Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Antimicrob Agents Chemother ; 60(7): 4361-3, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27114276

RESUMEN

The zoonotic malaria parasite Plasmodium knowlesi has recently been established in continuous in vitro culture. Here, the Plasmodium falciparum [(3)H]hypoxanthine uptake assay was adapted for P. knowlesi and used to determine the sensitivity of this parasite to chloroquine, cycloguanil, and clindamycin. The data demonstrate that P. knowlesi is sensitive to all drugs, with 50% inhibitory concentrations (IC50s) consistent with those obtained with P. falciparum This assay provides a platform to use P. knowlesi in vitro for drug discovery.


Asunto(s)
Hipoxantina/metabolismo , Malaria/fisiopatología , Plasmodium knowlesi/metabolismo , Animales , Antimaláricos/farmacología , Cloroquina/farmacología , Clindamicina/farmacología , Concentración 50 Inhibidora , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Plasmodium knowlesi/efectos de los fármacos , Proguanil/farmacología , Triazinas/farmacología
2.
ACS Infect Dis ; 8(1): 106-117, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-34985259

RESUMEN

Malaria, caused by Plasmodium parasites, results in >400,000 deaths annually. There is no effective vaccine, and new drugs with novel modes of action are needed because of increasing parasite resistance to current antimalarials. Histone deacetylases (HDACs) are epigenetic regulatory enzymes that catalyze post-translational protein deacetylation and are promising malaria drug targets. Here, we describe quantitative structure-activity relationship models to predict the antiplasmodial activity of hydroxamate-based HDAC inhibitors. The models incorporate P. falciparum in vitro activity data for 385 compounds containing a hydroxamic acid and were subject to internal and external validation. When used to screen 22 new hydroxamate-based HDAC inhibitors for antiplasmodial activity, model A7 (external accuracy 91%) identified three hits that were subsequently verified as having potent in vitro activity against P. falciparum parasites (IC50 = 6, 71, and 84 nM), with 8 to 51-fold selectivity for P. falciparum versus human cells.


Asunto(s)
Malaria , Parásitos , Animales , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Plasmodium falciparum , Relación Estructura-Actividad Cuantitativa
3.
Plasmid ; 66(1): 38-46, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21419794

RESUMEN

Plasmid DNA is one of the indispensable components in molecular biology research and a potential biomaterial for gene therapy and DNA vaccination. Both quality and quantity of extracted plasmid DNA are of the great interests in cloning and subsequent expression of genes in vitro and in vivo for basic research and therapeutic interventions. Bacteria with extremely short generation times are the valuable source of plasmid DNA that can be isolated through a number of existing techniques. However, the current methods have some limitations in isolating high quality plasmid DNA since the multimeric plasmid which is believed to be more efficiently transcribed by RNA polymerase than the monomeric form, is almost lost during the extraction process. Recently, we developed a rapid isolation technique for multimeric plasmid based on generation of a 'protein aggregate' using a zwitterionic detergent and alkali. Here we have investigated the roles of different parameters in the whole extraction process to optimise the production of high quality multimeric plasmid DNA. Moreover, we have showed the advantageous effects of nanoparticles to effectively sediment the 'protein aggregate' for smooth elution of multimeric plasmid DNA from it. Finally, quality assessment study has revealed that the isolated multimeric DNA is at least 10 times more transcriptionally active than the monomeric form isolated by the commercially available Qiaget kit.


Asunto(s)
Apatitas/química , ADN Bacteriano/aislamiento & purificación , Plásmidos/aislamiento & purificación , Compuestos de Amonio Cuaternario/química , Tampones (Química) , ADN Bacteriano/genética , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Células HeLa , Humanos , Nanopartículas/química , Plásmidos/química , Plásmidos/genética , Transfección , Transformación Bacteriana
4.
Int J Parasitol Drugs Drug Resist ; 17: 118-127, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34560571

RESUMEN

Malaria is caused by infection with Plasmodium parasites and results in significant health and economic impacts. Malaria eradication is hampered by parasite resistance to current drugs and the lack of a widely effective vaccine. Compounds that target epigenetic regulatory proteins, such as histone deacetylases (HDACs), may lead to new therapeutic agents with a different mechanism of action, thereby avoiding resistance mechanisms to current antimalarial drugs. The anticancer HDAC inhibitor AR-42, as its racemate (rac-AR-42), and 36 analogues were investigated for in vitro activity against P. falciparum. Rac-AR-42 and selected compounds were assessed for cytotoxicity against human cells, histone hyperacetylation, human HDAC1 inhibition and oral activity in a murine malaria model. Rac-AR-42 was tested for ex vivo asexual and in vitro exoerythrocytic stage activity against P. berghei murine malaria parasites. Rac-AR-42 and 13 achiral analogues were potent inhibitors of asexual intraerythrocytic stage P. falciparum 3D7 growth in vitro (IC50 5-50 nM), with four of these compounds having >50-fold selectivity for P. falciparum versus human cells (selectivity index 56-118). Rac-AR-42 induced in situ hyperacetylation of P. falciparum histone H4, consistent with PfHDAC(s) inhibition. Furthermore, rac-AR-42 potently inhibited P. berghei infected erythrocyte growth ex vivo (IC50 40 nM) and P. berghei exoerythrocytic forms in hepatocytes (IC50 1 nM). Oral administration of rac-AR-42 and two achiral analogues inhibited P. berghei growth in mice, with rac-AR-42 (50 mg/kg/day single dose for four days) curing all infections. These findings demonstrate curative properties for HDAC inhibitors in the oral treatment of experimental mouse malaria.


Asunto(s)
Antimaláricos , Malaria , Parásitos , Animales , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Malaria/tratamiento farmacológico , Ratones , Plasmodium berghei , Plasmodium falciparum
5.
Eur J Med Chem ; 161: 277-291, 2019 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-30366254

RESUMEN

Despite the recent reductions in the global burden of malaria, this disease remains a devastating cause of death in tropical and subtropical regions. As there is no broadly effective vaccine for malaria, prevention and treatment still rely on chemotherapy. Unfortunately, emerging resistance to the gold standard artemisinin combination therapies means that new drugs with novel modes of action are urgently needed. In this context, Plasmodium histone modifying enzymes have emerged as potential drug targets, prompting us to develop and optimize compounds directed against such epigenetic targets. A panel of 51 compounds designed to target different epigenetic enzymes were screened for activity against Plasmodium falciparum parasites. Based on in vitro activity against drug susceptible and drug-resistant P. falciparum lines, selectivity index criterion and favorable pharmacokinetic properties, four compounds, one HDAC inhibitor (1) and three DNMT inhibitors (37, 43 and 45), were selected for preclinical studies in a mouse model of malaria. In vivo data showed that 37, 43 and 45 exhibited oral efficacy in the mouse model of Plasmodium berghei infection. These compounds represent promising starting points for the development of novel antimalarial drugs.


Asunto(s)
Antimaláricos/farmacología , Plasmodium berghei/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Quinazolinas/farmacología , Animales , Antimaláricos/síntesis química , Antimaláricos/química , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/microbiología , Humanos , Ratones , Ratones Endogámicos BALB C , Conformación Molecular , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/crecimiento & desarrollo , Quinazolinas/síntesis química , Quinazolinas/química , Relación Estructura-Actividad
6.
Int J Parasitol Drugs Drug Resist ; 8(2): 189-193, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29631126

RESUMEN

Bromodomain-containing proteins (BDPs) are involved in the regulation of eukaryotic gene expression. Compounds that bind and/or inhibit BDPs are of interest as tools to better understand epigenetic regulation, and as possible drug leads for different diseases, including malaria. In this study, we assessed the activity of 42 compounds demonstrated or predicted (using virtual screening of a pharmacophore model) to bind/inhibit eukaryotic BDPs for activity against Plasmodium falciparum malaria parasites. In silico docking studies indicated that all compounds are predicted to participate in a typical hydrogen bond interaction with the conserved asparagine (Asn1436) of the P. falciparum histone acetyltransferase (PfGCN5) bromodomain and a conserved water molecule. Only one compound (the dimethylisoxazole SGC-CBP30; a selective inhibitor of CREBBP (CBP) and EP300 bromodomains) is also predicted to have a salt-bridge between the morpholine nitrogen and Glu1389. When tested for in vitro activity against asynchronous asexual stage P. falciparum Dd2 parasites, all compounds displayed 50% growth inhibitory concentrations (IC50) >10 µM. Further testing of the three most potent compounds using synchronous parasites for 72 h showed that SGC-CBP30 was the most active (IC50 3.2 µM). In vitro cytotoxicity assays showed that SGC-CBP30 has ∼7-fold better selectivity for the parasites versus a human cell line (HEK 293). Together these data provide a possible starting point for future investigation of these, or related compounds, as tools to understand epigenetic regulation or as potential new drug leads.


Asunto(s)
Estadios del Ciclo de Vida/efectos de los fármacos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/farmacología , Plasmodium falciparum/efectos de los fármacos , Antimaláricos/farmacología , Bencimidazoles/farmacología , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Descubrimiento de Drogas , Células HEK293 , Humanos , Concentración 50 Inhibidora , Isoxazoles/farmacología , Simulación del Acoplamiento Molecular/métodos , Relación Estructura-Actividad
7.
ChemMedChem ; 12(19): 1627-1636, 2017 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-28812327

RESUMEN

In this work we aimed to develop parasite-selective histone deacetylase inhibitors (HDAC) inhibitors with activity against the disease-causing asexual blood stages of Plasmodium as well as causal prophylactic and/or transmission blocking properties. We report the design, synthesis, and biological testing of a series of 13 terephthalic acid-based HDAC inhibitors. All compounds showed low cytotoxicity against human embryonic kidney (HEK293) cells (IC50 : 8->51 µm), with 11 also having sub-micromolar in vitro activity against drug-sensitive (3D7) and multidrug-resistant (Dd2) asexual blood-stage P. falciparum parasites (IC50 ≈0.1-0.5 µm). A subset of compounds were examined for activity against early- and late-stage P. falciparum gametocytes and P. berghei exo-erythrocytic-stage parasites. While only moderate activity was observed against gametocytes (IC50 >2 µm), the most active compound (N1 -((3,5-dimethylbenzyl)oxy)-N4 -hydroxyterephthalamide, 1 f) showed sub-micromolar activity against P. berghei exo-erythrocytic stages (IC50 0.18 µm) and >270-fold better activity for exo-erythrocytic forms than for HepG2 cells. This, together with asexual-stage in vitro potency (IC50 ≈0.1 µm) and selectivity of this compound versus human cells (SI>450), suggests that 1 f may be a valuable starting point for the development of novel antimalarial drug leads with low host cell toxicity and multi-stage anti-plasmodial activity.


Asunto(s)
Antimaláricos/síntesis química , Antimaláricos/farmacología , Diseño de Fármacos , Ácidos Ftálicos/química , Ácidos Ftálicos/farmacología , Plasmodium falciparum/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Células Hep G2 , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Concentración 50 Inhibidora , Estadios del Ciclo de Vida/efectos de los fármacos , Ácidos Ftálicos/síntesis química , Relación Estructura-Actividad
8.
Int J Parasitol Drugs Drug Resist ; 7(1): 42-50, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28107750

RESUMEN

Malaria, schistosomiasis and leishmaniases are among the most prevalent tropical parasitic diseases and each requires new innovative treatments. Targeting essential parasite pathways, such as those that regulate gene expression and cell cycle progression, is a key strategy for discovering new drug leads. In this study, four clinically approved anti-cancer drugs (Vorinostat, Belinostat, Panobinostat and Romidepsin) that target histone/lysine deacetylase enzymes were examined for in vitro activity against Plasmodium knowlesi, Schistosoma mansoni, Leishmania amazonensis and L. donovani parasites and two for in vivo activity in a mouse malaria model. All four compounds were potent inhibitors of P. knowlesi malaria parasites (IC50 9-370 nM), with belinostat, panobinostat and vorinostat having 8-45 fold selectivity for the parasite over human neonatal foreskin fibroblast (NFF) or human embryonic kidney (HEK 293) cells, while romidepsin was not selective. Each of the HDAC inhibitor drugs caused hyperacetylation of P. knowlesi histone H4. None of the drugs was active against Leishmania amastigote or promastigote parasites (IC50 > 20 µM) or S. mansoni schistosomula (IC50 > 10 µM), however romidepsin inhibited S. mansoni adult worm parings and egg production (IC50 ∼10 µM). Modest in vivo activity was observed in P. berghei infected mice dosed orally with vorinostat or panobinostat (25 mg/kg twice daily for four days), with a significant reduction in parasitemia observed on days 4-7 and 4-10 after infection (P < 0.05), respectively.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Leishmania/efectos de los fármacos , Plasmodium knowlesi/efectos de los fármacos , Schistosoma mansoni/efectos de los fármacos , Acetilación , Administración Oral , Animales , Depsipéptidos/farmacología , Células HEK293 , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Indoles/administración & dosificación , Indoles/farmacología , Indoles/uso terapéutico , Concentración 50 Inhibidora , Leishmania/crecimiento & desarrollo , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria/tratamiento farmacológico , Malaria/parasitología , Ratones , Panobinostat , Parasitemia/tratamiento farmacológico , Plasmodium berghei/efectos de los fármacos , Plasmodium knowlesi/crecimiento & desarrollo , Schistosoma mansoni/crecimiento & desarrollo , Sulfonamidas/farmacología , Vorinostat
9.
Int J Oncol ; 48(6): 2359-66, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27035628

RESUMEN

Treatment of breast cancer, the second leading cause of female deaths worldwide, with classical drugs is often accompanied by treatment failure and relapse of disease condition. Development of chemoresistance and drug toxicity compels compromising the drug concentration below the threshold level with the consequence of therapeutic inefficacy. Moreover, amplification and over-activation of proto-oncogenes in tumor cells make the treatment more challenging. The oncogene, ROS1 which is highly expressed in diverse types of cancers including breast carcinoma, functions as a survival protein aiding cancer progression. Thus we speculated that selective silencing of ROS1 gene by carrier-mediated delivery of siRNA might sensitize the cancer cells to the classical drugs at a relatively low concentration. In this investigation we showed that intracellular delivery of c-ROS1-targeting siRNA using pH-sensitive inorganic nanoparticles of carbonate apatite sensitizes mouse breast cancer cells (4T1) to doxorubicin, but not to cisplatin or paclitaxel, with the highest enhancement in chemosensitivity obtained at 40 nM of the drug concentration. Although intravenous administrations of ROS1-loaded nanoparticles reduced growth of the tumor, a further substantial effect on growth retardation was noted when the mice were treated with the siRNA- and Dox-bound particles, thus suggesting that silencing of ROS1 gene could sensitize the mouse breast cancer cells both in vitro and in vivo to doxorubicin as a result of synergistic effect of the gene knockdown and the drug action, eventually preventing activation of the survival pathway protein, AKT1. Our findings therefore provide valuable insight into the potential cross-talk between the pathways of ROS1 and doxorubicin for future development of effective therapeutics for breast cancer.


Asunto(s)
Doxorrubicina/farmacología , Neoplasias Mamarias Experimentales/terapia , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Apatitas/administración & dosificación , Línea Celular Tumoral , Cisplatino/farmacología , Sinergismo Farmacológico , Femenino , Técnicas de Silenciamiento del Gen , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Endogámicos BALB C , Paclitaxel/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Distribución Aleatoria
10.
Int J Nanomedicine ; 7: 2473-81, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22701315

RESUMEN

BACKGROUND: Multidrug resistance, a major impediment to successful cancer chemotherapy, is the result of overexpression of ATP-binding cassette (ABC) transporters extruding internalized drugs. Silencing of ABC transporter gene expression with small interfering RNA (siRNA) could be an attractive approach to overcome multidrug resistance of cancer, although delivery of siRNA remains a major hurdle to fully exploit the potential of siRNA-based therapeutics. Recently, we have developed pH-sensitive carbonate apatite nanoparticles to efficiently carry and transport siRNA across the cell membrane, enabling knockdown of the cyclin B1 gene and consequential induction of apoptosis in synergy with anti-cancer drugs. METHODS AND RESULTS: We report that carbonate apatite-mediated delivery of the siRNAs targeting ABCG2 and ABCB1 gene transcripts in human breast cancer cells which constitutively express both of the transporter genes dose-dependently enhanced chemosensitivity to doxorubicin, paclitaxel and cisplatin, the traditionally used chemotherapeutic agents. Moreover, codelivery of two specific siRNAs targeting ABCB1 and ABCG2 transcripts resulted in a more robust increase of chemosensitivity in the cancer cells, indicating the reversal of ABC transporter-mediated multidrug resistance. CONCLUSION: The delivery concept of multiple siRNAs against ABC transporter genes is highly promising for preclinical and clinical investigation in reversing the multidrug resistance phenotype of breast cancer.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Silenciador del Gen/efectos de los fármacos , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , Transportadoras de Casetes de Unión a ATP/metabolismo , Apatitas/farmacocinética , Apatitas/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Terapia Combinada , Relación Dosis-Respuesta a Droga , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Femenino , Humanos , Células MCF-7 , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacocinética
11.
J Control Release ; 147(1): 101-8, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20620182

RESUMEN

Gene therapy through intracellular delivery of a functional gene or a gene-silencing element is a promising approach to treat critical diseases. Elucidation of the genetic basis of human diseases with complete sequencing of human genome revealed many vital genes as possible targets in gene therapy programs. RNA interference (RNAi), a powerful tool in functional genomics to selectively silence messenger RNA (mRNA) expression, can be harnessed to rapidly develop novel drugs against any disease target. The ability of synthetic small interfering RNA (siRNA) to effectively silence genes in vitro and in vivo, has made them particularly well suited as a drug therapeutic. However, since naked siRNA is unable to passively diffuse through cellular membranes, delivery of siRNA remains the major hurdle to fully exploit the potential of siRNA technology. Here pH-sensitive carbonate apatite has been developed to efficiently deliver siRNA into the mammalian cells by virtue of its high affinity interactions with the siRNA and the desirable size of the resulting siRNA/apatite complex for effective cellular endocytosis. Moreover, following internalization by cells, siRNA was found to be escaped from the endosomes in a time-dependent manner and finally, more efficiently silenced reporter genes at a low dose than commercially available lipofectamine. Knockdown of cyclin B1 gene with only 10nM of siRNA delivered by carbonate apatite resulted in the significant death of cancer cells, suggesting that the new method of siRNA delivery is highly promising for pre-clinical and clinical cancer therapy.


Asunto(s)
Apatitas/química , Portadores de Fármacos/química , Técnicas de Silenciamiento del Gen/métodos , ARN Interferente Pequeño/administración & dosificación , Supervivencia Celular/genética , Clonación Molecular , Citomegalovirus/genética , Composición de Medicamentos , Silenciador del Gen , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Tamaño de la Partícula , Plásmidos , ARN Interferente Pequeño/genética , Solubilidad , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA