrana Celular/efectos de los fármacos , Membrana Celular/ultraestructura , Humanos , Microscopía Electrónica , Neutrófilos/ultraestructura
8.
J Rheumatol ; 15(2): 321-4, 1988 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2834554

RESUMEN

To determine the physical-chemical effects of [Fe++] and [Fe ] on calcium pyrophosphate dihydrate (CPPD) crystal formation de novo, we studied CPPD crystal formation in an established model aqueous solution mixture system using physiological concentrations of Na+, Mg++, Ca++, C1-. We found that [Fe++] greater than 1 microM or [Fe ] greater than or equal to 100 microM inhibits CPPD crystal formation under conditions of [Ca++] = 1.5 mM, [Pi] = 0.1 mM, and [PPi] = 0.1 mM that simulate CPPD formation in vivo. These experiments suggest that at biological concentrations, Fe++ acts to inhibit CPPD formation but that [Fe++] depletion therapy by removal of inhibition effects may facilitate CPPD crystal formation in articular tissues.


Asunto(s)
Pirofosfato de Calcio/antagonistas & inhibidores , Condrocalcinosis/metabolismo , Difosfatos/antagonistas & inhibidores , Compuestos Ferrosos/farmacología , Hemocromatosis/metabolismo , Pirofosfato de Calcio/biosíntesis , Cristalización , Compuestos Férricos/farmacología , Humanos
9.
Calcif Tissue Int ; 42(1): 46-52, 1988 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-2834030

RESUMEN

Proteoglycans are recognized to inhibit calcium pyrophosphate dihydrate (CPPD) crystal formation but the mechanisms are not known. To study the role of carboxylate (-CO2-) ligands, the possible inhibitor effects of sodium acetate, sodium D-glucuronate, disodium malate, and trisodium citrate were studied using solution mixtures containing [Ca2+] = 1.5 mM, [Mg2+] = 0.5 mM, [PPi] = 0.1 mM, [Pi] = 0.1 mM, [Na+] = 140 mM, 37 degrees C, pH 7.4 with or without 9.5 +/- 0.5 mg CPPD (seed) crystals. These studies showed that monocarboxylates (acetate, glucuronate) have little inhibitive effect. Progressively greater inhibition was found with dicarboxylate (malate) and tricarboxylate (citrate) indicating that the arrangement of carboxylate (and sulfate) ligands on proteoglycan is more important than the inhibitory effect of individual ligands.


Asunto(s)
Pirofosfato de Calcio/metabolismo , Ácidos Carboxílicos/farmacología , Difosfatos/metabolismo , Calcio/análisis , Pirofosfato de Calcio/antagonistas & inhibidores , Cristalización , Iones , Ligandos , Proteoglicanos/farmacología
10.
Br J Pharmacol ; 90(2): 413-9, 1987 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3030485

RESUMEN

Monosodium urate (MSU) and calcium pyrophosphate dihydrate (CPPD) crystals initiated acute inflammatory reactions characterized by increased plasma extravasation and polymorphonuclear leukocyte (PMNL) accumulation in the rat subcutaneous air-pouch. Pretreatment of rats with colchicine (1 mg kg-1, s.c.) inhibited PMNL accumulation induced by either crystal type but had a greater inhibitory effect on MSU-induced plasma extravasation compared with that induced by CPPD crystals. Colchicine (1 mg kg-1, s.c.) did not reduce histamine-induced plasma extravasation in the air-pouch. The lipoxygenase product of arachidonic acid metabolism, leukotriene B4 (LTB4), was detected in MSU-induced exudates but not in CPPD-induced exudates. Pretreatment of rats with colchicine (1 mg kg-1, s.c.) inhibited LTB4 production in MSU-induced exudates.


Asunto(s)
Antiinflamatorios , Pirofosfato de Calcio/toxicidad , Colchicina/farmacología , Difosfatos/toxicidad , Inflamación/inducido químicamente , Ácido Úrico/toxicidad , Animales , Pirofosfato de Calcio/antagonistas & inhibidores , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/prevención & control , Leucotrieno B4/metabolismo , Masculino , Neutrófilos/efectos de los fármacos , Ratas , Ratas Endogámicas , Ácido Úrico/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA