Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Annu Rev Biochem ; 81: 533-59, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22404633

RESUMEN

Nitric oxide (NO) is an essential signaling molecule in biological systems. In mammals, the diatomic gas is critical to the cyclic guanosine monophosphate (cGMP) pathway as it functions as the primary activator of soluble guanylate cyclase (sGC). NO is synthesized from l-arginine and oxygen (O(2)) by the enzyme nitric oxide synthase (NOS). Once produced, NO rapidly diffuses across cell membranes and binds to the heme cofactor of sGC. sGC forms a stable complex with NO and carbon monoxide (CO), but not with O(2). The binding of NO to sGC leads to significant increases in cGMP levels. The second messenger then directly modulates phosphodiesterases (PDEs), ion-gated channels, or cGMP-dependent protein kinases to regulate physiological functions, including vasodilation, platelet aggregation, and neurotransmission. Many studies are focused on elucidating the molecular mechanism of sGC activation and deactivation with a goal of therapeutic intervention in diseases involving the NO/cGMP-signaling pathway. This review summarizes the current understanding of sGC structure and regulation as well as recent developments in NO signaling.


Asunto(s)
Guanilato Ciclasa/química , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Animales , GMP Cíclico/metabolismo , Guanilato Ciclasa/aislamiento & purificación , Guanilato Ciclasa/metabolismo , Humanos , Isoenzimas/metabolismo , Receptores Citoplasmáticos y Nucleares/aislamiento & purificación , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Guanilil Ciclasa Soluble
2.
Biochemistry ; 62(17): 2677-2688, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37556730

RESUMEN

Polyketide synthases (PKSs) are megaenzymes that form chemically diverse polyketides and are found within the genomes of nearly all classes of life. We recently discovered the type I PKS from the apicomplexan parasite Toxoplasma gondii, TgPKS2, which contains a unique putative chain release mechanism that includes ketosynthase (KS) and thioester reductase (TR) domains. Our bioinformatic analysis of the thioester reductase of TgPKS2, TgTR, suggests differences compared to other systems and hints at a possibly conserved release mechanism within the apicomplexan subclass Coccidia. To evaluate this release module, we first isolated TgTR and observed that it is capable of 4 electron (4e-) reduction of octanoyl-CoA to the primary alcohol, octanol, utilizing NADH. TgTR was also capable of generating octanol in the presence of octanal and NADH, but no reactions were observed when NADPH was supplied as a cofactor. To biochemically characterize the protein, we measured the catalytic efficiency of TgTR using a fluorescence assay and determined the TgTR binding affinity for cofactor and substrates using isothermal titration calorimetry (ITC). We additionally show that TgTR is capable of reducing an acyl carrier protein (ACP)-tethered substrate by liquid chromatography mass spectrometry and determine that TgTR binds to holo-TgACP4, its predicted cognate ACP, with a KD of 5.75 ± 0.77 µM. Finally, our transcriptional analysis shows that TgPKS2 is upregulated ∼4-fold in the parasite's cyst-forming bradyzoite stage compared to tachyzoites. Our study identifies features that distinguish TgPKS2 from well-characterized systems in bacteria and fungi and suggests it aids the T. gondii cyst stage.


Asunto(s)
NAD , Sintasas Poliquetidas , Sintasas Poliquetidas/química , NAD/metabolismo , Proteína Transportadora de Acilo , Oxidorreductasas/metabolismo
3.
Chembiochem ; 24(17): e202300263, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37171468

RESUMEN

Natural product discovery has traditionally relied on the isolation of small molecules from producing species, but genome-sequencing technology and advances in molecular biology techniques have expanded efforts to a wider array of organisms. Protists represent an underexplored kingdom for specialized metabolite searches despite bioinformatic analysis that suggests they harbor distinct biologically active small molecules. Specifically, pathogenic apicomplexan parasites, responsible for billions of global infections, have been found to possess multiple biosynthetic gene clusters, which hints at their capacity to produce polyketide metabolites. Biochemical studies have revealed unique features of apicomplexan polyketide synthases, but to date, the identity and function of the polyketides synthesized by these megaenzymes remains unknown. Herein, we discuss the potential for specialized metabolite production in protists and the possible evolution of polyketide biosynthetic gene clusters in apicomplexan parasites. We then focus on a polyketide synthase from the apicomplexan Toxoplasma gondii to discuss the unique domain architecture and properties of these proteins when compared to previously characterized systems, and further speculate on the possible functions for polyketides in these pathogenic parasites.


Asunto(s)
Apicomplexa , Policétidos , Metabolismo Secundario , Sintasas Poliquetidas/genética , Sintasas Poliquetidas/metabolismo , Biología Computacional , Apicomplexa/genética , Apicomplexa/metabolismo , Policétidos/química
4.
Bioorg Med Chem Lett ; 92: 129410, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37478957

RESUMEN

A collection of ß-carbolines based on the natural product harmine, a compound known to target the heat shock 90 protein of Plasmodium falciparum, was synthesized and tested for antimalarial activity and potential toxicity. Several of these novel compounds display promising bioactivity, providing a new potential therapeutic with a mode of action that differs versus any currently available clinical treatment.


Asunto(s)
Antimaláricos , Antimaláricos/farmacología , Plasmodium falciparum , Carbolinas/farmacología , Respuesta al Choque Térmico
5.
Proc Natl Acad Sci U S A ; 117(11): 5810-5817, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32127489

RESUMEN

The antihistamine clemastine inhibits multiple stages of the Plasmodium parasite that causes malaria, but the molecular targets responsible for its parasite inhibition were unknown. Here, we applied parallel chemoproteomic platforms to discover the mechanism of action of clemastine and identify that clemastine binds to the Plasmodium falciparum TCP-1 ring complex or chaperonin containing TCP-1 (TRiC/CCT), an essential heterooligomeric complex required for de novo cytoskeletal protein folding. Clemastine destabilized all eight P. falciparum TRiC subunits based on thermal proteome profiling (TPP). Further analysis using stability of proteins from rates of oxidation (SPROX) revealed a clemastine-induced thermodynamic stabilization of the Plasmodium TRiC delta subunit, suggesting an interaction with this protein subunit. We demonstrate that clemastine reduces levels of the major TRiC substrate tubulin in P. falciparum parasites. In addition, clemastine treatment leads to disorientation of Plasmodium mitotic spindles during the asexual reproduction and results in aberrant tubulin morphology suggesting protein aggregation. This clemastine-induced disruption of TRiC function is not observed in human host cells, demonstrating a species selectivity required for targeting an intracellular human pathogen. Our findings encourage larger efforts to apply chemoproteomic methods to assist in target identification of antimalarial drugs and highlight the potential to selectively target Plasmodium TRiC-mediated protein folding for malaria intervention.


Asunto(s)
Chaperonina con TCP-1/metabolismo , Clemastina/farmacología , Antagonistas de los Receptores Histamínicos/farmacología , Proteínas Protozoarias/metabolismo , Sitios de Unión , Línea Celular , Chaperonina con TCP-1/química , Humanos , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Unión Proteica , Proteínas Protozoarias/química , Huso Acromático/efectos de los fármacos
6.
Nature ; 538(7625): 344-349, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27602946

RESUMEN

Antimalarial drugs have thus far been chiefly derived from two sources-natural products and synthetic drug-like compounds. Here we investigate whether antimalarial agents with novel mechanisms of action could be discovered using a diverse collection of synthetic compounds that have three-dimensional features reminiscent of natural products and are underrepresented in typical screening collections. We report the identification of such compounds with both previously reported and undescribed mechanisms of action, including a series of bicyclic azetidines that inhibit a new antimalarial target, phenylalanyl-tRNA synthetase. These molecules are curative in mice at a single, low dose and show activity against all parasite life stages in multiple in vivo efficacy models. Our findings identify bicyclic azetidines with the potential to both cure and prevent transmission of the disease as well as protect at-risk populations with a single oral dose, highlighting the strength of diversity-oriented synthesis in revealing promising therapeutic targets.


Asunto(s)
Antimaláricos/síntesis química , Antimaláricos/farmacología , Azetidinas/uso terapéutico , Descubrimiento de Drogas , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Animales , Antimaláricos/administración & dosificación , Antimaláricos/uso terapéutico , Compuestos de Azabiciclo/administración & dosificación , Compuestos de Azabiciclo/síntesis química , Compuestos de Azabiciclo/farmacología , Compuestos de Azabiciclo/uso terapéutico , Azetidinas/administración & dosificación , Azetidinas/efectos adversos , Azetidinas/farmacología , Citosol/enzimología , Modelos Animales de Enfermedad , Femenino , Hígado/efectos de los fármacos , Hígado/parasitología , Macaca mulatta/parasitología , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Masculino , Ratones , Fenilalanina-ARNt Ligasa/antagonistas & inhibidores , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/síntesis química , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Plasmodium falciparum/citología , Plasmodium falciparum/enzimología , Seguridad
7.
Chembiochem ; 22(16): 2591-2599, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-33999499

RESUMEN

Emerging Plasmodium parasite drug resistance is threatening progress towards malaria control and elimination. While recent efforts in cell-based, high-throughput drug screening have produced first-in-class drugs with promising activities against different Plasmodium life cycle stages, most of these antimalarial agents have elusive mechanisms of action. Though challenging to address, target identification can provide valuable information to facilitate lead optimization and preclinical drug prioritization. Recently, proteome-wide methods for direct assessment of drug-protein interactions have emerged as powerful tools in a number of systems, including Plasmodium. In this review, we will discuss current chemoproteomic strategies that have been adapted to antimalarial drug target discovery, including affinity- and activity-based protein profiling and the energetics-based techniques thermal proteome profiling and stability of proteins from rates of oxidation. The successful application of chemoproteomics to the Plasmodium blood stage highlights the potential of these methods to link inhibitors to their molecular targets in more elusive Plasmodium life stages and intracellular pathogens in the future.


Asunto(s)
Parásitos , Animales
8.
Biochemistry ; 59(8): 911-920, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32073254

RESUMEN

There is a pressing need for compounds with broad-spectrum activity against malaria parasites at various life cycle stages to achieve malaria elimination. However, this goal cannot be accomplished without targeting the tenacious dormant liver-stage hypnozoite that causes multiple relapses after the first episode of illness. In the search for the magic bullet to radically cure Plasmodium vivax malaria, tafenoquine outperformed other candidate drugs and was approved by the U.S. Food and Drug Administration in 2018. Tafenoquine is an 8-aminoquinoline that inhibits multiple life stages of various Plasmodium species. Additionally, its much longer half-life allows for single-dose treatment, which will improve the compliance rate. Despite its approval and the long-time use of other 8-aminoquinolines, the mechanisms behind tafenoquine's activity and adverse effects are still largely unknown. In this Perspective, we discuss the plausible underlying mechanisms of tafenoquine's antiparasitic activity and highlight its role as a cellular stressor. We also discuss potential drug combinations and the development of next-generation 8-aminoquinolines to further improve the therapeutic index of tafenoquine for malaria treatment and prevention.


Asunto(s)
Aminoquinolinas/uso terapéutico , Antimaláricos/uso terapéutico , Malaria Vivax/tratamiento farmacológico , Aminoquinolinas/efectos adversos , Anemia Hemolítica/inducido químicamente , Animales , Antimaláricos/efectos adversos , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/metabolismo , Técnicas de Silenciamiento del Gen , Deficiencia de Glucosafosfato Deshidrogenasa/metabolismo , Haplorrinos , Humanos , Metahemoglobinemia/inducido químicamente , Ratones , Plasmodium cynomolgi/efectos de los fármacos , Plasmodium vivax/efectos de los fármacos , Pez Cebra
9.
Chembiochem ; 21(9): 1279-1284, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-31845464

RESUMEN

Anopheles mosquito microbiomes are intriguing ecological niches. Within the gut, microbes adapt to oxidative stress due to heme and iron after blood meals. Although metagenomic sequencing has illuminated spatial and temporal fluxes of microbiome populations, limited data exist on microbial growth dynamics. Here, we analyze growth interactions between a dominant microbiome species, Elizabethkingia anophelis, and other Anopheles-associated bacteria. We find E. anophelis inhibits a Pseudomonas sp. via an antimicrobial-independent mechanism and observe biliverdins, heme degradation products, upregulated in cocultures. Purification and characterization of E. anophelis HemS demonstrates heme degradation, and we observe hemS expression is upregulated when cocultured with Pseudomonas sp. This study reveals a competitive microbial interaction between mosquito-associated bacteria and characterizes the stimulation of heme degradation in E. anophelis when grown with Pseudomonas sp.


Asunto(s)
Anopheles/microbiología , Proteínas Bacterianas/metabolismo , Flavobacteriaceae/metabolismo , Hemo/metabolismo , Microbiota , Virulencia , Animales , Técnicas de Cocultivo , Flavobacteriaceae/crecimiento & desarrollo , Genoma Bacteriano , Filogenia , Análisis de Secuencia de ADN
10.
PLoS Pathog ; 14(5): e1007057, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29775485

RESUMEN

Within the liver a single Plasmodium parasite transforms into thousands of blood-infective forms to cause malaria. Here, we use RNA-sequencing to identify host genes that are upregulated upon Plasmodium berghei infection of hepatocytes with the hypothesis that host pathways are hijacked to benefit parasite development. We found that expression of aquaporin-3 (AQP3), a water and glycerol channel, is significantly induced in Plasmodium-infected hepatocytes compared to uninfected cells. This aquaglyceroporin localizes to the parasitophorous vacuole membrane, the compartmental interface between the host and pathogen, with a temporal pattern that correlates with the parasite's expansion in the liver. Depletion or elimination of host AQP3 expression significantly reduces P. berghei parasite burden during the liver stage and chemical disruption by a known AQP3 inhibitor, auphen, reduces P. falciparum asexual blood stage and P. berghei liver stage parasite load. Further use of this inhibitor as a chemical probe suggests that AQP3-mediated nutrient transport is an important function for parasite development. This study reveals a previously unknown potential route for host-dependent nutrient acquisition by Plasmodium which was discovered by mapping the transcriptional changes that occur in hepatocytes throughout P. berghei infection. The dataset reported may be leveraged to identify additional host factors that are essential for Plasmodium liver stage infection and highlights Plasmodium's dependence on host factors within hepatocytes.


Asunto(s)
Acuaporina 3/metabolismo , Plasmodium berghei/metabolismo , Animales , Acuaporina 3/fisiología , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/parasitología , Humanos , Hígado/metabolismo , Hígado/parasitología , Hepatopatías , Malaria/parasitología , Ratones , Parásitos/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/parasitología , Proteínas Protozoarias/metabolismo , Análisis de Secuencia de ARN/métodos , Esporozoítos/metabolismo , Vacuolas/metabolismo
11.
Molecules ; 25(3)2020 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-32023950

RESUMEN

The discovery of natural products continues to interest chemists and biologists for their utility in medicine as well as facilitating our understanding of signaling, pathogenesis, and evolution. Despite an attenuation in the discovery rate of new molecules, the current genomics and transcriptomics revolution has illuminated the untapped biosynthetic potential of many diverse organisms. Today, natural product discovery can be driven by biosynthetic gene cluster (BGC) analysis, which is capable of predicting enzymes that catalyze novel reactions and organisms that synthesize new chemical structures. This approach has been particularly effective in mining bacterial and fungal genomes where it has facilitated the discovery of new molecules, increased the understanding of metabolite assembly, and in some instances uncovered enzymes with intriguing synthetic utility. While relatively less is known about the biosynthetic potential of non-fungal eukaryotes, there is compelling evidence to suggest many encode biosynthetic enzymes that produce molecules with unique bioactivities. In this review, we highlight how the advances in genomics and transcriptomics have aided natural product discovery in sources from eukaryotic lineages. We summarize work that has successfully connected genes to previously identified molecules and how advancing these techniques can lead to genetics-guided discovery of novel chemical structures and reactions distributed throughout the tree of life. Ultimately, we discuss the advantage of increasing the known biosynthetic space to ease access to complex natural and non-natural small molecules.


Asunto(s)
Productos Biológicos/metabolismo , Vías Biosintéticas , Descubrimiento de Drogas , Eucariontes , Perfilación de la Expresión Génica , Genómica , Familia de Multigenes
12.
Chemistry ; 25(26): 6500-6504, 2019 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-30912197

RESUMEN

As traditional small-molecule drug discovery programs focus on a relatively narrow range of chemical space, most human proteins are viewed as unreachable targets. Consequently, there is a strong interest in expanding the chemical space in drug discovery beyond traditional small molecules. Here, a strategy for the preparation of a broad natural-product-like macrocyclic library by using the tandem allylic oxidation/oxa-conjugate addition and macrocyclization reactions is reported. Cheminformatic analyses demonstrate that this tetrahydropyran-containing macrocyclic library shows a significant overlap with natural products in the chemical space. This approach can be used for designing libraries that may probe more deeply into natural-product-like space.

13.
Biochemistry ; 57(4): 365-375, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-28976181

RESUMEN

Apicomplexan parasites encompass a diverse group of eukaryotic intracellular pathogens that infect various animal hosts to cause disease. Intriguingly, apicomplexans possess a unique organelle of algal origin, the apicoplast, which phylogenetically links these parasites to dinoflagellates and photosynthetic, coral-associated organisms. While production of secondary metabolites in closely related organisms has been thoroughly examined, it remains widely unexplored in apicomplexans. In this Perspective, we discuss previous work toward understanding secondary metabolite building block biosynthesis in apicomplexans and highlight the unexplored enzymology and biosynthetic potential of these parasites in the context of evolution.


Asunto(s)
Apicomplexa/metabolismo , Apicoplastos/metabolismo , Evolución Biológica , Interacciones Huésped-Parásitos , Estadios del Ciclo de Vida , Filogenia , Proteínas Protozoarias/metabolismo , Metabolismo Secundario
14.
Artículo en Inglés | MEDLINE | ID: mdl-29339390

RESUMEN

Malaria remains a global health burden partly due to Plasmodium parasite resistance to first-line therapeutics. The molecular chaperone heat shock protein 90 (Hsp90) has emerged as an essential protein for blood-stage Plasmodium parasites, but details about its function during malaria's elusive liver stage are unclear. We used target-based screens to identify compounds that bind to Plasmodium falciparum and human Hsp90, which revealed insights into chemotypes with species-selective binding. Using cell-based malaria assays, we demonstrate that all identified Hsp90-binding compounds are liver- and blood-stage Plasmodium inhibitors. Additionally, the Hsp90 inhibitor SNX-0723 in combination with the phosphatidylinositol 3-kinase inhibitor PIK-75 synergistically reduces the liver-stage parasite load. Time course inhibition studies with the Hsp90 inhibitors and expression analysis support a role for Plasmodium Hsp90 in late-liver-stage parasite development. Our results suggest that Plasmodium Hsp90 is essential to liver- and blood-stage parasite infections and highlight an attractive route for development of species-selective PfHsp90 inhibitors that may act synergistically in combination therapies to prevent and treat malaria.


Asunto(s)
Antimaláricos/uso terapéutico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Benzamidas/uso terapéutico , Proteínas HSP90 de Choque Térmico/metabolismo , Interacciones Huésped-Patógeno , Humanos , Hidrazonas/uso terapéutico , Indoles/uso terapéutico , Malaria/tratamiento farmacológico , Malaria/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Sulfonamidas/uso terapéutico , ortoaminobenzoatos/uso terapéutico
15.
Chembiochem ; 19(15): 1590-1594, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29700993

RESUMEN

The Anopheles mosquito that harbors the Plasmodium parasite contains a microbiota that can influence both the vector and the parasite. In recent years, insect-associated microbes have highlighted the untapped potential of exploiting interspecies interactions to discover bioactive compounds. In this study, we report the discovery of nonribosomal lipodepsipeptides that are produced by a Serratia sp. within the midgut and salivary glands of Anopheles stephensi mosquitoes. The lipodepsipeptides, stephensiolides A-K, have antibiotic activity and facilitate bacterial surface motility. Bioinformatic analyses indicate that the stephensiolides are ubiquitous in nature and are likely important for Serratia spp. colonization within mosquitoes, humans, and other ecological niches. Our results demonstrate the usefulness of probing insect-microbiome interactions, enhance our understanding of the chemical ecology within Anopheles mosquitoes, and provide a secondary-metabolite scaffold for further investigate of this complex relationship.


Asunto(s)
Anopheles/microbiología , Antiinfecciosos/metabolismo , Depsipéptidos/metabolismo , Lipopéptidos/metabolismo , Mosquitos Vectores/microbiología , Serratia/metabolismo , Animales , Antiinfecciosos/química , Antiinfecciosos/aislamiento & purificación , Antiinfecciosos/farmacología , Bacterias/efectos de los fármacos , Depsipéptidos/química , Depsipéptidos/aislamiento & purificación , Depsipéptidos/farmacología , Células Hep G2 , Humanos , Lipopéptidos/química , Lipopéptidos/aislamiento & purificación , Lipopéptidos/farmacología , Malaria/parasitología , Malaria/transmisión , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Plasmodium falciparum/efectos de los fármacos
16.
Antimicrob Agents Chemother ; 60(3): 1430-7, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26666931

RESUMEN

Malaria remains a major global health problem, with more than half of the world population at risk of contracting the disease and nearly a million deaths each year. Here, we report the discovery of inhibitors that target multiple stages of malaria parasite growth. To identify these inhibitors, we took advantage of the Tres Cantos Antimalarial Compound Set (TCAMS) small-molecule library, which is comprised of diverse and potent chemical scaffolds with activities against the blood stage of the malaria parasite, and investigated their effects against the elusive liver stage of the malaria parasite using a forward chemical screen. From a screen of nearly 14,000 compounds, we identified and confirmed 103 compounds as dual-stage malaria inhibitors. Interestingly, these compounds show preferential inhibition of parasite growth in liver- versus blood-stage malaria parasite assays, highlighting the drug susceptibility of this parasite form. Mode-of-action studies were completed using genetically modified and drug-resistant Plasmodium parasite strains. While we identified some compound targets as classical antimalarial pathways, such as the mitochondrial electron transport chain through cytochrome bc1 complex inhibition or the folate biosynthesis pathway, most compounds induced parasite death through as yet unknown mechanisms of action. Importantly, the identification of new chemotypes with different modes of action in killing Plasmodium parasites represents a promising opportunity for probing essential and novel molecular processes that remain to be discovered. The chemical scaffolds identified with activity against drug-resistant Plasmodium parasites represent starting points for dual-stage antimalarial development to surmount the threat of malaria parasite drug resistance.


Asunto(s)
Antimaláricos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Plasmodium berghei/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Animales Modificados Genéticamente , Anopheles/parasitología , Dihidroorotato Deshidrogenasa , Células Hep G2/efectos de los fármacos , Células Hep G2/parasitología , Humanos , Terapia Molecular Dirigida/métodos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo
17.
Proc Natl Acad Sci U S A ; 109(22): 8511-6, 2012 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-22586124

RESUMEN

Human malaria infection begins with a one-time asymptomatic liver stage followed by a cyclic symptomatic blood stage. All high-throughput malaria drug discovery efforts have focused on the cyclic blood stage, which has limited potential for the prophylaxis, transmission blocking, and eradication efforts that will be needed in the future. To address these unmet needs, a high-throughput phenotypic liver-stage Plasmodium parasite screen was developed to systematically identify molecules with liver-stage efficacy. The screen recapitulates liver-stage infection by isolating luciferase-expressing Plasmodium berghei parasites directly from the salivary glands of infected mosquitoes, adding them to confluent human liver cells in 384-well plates, and measuring luciferase activity after a suitable incubation period. Screening 5,375 known bioactive compounds identified 37 liver-stage malaria inhibitors with diverse modes of action, as shown by inhibition time course experiments. Further analysis of the hits in the Food and Drug Administration-approved drug subset revealed compounds that seem to act specifically on the liver stage of infection, suggesting that this phase of the parasite's life cycle presents a promising area for new drug discovery. Notably, many active compounds in this screen have molecular structures and putative targets distinctly different from those of known antimalarial agents.


Asunto(s)
Antimaláricos/farmacología , Hígado/efectos de los fármacos , Malaria/prevención & control , Plasmodium berghei/efectos de los fármacos , Animales , Anopheles/parasitología , Antimaláricos/clasificación , Evaluación Preclínica de Medicamentos , Células Hep G2 , Humanos , Concentración 50 Inhibidora , Insectos Vectores/parasitología , Estadios del Ciclo de Vida , Hígado/parasitología , Hígado/patología , Malaria/parasitología , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/aislamiento & purificación , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Resultado del Tratamiento
18.
Antimicrob Agents Chemother ; 58(3): 1516-22, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24366746

RESUMEN

Drugs that target both the liver and blood stages of malaria will be needed to reduce the disease's substantial worldwide morbidity and mortality. Evaluation of a 259-member library of compounds that block proliferation of the blood stage of malaria revealed several scaffolds--dihydroquinazolinones, phenyldiazenylpyridines, piperazinyl methyl quinolones, and bis-benzimidazoles--with promising activity against the liver stage. Focused structure-activity studies on the dihydroquinazolinone scaffold revealed several molecules with excellent potency against both blood and liver stages. One promising early lead with dual activity is 2-(p-bromophenyl)-3-(2-(diethylamino)ethyl)-2,3-dihydroquinazolin-4(1H)-one with 50% effective concentrations (EC50s) of 0.46 µM and 0.34 µM against liver stage Plasmodium berghei ANKA and blood stage Plasmodium falciparum 3D7 parasites, respectively. Structure-activity relationships revealed that liver stage activity for this compound class requires a 3-dialkyl amino ethyl group and is abolished by substitution at the ortho-position of the phenyl moiety. These compounds have minimal toxicity to mammalian cells and are thus attractive compounds for further development.


Asunto(s)
Antimaláricos/farmacología , Hígado/parasitología , Plasmodium/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria/sangre , Malaria/tratamiento farmacológico , Malaria/parasitología , Plasmodium/crecimiento & desarrollo , Plasmodium berghei/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Quinazolinas/antagonistas & inhibidores , Relación Estructura-Actividad
19.
Chembiochem ; 15(13): 1920-30, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25111632

RESUMEN

Malaria, an infectious disease caused by eukaryotic parasites of the genus Plasmodium, afflicts hundreds of millions of people every year. Both the parasite and its host utilize protein kinases to regulate essential cellular processes. Bioinformatic analyses of parasite genomes predict at least 65 protein kinases, but their biological functions and therapeutic potential are largely unknown. We profiled 1358 small-molecule kinase inhibitors to evaluate the role of both the human and the malaria kinomes in Plasmodium infection of liver cells, the parasites' obligatory but transient developmental stage that precedes the symptomatic blood stage. The screen identified several small molecules that inhibit parasite load in liver cells, some with nanomolar efficacy, and each compound was subsequently assessed for activity against blood-stage malaria. Most of the screening hits inhibited both liver- and blood-stage malaria parasites, which have dissimilar gene expression profiles and infect different host cells. Evaluation of existing kinase activity profiling data for the library members suggests that several kinases are essential to malaria parasites, including cyclin-dependent kinases (CDKs), glycogen synthase kinases, and phosphoinositide-3-kinases. CDK inhibitors were found to bind to Plasmodium protein kinase 5, but it is likely that these compounds target multiple parasite kinases. The dual-stage inhibition of the identified kinase inhibitors makes them useful chemical probes and promising starting points for antimalarial development.


Asunto(s)
Genoma de Protozoos/genética , Malaria/genética , Plasmodium/genética , Proteínas Quinasas/genética , Animales , Antimaláricos/química , Biología Computacional , Evaluación Preclínica de Medicamentos , Humanos , Hígado/parasitología , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Plasmodium/enzimología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Bibliotecas de Moléculas Pequeñas
20.
Cell Chem Biol ; 31(4): 625-626, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38640899

RESUMEN

In an interview with Samantha Nelson, a scientific editor of Cell Chemical Biology, the first and corresponding authors of the research article entitled Selective targeting of Plasmodium falciparum Hsp90 disrupts the 26S proteasome share more about the project and their perspectives on the field.


Asunto(s)
Plasmodium falciparum , Complejo de la Endopetidasa Proteasomal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA