Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 339
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Chem Rev ; 123(10): 6612-6667, 2023 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-37071737

RESUMEN

The gold drugs, gold sodium thiomalate (Myocrisin), aurothioglucose (Solganal), and the orally administered auranofin (Ridaura), are utilized in modern medicine for the treatment of inflammatory arthritis including rheumatoid and juvenile arthritis; however, new gold agents have been slow to enter the clinic. Repurposing of auranofin in different disease indications such as cancer, parasitic, and microbial infections in the clinic has provided impetus for the development of new gold complexes for biomedical applications based on unique mechanistic insights differentiated from auranofin. Various chemical methods for the preparation of physiologically stable gold complexes and associated mechanisms have been explored in biomedicine such as therapeutics or chemical probes. In this Review, we discuss the chemistry of next generation gold drugs, which encompasses oxidation states, geometry, ligands, coordination, and organometallic compounds for infectious diseases, cancer, inflammation, and as tools for chemical biology via gold-protein interactions. We will focus on the development of gold agents in biomedicine within the past decade. The Review provides readers with an accessible overview of the utility, development, and mechanism of action of gold-based small molecules to establish context and basis for the thriving resurgence of gold in medicine.


Asunto(s)
Artritis Reumatoide , Auranofina , Humanos , Auranofina/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Antiinflamatorios/farmacología , Oro , Aurotioglucosa/farmacología , Aurotioglucosa/uso terapéutico , Tiomalato Sódico de Oro/farmacología , Tiomalato Sódico de Oro/uso terapéutico
2.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34502400

RESUMEN

Giardiasis represents a latent problem in public health due to the exceptionally pathogenic strategies of the parasite Giardia lamblia for evading the human immune system. Strains resistant to first-line drugs are also a challenge. Therefore, new antigiardial therapies are urgently needed. Here, we tested giardial arginine deiminase (GlADI) as a target against giardiasis. GlADI belongs to an essential pathway in Giardia for the synthesis of ATP, which is absent in humans. In silico docking with six thiol-reactive compounds was performed; four of which are approved drugs for humans. Recombinant GlADI was used in enzyme inhibition assays, and computational in silico predictions and spectroscopic studies were applied to follow the enzyme's structural disturbance and identify possible effective drugs. Inhibition by modification of cysteines was corroborated using Ellman's method. The efficacy of these drugs on parasite viability was assayed on Giardia trophozoites, along with the inhibition of the endogenous GlADI. The most potent drug against GlADI was assayed on Giardia encystment. The tested drugs inhibited the recombinant GlADI by modifying its cysteines and, potentially, by altering its 3D structure. Only rabeprazole and omeprazole decreased trophozoite survival by inhibiting endogenous GlADI, while rabeprazole also decreased the Giardia encystment rate. These findings demonstrate the potential of GlADI as a target against giardiasis.


Asunto(s)
Giardia lamblia/efectos de los fármacos , Giardiasis/tratamiento farmacológico , Hidrolasas/metabolismo , Animales , Antiprotozoarios/farmacología , Simulación por Computador , Cisteína/química , Evaluación Preclínica de Medicamentos/métodos , Reposicionamiento de Medicamentos/métodos , Giardia lamblia/patogenicidad , Giardiasis/inmunología , Tiomalato Sódico de Oro/farmacología , Humanos , Hidrolasas/efectos de los fármacos , Hidrolasas/ultraestructura , Omeprazol/farmacología , Inhibidores de la Bomba de Protones/farmacología , Rabeprazol , Tiamina/análogos & derivados , Tiamina/farmacología , Trofozoítos/efectos de los fármacos
3.
Bull Exp Biol Med ; 169(4): 426-430, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32889566

RESUMEN

Suppression of the production of granulocytic CSF under the effect of 5-fluorouracyl is related to disorders in the NF-κB-, cAMP-dependent signaling pathways and MAPK cascade. These secondary messengers are involved in the regulation of functional activity of nonadherent myelokaryocytes starting from day 10 of the experiment (initial period of the hemopoietic granulocytic stem regeneration after antimetabolite challenge). Granulocytic CSF does not play essential role in the formation of colony-stimulating activity of cells of the adherent and nonadherent fractions of the bone marrow. Only cAMP-dependent pathway is involved in the regulation of the realization of the granulocytic precursor growth potential in response to the challenge.


Asunto(s)
Citostáticos/farmacología , Fluorouracilo/farmacología , Factor Estimulante de Colonias de Granulocitos/genética , Granulocitos/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , FN-kappa B/genética , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Didesoxiadenosina/análogos & derivados , Didesoxiadenosina/farmacología , Regulación de la Expresión Génica , Tiomalato Sódico de Oro/farmacología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Granulocitos/citología , Granulocitos/metabolismo , Hematopoyesis/genética , Imidazoles/farmacología , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Piridinas/farmacología , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Bull Exp Biol Med ; 169(6): 759-764, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33098513

RESUMEN

The role of NF-κВ in the realization of the growth potential of neural progenitor cells from the subventricular area of cerebral hemispheres and secretion of neurotrophins by glial elements was studied under conditions of in vitro and in vivo modeled ethanol-induced neurodegeneration. It was found that this transcription factor does not participate in the regulation of mitotic activity of neural stem cells and neuronal-committed progenitors under optimal conditions and under the influence of ethanol in vitro. At the same time, NF-κВ suppresses differentiation/maturation of neural progenitor cells. Long-term peroral administration of ethanol to mice was accompanied by the inhibitory influence of NF-κВ on proliferation of progenitor cells. Blockade of NF-κВ in neural stem cells and committed neuronal precursors in animals with neurodegeneration induced cell cycle progression in these elements. The involvement of NF-κВ in the secretory function of astrocytes and oligodendrogliocytes was established. Inactivation of the nuclear transcription factor reduced the production of neurotrophins, in particular, in the case of ethanol exposure. At the same time, no changes in the function of microglia were noted.


Asunto(s)
Ventrículos Laterales/efectos de los fármacos , FN-kappa B/genética , Células-Madre Neurales/efectos de los fármacos , Enfermedades Neurodegenerativas/genética , Regeneración/efectos de los fármacos , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Etanol/farmacología , Regulación de la Expresión Génica , Tiomalato Sódico de Oro/farmacología , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , FN-kappa B/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Cultivo Primario de Células , Regeneración/genética , Transducción de Señal
5.
Bull Exp Biol Med ; 166(3): 344-347, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30627910

RESUMEN

The role of signaling molecules in synthesis of humoral regulators of granulocytopoiesis by the hematopoietic microenvironmental cells during stress was analyzed using specific inhibitors. The major role in stimulation of the synthesis of granulocytic CSF during stressful stimulation is played by PI3K/Akt signaling cascade. Nuclear transcription factor NF-κB plays an auxiliary role in the regulation of functional activity of the bone marrow mononuclears. However, this factor affects the synthesis of granulocytic CSF by CD4+ cells of the bone marrow in response to stressful stimulation. Different degree and specific character of involvement of the signaling proteins in the regulation of the production of humoral factors determining colony-stimulating activity are explained by changes in functional state of monocyte-derived macrophages in different periods of stress response.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/genética , Granulocitos/inmunología , FN-kappa B/genética , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal/inmunología , Estrés Psicológico/genética , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Cromonas/farmacología , Flavonoides/farmacología , Regulación de la Expresión Génica , Tiomalato Sódico de Oro/farmacología , Factor Estimulante de Colonias de Granulocitos/inmunología , Granulocitos/efectos de los fármacos , Granulocitos/patología , Imidazoles/farmacología , Inmovilización/métodos , Leucopoyesis/efectos de los fármacos , Leucopoyesis/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Morfolinas/farmacología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Piridinas/farmacología , Estrés Psicológico/inmunología , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
6.
Scand J Rheumatol ; 44(1): 74-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25314295

RESUMEN

OBJECTIVES: Microsomal prostaglandin E synthase-1 (mPGES-1) is a terminal enzyme in the production of prostaglandin E2 (PGE2) and its expression is upregulated during inflammation. mPGES-1 is considered as a potential drug target for the treatment of arthritis to reduce adverse effects related to the current non-steroidal anti-inflammatory drugs (NSAIDs). Our aim was to study the expression of mPGES-1 in primary human chondrocytes and whether the expression is affected by clinically used antirheumatic drugs. METHOD: Primary human chondrocytes were isolated from cartilage samples obtained from patients undergoing total knee replacement surgery. Expression of mPGES-1 was studied by quantitative real-time polymerase chain reaction (PCR) and Western blot analysis. PGE2 levels were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: mPGES-1 expression in primary human chondrocytes was enhanced when the cells were exposed to interleukin-1ß (IL-1ß) and mPGES-1 protein levels continued to increase up to the 96-h follow-up. Aurothiomalate inhibited mPGES-1 expression and PGE2 production in a dose-dependent manner, as did the anti-inflammatory steroid dexamethasone. Other disease-modifying antirheumatic drugs (DMARDs) studied (sulfasalazine, methotrexate, and hydroxychloroquine) did not alter mPGES-1 expression. CONCLUSIONS: The results introduce aurothiomalate as the first, and so far the only, DMARD found to be able to inhibit mPGES-1 expression. The effect is likely involved in the mechanisms of action of this gold-containing DMARD in rheumatic diseases. The results are implicated in the regulatory mechanisms of mPGES-1 expression, which are under intensive research.


Asunto(s)
Antirreumáticos/farmacología , Condrocitos/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Tiomalato Sódico de Oro/farmacología , Oxidorreductasas Intramoleculares/genética , Condrocitos/citología , Condrocitos/fisiología , Dexametasona/farmacología , Dinoprostona/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Glucocorticoides/farmacología , Humanos , Hidroxicloroquina/farmacología , Oxidorreductasas Intramoleculares/metabolismo , Metotrexato/farmacología , Osteoartritis de la Rodilla/tratamiento farmacológico , Cultivo Primario de Células , Prostaglandina-E Sintasas , Reacción en Cadena en Tiempo Real de la Polimerasa , Sulfasalazina/farmacología
7.
Bull Exp Biol Med ; 158(5): 624-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25778647

RESUMEN

We studied the role of NF-κB/IKK-mediated signaling in the stimulation of growth potential of mesenchymal progenitor cells by alkaloid songorine in vitro. Specific NF-κB inhibitor oridonin abolished activation of proliferation and differentiation of progenitor cells. Aurothiomalate, a selective blocker of IKK-2, also suppressed mitotic activity of fibroblast precursors, but had no effect on the rate of the differentiation.


Asunto(s)
Alcaloides/farmacología , Quinasa I-kappa B/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , FN-kappa B/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Diterpenos de Tipo Kaurano/farmacología , Tiomalato Sódico de Oro/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Ratones , Modelos Biológicos , FN-kappa B/antagonistas & inhibidores , Medicina Regenerativa
8.
Anticancer Drugs ; 25(3): 332-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24304691

RESUMEN

Osteosarcoma is a highly fatal cancer, with most patients ultimately succumbing to metastatic disease. The purpose of this study was to evaluate the effects of the antirheumatoid drug aurothiomalate on canine and human osteosarcoma cells and on canine osteosarcoma growth and metastasis in a mouse xenograft model. We hypothesized that aurothiomalate would decrease osteosarcoma cell survival, tumor cellular proliferation, tumor growth, and metastasis. After performing clonogenic assays, aurothiomalate or a placebo was administered to 54 mice inoculated with canine osteosarcoma. Survival, tumor growth, embolization, metastasis, histopathology, cell proliferation marker Ki67, and apoptosis marker caspase-3 were compared between groups. Statistical analysis was carried out using the Kaplan-Meier method with the log-rank test and one-way analysis of variance with the Tukey's test or Dunn's method. Aurothiomalate caused dose-dependent inhibition of osteosarcoma cell survival (P<0.001) and decreased tumor growth (P<0.001). Pulmonary macrometastasis and Ki67 labeling were reduced with low-dose aurothiomalate (P=0.033 and 0.005, respectively), and tumor emboli and pulmonary micrometastases were decreased with high-dose aurothiomalate (P=0.010 and 0.011, respectively). There was no difference in survival, tumor development, ulceration, mitotic indices, tumor necrosis, nonpulmonary metastases, and caspase-3 labeling. Aurothiomalate treatment inhibited osteosarcoma cell survival and reduced tumor cell proliferation, growth, embolization, and pulmonary metastasis. Given aurothiomalate's established utility in canine and human medicine, our results suggest that this compound may hold promise as an adjunctive therapy for osteosarcoma. Further translational research is warranted to better characterize the dose response of canine and human osteosarcoma to aurothiomalate.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Tiomalato Sódico de Oro/uso terapéutico , Osteosarcoma/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Óseas/patología , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Perros , Tiomalato Sódico de Oro/farmacología , Humanos , Antígeno Ki-67/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Ratones , Osteosarcoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Bull Exp Biol Med ; 157(1): 32-6, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24915945

RESUMEN

The study examines the role of signal pathways in triggering the compensatory reactions in the blood system in response to the cytostatic administration. In vitro experiments elucidated the involvement of protein kinase p38 in the synthesis of granulocytic CSF by cells of hemopoietic microenvironment. The important role of the transcriptional factor NF-κB and protein kinase p38 in limitation of the processes of maturation of the hemopoietic precursors was demonstrated.


Asunto(s)
Células de la Médula Ósea/metabolismo , Ciclofosfamida/farmacología , Agonistas Mieloablativos/farmacología , FN-kappa B/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Células Eritroides/patología , Regulación de la Expresión Génica , Tiomalato Sódico de Oro/farmacología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos/metabolismo , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Granulocitos/patología , Hematopoyesis/efectos de los fármacos , Imidazoles/farmacología , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos CBA , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Cultivo Primario de Células , Piridinas/farmacología , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
10.
Microbiol Spectr ; 12(2): e0296823, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38206030

RESUMEN

Auranofin, an FDA-approved drug for rheumatoid arthritis, has emerged as a promising antiparasitic medication in recent years. The gold(I) ion in auranofin is postulated to be responsible for its antiparasitic activity. Notably, aurothiomalate and aurothioglucose also contain gold(I), and, like auranofin, they were previously used to treat rheumatoid arthritis. Whether they have antiparasitic activity remains to be elucidated. Herein, we demonstrated that auranofin and similar derivatives, but not aurothiomalate and aurothioglucose, inhibited the growth of Toxoplasma gondii in vitro. We found that auranofin affected the T. gondii biological cycle (lytic cycle) by inhibiting T. gondii's invasion and triggering its egress from the host cell. However, auranofin could not prevent parasite replication once T. gondii resided within the host. Auranofin treatment induced apoptosis in T. gondii parasites, as demonstrated by its reduced size and elevated phosphatidylserine externalization (PS). Notably, the gold from auranofin enters the cytoplasm of T. gondii, as demonstrated by scanning transmission electron microscopy-energy dispersive X-ray spectroscopy (STEM-EDS) and Inductively Coupled Plasma-Mass Spectrometry (ICP-MS).IMPORTANCEToxoplasmosis, caused by Toxoplasma gondii, is a devastating disease affecting the brain and the eyes, frequently affecting immunocompromised individuals. Approximately 60 million people in the United States are already infected with T. gondii, representing a population at-risk of developing toxoplasmosis. Recent advances in treating cancer, autoimmune diseases, and organ transplants have contributed to this at-risk population's exponential growth. Paradoxically, treatments for toxoplasmosis have remained the same for more than 60 years, relying on medications well-known for their bone marrow toxicity and allergic reactions. Discovering new therapies is a priority, and repurposing FDA-approved drugs is an alternative approach to speed up drug discovery. Herein, we report the effect of auranofin, an FDA-approved drug, on the biological cycle of T. gondii and how both the phosphine ligand and the gold molecule determine the anti-parasitic activity of auranofin and other gold compounds. Our studies would contribute to the pipeline of candidate anti-T. gondii agents.


Asunto(s)
Artritis Reumatoide , Fosfinas , Toxoplasma , Toxoplasmosis , Humanos , Auranofina/farmacología , Auranofina/uso terapéutico , Oro/farmacología , Oro/uso terapéutico , Ligandos , Aurotioglucosa/farmacología , Aurotioglucosa/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Tiomalato Sódico de Oro/farmacología , Tiomalato Sódico de Oro/uso terapéutico , Antiparasitarios/farmacología , Antiparasitarios/uso terapéutico
11.
Inflammopharmacology ; 21(4): 291-300, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23686086

RESUMEN

This article discusses the bizarre and contrary effects of thiocyanate, the major detoxication product of hydrogen cyanide inhaled from tobacco smoke or liberated from cyanogenic foods, e.g. cassava. Thiocyanate both (1) promotes inflammatory disease in rats and (2) facilitates the anti-inflammatory action of historic metal therapies based on gold (Au) or silver (Ag) in three models of chronic polyarthritis in rats. Low doses of nanoparticulate metallic silver (NMS) preparations, i.e. zerovalent silver (Ag°) administered orally, suppressed the mycobacterial ('adjuvant')-induced arthritis (MIA) in rats. Similar doses of cationic silver, Ag(I), administered orally as silver oxide or soluble silver salts were inactive. By contrast, NMS only inhibited the development of the collagen-induced arthritis (CIA) and pristane-induced arthritis (PIA) in rats when thiocyanate was also co-administered in drinking water. These (a) arthritis-selective and (b) thiocyanate-inducible effects of Ag° were also observed in some previous, and now extended, studies with the classic anti-arthritic drug, sodium aurothiomalate (ATM, Myocrisin(®)) and its silver analogue (STM), administered subcutaneously to rats developing the same three forms of polyarthritis. In the absence of either Ag° or ATM, thiocyanate considerably increased the severity of the MIA, CIA and PIA, i.e. acting as a pro-pathogen. Hitherto, thiocyanate was considered relatively harmless. This may not be true in rats/people with immuno-inflammatory stress and concomitant leukocyte activation. Collectively, these findings show how the drug action of a xenobiotic might be determined by the nature (and severity) of the experimental inflammation, as an example of conditional pharmacology. They also suggest that an incipient toxicity, even of normobiotics such as thiocyanate, might likewise be modulated beneficially by well-chosen xenobiotics (drugs, nutritional supplements, etc.), i.e. conditional toxicology (Powanda 1995). Thus, both the disease and the environment may determine (1) the therapeutic action and/or (2) adverse effect(s) of xenobiotics--and even some normobiotics.


Asunto(s)
Antirreumáticos , Artritis Experimental/tratamiento farmacológico , Tiomalato Sódico de Oro , Plata , Tiocianatos , Animales , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Antirreumáticos/toxicidad , Artritis Experimental/inducido químicamente , Artritis Experimental/microbiología , Sinergismo Farmacológico , Quimioterapia Combinada , Tiomalato Sódico de Oro/farmacología , Tiomalato Sódico de Oro/uso terapéutico , Tiomalato Sódico de Oro/toxicidad , Nanopartículas , Ratas , Plata/farmacología , Plata/uso terapéutico , Plata/toxicidad , Especificidad de la Especie , Tiocianatos/farmacología , Tiocianatos/uso terapéutico , Tiocianatos/toxicidad
12.
Bull Exp Biol Med ; 153(4): 472-4, 2012 Aug.
Artículo en Inglés, Ruso | MEDLINE | ID: mdl-22977847

RESUMEN

We compared the effect of NF-κB inhibitor aurothiomalate and voltaren on local inflammation in different types of immune response. Both substances reduced edema caused by sheep erythrocytes (Th1-type immune response) and local immediate-type hypersensitivity response induced with ovalbumin (Th2-dependent response). The anti-inflammatory effects of aurothiomalate were similar to those of voltaren during Th1-type immune response.


Asunto(s)
Tiomalato Sódico de Oro/farmacología , Hipersensibilidad Tardía/tratamiento farmacológico , Hipersensibilidad Inmediata/tratamiento farmacológico , Inmunidad Celular/inmunología , FN-kappa B/antagonistas & inhibidores , Células TH1/inmunología , Células Th2/inmunología , Análisis de Varianza , Animales , Citocinas/inmunología , Diclofenaco/farmacología , Diclofenaco/uso terapéutico , Eritrocitos , Femenino , Tiomalato Sódico de Oro/uso terapéutico , Hipersensibilidad Tardía/inducido químicamente , Hipersensibilidad Inmediata/inducido químicamente , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/toxicidad
13.
Acta Parasitol ; 67(2): 640-647, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35380401

RESUMEN

PURPOSE: Leishmaniasis is a major public health problem worldwide in many parts of the world. Current anti-leishmanial drugs have only limited clinical efficacy. Aurothiomalate derivatives are useful for treating rheumatoid arthritis, but have emerged as a promising therapeutic candidate for leishmaniasis. This paper gives a review of the literature about the usefulness of aurothiomalate derivatives against leishmaniasis. METHODS: In this study, we reviewed the proposed mechanisms of action of aurothiomalate and related compounds on the metabolism of L. major and collected data by searching relevant articles. RESULTS: Aurothiomalate-based drugs could be effective against leishmaniasis through two direct and indirect mechanisms: first, cytotoxic effects on parasites via thiomalate's false substrate role in the citric acid cycle against malate; and second, immunosuppressive and anti-inflammatory effects of aurothiomalate derivatives with prostaglandin production inhibitory effects. CONCLUSIONS: The current study documented that aurothiomalate-based drugs could be effective against leishmaniasis through two direct and indirect mechanisms of action. Gold thiomalate as a promising hit should be evaluated against L. major in vitro and in vivo conditions in the future.


Asunto(s)
Leishmania major , Leishmaniasis , Tiomalato Sódico de Oro/metabolismo , Tiomalato Sódico de Oro/farmacología , Tiomalato Sódico de Oro/uso terapéutico , Humanos , Leishmaniasis/tratamiento farmacológico
14.
Arthritis Rheum ; 62(6): 1650-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20178133

RESUMEN

OBJECTIVE: Aurothiomalate is a disease-modifying antirheumatic drug that suppresses inflammation and retards cartilage degradation and bone erosion in arthritis. The molecular mechanisms of action of aurothiomalate are not known in detail. MAPK pathways are major signaling pathways in inflammation that regulate the production of many inflammatory and destructive factors in arthritis. The purpose of the present study was to investigate the effects of aurothiomalate on the activity of p38 MAPK and on the expression of MAPK phosphatase 1 (MKP-1), cyclooxygenase 2 (COX-2), matrix metalloproteinase 3 (MMP-3), and interleukin-6 (IL-6) in immortalized murine H4 chondrocytes and in intact human and murine cartilage. METHODS: Protein expression was examined by Western blotting or by enzyme-linked immunosorbent assay, and messenger RNA (mRNA) expression was examined by real-time reverse transcription-polymerase chain reaction analysis. The mediator role of MKP-1 was investigated by using small interfering RNA (siRNA) methods to down-regulated MKP-1 expression in chondrocytes in culture and by comparing the responses in intact cartilage from MKP-1-deficient and wild-type mice. The effects of aurothiomalate were also confirmed in human rheumatoid cartilage by using tissue samples obtained at the time of total knee replacement surgery. RESULTS: Aurothiomalate inhibited IL-1beta-induced COX-2 expression and prostaglandin E(2) production by destabilizing COX-2 mRNA, as did the p38 MAPK inhibitor SB203580. Interestingly, aurothiomalate also increased the expression of MKP-1 and reduced the IL-1beta-induced phosphorylation of p38 MAPK. Knockdown of MKP-1 by siRNA significantly impaired the ability of aurothiomalate to inhibit the phosphorylation of p38 MAPK and the expression of COX-2, MMP-3, and IL-6. Likewise, aurothiomalate reduced COX-2, MMP-3, and IL-6 expression in articular cartilage from patients with rheumatoid arthritis, as well as in articular cartilage from wild-type mice but not from MKP-1(-/-) mice. CONCLUSION: Our findings indicate a novel mechanism for the antiinflammatory and antierosive actions of aurothiomalate, through increased expression of MKP-1, which leads to reduced activation of p38 MAPK and suppressed expression of COX-2, MMP-3, and IL-6. The results suggest that manipulation of MKP-1 levels is a promising new mechanism to be directed in the search and development of novel antiinflammatory and antierosive compounds that have the good efficacy of gold compounds but not their toxicity.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Ciclooxigenasa 2/metabolismo , Fosfatasa 1 de Especificidad Dual/metabolismo , Tiomalato Sódico de Oro/farmacología , Interleucina-6/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Análisis de Varianza , Animales , Antirreumáticos/farmacología , Western Blotting , Cartílago Articular/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Ciclooxigenasa 2/genética , Fosfatasa 1 de Especificidad Dual/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Regulación de la Expresión Génica , Humanos , Interleucina-6/genética , Metaloproteinasa 3 de la Matriz/genética , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Radioinmunoensayo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas p38 Activadas por Mitógenos/genética
15.
Chem Biodivers ; 8(7): 1274-83, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21766448

RESUMEN

The potential anti- or pro-oxidative effects of a disease-modifying antirheumatic drug, aurothiomalate, to protect high-molar-mass hyaluronan against radical degradation were investigated along with L-glutathione - tested in similar functions. Hyaluronan degradation was induced by the oxidative system Cu(II) plus ascorbate known as the Weissberger's oxidative system. The time- and dose-dependent changes of the dynamic viscosity of the hyaluronan solutions were studied by the method of rotational viscometry. Additionally, the antioxidative activity of aurothiomalate expressed as a radical-scavenging capacity based on a decolorization 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) assay was inspected. At the higher concentrations tested, L-glutathione showed excellent scavenging of (.) OH and peroxyl-type radicals, however, at the lowest concentration applied, its pro-oxidative effect was revealed. The effects of aurothiomalate on hyaluronan degradation were similar to that of L-glutathione, however, at the lowest concentration tested, no significant pro-oxidant effect was observed.


Asunto(s)
Antioxidantes/farmacología , Antirreumáticos/farmacología , Tiomalato Sódico de Oro/farmacología , Ácido Hialurónico/metabolismo , Benzotiazoles , Secuencia de Carbohidratos , Glutatión/farmacología , Ácido Hialurónico/química , Datos de Secuencia Molecular , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Ácidos Sulfónicos/metabolismo , Tiazoles/metabolismo
16.
Bull Exp Biol Med ; 151(2): 190-3, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22238747

RESUMEN

We compared the effects of NF-κB inhibitor aurothiomalate and voltaren on NO production by mouse macrophages in vitro, their ability to cause local edema at the site of injection, and their effect on carrageenan-induced inflammation. High concentrations of aurothiomalate reduced NO production, while in low concentrations both aurothiomalate and voltaren stimulated this process. When injected into mouse footpad, aurothiomalate in a dose >1 mM and voltaren in a dose >1.6 µM induce paw edema. Both compounds suppressed carrageenan-induced inflammation, but the efficacy of aurothiomalate 2-fold exceeded that of voltaren.


Asunto(s)
Antiinflamatorios/farmacología , Tiomalato Sódico de Oro/farmacología , Inflamación/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Animales , Antiinflamatorios/uso terapéutico , Diclofenaco/farmacología , Diclofenaco/uso terapéutico , Femenino , Tiomalato Sódico de Oro/uso terapéutico , Inflamación/inducido químicamente , Inflamación/patología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo
17.
J Exp Med ; 138(2): 461-72, 1973 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-4124211

RESUMEN

The granule fraction of human leukocytes contains neutral protease capable of degrading the noncollagenous protein mucopolysaccharide matrix of cartilage at neutral pH in physiological salt solution. Cartilage degradation was monitored by quantitating the release of (35)S from labeled rabbit ear cartilage. Degradation of cartilage matrix occurs when intact viable human leukocytes are incubated with cartilage opsonized with aggregated human gamma globulin (AHGG). During a similar 4 h incubation period cells did not degrade uncoated cartilage or cartilage coated with nonaggregated gamma globulin. Cells remain viable during the enzyme release process as evidenced by the absence of a cytoplasmic enzyme marker (lactic dehydrogenase) in the supernatant and dye exclusion studies. The release of (35)S from labeled cartilage by human leukocytes in the presence of cartilage coated with AHGG (nonphagocytic enzyme release) was compared with the cartilage degrading activity of the supernatant from the same number of cells preincubated with a suspension of AHGG (phagocytic enzyme release). Nonphagocytic enzyme release by 5 x 10(6) cells provoked two to four times more (35)S and beta-glucuronidase (beta-G) release from cartilage than phagocytic enzyme release conditions. beta-glucuronidase was used as an indicator of the release of lysosomal granule enzymes. By the use of selected pharmacological agents it was possible to dissociate the enzyme release process from intrinsic enzyme (neutral protease) activity. Neutral protease and beta-G release by human cells in the presence of AHGG-coated cartilage was inhibited by 10(-5)M colchicine, whereas the protease activity, but not the release process, was inhibited by 10(-6)M gold thiomalate and 10% human serum. It is suggested that the release of a cartilage degrading neutral protease by viable human cells when exposed to AHGG might be a relevant model for the study of cartilage destruction as it occurs in rheumatoid arthritis.


Asunto(s)
Leucocitos/enzimología , Péptido Hidrolasas/metabolismo , Cartílago , Colchicina/farmacología , Glucuronidasa/análisis , Glicosaminoglicanos/metabolismo , Tiomalato Sódico de Oro/farmacología , Humanos , Concentración de Iones de Hidrógeno , L-Lactato Deshidrogenasa/análisis , Lisosomas/enzimología , Modelos Biológicos , Proteínas Opsoninas , Fagocitosis , Isótopos de Azufre , gammaglobulinas
18.
Mol Med ; 16(9-10): 343-51, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20386869

RESUMEN

Several HMGB1-specific antagonists have provided beneficial results in multiple models of inflammatory disease-preclinical trials including arthritis. Since no HMGB1-specific targeted therapy has yet reached the clinic, we have performed in vitro studies to investigate whether any of a selection of well-established antirheumatic drugs inhibit HMGB1 release as part of its mode of action. Freshly purified peripheral blood monocytes from healthy donors were stimulated in cultures with LPS and IFNγ to cause HMGB1 and TNF release detected in ELISPOT assays. Effects on the secretion were assessed in cultures supplemented with dexamethasone, cortisone, chloroquine, gold sodium thiomalate, methotrexate, colchicine, etanercept or anakinra. Pharmacologically relevant doses of dexamethasone, gold sodium thiomalate and chloroquine inhibited the extracellular release of HMGB1 in a dose-dependent mode. Immunostaining demonstrated that dexamethasone caused intracellular HMGB1 retention. No effects on HMGB1 secretion were observed in cultures with activated monocytes by any of the other studied agents. TNF production in LPS/IFNγ-activated monocytes was readily downregulated by dexamethasone and, to some extent, by chloroquine and etanercept. We conclude that dexamethasone, gold sodium thiomalate and chloroquine share a capacity to inhibit HMGB1 release from activated monocytes.


Asunto(s)
Proteína HMGB1/metabolismo , Factores Inmunológicos/farmacología , Monocitos/citología , Monocitos/metabolismo , Animales , Células Cultivadas , Cloroquina/farmacología , Colchicina/farmacología , Cortisona/farmacología , Dexametasona/farmacología , Regulación hacia Abajo/efectos de los fármacos , Tiomalato Sódico de Oro/farmacología , Humanos , Interleucina-1beta/antagonistas & inhibidores , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Metotrexato/farmacología , Ratones , Monocitos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
19.
Malar J ; 9: 118, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20459650

RESUMEN

BACKGROUND: Premature death of Plasmodium-infected erythrocytes is considered to favourably influence the clinical course of malaria. Aurothiomalate has previously been shown to trigger erythrocyte death or eryptosis, which is characterized by cell membrane scrambling leading to phosphatidylserine exposure at the cell surface. Phosphatidylserine-exposing cells are rapidly cleared from circulating blood. The present study thus tested whether sodium aurothiomalate influences the intraerythrocytic parasite development in vitro and the clinical course of murine malaria in vivo. METHODS: Human erythrocytes were infected with Plasmodium falciparum BinH in vitro and mice were infected (intraperitoneal injection of 1 x 106 parasitized murine erythrocytes) with Plasmodium berghei ANKA in vivo. RESULTS: Exposure to aurothiomalate significantly decreased the in vitro parasitemia of P. falciparum-infected human erythrocytes without influencing the intraerythrocytic DNA/RNA content. Administration of sodium aurothiomalate in vivo (daily 10 mg/kg b.w. s.c. from the 8th day of infection) enhanced the percentage of phosphatidylserine-exposing infected and noninfected erythrocytes in blood. All nontreated mice died within 30 days of infection. Aurothiomalate-treatment delayed the lethal course of malaria leading to survival of more than 50% of the mice 30 days after infection. CONCLUSIONS: Sodium aurothiomalate influences the survival of Plasmodium berghei-infected mice, an effect only partially explained by stimulation of eryptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Tiomalato Sódico de Oro/farmacología , Plasmodium berghei/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Animales , Recuento de Eritrocitos , Eritrocitos/parasitología , Femenino , Humanos , Malaria/tratamiento farmacológico , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Parasitemia/tratamiento farmacológico , Parasitemia/parasitología , Fosfatidilserinas/metabolismo , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/fisiología , Plasmodium falciparum/crecimiento & desarrollo , Análisis de Supervivencia
20.
J Leukoc Biol ; 83(1): 31-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17913975

RESUMEN

Gold compounds such as gold sodium thiomalate (GST) can reduce the symptoms of rheumatoid arthritis (RA), although their mechanism of action is not well defined. As the proinflammatory mediator high mobility group box chromosomal protein 1 (HMGB1) may play a role in the pathogenesis of RA, we have performed in vitro studies to investigate whether GST inhibits HMGB1 release as the basis of its mode of action. Murine RAW 264.7 or human THP-1 macrophage cells were stimulated in culture with agents causing extracellular HMGB1 release, including LPS, IFN-gamma, polyinosinic:polycytidylic acid, IFN-beta, or NO in the presence of GST, ranging from 0 microM to 250 microM. Secretion and intracellular location of HMGB1 were assessed by Western blotting, HMGB1-specific ELISPOT assay, and immunofluorescent staining. In parallel, TNF and IFN-beta levels were analyzed by ELISPOT and/or ELISA. Supernatant NO production was analyzed by the Griess method. At pharmacologically relevant doses, GST inhibited the extracellular release of HMGB1 from activated macrophages and caused the nuclear retention of this protein; in contrast, no effects were observed on the secretion or production of TNF. Release of the key endogenous mediators of HMGB1 translocation, IFN-beta and NO, was inhibited by GST. This inhibition required gold, as sodium thiomalate did not affect the responses measured. Furthermore, gold chloride also inhibited release of HMGB1. Together, these results suggest a new mechanism for the anti-rheumatic effects of gold salts in RA and the potential of drugs, which interfere with intracellular HMGB1 transport mechanisms, as novel agents to treat RA.


Asunto(s)
Núcleo Celular/metabolismo , Tiomalato Sódico de Oro/farmacología , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/inmunología , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/inmunología , Tiomalato Sódico de Oro/uso terapéutico , Proteína HMGB1/inmunología , Humanos , Interferón beta/efectos de los fármacos , Interferón beta/metabolismo , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Óxido Nítrico/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA