Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
mSphere ; : e0044421, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34190588

RESUMEN

Egress from host cells is an essential step in the lytic cycle of T. gondii and other apicomplexan parasites; however, only a few parasite secretory proteins are known to affect this process. The putative metalloproteinase toxolysin 4 (TLN4) was previously shown to be an extensively processed microneme protein, but further characterization was impeded by the inability to genetically ablate TLN4. Here, we show that TLN4 has the structural properties of an M16 family metalloproteinase, that it possesses proteolytic activity on a model substrate, and that genetic disruption of TLN4 reduces the efficiency of egress from host cells. Complementation of the knockout strain with the TLN4 coding sequence significantly restored egress competency, affirming that the phenotype of the Δtln4 parasite was due to the absence of TLN4. This work identifies TLN4 as the first metalloproteinase and the second microneme protein to function in T. gondii egress. The study also lays a foundation for future mechanistic studies defining the precise role of TLN4 in parasite exit from host cells. IMPORTANCE After replicating within infected host cells, the single-celled parasite Toxoplasma gondii must rupture out of such cells in a process termed egress. Although it is known that T. gondii egress is an active event that involves disruption of host-derived membranes surrounding the parasite, very few proteins that are released by the parasite are known to facilitate egress. In this study, we identify a parasite secretory protease that is necessary for efficient and timely egress, laying the foundation for understanding precisely how this protease facilitates T. gondii exit from host cells.

2.
Nat Microbiol ; 2: 17035, 2017 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-28319084

RESUMEN

Argonaute (Ago) proteins in eukaryotes are known as key players in post-transcriptional gene silencing1, while recent studies on prokaryotic Agos hint at their role in the protection against invading DNA2,3. Here, we present crystal structures of the apo enzyme and a binary Ago-guide complex of the archaeal Methanocaldococcus jannaschii (Mj) Ago. Binding of a guide DNA leads to large structural rearrangements. This includes the structural transformation of a hinge region containing a switch helix, which has been shown for human Ago2 to be critical for the dynamic target search process4-6. To identify key residues crucial for MjAgo function, we analysed the effect of several MjAgo mutants. We observe that the nature of the 3' and 5' nucleotides in particular, as well as the switch helix, appear to impact MjAgo cleavage activity. In summary, we provide insights into the molecular mechanisms that drive DNA-guided DNA silencing by an archaeal Ago.


Asunto(s)
Proteínas Argonautas/química , Proteínas Argonautas/metabolismo , ADN de Archaea/genética , Silenciador del Gen , Proteínas Argonautas/genética , Cristalografía por Rayos X , ADN de Archaea/metabolismo , Humanos , Methanocaldococcus/genética , Methanocaldococcus/metabolismo , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Interferencia de ARN
3.
Proc Natl Acad Sci U S A ; 112(50): 15360-5, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26627714

RESUMEN

The lethal factor in stonefish venom is stonustoxin (SNTX), a heterodimeric cytolytic protein that induces cardiovascular collapse in humans and native predators. Here, using X-ray crystallography, we make the unexpected finding that SNTX is a pore-forming member of an ancient branch of the Membrane Attack Complex-Perforin/Cholesterol-Dependent Cytolysin (MACPF/CDC) superfamily. SNTX comprises two homologous subunits (α and ß), each of which comprises an N-terminal pore-forming MACPF/CDC domain, a central focal adhesion-targeting domain, a thioredoxin domain, and a C-terminal tripartite motif family-like PRY SPla and the RYanodine Receptor immune recognition domain. Crucially, the structure reveals that the two MACPF domains are in complex with one another and arranged into a stable early prepore-like assembly. These data provide long sought after near-atomic resolution insights into how MACPF/CDC proteins assemble into prepores on the surface of membranes. Furthermore, our analyses reveal that SNTX-like MACPF/CDCs are distributed throughout eukaryotic life and play a broader, possibly immune-related function outside venom.


Asunto(s)
Venenos de los Peces/química , Perforina/química , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , Colesterol/química , Complejo de Ataque a Membrana del Sistema Complemento/química , Cristalografía por Rayos X , Microscopía Electrónica de Transmisión , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Solubilidad , Homología Estructural de Proteína
4.
J Med Chem ; 56(12): 5213-7, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23713488

RESUMEN

The malaria parasite Plasmodium falciparum employs two metallo-aminopeptidases, PfA-M1 and PfA-M17, which are essential for parasite survival. Compounds that inhibit the activity of either enzyme represent leads for the development of new antimalarial drugs. Here we report the synthesis and structure-activity relationships of a small library of phosphonic acid arginine mimetics that probe the S1 pocket of both enzymes and map the necessary interactions that would be important for a dual inhibitor.


Asunto(s)
Arginina/química , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Antígenos CD13/antagonistas & inhibidores , Ácidos Fosforosos/química , Plasmodium falciparum/enzimología , Materiales Biomiméticos/síntesis química , Antígenos CD13/química , Dominio Catalítico , Técnicas de Química Sintética , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Modelos Moleculares , Relación Estructura-Actividad
5.
J Mol Biol ; 422(4): 495-507, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22709581

RESUMEN

The malarial aminopeptidases have emerged as promising new drug targets for the development of novel antimalarial drugs. The M18AAP of Plasmodium falciparum malaria is a metallo-aminopeptidase that we show demonstrates a highly restricted specificity for peptides with an N-terminal Glu or Asp residue. Thus, the enzyme may function alongside other aminopeptidases in effecting the complete degradation or turnover of proteins, such as host hemoglobin, which provides a free amino acid pool for the growing parasite. Inhibition of PfM18AAP's function using antisense RNA is detrimental to the intra-erythrocytic malaria parasite and, hence, it has been proposed as a potential novel drug target. We report the X-ray crystal structure of the PfM18AAP aminopeptidase and reveal its complex dodecameric assembly arranged via dimer and trimer units that interact to form a large tetrahedron shape that completely encloses the 12 active sites within a central cavity. The four entry points to the catalytic lumen are each guarded by 12 large flexible loops that could control substrate entry into the catalytic sites. PfM18AAP thus resembles a proteasomal-like machine with multiple active sites able to degrade peptide substrates that enter the central lumen. The Plasmodium enzyme shows significant structural differences around the active site when compared to recently determined structures of its mammalian and human homologs, which provides a platform from which a rational approach to inhibitor design of new malaria-specific drugs can begin.


Asunto(s)
Aminopeptidasas/química , Malaria Falciparum/enzimología , Plasmodium falciparum/enzimología , Proteínas Protozoarias/química , Aminoácidos/química , Aminoácidos/metabolismo , Aminopeptidasas/metabolismo , Animales , Dominio Catalítico , Cristalografía por Rayos X/métodos , Eritrocitos/metabolismo , Humanos , Malaria Falciparum/parasitología , Péptidos/química , Péptidos/metabolismo , Proteolisis , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
6.
J Med Chem ; 54(6): 1655-66, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21366301

RESUMEN

The malarial PfA-M1 metallo-aminopeptidase is considered a putative drug target. The natural product dipeptide mimetic, bestatin, is a potent inhibitor of PfA-M1. Herein we present a new, efficient, and high-yielding protocol for the synthesis of bestatin derivatives from natural and unnatural N-Boc-d-amino acids. A diverse library of bestatin derivatives was synthesized with variants at the side chain of either the α-hydroxy-ß-amino acid (P1) or the adjacent natural α-amino acid (P1'). Surprisingly, we found that extended aromatic side chains at the P1 position resulted in potent inhibition against PfA-M1. To understand these data, we determined the X-ray cocrystal structures of PfA-M1 with two derivatives having either a Tyr(OMe) 15 or Tyr(OBzl) 16 at the P1 position and observed substantial inhibitor-induced rearrangement of the primary loop within the PfA-M1 pocket that interacts with the P1 side chain. Our data provide important insights for the rational design of more potent and selective inhibitors of this enzyme that may eventually lead to new therapies for malaria.


Asunto(s)
Antimaláricos/síntesis química , Antígenos CD13/antagonistas & inhibidores , Leucina/análogos & derivados , Plasmodium falciparum/efectos de los fármacos , Antimaláricos/química , Antimaláricos/farmacología , Sitios de Unión , Antígenos CD13/química , Cristalografía por Rayos X , Leucina/síntesis química , Leucina/química , Leucina/farmacología , Modelos Moleculares , Estructura Molecular , Plasmodium falciparum/enzimología , Unión Proteica , Bibliotecas de Moléculas Pequeñas , Estereoisomerismo , Relación Estructura-Actividad
7.
Proc Natl Acad Sci U S A ; 107(6): 2449-54, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20133789

RESUMEN

Current therapeutics and prophylactics for malaria are under severe challenge as a result of the rapid emergence of drug-resistant parasites. The human malaria parasite Plasmodium falciparum expresses two neutral aminopeptidases, PfA-M1 and PfA-M17, which function in regulating the intracellular pool of amino acids required for growth and development inside the red blood cell. These enzymes are essential for parasite viability and are validated therapeutic targets. We previously reported the X-ray crystal structure of the monomeric PfA-M1 and proposed a mechanism for substrate entry and free amino acid release from the active site. Here, we present the X-ray crystal structure of the hexameric leucine aminopeptidase, PfA-M17, alone and in complex with two inhibitors with antimalarial activity. The six active sites of the PfA-M17 hexamer are arranged in a disc-like fashion so that they are orientated inwards to form a central catalytic cavity; flexible loops that sit at each of the six entrances to the catalytic cavern function to regulate substrate access. In stark contrast to PfA-M1, PfA-M17 has a narrow and hydrophobic primary specificity pocket which accounts for its highly restricted substrate specificity. We also explicate the essential roles for the metal-binding centers in these enzymes (two in PfA-M17 and one in PfA-M1) in both substrate and drug binding. Our detailed understanding of the PfA-M1 and PfA-M17 active sites now permits a rational approach in the development of a unique class of two-target and/or combination antimalarial therapy.


Asunto(s)
Aminopeptidasas/química , Diseño de Fármacos , Plasmodium falciparum/enzimología , Proteínas Protozoarias/química , Aminopeptidasas/antagonistas & inhibidores , Aminopeptidasas/metabolismo , Antimaláricos/química , Antimaláricos/metabolismo , Antimaláricos/farmacología , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Interacciones Hidrofóbicas e Hidrofílicas , Metales/química , Metales/metabolismo , Modelos Moleculares , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...