Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Trends Biochem Sci ; 47(2): 149-159, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34887149

RESUMO

The difficulty of faithfully recapitulating malarial protein complexes in heterologous expression systems has long impeded structural study for much of the Plasmodium falciparum proteome. However, recent advances in single-particle cryo electron microscopy (cryoEM) now enable structure determination at atomic resolution with significantly reduced requirements for both sample quantity and purity. Combined with recent developments in gene editing, these advances open the door to structure determination and structural proteomics of macromolecular complexes enriched directly from P. falciparum parasites. Furthermore, the combination of cryoEM with the rapidly emerging use of in situ cryo electron tomography (cryoET) to directly visualize ultrastructures and protein complexes in the native cellular context will yield exciting new insights into the molecular machinery underpinning malaria parasite biology and pathogenesis.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Microscopia Crioeletrônica/métodos , Malária Falciparum/parasitologia , Plasmodium falciparum/metabolismo
2.
Nature ; 561(7721): 70-75, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30150771

RESUMO

The putative Plasmodium translocon of exported proteins (PTEX) is essential for transport of malarial effector proteins across a parasite-encasing vacuolar membrane into host erythrocytes, but the mechanism of this process remains unknown. Here we show that PTEX is a bona fide translocon by determining structures of the PTEX core complex at near-atomic resolution using cryo-electron microscopy. We isolated the endogenous PTEX core complex containing EXP2, PTEX150 and HSP101 from Plasmodium falciparum in the 'engaged' and 'resetting' states of endogenous cargo translocation using epitope tags inserted using the CRISPR-Cas9 system. In the structures, EXP2 and PTEX150 interdigitate to form a static, funnel-shaped pseudo-seven-fold-symmetric protein-conducting channel spanning the vacuolar membrane. The spiral-shaped AAA+ HSP101 hexamer is tethered above this funnel, and undergoes pronounced compaction that allows three of six tyrosine-bearing pore loops lining the HSP101 channel to dissociate from the cargo, resetting the translocon for the next threading cycle. Our work reveals the mechanism of P. falciparum effector export, and will inform structure-based design of drugs targeting this unique translocon.


Assuntos
Microscopia Crioeletrônica , Plasmodium falciparum/ultraestrutura , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/ultraestrutura , Animais , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Modelos Biológicos , Modelos Moleculares , Terapia de Alvo Molecular/tendências , Movimento , Plasmodium falciparum/química , Plasmodium falciparum/metabolismo , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Transporte Proteico , Proteínas de Protozoários/química , Vacúolos/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34446549

RESUMO

The RhopH complex is implicated in malaria parasites' ability to invade and create new permeability pathways in host erythrocytes, but its mechanisms remain poorly understood. Here, we enrich the endogenous RhopH complex in a native soluble form, comprising RhopH2, CLAG3.1, and RhopH3, directly from parasite cell lysates and determine its atomic structure using cryo-electron microscopy (cryo-EM), mass spectrometry, and the cryoID program. CLAG3.1 is positioned between RhopH2 and RhopH3, which both share substantial binding interfaces with CLAG3.1 but make minimal contacts with each other. The forces stabilizing individual subunits include 13 intramolecular disulfide bonds. Notably, CLAG3.1 residues 1210 to 1223, previously predicted to constitute a transmembrane helix, are embedded within a helical bundle formed by residues 979 to 1289 near the C terminus of CLAG3.1. Buried in the core of the RhopH complex and largely shielded from solvent, insertion of this putative transmembrane helix into the erythrocyte membrane would likely require a large conformational rearrangement. Given the unusually high disulfide content of the complex, it is possible that such a rearrangement could be initiated by the breakage of allosteric disulfide bonds, potentially triggered by interactions at the erythrocyte membrane. This first direct observation of an exported Plasmodium falciparum transmembrane protein-in a soluble, trafficking state and with atomic details of buried putative membrane-insertion helices-offers insights into the assembly and trafficking of RhopH and other parasite-derived complexes to the erythrocyte membrane. Our study demonstrates the potential the endogenous structural proteomics approach holds for elucidating the molecular mechanisms of hard-to-isolate complexes in their native, functional forms.


Assuntos
Membrana Eritrocítica/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/química , Permeabilidade da Membrana Celular , Microscopia Crioeletrônica , Membrana Eritrocítica/parasitologia , Humanos , Modelos Moleculares , Nutrientes/metabolismo , Conformação Proteica , Proteômica , Proteínas de Protozoários/fisiologia , Proteínas de Protozoários/ultraestrutura , Relação Estrutura-Atividade
4.
Nat Methods ; 17(1): 79-85, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31768063

RESUMO

X-ray crystallography often requires non-native constructs involving mutations or truncations, and is challenged by membrane proteins and large multicomponent complexes. We present here a bottom-up endogenous structural proteomics approach whereby near-atomic-resolution cryo electron microscopy (cryoEM) maps are reconstructed ab initio from unidentified protein complexes enriched directly from the endogenous cellular milieu, followed by identification and atomic modeling of the proteins. The proteins in each complex are identified using cryoID, a program we developed to identify proteins in ab initio cryoEM maps. As a proof of principle, we applied this approach to the malaria-causing parasite Plasmodium falciparum, an organism that has resisted conventional structural-biology approaches, to obtain atomic models of multiple protein complexes implicated in intraerythrocytic survival of the parasite. Our approach is broadly applicable for determining structures of undiscovered protein complexes enriched directly from endogenous sources.


Assuntos
Microscopia Crioeletrônica/métodos , Eritrócitos/parasitologia , Processamento de Imagem Assistida por Computador/métodos , Complexos Multiproteicos/química , Plasmodium falciparum/metabolismo , Proteômica/métodos , Proteínas de Protozoários/química , Secretases da Proteína Precursora do Amiloide/metabolismo , Humanos , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Espectrometria de Massas , Modelos Moleculares , Complexos Multiproteicos/ultraestrutura , Plasmodium falciparum/isolamento & purificação , Conformação Proteica , Proteínas de Protozoários/ultraestrutura
5.
PLoS Pathog ; 17(4): e1009394, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33793667

RESUMO

Obligate intracellular malaria parasites reside within a vacuolar compartment generated during invasion which is the principal interface between pathogen and host. To subvert their host cell and support their metabolism, these parasites coordinate a range of transport activities at this membrane interface that are critically important to parasite survival and virulence, including nutrient import, waste efflux, effector protein export, and uptake of host cell cytosol. Here, we review our current understanding of the transport mechanisms acting at the malaria parasite vacuole during the blood and liver-stages of development with a particular focus on recent advances in our understanding of effector protein translocation into the host cell by the Plasmodium Translocon of EXported proteins (PTEX) and small molecule transport by the PTEX membrane-spanning pore EXP2. Comparison to Toxoplasma gondii and other related apicomplexans is provided to highlight how similar and divergent mechanisms are employed to fulfill analogous transport activities.


Assuntos
Transporte Biológico/fisiologia , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita/imunologia , Malária Falciparum/parasitologia , Plasmodium falciparum/metabolismo , Animais , Humanos , Malária/metabolismo
6.
J Am Chem Soc ; 142(9): 4445-4455, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32064871

RESUMO

The lipopolysaccharide biosynthesis pathway is considered an attractive drug target against the rising threat of multi-drug-resistant Gram-negative bacteria. Here, we report two novel small-molecule inhibitors (compounds 1 and 2) of the acyltransferase LpxA, the first enzyme in the lipopolysaccharide biosynthesis pathway. We show genetically that the antibacterial activities of the compounds against efflux-deficient Escherichia coli are mediated by LpxA inhibition. Consistently, the compounds inhibited the LpxA enzymatic reaction in vitro. Intriguingly, using biochemical, biophysical, and structural characterization, we reveal two distinct mechanisms of LpxA inhibition; compound 1 is a substrate-competitive inhibitor targeting apo LpxA, and compound 2 is an uncompetitive inhibitor targeting the LpxA/product complex. Compound 2 exhibited more favorable biological and physicochemical properties than compound 1 and was optimized using structural information to achieve improved antibacterial activity against wild-type E. coli. These results show that LpxA is a promising antibacterial target and imply the advantages of targeting enzyme/product complexes in drug discovery.


Assuntos
Aciltransferases/antagonistas & inibidores , Antibacterianos/farmacologia , Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , Pirazóis/farmacologia , Aciltransferases/metabolismo , Antibacterianos/metabolismo , Cristalografia por Raios X , Inibidores Enzimáticos/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/enzimologia , Imidazóis/metabolismo , Testes de Sensibilidade Microbiana , Ligação Proteica , Pirazóis/metabolismo
7.
Proc Natl Acad Sci U S A ; 107(21): 9638-43, 2010 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-20457942

RESUMO

In humans, NH(3) transport across cell membranes is facilitated by the Rh (rhesus) family of proteins. Human Rh C glycoprotein (RhCG) forms a trimeric complex that plays an essential role in ammonia excretion and renal pH regulation. The X-ray crystallographic structure of human RhCG, determined at 2.1 A resolution, reveals the mechanism of ammonia transport. Each monomer contains 12 transmembrane helices, one more than in the bacterial homologs. Reconstituted into proteoliposomes, RhCG conducts NH(3) to raise internal pH. Models of the erythrocyte Rh complex based on our RhCG structure suggest that the erythrocytic Rh complex is composed of stochastically assembled heterotrimers of RhAG, RhD, and RhCE.


Assuntos
Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Sistema do Grupo Sanguíneo Rh-Hr/química , Sistema do Grupo Sanguíneo Rh-Hr/metabolismo , Amônia/metabolismo , Transporte Biológico , Linhagem Celular , Cristalografia por Raios X , Eritrócitos/química , Eritrócitos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
8.
mSphere ; 8(2): e0061522, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-36892278

RESUMO

Drs. Monica Mugnier and Chi-Min Ho work in the field of parasitology. In this mSphere of Influence article, they share their experience as cochairs of the Young Investigators in Parasitology (YIPs) meeting, a 2-day biennial meeting for new PIs in parasitology. Setting up a new lab can be a daunting task. YIPS is designed to make the transition a little easier. YIPs is both a crash course in the skills needed to run a successful research lab and a way to build community among new group leaders in parasitology. In this perspective, they describe YIPs and the benefit it has had on the molecular parasitology community. They also provide some tips for building and running a meeting like YIPs, in the hopes that other fields might replicate their model.


Assuntos
Parasitologia
9.
Acta Crystallogr D Struct Biol ; 77(Pt 4): 457-462, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33825706

RESUMO

Using single-particle electron cryo-microscopy (cryo-EM), it is possible to obtain multiple reconstructions showing the 3D structures of proteins imaged as a mixture. Here, it is shown that automatic map interpretation based on such reconstructions can be used to create atomic models of proteins as well as to match the proteins to the correct sequences and thereby to identify them. This procedure was tested using two proteins previously identified from a mixture at resolutions of 3.2 Å, as well as using 91 deposited maps with resolutions between 2 and 4.5 Å. The approach is found to be highly effective for maps obtained at resolutions of 3.5 Šand better, and to have some utility at resolutions as low as 4 Å.


Assuntos
Microscopia Crioeletrônica/métodos , Modelos Moleculares , Proteínas/química , Conformação Proteica , Software
10.
ACS Infect Dis ; 6(6): 1480-1489, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-31402665

RESUMO

LpxD, acyl-ACP-dependent N-acyltransferase, is the third enzyme of lipid A biosynthesis in Gram-negative bacteria. A recent probe-based screen identified several compounds, including 6359-0284 (compound 1), that inhibit the enzymatic activity of Escherichia coli (E. coli) LpxD. Here, we use these inhibitors to chemically validate LpxD as an attractive antibacterial target. We first found that compound 1 was oxidized in solution to the more stable aromatized tetrahydro-pyrazolo-quinolinone compound 1o. From the Escherichia coli strain deficient in efflux, we isolated a mutant that was less susceptible to compound 1o and had an lpxD missense mutation (Gly268Cys), supporting the cellular on-target activity. Using surface plasma resonance, we showed direct binding to E. coli LpxD for compound 1o and other reported LpxD inhibitors in vitro. Furthermore, we determined eight cocrystal structures of E. coli LpxD/inhibitor complexes. These costructures pinpointed the 4'-phosphopantetheine binding site as the common ligand binding hotspot, where hydrogen bonds to Gly269 and/or Gly287 were important for inhibitor binding. In addition, the LpxD/compound 1o costructure rationalized the reduced activity of compound 1o in the LpxDGly268Cys mutant. Moreover, we obtained the LpxD structure in complex with a previously reported LpxA/LpxD dual targeting peptide inhibitor, RJPXD33, providing structural rationale for the unique dual targeting properties of this peptide. Given that the active site residues of LpxD are conserved in multidrug resistant Enterobacteriaceae, this work paves the way for future LpxD drug discovery efforts combating these Gram-negative pathogens.


Assuntos
Aciltransferases , Proteínas de Escherichia coli , Escherichia coli , Aciltransferases/antagonistas & inibidores , Aciltransferases/genética , Sítios de Ligação , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/antagonistas & inibidores , Lipídeo A , Lipopolissacarídeos
11.
J Mol Biol ; 385(3): 820-30, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-19061901

RESUMO

A medium-throughput approach is used to rapidly identify membrane proteins from a eukaryotic organism that are most amenable to expression in amounts and quality adequate to support structure determination. The goal was to expand knowledge of new membrane protein structures based on proteome-wide coverage. In the first phase, membrane proteins from the budding yeast Saccharomyces cerevisiae were selected for homologous expression in S. cerevisiae, a system that can be adapted to expression of membrane proteins from other eukaryotes. We performed medium-scale expression and solubilization tests on 351 rationally selected membrane proteins from S. cerevisiae. These targets are inclusive of all annotated and unannotated membrane protein families within the organism's membrane proteome. Two hundred seventy-two targets were expressed, and of these, 234 solubilized in the detergent n-dodecyl-beta-D-maltopyranoside. Furthermore, we report the identity of a subset of targets that were purified to homogeneity to facilitate structure determinations. The extensibility of this approach is demonstrated with the expression of 10 human integral membrane proteins from the solute carrier superfamily. This discovery-oriented pipeline provides an efficient way to select proteins from particular membrane protein classes, families, or organisms that may be more suited to structure analysis than others.


Assuntos
Proteínas de Membrana/química , Proteínas de Saccharomyces cerevisiae/química , Cromatografia de Afinidade , Cromatografia em Gel , Humanos , Proteínas de Membrana/isolamento & purificação , Plasmídeos , Sinais Direcionadores de Proteínas , Proteínas de Saccharomyces cerevisiae/isolamento & purificação , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA