Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34706941

RESUMO

Type III secretion systems are multiprotein molecular machines required for the virulence of several important bacterial pathogens. The central element of these machines is the injectisome, a ∼5-Md multiprotein structure that mediates the delivery of bacterially encoded proteins into eukaryotic target cells. The injectisome is composed of a cytoplasmic sorting platform, and a membrane-embedded needle complex, which is made up of a multiring base and a needle-like filament that extends several nanometers from the bacterial surface. The needle filament is capped at its distal end by another substructure known as the tip complex, which is crucial for the translocation of effector proteins through the eukaryotic cell plasma membrane. Here we report the cryo-EM structure of the Salmonella Typhimurium needle tip complex docked onto the needle filament tip. Combined with a detailed analysis of structurally guided mutants, this study provides major insight into the assembly and function of this essential component of the type III secretion protein injection machine.


Assuntos
Salmonella typhimurium/ultraestrutura , Sistemas de Secreção Tipo III/ultraestrutura , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Sistemas de Secreção Bacterianos/metabolismo , Sistemas de Secreção Bacterianos/ultraestrutura , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Microscopia Crioeletrônica/métodos , Citoesqueleto/metabolismo , Citosol/metabolismo , Transporte Proteico/fisiologia , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidade , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/fisiologia
2.
PLoS Pathog ; 16(2): e1008263, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32092125

RESUMO

The Type III Secretion Systems (T3SS) needle complex is a conserved syringe-shaped protein translocation nanomachine with a mass of about 3.5 MDa essential for the survival and virulence of many Gram-negative bacterial pathogens. This system is composed of a membrane-embedded basal body and an extracellular needle that deliver effector proteins into host cells. High-resolution structures of the T3SS from different organisms and infection stages are needed to understand the underlying molecular mechanisms of effector translocation. Here, we present the cryo-electron microscopy structure of the isolated Shigella T3SS needle complex. The inner membrane (IM) region of the basal body adopts 24-fold rotational symmetry and forms a channel system that connects the bacterial periplasm with the export apparatus cage. The secretin oligomer adopts a heterogeneous architecture with 16- and 15-fold cyclic symmetry in the periplasmic N-terminal connector and C-terminal outer membrane ring, respectively. Two out of three IM subunits bind the secretin connector via a ß-sheet augmentation. The cryo-EM map also reveals the helical architecture of the export apparatus core, the inner rod, the needle and their intervening interfaces.


Assuntos
Proteínas de Bactérias/ultraestrutura , Membrana Celular/ultraestrutura , Microscopia Crioeletrônica , Shigella/ultraestrutura , Sistemas de Secreção Tipo III/ultraestrutura , Proteínas de Bactérias/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Conformação Proteica em Folha beta , Domínios Proteicos , Shigella/genética , Shigella/metabolismo , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo
3.
PLoS Biol ; 17(7): e3000351, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31260457

RESUMO

Type III protein-secretion machines are essential for the interactions of many pathogenic or symbiotic bacterial species with their respective eukaryotic hosts. The core component of these machines is the injectisome, a multiprotein complex that mediates the selection of substrates, their passage through the bacterial envelope, and ultimately their delivery into eukaryotic target cells. The injectisome is composed of a large cytoplasmic complex or sorting platform, a multiring base embedded in the bacterial envelope, and a needle-like filament that protrudes several nanometers from the bacterial surface and is capped at its distal end by the tip complex. A characteristic feature of these machines is that their activity is stimulated by contact with target host cells. The sensing of target cells, thought to be mediated by the distal tip of the needle filament, generates an activating signal that must be transduced to the secretion machine by the needle filament. Here, through a multidisciplinary approach, including solid-state NMR (SSNMR) and cryo electron microscopy (cryo-EM) analyses, we have identified critical residues of the needle filament protein of a Salmonella Typhimurium type III secretion system that are involved in the regulation of the activity of the secretion machine. We found that mutations in the needle filament protein result in various specific phenotypes associated with different steps in the type III secretion process. More specifically, these studies reveal an important role for a polymorphic helix of the needle filament protein and the residues that line the lumen of its central channel in the control of type III secretion.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/metabolismo , Complexos Multiproteicos/metabolismo , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sistemas de Secreção Bacterianos/química , Sistemas de Secreção Bacterianos/genética , Microscopia Crioeletrônica , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/ultraestrutura , Mutação , Conformação Proteica , Transporte Proteico/genética , Salmonella typhimurium/genética , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/ultraestrutura
4.
Proc Natl Acad Sci U S A ; 116(49): 24786-24795, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31744874

RESUMO

Type III protein secretion systems are essential virulence factors for many important pathogenic bacteria. The entire protein secretion machine is composed of several substructures that organize into a holostructure or injectisome. The core component of the injectisome is the needle complex, which houses the export apparatus that serves as a gate for the passage of the secreted proteins through the bacterial inner membrane. Here, we describe a high-resolution structure of the export apparatus of the Salmonella type III secretion system in association with the needle complex and the underlying bacterial membrane, both in isolation and in situ. We show the precise location of the core export apparatus components within the injectisome and bacterial envelope and demonstrate that their deployment results in major membrane remodeling and thinning, which may be central for the protein translocation process. We also show that InvA, a critical export apparatus component, forms a multiring cytoplasmic conduit that provides a pathway for the type III secretion substrates to reach the entrance of the export gate. Combined with structure-guided mutagenesis, our studies provide major insight into potential mechanisms of protein translocation and injectisome assembly.


Assuntos
Proteínas de Bactérias/ultraestrutura , Membrana Celular/ultraestrutura , Salmonella typhimurium/ultraestrutura , Via Secretória , Sistemas de Secreção Tipo III/ultraestrutura , Proteínas de Bactérias/metabolismo , Membrana Celular/metabolismo , Microscopia Crioeletrônica , Simulação de Acoplamento Molecular , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/metabolismo
5.
Curr Top Microbiol Immunol ; 427: 67-90, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31667599

RESUMO

The type III secretion system (T3SS) is an essential virulence factor of many pathogenic bacterial species including Salmonella, Yersinia, Shigella and enteropathogenic Escherichia coli (EPEC). It is an intricate molecular machine that spans the bacterial membranes and injects effector proteins into target host cells, enabling bacterial infection. The T3SS needle complex comprises of proteinaceous rings supporting a needle filament which extends out into the extracellular environment. It serves as the central conduit for translocating effector proteins. Multiple laboratories have dedicated a remarkable effort to decipher the structure and function of the needle complex. A combination of structural biology techniques such as cryo-electron microscopy (cryoEM), X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy and computer modelling have been utilized to study different structural components at progressively higher resolutions. This chapter will provide an overview of the structural details of the T3SS needle complex, shedding light on this essential component of this fascinating bacterial system.


Assuntos
Sistemas de Secreção Tipo III/química , Proteínas de Bactérias , Simulação por Computador , Microscopia Crioeletrônica , Cristalografia por Raios X , Ressonância Magnética Nuclear Biomolecular , Sistemas de Secreção Tipo III/ultraestrutura , Fatores de Virulência
6.
Curr Top Microbiol Immunol ; 427: 109-131, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31974760

RESUMO

Among the Gram-negative bacterial secretion systems, type III secretion systems (T3SS) possess a unique extracellular molecular apparatus called the needle. This macromolecular protein assembly is a nanometre-size filament formed by the helical arrangement of hundreds of copies of a single, small protein, which is highly conserved between T3SSs from animal to plant bacterial pathogens. The needle filament forms a hollow tube with a channel ~20 Å in diameter that serves as a conduit for proteins secreted into the targeted host cell. In the past ten years, technical breakthroughs in biophysical techniques such as cryo-electron microscopy (cryo-EM) and solid-state NMR (SSNMR) spectroscopy have uncovered atomic resolution details about the T3SS needle assembly. Several high-resolution structures of Salmonella typhimurium and Shigella flexneri T3SS needles have been reported demonstrating a common structural fold. These structural models have been used to explain the active role of the needle in transmitting the host-cell contact signal from the tip to the base of the T3SS through conformational changes as well as during the injection of effector proteins. In this chapter, we summarize the current knowledge about the structure and the role of the T3SS needle during T3SS assembly and effector secretion.


Assuntos
Sistemas de Secreção Tipo III/química , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Microscopia Crioeletrônica , Ressonância Magnética Nuclear Biomolecular , Salmonella typhimurium/química , Shigella flexneri/química , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/ultraestrutura
7.
PLoS Biol ; 16(11): e3000050, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30412577

RESUMO

Periplasmic flagella are essential for the distinct morphology and motility of spirochetes. A flagella-specific type III secretion system (fT3SS) composed of a membrane-bound export apparatus and a cytosolic ATPase complex is responsible for the assembly of the periplasmic flagella. Here, we deployed cryo-electron tomography (cryo-ET) to visualize the fT3SS machine in the Lyme disease spirochete Borrelia burgdorferi. We show, for the first time, that the cytosolic ATPase complex is attached to the flagellar C-ring through multiple spokes to form the "spoke and hub" structure in B. burgdorferi. This structure not only strengthens structural rigidity of the round-shaped C-ring but also appears to rotate with the C-ring. Our studies provide structural insights into the unique mechanisms underlying assembly and rotation of the periplasmic flagella and may provide the basis for the development of novel therapeutic strategies against several pathogenic spirochetes.


Assuntos
Adenosina Trifosfatases/ultraestrutura , Borrelia burgdorferi/fisiologia , Flagelos/fisiologia , Adenosina Trifosfatases/química , Adenosina Trifosfatases/fisiologia , Proteínas de Bactérias/química , Borrelia burgdorferi/metabolismo , Citoplasma , Tomografia com Microscopia Eletrônica/métodos , Flagelos/metabolismo , Flagelos/ultraestrutura , Periplasma/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/ultraestrutura
8.
Proc Natl Acad Sci U S A ; 114(23): 6098-6103, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28533372

RESUMO

Type III protein secretion machines have evolved to deliver bacterially encoded effector proteins into eukaryotic cells. Although electron microscopy has provided a detailed view of these machines in isolation or fixed samples, little is known about their organization in live bacteria. Here we report the visualization and characterization of the Salmonella type III secretion machine in live bacteria by 2D and 3D single-molecule switching superresolution microscopy. This approach provided access to transient components of this machine, which previously could not be analyzed. We determined the subcellular distribution of individual machines, the stoichiometry of the different components of this machine in situ, and the spatial distribution of the substrates of this machine before secretion. Furthermore, by visualizing this machine in Salmonella mutants we obtained major insights into the machine's assembly. This study bridges a major resolution gap in the visualization of this nanomachine and may serve as a paradigm for the examination of other bacterially encoded molecular machines.


Assuntos
Imagem Individual de Molécula/métodos , Sistemas de Secreção Tipo III/fisiologia , Sistemas de Secreção Tipo III/ultraestrutura , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/metabolismo , Análise por Conglomerados , Modelos Moleculares , Transporte Proteico , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/química
9.
PLoS Pathog ; 12(12): e1006071, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27977800

RESUMO

Bacterial type III protein secretion systems inject effector proteins into eukaryotic host cells in order to promote survival and colonization of Gram-negative pathogens and symbionts. Secretion across the bacterial cell envelope and injection into host cells is facilitated by a so-called injectisome. Its small hydrophobic export apparatus components SpaP and SpaR were shown to nucleate assembly of the needle complex and to form the central "cup" substructure of a Salmonella Typhimurium secretion system. However, the in vivo placement of these components in the needle complex and their function during the secretion process remained poorly defined. Here we present evidence that a SpaP pentamer forms a 15 Å wide pore and provide a detailed map of SpaP interactions with the export apparatus components SpaQ, SpaR, and SpaS. We further refine the current view of export apparatus assembly, consolidate transmembrane topology models for SpaP and SpaR, and present intimate interactions of the periplasmic domains of SpaP and SpaR with the inner rod protein PrgJ, indicating how export apparatus and needle filament are connected to create a continuous conduit for substrate translocation.


Assuntos
Salmonella typhimurium/metabolismo , Salmonella typhimurium/ultraestrutura , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/ultraestrutura , Cromatografia em Gel , Processamento de Imagem Assistida por Computador , Immunoblotting , Espectrometria de Massas , Microscopia Eletrônica
10.
Nat Commun ; 12(1): 4223, 2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34244518

RESUMO

The bacterial flagellar MS ring is a transmembrane complex acting as the core of the flagellar motor and template for flagellar assembly. The C ring attached to the MS ring is involved in torque generation and rotation switch, and a large symmetry mismatch between these two rings has been a long puzzle, especially with respect to their role in motor function. Here, using cryoEM structural analysis of the flagellar basal body and the MS ring formed by full-length FliF from Salmonella enterica, we show that the native MS ring is formed by 34 FliF subunits with no symmetry variation. Symmetry analysis of the C ring shows a variation with a peak at 34-fold, suggesting flexibility in C ring assembly. Finally, our data also indicate that FliF subunits assume two different conformations, contributing differentially to the inner and middle parts of the M ring and thus resulting in 23- and 11-fold subsymmetries in the inner and middle M ring, respectively. The internal core of the M ring, formed by 23 subunits, forms a hole of the right size to accommodate the protein export gate.


Assuntos
Proteínas de Bactérias/ultraestrutura , Flagelos/ultraestrutura , Proteínas de Membrana/ultraestrutura , Sistemas de Secreção Tipo III/ultraestrutura , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/metabolismo , Fracionamento Celular , Microscopia Crioeletrônica , Flagelos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Proteínas de Membrana/metabolismo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Conformação Proteica , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Salmonella typhimurium/ultraestrutura , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo
11.
PLoS One ; 16(6): e0252800, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34143799

RESUMO

Type three secretion is the mechanism of protein secretion found in bacterial flagella and injectisomes. At its centre is the export apparatus (EA), a complex of five membrane proteins through which secretion substrates pass the inner membrane. While the complex formed by four of the EA proteins has been well characterised structurally, little is known about the structure of the membrane domain of the largest subunit, FlhA in flagella, SctV in injectisomes. Furthermore, the biologically relevant nonameric assembly of FlhA/SctV has been infrequently observed and differences in conformation of the cytoplasmic portion of FlhA/SctV between open and closed states have been suggested to reflect secretion system specific differences. FlhA has been shown to bind to chaperone-substrate complexes in an open state, but in previous assembled ring structures, SctV is in a closed state. Here, we identify FlhA and SctV homologues that can be recombinantly produced in the oligomeric state and study them using cryo-electron microscopy. The structures of the cytoplasmic domains from both FlhA and SctV are in the open state and we observe a conserved interaction between a short stretch of residues at the N-terminus of the cytoplasmic domain, known as FlhAL/SctVL, with a groove on the adjacent protomer's cytoplasmic domain, which stabilises the nonameric ring assembly.


Assuntos
Proteínas de Bactérias/metabolismo , Flagelos/metabolismo , Proteínas de Membrana/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Microscopia Crioeletrônica/métodos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Microscopia de Fluorescência/métodos , Modelos Moleculares , Conformação Proteica , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/ultraestrutura , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/metabolismo , Yersinia enterocolitica/genética , Yersinia enterocolitica/metabolismo
12.
J Mol Biol ; 433(21): 167188, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34454944

RESUMO

Type III protein secretion is widespread in Gram-negative pathogens. It comprises the injectisome with a surface-exposed needle and an inner membrane translocase. The translocase contains the SctRSTU export channel enveloped by the export gate subunit SctV that binds chaperone/exported clients and forms a putative ante-chamber. We probed the assembly, function, structure and dynamics of SctV from enteropathogenic E. coli (EPEC). In both EPEC and E. coli lab strains, SctV forms peripheral oligomeric clusters that are detergent-extracted as homo-nonamers. Membrane-embedded SctV9 is necessary and sufficient to act as a receptor for different chaperone/exported protein pairs with distinct C-domain binding sites that are essential for secretion. Negative staining electron microscopy revealed that peptidisc-reconstituted His-SctV9 forms a tripartite particle of ∼22 nm with a N-terminal domain connected by a short linker to a C-domain ring structure with a ∼5 nm-wide inner opening. The isolated C-domain ring was resolved with cryo-EM at 3.1 Å and structurally compared to other SctV homologues. Its four sub-domains undergo a three-stage "pinching" motion. Hydrogen-deuterium exchange mass spectrometry revealed this to involve dynamic and rigid hinges and a hyper-flexible sub-domain that flips out of the ring periphery and binds chaperones on and between adjacent protomers. These motions are coincident with local conformational changes at the pore surface and ring entry mouth that may also be modulated by the ATPase inner stalk. We propose that the intrinsic dynamics of the SctV protomer are modulated by chaperones and the ATPase and could affect allosterically the other subunits of the nonameric ring during secretion.


Assuntos
Adenosina Trifosfatases/química , Escherichia coli Enteropatogênica/ultraestrutura , Proteínas de Escherichia coli/química , Flagelos/ultraestrutura , Canais de Translocação SEC/química , Sistemas de Secreção Tipo III/ultraestrutura , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Regulação Alostérica , Sítios de Ligação , Clonagem Molecular , Microscopia Crioeletrônica , Medição da Troca de Deutério , Escherichia coli Enteropatogênica/genética , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Flagelos/genética , Flagelos/metabolismo , Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Cinética , Espectrometria de Massas , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canais de Translocação SEC/genética , Canais de Translocação SEC/metabolismo , Especificidade por Substrato , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/metabolismo
13.
Curr Opin Struct Biol ; 61: 71-78, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31821956

RESUMO

The T3SS is a syringe-shaped nanomachine essential for the progression of many Gram-negative bacterial infections including plague, typhoid fever, and dysentery. It spans both bacterial membranes and that of the host allowing delivery of proteins that modulate cell function to aid bacterial survival. Its structure has been the focus of scrutiny for 20 years; however, limitations in purification and structure determination techniques have restricted understanding to atomic structures of individual components and subcomplexes or lower resolution information of the more complete assembly. The recent cryo-EM resolution revolution has facilitated dramatic advances in our structural understanding of the T3SS with complimentary techniques of single particle cryo-EM and cryo-ET revealing structures of isolated complexes to near-atomic resolutions or the architecture of the entire T3SS in its native cellular environment. Here we present an overview of these advances and discuss how these structures further understanding of the dynamic process of injectisome assembly.


Assuntos
Sistemas de Secreção Tipo III/química , Sistemas de Secreção Tipo III/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Microscopia Crioeletrônica , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Secretina/química , Secretina/metabolismo , Relação Estrutura-Atividade , Sistemas de Secreção Tipo III/ultraestrutura
14.
J Mol Biol ; 432(24): 166693, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-33122003

RESUMO

Many Gram-negative bacterial pathogens use type III secretion systems (T3SS) to inject proteins into eukaryotic cells to subvert normal cellular functions. The T3SS apparatus (injectisome) shares a common architecture in all systems studied thus far, comprising three major components - the cytoplasmic sorting platform, envelope-spanning basal body and external needle with tip complex. The sorting platform consists of an ATPase (SctN) connected to "pods" (SctQ) having six-fold symmetry via radial spokes (SctL). These pods interface with the 24-fold symmetric SctD inner membrane ring (IR) via an adaptor protein (SctK). Here we report the first high-resolution structure of a SctK protein family member, PscK from Pseudomonas aeruginosa, as well as the structure of its interacting partner, the cytoplasmic domain of PscD (SctD). The cytoplasmic domain of PscD forms a forkhead-associated (FHA) fold, like that of its homologues from other T3SS. PscK, on the other hand, forms a helix-rich structure that does not resemble any known protein fold. Based on these structural findings, we present the first model for an interaction between proteins from the sorting platform and the IR. We also test the importance of the PscD residues predicted to mediate this electrostatic interaction using a two-hybrid analysis. The functional need for these residues in vivo was then confirmed by monitoring secretion of the effector ExoU. These structures will contribute to the development of atomic-resolution models of the entire sorting platform and to our understanding of the mechanistic interface between the sorting platform and the basal body of the injectisome.


Assuntos
Adenosina Trifosfatases/ultraestrutura , Proteínas de Bactérias/ultraestrutura , Pseudomonas aeruginosa/ultraestrutura , Sistemas de Secreção Tipo III/ultraestrutura , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Corpos Basais/enzimologia , Corpos Basais/ultraestrutura , Citoplasma/química , Citoplasma/genética , Citoplasma/ultraestrutura , Citosol/ultraestrutura , Transporte Proteico/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Sistemas de Secreção Tipo III/química , Sistemas de Secreção Tipo III/genética
15.
Nat Commun ; 10(1): 626, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733444

RESUMO

Many Gram-negative bacteria, including causative agents of dysentery, plague, and typhoid fever, rely on a type III secretion system - a multi-membrane spanning syringe-like apparatus - for their pathogenicity. The cytosolic ATPase complex of this injectisome is proposed to play an important role in energizing secretion events and substrate recognition. We present the 3.3 Å resolution cryo-EM structure of the enteropathogenic Escherichia coli ATPase EscN in complex with its central stalk EscO. The structure shows an asymmetric pore with different functional states captured in its six catalytic sites, details directly supporting a rotary catalytic mechanism analogous to that of the heterohexameric F1/V1-ATPases despite its homohexameric nature. Situated at the C-terminal opening of the EscN pore is one molecule of EscO, with primary interaction mediated through an electrostatic interface. The EscN-EscO structure provides significant atomic insights into how the ATPase contributes to type III secretion, including torque generation and binding of chaperone/substrate complexes.


Assuntos
Microscopia Crioeletrônica/métodos , ATPases Translocadoras de Prótons/metabolismo , ATPases Translocadoras de Prótons/ultraestrutura , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/ultraestrutura , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestrutura , Estrutura Secundária de Proteína
16.
Nat Commun ; 9(1): 3840, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30242280

RESUMO

The bacterial type III secretion system, or injectisome, is a syringe shaped nanomachine essential for the virulence of many disease causing Gram-negative bacteria. At the core of the injectisome structure is the needle complex, a continuous channel formed by the highly oligomerized inner and outer membrane hollow rings and a polymerized helical needle filament which spans through and projects into the infected host cell. Here we present the near-atomic resolution structure of a needle complex from the prototypical Salmonella Typhimurium SPI-1 type III secretion system, with local masking protocols allowing for model building and refinement of the major membrane spanning components of the needle complex base in addition to an isolated needle filament. This work provides significant insight into injectisome structure and assembly and importantly captures the molecular basis for substrate induced gating in the giant outer membrane secretin portal family.


Assuntos
Sistemas de Secreção Tipo III/ultraestrutura , Microscopia Crioeletrônica , Salmonella
17.
Elife ; 72018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30015613

RESUMO

Type III protein secretion systems (T3SS) are encoded by several pathogenic or symbiotic bacteria. The central component of this nanomachine is the needle complex. Here we show in a Salmonella Typhimurium T3SS that assembly of the needle filament of this structure requires OrgC, a protein encoded within the T3SS gene cluster. Absence of OrgC results in significantly reduced number of needle substructures but does not affect needle length. We show that OrgC is secreted by the T3SS and that exogenous addition of OrgC can complement a ∆orgC mutation. We also show that OrgC interacts with the needle filament subunit PrgI and accelerates its polymerization into filaments in vitro. The structure of OrgC shows a novel fold with a shared topology with a domain from flagellar capping proteins. These findings identify a novel component of T3SS and provide new insight into the assembly of the type III secretion machine.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Salmonella typhimurium/metabolismo , Sistemas de Secreção Tipo III/química , Sistemas de Secreção Tipo III/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/ultraestrutura , Ilhas de CpG , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Mutação/genética , Polimerização , Ligação Proteica , Estrutura Secundária de Proteína , Salmonella typhimurium/citologia , Salmonella typhimurium/ultraestrutura , Sistemas de Secreção Tipo III/ultraestrutura
18.
Nat Struct Mol Biol ; 25(7): 583-590, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29967543

RESUMO

Export of proteins through type III secretion systems is critical for motility and virulence of many major bacterial pathogens. Three putative integral membrane proteins (FliP, FliQ, FliR) are suggested to form the core of an export gate in the inner membrane, but their structure, assembly and location within the final nanomachine remain unclear. Here, we present the cryoelectron microscopy structure of the Salmonella Typhimurium FliP-FliQ-FliR complex at 4.2 Å. None of the subunits adopt canonical integral membrane protein topologies, and common helix-turn-helix structural elements allow them to form a helical assembly with 5:4:1 stoichiometry. Fitting of the structure into reconstructions of intact secretion systems, combined with cross-linking, localize the export gate as a core component of the periplasmic portion of the machinery. This study thereby identifies the export gate as a key element of the secretion channel and implies that it primes the helical architecture of the components assembling downstream.


Assuntos
Sistemas de Secreção Tipo III/química , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Microscopia Crioeletrônica , Proteínas de Membrana/química , Proteínas de Membrana/ultraestrutura , Modelos Moleculares , Estrutura Quaternária de Proteína , Subunidades Proteicas , Salmonella typhimurium/química , Salmonella typhimurium/ultraestrutura , Sistemas de Secreção Tipo III/ultraestrutura
19.
Nat Rev Microbiol ; 15(6): 323-337, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28392566

RESUMO

Type III secretion systems (T3SSs) are protein transport nanomachines that are found in Gram-negative bacterial pathogens and symbionts. Resembling molecular syringes, T3SSs form channels that cross the bacterial envelope and the host cell membrane, which enable bacteria to inject numerous effector proteins into the host cell cytoplasm and establish trans-kingdom interactions with diverse hosts. Recent advances in cryo-electron microscopy and integrative imaging have provided unprecedented views of the architecture and structure of T3SSs. Furthermore, genetic and molecular analyses have elucidated the functions of many effectors and key regulators of T3SS assembly and secretion hierarchy, which is the sequential order by which the protein substrates are secreted. As essential virulence factors, T3SSs are attractive targets for vaccines and therapeutics. This Review summarizes our current knowledge of the structure and function of this important protein secretion machinery. A greater understanding of T3SSs should aid mechanism-based drug design and facilitate their manipulation for biotechnological applications.


Assuntos
Membrana Celular/metabolismo , Bactérias Gram-Negativas/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/ultraestrutura , Microscopia Crioeletrônica , Flagelos/metabolismo , Transporte Proteico/fisiologia
20.
Nat Commun ; 6: 10114, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26656452

RESUMO

Many bacterial pathogens of animals and plants use a conserved type III secretion system (T3SS) to inject virulence effector proteins directly into eukaryotic cells to subvert host functions. Contact with host membranes is critical for T3SS activation, yet little is known about T3SS architecture in this state or the conformational changes that drive effector translocation. Here we use cryo-electron tomography and sub-tomogram averaging to derive the intact structure of the primordial Chlamydia trachomatis T3SS in the presence and absence of host membrane contact. Comparison of the averaged structures demonstrates a marked compaction of the basal body (4 nm) occurs when the needle tip contacts the host cell membrane. This compaction is coupled to a stabilization of the cytosolic sorting platform-ATPase. Our findings reveal the first structure of a bacterial T3SS from a major human pathogen engaged with a eukaryotic host, and reveal striking 'pump-action' conformational changes that underpin effector injection.


Assuntos
Membrana Celular/fisiologia , Chlamydia trachomatis/fisiologia , Sistemas de Secreção Tipo III/fisiologia , Linhagem Celular , Membrana Celular/ultraestrutura , Chlamydia trachomatis/ultraestrutura , Microscopia Crioeletrônica , Humanos , Sistemas de Secreção Tipo III/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA