Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Parasitol Res ; 123(4): 180, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38592544

RESUMO

Because of recent reports of praziquantel resistance in schistosome infections, there have been suggestions to employ ivermectin as a possible alternative, especially as its chemical composition is different from that of praziquantel, so cross-resistance is not expected. In order to ascertain possible damage and elimination of worms, we used ivermectin by oral gavage in infected mice, at a high dose (30.1 mg/kg, bordering toxicity). We also tested the efficacy of the drug at various times postinfection (PI), to check on possible effect on young and mature stages of the parasites. Thus, we treated mice on days 21 and 22 or on days 41 and 42 and even on days 21, 22, 41, and 42 PI. None of the treatment regimens resulted in cure rates or signs of lessened pathology in the mice. We also compared the effect of ivermectin to that of artemisone, an artemisinin derivative which had served us in the past as an effective anti-schistosome drug, and there was a stark difference in the artemisone's efficacy compared to that of ivermectin; while ivermectin was not effective, artemisone eliminated most of the worms, prevented egg production and granulomatous inflammatory response. We assume that the reported lack of activity of ivermectin, in comparison with praziquantel and artemisinins, originates from the difference in their mode of action. In wake of our results, we suggest that ivermectin is not a suitable drug for treatment of schistosomiasis.


Assuntos
Artemisininas , Schistosomatidae , Esquistossomose , Animais , Camundongos , Praziquantel/uso terapêutico , Ivermectina/uso terapêutico , Esquistossomose/tratamento farmacológico
2.
Glob Chall ; 7(12): 2300030, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38094863

RESUMO

Employing new therapeutic indications for drugs that are already approved for human use has obvious advantages, including reduced costs and timelines, because some routine steps of drug development and regulation are not required. This work concentrates on the redirection of artemisinins (ARTS) that already are approved for clinical use, or investigated, for malaria treatment. Several mechanisms of action are suggested for ARTS, among which only a few have been successfully examined in vivo, mainly the induction of oxidant stress and anti-inflammatory effects. Despite these seemingly contradictory effects, ARTS are proposed for repurposing in treatment of inflammatory disorders and diverse types of diseases caused by viral, bacterial, fungal, and parasitic infections. When pathogens are treated the expected outcome is diminution of the causative agents and/or their inflammatory damage. In general, repurposing ARTS is successful in only a very few cases, specifically when a valid mechanism can be targeted using an additional therapeutic agent and appropriate drug delivery. Investigation of repurposing should include optimization of drug combinations followed by examination in relevant cell lines, organoids, and animal models, before moving to clinical trials.

3.
Front Pharmacol ; 11: 846, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32595499

RESUMO

Malaria caused by Plasmodium falciparum causes numerous cases of morbidity with about 400,000 deaths yearly owing, mainly, to inflammation leading to cerebral malaria (CM). CM conventionally is treated by repetitive administration of anti-plasmodial drugs and supportive non-specific drugs, for about a week. A mouse model of CM caused by Plasmodium berghei ANKA, in which brain and systemic clinical pathologies occur followed by sudden death within about a week, was used to study the effect of artemisone, a relatively new artemisinin, within an injectable pasty polymer formulated for its controlled release. The parasites were exposed to the drug over several days at a non-toxic concentrations for the mice but high enough to affect the parasites. Artemisone was also tested in cultures of bacteria, cancer cells and P. falciparum to evaluate the specificity and suitability of these cells for examining the release of artemisone from its carrier. Cultures of P. falciparum were the most suitable. Artemisone released from subcutaneous injected poly(sebacic acid-ricinoleic acid) (PSARA) pasty polymer, reduced parasitemias in infected mice, prolonged survival and prevented death in most of the infected mice. Successful prophylactic treatment before infection proved that there was a slow release of the drug for about a week, which contrasts with the three hour half-life that occurs after injection of just the drug. Treatment with artemisone within the polymer, even at a late stage of the disease, helped to prevent or, at least, delay accompanying severe symptoms. In some cases, treatment prevented death of CM and the mice died later of anemia. Postponing the severe clinical symptoms is also beneficial in cases of human malaria, giving more time for an appropriate diagnosis and treatment before severe symptoms appear. The method presented here may also be useful for combination therapy of anti-plasmodial and immunomodulatory drugs.

4.
Pharmaceutics ; 12(6)2020 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-32503130

RESUMO

Artemisone is an innovative artemisinin derivative with applications in the treatment of malaria, schistosomiasis and other diseases. However, its low aqueous solubility and tendency to degrade after solubilisation limits the translation of this drug into clinical practice. We developed a self-microemulsifying drug delivery system (SMEDDS), which is easy to produce (simple mixing) with a high drug load. In addition to known pharmaceutical excipients (Capmul MCM, Kolliphor HS15, propylene glycol), we identified Polysorb ID 46 as a beneficial new additional excipient. The physicochemical properties were characterized by dynamic light scattering, conductivity measurements, rheology and electron microscopy. High storage stability, even at 30 °C, was achieved. The orally administrated artemisone SMEDDS formulation was highly active in vivo in S. mansoni infected mice. Thorough elimination of the adult worms, their eggs and prevention of the deleterious granuloma formation in the livers of infected mice was observed even at a relatively low dose of the drug. The new formulation has a high potential to accelerate the clinical use of artemisone in schistosomiasis and malaria.

5.
Int J Parasitol Drugs Drug Resist ; 7(2): 241-247, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28511056

RESUMO

The current treatment of schistosomiasis is based on the anti-helminthic drug praziquantel (PZQ). PZQ affects only the adult stages of schistosomes. In addition, resistance to PZQ is emerging. We suggest a drug, which could serve as a potential alternative or complement to PZQ, and as a means of treating infections at earlier, pre-granuloma stage. Derivatives of the peroxidic antimalarial drug artemisinin have been indicated as alternatives, because both plasmodia and schistosomes are blood-feeding parasites. The mechanism of action of artemisinins is related to oxidative effects of the artemisinins on intracellular reductants leading to formation of cytotoxic reactive oxygen species. We used artemisone, which has improved pharmacokinetics and anti-plasmodial activity, and reduced toxicity compared to other artemisinins in clinical use against malaria. We infected adult mice by subcutaneous injection of S. mansoni cercariae (about 200) and treated them at various times post infection by the following methods: i. artemisone suspension administered by gavage (400-450 mg/kg); ii. subcutaneous injection of a gel containing a known concentration of artemisone (115-120 mg/kg); iii. subcutaneous insertion of the drug incorporated in a solid polymer (56-60 mg/kg); iv. intraperitoneal injection of the drug solubilized in DMSO (115-120 mg/kg). Drug administration in polymers was performed to enable slow release of the artemisone that was verified in vivo and in vitro bioassays using drug-sensitive malaria parasites. We found superior strong anti-schistosome effects up to a total reduction of worm number, mainly following repetitive treatments with the drug absorbed in the polymers (73.1% and 95.9% reduction in mice treated with artemisone in gel 7 and 14, and 21, 28 and 35 days post infection, respectively). The results indicate that artemisone has a potent anti-schistosome activity. Its main importance in this context is its effectiveness in treating hosts harboring juvenile schistosomes, before egg-deposition and induction of deleterious immune responses.


Assuntos
Anti-Helmínticos/administração & dosagem , Artemisininas/administração & dosagem , Preparações de Ação Retardada/administração & dosagem , Schistosoma mansoni/efeitos dos fármacos , Esquistossomose mansoni/tratamento farmacológico , Administração Oral , Animais , Modelos Animais de Doenças , Injeções Intraperitoneais , Injeções Subcutâneas , Camundongos , Resultado do Tratamento
6.
Nanomedicine ; 11(5): 1057-64, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25791809

RESUMO

Intraoperative ureter identification can assist in the prevention of ureteral injury and consequently improve surgery outcomes. Our aim was to take advantage of the altered pharmacokinetics of liposomal indocyanine green (ICG), the only FDA-approved near-infrared (NIR) dye, for imaging of ureters during surgeries. ICG was passively adsorbed to liposomes. NIR whole mice body and isolated tissue imaging were used to study liposomal ICG properties vs. free ICG. In vivo, the urinary bladder could be clearly observed in most of the liposome-treated mice. Liposomal encapsulation of ICG enhanced ureteral emission up to 1.9 fold compared to free ICG (P<0.01). Increase in liposomal micropolarity and microviscosity and differential scanning calorimetry supported ICG localization within the liposomal bilayer. Our findings suggest that liposomal ICG could be utilized for ureteral imaging intra-operatively, thus potentially improving surgical outcomes. FROM THE CLINICAL EDITOR: Iatrogenic ureteral injury is a serious complication of abdominal surgery and intra-operative recognition of the ureters is usually the best method of injury prevention. In this article, the authors developed liposomal indocyanine green, which could be excreted via the urinary system and investigated its in-vivo use in mice.


Assuntos
Corantes/administração & dosagem , Verde de Indocianina/administração & dosagem , Imagem Óptica/métodos , Ureter/patologia , Bexiga Urinária/patologia , Animais , Corantes/farmacocinética , Feminino , Verde de Indocianina/farmacocinética , Raios Infravermelhos , Lipossomos , Camundongos , Ureter/lesões , Bexiga Urinária/lesões
7.
Antimicrob Agents Chemother ; 58(8): 4745-54, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24913162

RESUMO

The decreasing effectiveness of antimalarial therapy due to drug resistance necessitates constant efforts to develop new drugs. Artemisinin derivatives are the most recent drugs that have been introduced and are considered the first line of treatment, but there are already indications of Plasmodium falciparum resistance to artemisinins. Consequently, drug combinations are recommended for prevention of the induction of resistance. The research here demonstrates the effects of novel combinations of the new artemisinin derivative, artemisone, a recently described 10-alkylamino artemisinin derivative with improved antimalarial activity and reduced neurotoxicity. We here investigate its ability to kill P. falciparum in a high-throughput in vitro assay and to protect mice against lethal cerebral malaria caused by Plasmodium berghei ANKA when used alone or in combination with established antimalarial drugs. Artemisone effects against P. falciparum in vitro were synergistic with halofantrine and mefloquine, and additive with 25 other drugs, including chloroquine and doxycycline. The concentrations of artemisone combinations that were toxic against THP-1 cells in vitro were much higher than their effective antimalarial concentration. Artemisone, mefloquine, chloroquine, or piperaquine given individually mostly protected mice against cerebral malaria caused by P. berghei ANKA but did not prevent parasite recrudescence. Combinations of artemisone with any of the other three drugs did completely cure most mice of malaria. The combination of artemisone and chloroquine decreased the ratio of proinflammatory (gamma interferon, tumor necrosis factor) to anti-inflammatory (interleukin 10 [IL-10], IL-4) cytokines in the plasma of P. berghei-infected mice. Thus, artemisone in combinations with other antimalarial drugs might have a dual action, both killing parasites and limiting the potentially deleterious host inflammatory response.


Assuntos
Antimaláricos/farmacologia , Artemisininas/farmacologia , Malária Cerebral/tratamento farmacológico , Plasmodium berghei/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cloroquina/farmacologia , Doxiciclina/farmacologia , Sinergismo Farmacológico , Quimioterapia Combinada , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Humanos , Interleucina-10/antagonistas & inibidores , Interleucina-10/biossíntese , Interleucina-4/antagonistas & inibidores , Interleucina-4/biossíntese , Malária Cerebral/imunologia , Malária Cerebral/parasitologia , Mefloquina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Monócitos/efeitos dos fármacos , Testes de Sensibilidade Parasitária , Fenantrenos/farmacologia , Plasmodium berghei/fisiologia , Plasmodium falciparum/fisiologia
8.
Chemother Res Pract ; 2011: 175054, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22295203

RESUMO

Local delivery of chemotherapeutic drugs has long been recognized as a potential method for reaching high drug doses at the target site while minimizing systemic exposure. Cisplatin is one of the most effective chemotherapeutic agents for the treatment of various tumors; however, its systemic toxicity remains the primary dose-limiting factor. Here we report that incorporation of cisplatin into a fatty acid-based polymer carrier followed by a local injection into the solid tumor resulted in a successful tumor growth inhibition in heterotopic mouse bladder tumor model in mice. Platinum concentration in the tumor tissue surrounding the injected implant remained above the therapeutic level up to 14 days after the injection, while the plasma levels were several orders of magnitude lower comparing to systemic delivery. The reported delivery system increased the maximum tolerated dose of cisplatin 5 times compared to systemic delivery, thus potentially improving antitumor efficacy of cisplatin in solid tumor model.

9.
Infect Immun ; 78(10): 4195-205, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660606

RESUMO

Protein kinase C θ (PKCθ) functions as a core component of the immunological synapse and serves as a key protein in the integrated T-cell antigen receptor (TCR)/CD28-induced signaling cascade leading to T-cell activation. However, the involvement of PKCθ in host-mediated immune responses to pathogens has not been thoroughly investigated. We tested the consequences of PKCθ ablation on the host response to infection by Plasmodium berghei ANKA (PbA). We found that both PKCθ(+/+) and PKCθ(-/-) C57BL/6J mice are susceptible to infection with PbA. However, despite a similar parasite burden, PKCθ(+/+) mice had an earlier onset of neurological signs, characteristics of experimental cerebral malaria (ECM), resulting in an earlier death. These mice suffered from an early and pronounced splenomegaly with a concomitant increase in the total number of CD4(+) splenic T cells. In contrast, a large proportion of PbA-infected PKCθ(-/-) mice overcame the acute phase characterized by neurological symptoms and survived longer than PKCθ(+/+) mice. The partial resistance of PKCθ(-/-) mice to ECM was associated with an impaired production of Th1-type cytokines, including gamma interferon and tumor necrosis factor alpha/lymphotoxin-α, which are known to exacerbate symptoms leading to ECM. In addition, PbA infection-induced LFA-1 expression in CD8(+) T cells was suppressed in PKCθ-deficient T cells, suggesting a diminished ability to adhere to endothelial cells and sequester in brain microvasculature, which may explain the decrease in neurological symptoms. These data implicate PKCθ in CD4(+) Th1(+) and CD8(+) T-cell-mediated immune responses during PbA infection that contribute to the development of ECM.


Assuntos
Isoenzimas/genética , Malária Cerebral/imunologia , Plasmodium berghei/imunologia , Proteína Quinase C/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Regulação da Expressão Gênica/imunologia , Predisposição Genética para Doença , Isoenzimas/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Malária/imunologia , Malária/parasitologia , Malária Cerebral/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Proteína Quinase C/metabolismo , Proteína Quinase C-theta , Baço/patologia , Células Th1/imunologia , Regulação para Cima
10.
Int J Pharm ; 358(1-2): 114-20, 2008 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-18406086

RESUMO

PURPOSE: The aim of this study was to investigate the effectiveness of paclitaxel controlled release from intratumorally injected polymer. METHODS: The effectiveness of paclitaxel-polymer formulation injected intratumorally was tested in mouse bladder tumor model. To determine paclitaxel biodistribution in tumor at predetermined time periods the tumor was excised, frozen and sectioned, and the paclitaxel concentrations were determined in the tumor tissue and in plasma by HPLC. Histopathological evaluation of the necrosis and inflammation was performed on tumor sections. RESULTS: In the paclitaxel/polymer group mice were injected intratumorally with 0.2 ml of the 10% (w/w) paclitaxel formulation, the tumor disappeared completely 5 days after injection, and mice survived till the end of the study (50 days post-tumor cells inoculation). In biodistribution studies, the highest paclitaxel concentration in the tumor tissue was 40 microg/mg 1 day after the intratumoral injection and decreased gradually during 10 days to 5 microg/mg that is still high enough to induce cytotoxic effect, and the necrotic effect of paclitaxel on the tumors was confirmed by histopathology. CONCLUSIONS: Treatment with local injection of polymer-paclitaxel formulation inhibited the growth of solid tumors. Distribution studies of paclitaxel after intratumoral injection showed high and effective drug concentrations in tumor.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Paclitaxel/farmacocinética , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/sangue , Linhagem Celular Tumoral , Sobrevivência Celular , Química Farmacêutica , Cromatografia Líquida de Alta Pressão , Ácidos Decanoicos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Injeções , Camundongos , Camundongos Endogâmicos C3H , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Paclitaxel/administração & dosagem , Paclitaxel/sangue , Polímeros , Ácidos Ricinoleicos
11.
J Control Release ; 117(1): 90-6, 2007 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-17150275

RESUMO

Gentamicin sulfate, a potent antibiotic agent, is currently used for treatment of osteomyelitis mainly by intravenous injection with a long-term indwelling catheter, local implant of antibiotic containing polymethylmethacrylate beads or calcium phosphate (bone cements). Searching for more effective treatments, this study was designed to evaluate biodegradable injectable gelling polymeric devices for the controlled release of gentamicin sulfate in the treatment of invasive bacterial infections. Gentamicin sulfate was incorporated in poly(sebacic-co-ricinoleic-ester-anhydride P(SA-RA)) paste at 10-20% w/w and its release in buffer solution was monitored. The in vitro activity of the formulations was determined against Staphylococcus aureus. A constant release of active gentamicin for over 28 days was found. The stability of the formulation was determined under different storage conditions. The formulations were stable to sterilization by gamma-irradiation and long term storage under freezing. The toxicity of the polymer and the formulations with gentamicin was examined by subcutaneous injection to rats. Four weeks after implantation, histopathological examination of the tissues surrounding the implant showed no inflammation. A preliminary study revealed positive effect of gentamicin containing P(SA-RA) on established osteomyelitis in a rat model. In conclusion this study suggests that poly(sebacic-co-ricinoleic-ester-anhydride) 3:7 loaded with 10%-20% gentamicin sulfate, might be used as an injectable biodegradable device for in situ treatment of osteomyelitis induced by S. aureus.


Assuntos
Antibacterianos/administração & dosagem , Gentamicinas/administração & dosagem , Animais , Antibacterianos/farmacocinética , Antibacterianos/toxicidade , Química Farmacêutica , Ácidos Decanoicos , Preparações de Ação Retardada , Implantes de Medicamento , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Raios gama , Gentamicinas/farmacocinética , Gentamicinas/toxicidade , Injeções , Masculino , Testes de Sensibilidade Microbiana , Peso Molecular , Osteomielite/tratamento farmacológico , Osteomielite/microbiologia , Polímeros , Ratos , Ratos Wistar , Ácidos Ricinoleicos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Viscosidade
12.
Cell Immunol ; 250(1-2): 24-30, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18275944

RESUMO

Strategies using epitope-based vaccination are being considered for melanoma immunotherapy, in an attempt to overcome failure of other modalities. In the present study, we designed and produced a multiepitope polypeptide for melanoma (MEP-mel), which contains three repeats of four antigenic epitopes (gp100: 209-217 (210M); gp100: 280-288 (288V); Mart1: 26-35 (27L); tyrosinase: 368-376 (370D). The peptides were attached to each other by linkers containing sequences recognized by the proteasome, to improve protein cleavage and antigen presentation. The results show that peptide-specific T cells produced IFN-gamma when stimulated with MEP-mel-transfected dendritic cells. The presentation of peptides by MEP-mel-transfected dendritic cells was proteasome-dependent and was more long-lasting than the presentation of exogenously delivered native peptides. When dendritic cells were loaded with MEP-mel protein, weak cross presentation was induced. The production of multiepitope molecules based on several peptides linked by sequences sensitive to proteasomal cleavage represents a promising new tool for the improvement of cancer immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Melanoma/imunologia , Peptídeos/imunologia , Proteínas/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Separação Celular , Células Cultivadas , DNA Complementar/genética , DNA Complementar/farmacocinética , Células Dendríticas/efeitos dos fármacos , Desenho de Fármacos , Eletroporação , Inibidores Enzimáticos/farmacologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/farmacologia , Escherichia/genética , Humanos , Imunoterapia/métodos , Interferon gama/metabolismo , Melanoma/genética , Peptídeos/genética , Peptídeos/farmacologia , Plasmídeos/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Proteínas/genética , Proteínas/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia
13.
Int J Parasitol ; 36(14): 1427-41, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17005183

RESUMO

Artemisinin derivatives are the most recent single drugs approved and introduced for public antimalarial treatment. Although their recommended use is for treatment of Plasmodium falciparum infection, these drugs also act against other parasites, as well as against tumor cells. The mechanisms of action attributed to artemisinin include interference with parasite transport proteins, disruption of parasite mitochondrial function, modulation of host immune function and inhibition of angiogenesis. Artemisinin combination therapies are currently the preferred treatment for malaria. These combinations may prevent the induction of parasite drug resistance. However, in view of the multiple mechanisms involved, especially when additional drugs are used, the combined therapy should be carefully examined for antagonistic effects. It is now a general theory that the crucial mechanism is interference with plasmodial SERCA. Therefore, future development of resistance may be associated with overproduction or mutations of this transporter. However, a general mechanism, such as alterations in general drug transport pathways, is feasible. In this article, we review the evidence for each mechanism of action suggested.


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Malária/tratamento farmacológico , Animais , Antimaláricos/efeitos adversos , Antimaláricos/metabolismo , Artemisininas/efeitos adversos , Artemisininas/metabolismo , Resistência a Medicamentos/efeitos dos fármacos , Transporte de Elétrons/efeitos dos fármacos , Humanos , Sistema Imunitário/efeitos dos fármacos , Malária Falciparum/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Plasmodium falciparum/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
14.
J Parasitol ; 92(1): 170-7, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16629332

RESUMO

Iron chelating agents, which permeate through erythrocytic and parasite membranes, are effective against Plasmodium falciparum in vitro. However, the protective effect in humans is transient. We examined the antiplasmodial capacity of several iron chelators in vitro and in vivo. The chelators 3/3hb/2m and 3/2hb/b (together, MoB) were more effective against P. falciparum in vitro than desferrioxamine (DFO) and Salicylaldehyde isonicotinoyl hydrazone (SIH) (together, DoS). Despite similar pharmacokinetics of all iron chelators, mice infected with Plasmodium vinckei and treated with MoB succumbed to malaria, whereas DoS-treated mice survived. However, even in the surviving mice, peak parasitemias were above 30%. These results indicate that the direct effects of the drugs on the parasites were not responsible alone for the complete recovery of the mice. We suggest that the recovery is related to differential effects of the drugs on various immune functions. We concentrated on the effect of the iron chelators on B cell and T cell proliferation and on allogeneic stimulation (MLR), interleukin-10 (IL-10), gamma-interferon (gamma-IFN), tumor necrosis factor-alpha (TNF-alpha), and radical production. All the iron chelators examined inhibited the in vitro proliferation of B cells and T cells, and MLR. This may explain why iron chelators are only slightly efficient in treating human malaria. However, the inhibitory effects of MoB on B cell and T cell proliferation and on MLR were more pronounced than those of DoS. In addition, the release of free radicals by effector cells was inhibited to a greater extent by MoB than by DoS. These results may explain why MoB, which was more efficient in vitro, was not effective in vivo. The DoS effects on the in vitro secretion of cytokines correlate with their in vivo effect; there was a decrease of IL-10 and a parallel increase in gamma-IFN and TNF-alpha production by human mononuclear cells. MoB, which could not rescue the animals from malaria, did not affect IL-10 and TNF-alpha, but reduced gamma-IFN levels. Identical results were obtained when using monocytes instead of mononuclear cells (except for gamma-IFN, which is not produced by monocytes). Our results indicate that an iron chelator, or any antiparasitic drug that kills the parasites in vitro, should also be selected for further evaluation on the basis of its reaction with immune components; it should not interfere with crucial protective immunological processes, but it may still alleviate parasitemia by positive immune modulation.


Assuntos
Quelantes de Ferro/farmacologia , Malária/tratamento farmacológico , Plasmodium/efeitos dos fármacos , Plasmodium/imunologia , Aldeídos/química , Aldeídos/farmacologia , Animais , Linfócitos B/efeitos dos fármacos , Células Cultivadas , Desferroxamina/química , Desferroxamina/farmacologia , Modelos Animais de Doenças , Humanos , Hidrazonas/química , Hidrazonas/farmacologia , Interleucina-10/biossíntese , Quelantes de Ferro/química , Leucócitos Mononucleares/efeitos dos fármacos , Luminescência , Malária/imunologia , Camundongos , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/imunologia , Análise de Sobrevida , Linfócitos T/efeitos dos fármacos , Fatores de Tempo , Fator de Necrose Tumoral alfa/biossíntese
15.
J Parasitol ; 91(1): 158-63, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15856892

RESUMO

The effects of a water-soluble amphotericin B (AmB)-arabinogalactan (AG) conjugate on several immune functions were investigated. The experiments measured the effects of AmB-AG on (1) release of tumor necrosis factor-alpha (TNF-alpha), nitric oxide (NO), and interferon-gamma (IFN-gamma) from phagocytic cells and (2) cell-mediated immune responses. AmB-AG increased TNF-alpha release from mouse peritoneal macrophages and human monocytes but had no effect on IFN-gamma and NO release. A commercial preparation of nonconjugated AmB (Fungizone) also increased TNF-alpha production, but to a lesser extent than AmB-AG. AG alone had no effect on TNF-alpha production, proving that AmB caused the increased TNF-alpha production. AmB-AG and Fungizone were also tested for their effect on B- and T-cell proliferation. Neither compound altered T-lymphocyte responses to concanavalin A, but both inhibited the stimulation of B lymphocytes by lipopolysaccharides. However, Fungizone showed a stronger inhibitory effect on B cells. Allocytotoxicity was also inhibited by AmB-AG and more strongly by Fungizone. The increased production of TNF-alpha by cells treated with AmB-AG and the lower inhibitory effect of AmB-AG on lymphocyte stimulation and allocytotoxicity, as compared with Fungizone, explain the better therapeutic efficacy of the AmB-polysaccharide conjugate. AmB is active because of its preferential binding to ergosterol rather than cholesterol, the former sterol preferentially present in parasite surface membranes. This is also valid for the axenic amastigotes, which were sensitive to the AmB-AG. Overall, our results suggest that the antileishmanial activity of AmB-AG is mediated both directly and via modulation of immune functions.


Assuntos
Anfotericina B/análogos & derivados , Anfotericina B/farmacologia , Antiprotozoários/farmacologia , Galactanos/farmacologia , Imunidade Celular/efeitos dos fármacos , Leishmania tropica/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Bioensaio , Humanos , Interferon gama/metabolismo , Leishmania tropica/crescimento & desenvolvimento , Leishmania tropica/imunologia , Medições Luminescentes , Ativação Linfocitária/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Monócitos/imunologia , Monócitos/parasitologia , Óxido Nítrico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/metabolismo
16.
Biochem Pharmacol ; 66(5): 809-17, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12948862

RESUMO

Ferriprotoporphyrin IX (FP) is released inside the food vacuole of the malaria parasite during the digestion of host cell hemoglobin. FP is detoxified by its biomineralization to hemozoin. This process is effectively inhibited by 4-aminoquinolines. As a result FP accumulates in the membrane fraction and associates with enzymes of infected cells in parallel with parasite killing. Free FP is degraded by reduced glutathione (GSH). This degradation is inhibited by chloroquine (CQ) and amodiaquine (AQ) but not by quinine (Q) or mefloquine (MQ). Increased GSH levels in Plasmodium falciparum-infected cells confer resistance to CQ and vice versa, and sensitize CQ-resistant Plasmodium berghei by inhibiting the synthesis of glutathione. Some drugs are known to reduce GSH in body tissues when used in excess, either due to their pro-oxidant activity or their ability to form conjugates with GSH. We show that acetaminophen, indomethacin and disulfiram were able to potentiate the antimalarial action of sub-curative doses of CQ and AQ in P. berghei- or Plasmodium vinckei petteri-infected mice, but not that of Q and MQ. In contrast, N-acetyl-cysteine which is expected to increase the cellular levels of GSH, antagonized the action of CQ. Although these results imply that alteration in GSH are involved, measurement of total glutathione either in uninfected or P. berghei-infected mice, treated with these drugs did not reveal major changes. In conclusion, experimental evidences provided in this study suggest that some off the counter drugs can be used in combination with some antimalarials to which the parasite has become resistant.


Assuntos
Acetaminofen/farmacologia , Antimaláricos/uso terapêutico , Cloroquina/uso terapêutico , Glutationa/metabolismo , Malária/tratamento farmacológico , Acetilcisteína/farmacologia , Aminoquinolinas/farmacologia , Animais , Dissulfiram/farmacologia , Sinergismo Farmacológico , Indometacina/farmacologia , Camundongos , Plasmodium berghei , Plasmodium falciparum
18.
Redox Rep ; 8(5): 276-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14962364

RESUMO

Ferriprotoporphyrin IX (FP) is released inside the food vacuole of the malaria parasite during the digestion of host cell hemoglobin. FP is detoxified by its biomineralization to hemozoin. This process is effectively inhibited by chloroquine (CQ) and amodiaquine (AQ). Undegraded FP accumulates in the membrane fraction and inhibits enzymes of infected cells in parallel with parasite killing. FP is demonstrably degraded by reduced glutathione (GSH) in a radical-mediated mechanism. This degradation is inhibited by CQ and AQ in a competitive manner, thus explaining the ability of increased GSH levels in Plasmodium falciparum-infected cells to increase resistance to CQ and vice versa, and to render Plasmodium berghei that were selected for CQ resistance in vivo sensitive to the CQ when glutathione synthesis is inhibited. Some over-the-counter drugs that are known to reduce GSH in body tissues when used in excess were found to enhance the antimalarial action of CQ and AQ in mice infected either with P. berghei or Plasmodium vinckei. In contrast, N-acetyl-cysteine which is expected to increase the cellular levels of GSH, antagonized the action of CQ. These results suggest that some over-the-counter drugs can be used in combination with some antimalarials to which the parasite has become resistant.


Assuntos
Antimaláricos/farmacologia , Cloroquina/farmacologia , Glutationa/metabolismo , Plasmodium/efeitos dos fármacos , Acetilcisteína/farmacologia , Amodiaquina/farmacologia , Animais , Resistência a Medicamentos , Hemina/metabolismo , Humanos , Malária/tratamento farmacológico , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA