Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Planta Med ; 88(13): 1123-1131, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34763354

RESUMO

cis-Aconitic acid is a constituent from the leaves of Echinodorus grandiflorus, a medicinal plant traditionally used in Brazil to treat inflammatory conditions, including arthritic diseases. The present study aimed to investigate the anti-arthritic effect of cis-aconitic acid in murine models of antigen-induced arthritis and monosodium urate-induced gout. The possible underlying mechanisms of action was evaluated in THP-1 macrophages. Oral treatment with cis-aconitic acid (10, 30, and 90 mg/kg) reduced leukocyte accumulation in the joint cavity and C-X-C motif chemokine ligand 1 and IL-1ß levels in periarticular tissue. cis-Aconitic acid treatment reduced joint inflammation in tissue sections of antigen-induced arthritis mice and these effects were associated with decreased mechanical hypernociception. Administration of cis-aconitic acid (30 mg/kg p. o.) also reduced leukocyte accumulation in the joint cavity after the injection of monosodium urate crystals. cis-Aconitic acid reduced in vitro the release of TNF-α and phosphorylation of IκBα in lipopolysaccharide-stimulated THP-1 macrophages, suggesting that inhibition of nuclear factor kappa B activation was an underlying mechanism of cis-aconitic acid-induced anti-inflammatory effects. In conclusion, cis-aconitic acid has significant anti-inflammatory effects in antigen-induced arthritis and monosodium urate-induced arthritis in mice, suggesting its potential for the treatment of inflammatory diseases of the joint in humans. Additionally, our findings suggest that this compound may contribute to the anti-inflammatory effect previously reported for E. grandiflorus extracts.


Assuntos
Alismataceae , Gota , Humanos , Camundongos , Animais , Ácido Aconítico/farmacologia , Inibidor de NF-kappaB alfa , Ácido Úrico , Lipopolissacarídeos , NF-kappa B , Fator de Necrose Tumoral alfa , Ligantes , Alismataceae/química , Gota/induzido quimicamente , Gota/tratamento farmacológico , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Quimiocinas , Inflamação
2.
Angew Chem Int Ed Engl ; 58(14): 4632-4637, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30695128

RESUMO

In cancer treatment, the unsatisfactory solid-tumor penetration of nanomaterials limits their therapeutic efficacy. We employed an in vivo self-assembly strategy and designed polymer-peptide conjugates (PPCs) that underwent an acid-induced hydrophobicity increase with a narrow pH-response range (from 7.4 to 6.5). In situ self-assembly in the tumor microenvironment at appropriate molecular concentrations (around the IC50 values of PPCs) enabled drug delivery deeper into the tumor. A cytotoxic peptide KLAK, decorated with the pH-sensitive moiety cis-aconitic anhydride (CAA), and a cell-penetrating peptide TAT were conjugated onto poly(ß-thioester) backbones to produce PT-K-CAA, which can penetrate deeply into solid tumors owing to its small size as a single chain. During penetration in vivo, CAA responds to the weak acid, leading to the self-assembly of PPCs and the recovery of therapeutic activity. Therefore, a deep-penetration ability for enhanced cancer therapy is provided by this in vivo assembly strategy.


Assuntos
Antineoplásicos/farmacologia , Peptídeos/farmacologia , Polímeros/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Ácido Aconítico/administração & dosagem , Ácido Aconítico/análogos & derivados , Ácido Aconítico/química , Ácido Aconítico/farmacologia , Administração Intravenosa , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Tamanho da Partícula , Peptídeos/administração & dosagem , Peptídeos/química , Polímeros/administração & dosagem , Polímeros/química , Propriedades de Superfície
3.
Mol Pharmacol ; 62(4): 888-900, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12237336

RESUMO

Anthracyclines are effective antineoplastic agents. However, the interaction of these drugs with iron (Fe) is an important cause of myocardial toxicity, limiting their therapeutic use (J Lab Clin Med 122:245-251, 1993). To overcome this limitation, it is crucial to understand how anthracyclines interact with the Fe metabolism of myocardial and neoplastic cells. Iron-regulatory proteins (IRPs) play vital roles in regulating cellular Fe metabolism via their mRNA-binding activity. We showed that doxorubicin (DOX) and its analogs interfere with tumor and myocardial cell Fe metabolism by affecting the RNA-binding activity of IRPs. Unexpectedly, experiments with the free radical scavengers, catalase, superoxide dismutase, ebselen, and Mn(III) tetrakis (4-benzoic acid) porphyrin complex, suggested that the effects of DOX on IRP-RNA-binding activity were not due to anthracycline-mediated free radical production. In contrast to previous studies, we showed that the DOX metabolite, doxorubicinol, had no effect on IRP-RNA-binding activity. Rather, the anthracycline-Fe and -copper (Cu) complexes decreased IRP-RNA-binding activity, indicating that formation of anthracycline-metal complexes may affect cellular Fe metabolism. In addition, anthracyclines prevented the response of IRPs to the depletion of intracellular Fe by chelators. This information may be useful in designing novel therapeutic strategies against tumor cells by combining chelators and anthracyclines. Interestingly, the effect of DOX on primary cultures of cardiomyocytes was similar to that observed using neoplastic cells, and particularly notable was the decrease in IRP2-RNA-binding activity. Our results add significant new information regarding the effects of anthracyclines on Fe metabolism that may lead to the design of more effective treatments.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Cobre/farmacologia , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Proteínas Ferro-Enxofre/metabolismo , Ferro/farmacologia , Proteínas de Ligação a RNA/metabolismo , Aconitato Hidratase/metabolismo , Ácido Aconítico/farmacologia , Quelantes/farmacologia , Citoplasma/enzimologia , Citoplasma/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Radicais Livres/metabolismo , Coração/efeitos dos fármacos , Humanos , Proteína 2 Reguladora do Ferro , Proteínas Reguladoras de Ferro , Proteínas Ferro-Enxofre/efeitos dos fármacos , Miocárdio/metabolismo , RNA/efeitos dos fármacos , RNA/metabolismo , Proteínas de Ligação a RNA/efeitos dos fármacos , Fatores de Tempo , Células Tumorais Cultivadas
4.
Biochem Med Metab Biol ; 42(3): 171-8, 1989 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2597432

RESUMO

TAA, an inhibitor of the enzyme aconitase, inhibits the growth of L. donovani promastigotes. Morphogenic transformation of the amastigote to the promastigote (table; see text) form in vitro was also inhibited by 2 mM TAA. TAA also reduced multiplication of the parasite in macrophage culture. In the hamster model of leishmania, TAA significantly reduced the parasitic burden of liver. In acute toxicity tests with BALB/c mice no deaths were recorded even at a dose level of 2 g/kg body wt/day.


Assuntos
Ácido Aconítico/farmacologia , Leishmania donovani/efeitos dos fármacos , Ácidos Tricarboxílicos/farmacologia , Ácido Aconítico/uso terapêutico , Ácido Aconítico/toxicidade , Animais , Células Cultivadas , Cricetinae , Leishmania donovani/crescimento & desenvolvimento , Leishmaniose Visceral/tratamento farmacológico , Leishmaniose Visceral/parasitologia , Macrófagos/parasitologia , Masculino , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C
5.
Acta Biochim Pol ; 23(2-3): 185-92, 1976.
Artigo em Inglês | MEDLINE | ID: mdl-970033

RESUMO

1. 2-Oxoglutarate, succinate, fumarate, malate and citrate, cis-aconitate and isocitrate stimulate conversion of cholesterol to progesterone in human placental mitochondria. 2. The stimulatory effect of dicarboxylic and tricarboxylic acids depends on the activity of malate dehydrogenase (decarboxylating) (NADP+) (EC 1.1.1.40) and isocitrate dehydrogenase (NADP+) (EC 1.1.1.42), respectively.


Assuntos
Ciclo do Ácido Cítrico , Mitocôndrias/metabolismo , Placenta/metabolismo , Progesterona/biossíntese , Ácido Aconítico/farmacologia , Citratos/farmacologia , Sinergismo Farmacológico , Feminino , Fumaratos/farmacologia , Humanos , Isocitrato Desidrogenase/metabolismo , Isocitratos/farmacologia , Ácidos Cetoglutáricos/farmacologia , Malato Desidrogenase/metabolismo , Malatos/farmacologia , Malonatos/farmacologia , Manganês/farmacologia , Mitocôndrias/efeitos dos fármacos , Gravidez , Succinatos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA