Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
mBio ; 15(5): e0060724, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38572992

RESUMEN

Salmonella enterica serovar Typhi and Paratyphi A are the cause of typhoid and paratyphoid fever in humans, which are systemic life-threatening illnesses. Both serovars are exclusively adapted to the human host, where they can cause life-long persistent infection. A distinct feature of these serovars is the presence of a relatively high number of degraded coding sequences coding for metabolic pathways, most likely a consequence of their adaptation to a single host. As a result of convergent evolution, these serovars shared many of the degraded coding sequences although often affecting different genes in the same metabolic pathway. However, there are several coding sequences that appear intact in one serovar while clearly degraded in the other, suggesting differences in their metabolic capabilities. Here, we examined the functionality of metabolic pathways that appear intact in S. Typhi but that show clear signs of degradation in S. Paratyphi A. We found that, in all cases, the existence of single amino acid substitutions in S. Typhi metabolic enzymes, transporters, or transcription regulators resulted in the inactivation of these metabolic pathways. Thus, the inability of S. Typhi to metabolize Glucose-6-Phosphate or 3-phosphoglyceric acid is due to the silencing of the expression of the genes encoding the transporters for these compounds due to point mutations in the transcriptional regulatory proteins. In contrast, its inability to utilize glucarate or galactarate is due to the presence of point mutations in the transporter and enzymes necessary for the metabolism of these sugars. These studies provide additional support for the concept of adaptive convergent evolution of these two human-adapted S. enterica serovars and highlight a limitation of bioinformatic approaches to predict metabolic capabilities. IMPORTANCE: Salmonella enterica serovar Typhi and Paratyphi A are the cause of typhoid and paratyphoid fever in humans, which are systemic life-threatening illnesses. Both serovars can only infect the human host, where they can cause life-long persistent infection. Because of their adaptation to the human host, these bacterial pathogens have changed their metabolism, leading to the loss of their ability to utilize certain nutrients. In this study we examined the functionality of metabolic pathways that appear intact in S. Typhi but that show clear signs of degradation in S. Paratyphi A. We found that, in all cases, the existence of single amino acid substitutions in S. Typhi metabolic enzymes, transporters, or transcription regulators resulted in the inactivation of these metabolic pathways. These studies provide additional support for the concept of adaptive convergent evolution of these two human-adapted S. enterica serovars.


Asunto(s)
Redes y Vías Metabólicas , Salmonella typhi , Redes y Vías Metabólicas/genética , Salmonella typhi/genética , Salmonella typhi/metabolismo , Humanos , Genoma Bacteriano , Salmonella paratyphi A/genética , Salmonella paratyphi A/metabolismo , Mutación con Pérdida de Función , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fiebre Tifoidea/microbiología , Serogrupo
2.
bioRxiv ; 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38405738

RESUMEN

Salmonella enterica serovar Typhi and Paratyphi A are the cause of typhoid and paratyphoid fever in humans, which are systemic life-threatening illnesses. Both serovars are exclusively adapted to the human host, where they can cause life-long persistent infection. A distinct feature of these serovars is the presence of a relatively high number of degraded coding sequences coding for metabolic pathways, most likely a consequence of their adaptation to a single host. As a result of convergent evolution, these serovars shared many of the degraded coding sequences although often affecting different genes in the same metabolic pathway. However, there are several coding sequences that appear intact in one serovar while clearly degraded in the other, suggesting differences in their metabolic capabilities. Here, we examined the functionality of metabolic pathways that appear intact in S . Typhi but that show clear signs of degradation in S . Paratyphi A. We found that, in all cases, the existence of single amino acid substitutions in S. Typhi metabolic enzymes, transporters, or transcription regulators resulted in the inactivation of these metabolic pathways. Thus, the inability of S . Typhi to metabolize Glucose-6-Phosphate or 3-phosphoglyceric acid is due to the silencing of the expression of the genes encoding the transporters for these compounds due to point mutations in the transcriptional regulatory proteins. In contrast, its inability to utilize glucarate or galactarate is due to the presence of point mutations in the transporter and enzymes necessary for the metabolism of these sugars. These studies provide additional support for the concept of adaptive convergent evolution of these two human-adapted Salmonella enterica serovars and highlight a limitation of bioinformatic approaches to predict metabolic capabilities.

3.
bioRxiv ; 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38187563

RESUMEN

The bacterial pathogen Salmonella spp. modulates cellular processes by delivering effector proteins through its type III secretion systems. Among these effectors, SipA facilitates bacterial invasion and promotes intestinal inflammation. The mechanisms by which this effector carries out these functions are incompletely understood although SipA's ability to modulate actin dynamics is central to some of these activities. Here we report the cryo-EM structure of SipA bound to filamentous actin. We show that this effector stabilizes actin filaments through unique interactions of its carboxy terminal domain with four actin subunits. Furthermore, our structure-function studies revealed that SipA's actin-binding activity is independent from its ability to stimulate intestinal inflammation. Overall, these studies illuminate critical aspects of Salmonella pathogenesis, and provide unique insight into the mechanisms by which a bacterial effector modulates actin dynamics.

4.
Nat Microbiol ; 8(10): 1880-1895, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37640963

RESUMEN

Cell-intrinsic defences constitute the first line of defence against intracellular pathogens. The guanosine triphosphatase RAB32 orchestrates one such defence response against the bacterial pathogen Salmonella, through delivery of antimicrobial itaconate. Here we show that the Parkinson's disease-associated leucine-rich repeat kinase 2 (LRRK2) orchestrates this defence response by scaffolding a complex between RAB32 and aconitate decarboxylase 1, which synthesizes itaconate from mitochondrial precursors. Itaconate delivery to Salmonella-containing vacuoles was impaired and Salmonella replication increased in LRRK2-deficient cells. Loss of LRRK2 also restored virulence of a Salmonella mutant defective in neutralizing this RAB32-dependent host defence pathway in mice. Cryo-electron tomography revealed tether formation between Salmonella-containing vacuoles and host mitochondria upon Salmonella infection, which was significantly impaired in LRRK2-deficient cells. This positions LRRK2 centrally within a host defence mechanism, which may have favoured selection of a common familial Parkinson's disease mutant allele in the human population.


Asunto(s)
Enfermedad de Parkinson , Infecciones por Salmonella , Humanos , Ratones , Animales , Enfermedad de Parkinson/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Infecciones por Salmonella/microbiología , Salmonella/metabolismo
5.
Proc Natl Acad Sci U S A ; 119(51): e2218010119, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36512499

RESUMEN

Type III secretion systems are bacterial nanomachines specialized in protein delivery into target eukaryotic cells. The structural and functional complexity of these machines demands highly coordinated mechanisms for their assembly and operation. The sorting platform is a critical component of type III secretion machines that ensures the timely engagement and secretion of proteins destined to travel this export pathway. However, the mechanisms that lead to the assembly of this multicomponent structure have not been elucidated. Herein, employing an extensive in vivo cross-linking strategy aided by structure modeling, we provide a detailed intersubunit contact survey of the entire sorting platform complex. Using the identified cross-links as signatures for pairwise intersubunit interactions in combination with systematic genetic deletions, we mapped the assembly process of this unique bacterial structure. Insights generated by this study could serve as the bases for the rational development of antivirulence strategies to combat several medically important bacterial pathogens.


Asunto(s)
Proteínas Bacterianas , Salmonella typhimurium , Salmonella typhimurium/metabolismo , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Transporte de Proteínas
6.
Elife ; 112022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35579416

RESUMEN

Typhoid toxin is an essential virulence factor for Salmonella Typhi, the cause of typhoid fever in humans. This toxin has an unusual biology in that it is produced by Salmonella Typhi only when located within host cells. Once synthesized, the toxin is secreted to the lumen of the Salmonella-containing vacuole from where it is transported to the extracellular space by vesicle carrier intermediates. Here, we report the identification of the typhoid toxin sorting receptor and components of the cellular machinery that packages the toxin into vesicle carriers, and exports it to the extracellular space. We found that the cation-independent mannose-6-phosphate receptor serves as typhoid toxin sorting receptor and that the coat protein COPII and the GTPase Sar1 mediate its packaging into vesicle carriers. Formation of the typhoid toxin carriers requires the specific environment of the Salmonella Typhi-containing vacuole, which is determined by the activities of specific effectors of its type III protein secretion systems. We also found that Rab11B and its interacting protein Rip11 control the intracellular transport of the typhoid toxin carriers, and the SNARE proteins VAMP7, SNAP23, and Syntaxin 4 their fusion to the plasma membrane. Typhoid toxin's cooption of specific cellular machinery for its transport to the extracellular space illustrates the remarkable adaptation of an exotoxin to exert its function in the context of an intracellular pathogen.


Asunto(s)
Inmunotoxinas , Fiebre Tifoidea , Humanos , Inmunotoxinas/metabolismo , Salmonella , Salmonella typhi/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34706941

RESUMEN

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.


Asunto(s)
Salmonella typhimurium/ultraestructura , Sistemas de Secreción Tipo III/ultraestructura , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Sistemas de Secreción Bacterianos/metabolismo , Sistemas de Secreción Bacterianos/ultraestructura , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Microscopía por Crioelectrón/métodos , Citoesqueleto/metabolismo , Citosol/metabolismo , Transporte de Proteínas/fisiología , Salmonella typhimurium/metabolismo , Salmonella typhimurium/patogenicidad , Sistemas de Secreción Tipo III/metabolismo , Sistemas de Secreción Tipo III/fisiología
8.
Nat Rev Microbiol ; 19(11): 716-725, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34012042

RESUMEN

Microbial infections are controlled by host inflammatory responses that are initiated by innate immune receptors after recognition of conserved microbial products. As inflammation can also lead to disease, tissues that are exposed to microbial products such as the intestinal epithelium are subject to stringent regulatory mechanisms to prevent indiscriminate signalling through innate immune receptors. The enteric pathogen Salmonella enterica subsp. enterica serovar Typhimurium, which requires intestinal inflammation to sustain its replication in the intestinal tract, uses effector proteins of its type III secretion systems to trigger an inflammatory response without the engagement of innate immune receptors. Furthermore, S. Typhimurium uses a different set of effectors to restrict the inflammatory response to preserve host homeostasis. The S. Typhimurium-host interface is a remarkable example of the unique balance that emerges from the co-evolution of a pathogen and its host.


Asunto(s)
Inflamación/microbiología , Infecciones por Salmonella/microbiología , Salmonella typhimurium/metabolismo , Animales , Interacciones Huésped-Patógeno , Humanos , Inflamación/patología , Infecciones por Salmonella/patología
9.
Nat Microbiol ; 6(5): 658-671, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33603205

RESUMEN

The food-borne bacterial pathogen Salmonella Typhimurium uses a type III protein secretion system to deliver multiple proteins into host cells. These secreted effectors modulate the functions of host cells and activate specific signalling cascades that result in the production of pro-inflammatory cytokines and intestinal inflammation. Some of the Salmonella-encoded effectors counteract this inflammatory response and help to preserve host homeostasis. Here, we demonstrate that the Salmonella effector protein SopD, which is required for pathogenesis, functions to both activate and inhibit the inflammatory response by targeting the Rab8 GTPase, which is a negative regulator of inflammation. We show that SopD has GTPase-activating protein activity for Rab8 and, therefore, inhibits this GTPase and stimulates inflammation. We also show that SopD activates Rab8 by displacing it from its cognate guanosine dissociation inhibitor, resulting in the stimulation of a signalling cascade that suppresses inflammation. We solved the crystal structure of SopD in association with Rab8 to a resolution of 2.3 Å, which reveals a unique contact interface that underlies these complex interactions. These findings show the remarkable evolution of a bacterial effector protein to exert both agonistic and antagonistic activities towards the same host cellular target to modulate the inflammatory response.


Asunto(s)
Proteínas Bacterianas/inmunología , Interacciones Huésped-Patógeno , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Proteínas de Unión al GTP rab/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Humanos , Unión Proteica , Infecciones por Salmonella/genética , Infecciones por Salmonella/microbiología , Salmonella typhimurium/química , Salmonella typhimurium/genética , Proteínas de Unión al GTP rab/química , Proteínas de Unión al GTP rab/genética
10.
Science ; 369(6502): 450-455, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32703879

RESUMEN

The guanosine triphosphatase (GTPase) Rab32 coordinates a cell-intrinsic host defense mechanism that restricts the replication of intravacuolar pathogens such as Salmonella Here, we show that this mechanism requires aconitate decarboxylase 1 (IRG1), which synthesizes itaconate, a metabolite with antimicrobial activity. We find that Rab32 interacts with IRG1 on Salmonella infection and facilitates the delivery of itaconate to the Salmonella-containing vacuole. Mice defective in IRG1 rescued the virulence defect of a S. enterica serovar Typhimurium mutant specifically defective in its ability to counter the Rab32 defense mechanism. These studies provide a link between a metabolite produced in the mitochondria after stimulation of innate immune receptors and a cell-autonomous defense mechanism that restricts the replication of an intracellular bacterial pathogen.


Asunto(s)
Hidroliasas/inmunología , Infecciones por Salmonella/inmunología , Salmonella enterica , Salmonella typhimurium , Proteínas de Unión al GTP rab/inmunología , Animales , Línea Celular , Interacciones Huésped-Patógeno , Humanos , Hidroliasas/metabolismo , Ratones , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Succinatos , Virulencia , Proteínas de Unión al GTP rab/metabolismo
11.
Infect Immun ; 88(10)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32661121

RESUMEN

Typhoid toxin is a virulence factor of Salmonella enterica serovar Typhi, the causative agent of typhoid fever, and is thought to be responsible for the symptoms of severe disease. This toxin has a unique A2B5 architecture with two active subunits, the ADP ribosyl transferase PltA and the DNase CdtB, linked to a pentameric B subunit, which is alternatively made of PltB or PltC. Here, we describe the generation and characterization of typhoid toxin-neutralizing human monoclonal antibodies by immunizing genetically engineered mice that have a full set of human immunoglobulin variable region genes. We identified several monoclonal antibodies with strong in vitro and in vivo toxin-neutralizing activity and different mechanisms of toxin neutralization. These antibodies could serve as the basis for the development of novel therapeutic strategies against typhoid fever.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/inmunología , Toxinas Bacterianas/inmunología , Salmonella typhi/inmunología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular , Humanos , Ratones , Ratones Transgénicos , Pruebas de Neutralización , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Salmonella typhi/genética , Fiebre Tifoidea/prevención & control
12.
Elife ; 92020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31958059

RESUMEN

Typhoid toxin is a virulence factor for the bacterial pathogen Salmonella Typhi, which causes typhoid fever in humans. After its synthesis by intracellular bacteria, typhoid toxin is secreted into the lumen of the Salmonella-containing vacuole by a secretion mechanism strictly dependent on TtsA, a specific muramidase that facilitates toxin transport through the peptidoglycan layer. Here we show that substrate recognition by TtsA depends on a discrete domain within its carboxy terminus, which targets the enzyme to the bacterial poles to recognize YcbB-edited peptidoglycan. Comparison of the atomic structures of TtsA bound to its substrate and that of a close homolog with different specificity identified specific determinants involved in substrate recognition. Combined with structure-guided mutagenesis and in vitro and in vivo crosslinking experiments, this study provides an unprecedented view of the mechanisms by which a muramidase recognizes its peptidoglycan substrate to facilitate protein secretion.


Asunto(s)
Toxinas Bacterianas/metabolismo , Muramidasa/metabolismo , Salmonella typhi/metabolismo , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Modelos Moleculares , Estructura Molecular , Mutación , Peptidoglicano/metabolismo , Transporte de Proteínas , Salmonella typhi/enzimología , Especificidad por Sustrato , Factores de Virulencia/metabolismo
13.
Cell Microbiol ; 22(5): e13157, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31891220

RESUMEN

Many human pathogens use Type III, Type IV, and Type VI secretion systems to deliver effectors into their target cells. The contribution of these secretion systems to microbial virulence was the main focus of a workshop organised by the International University of Andalusia in Spain. The meeting addressed structure-function, substrate recruitment, and translocation processes, which differ widely on the different secretion machineries, as well as the nature of the translocated effectors and their roles in subverting the host cell. An excellent panel of worldwide speakers presented the state of the art of the field, highlighting the involvement of bacterial secretion in human disease and discussing mechanistic aspects of bacterial pathogenicity, which can provide the bases for the development of novel antivirulence strategies.


Asunto(s)
Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/metabolismo , Bacterias/genética , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos/química , Sistemas de Secreción Bacterianos/genética , Humanos , Transporte de Proteínas , España , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo , Sistemas de Secreción Tipo IV/genética , Sistemas de Secreción Tipo IV/metabolismo , Sistemas de Secreción Tipo VI/genética , Sistemas de Secreción Tipo VI/metabolismo , Virulencia
14.
Proc Natl Acad Sci U S A ; 116(49): 24786-24795, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31744874

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Membrana Celular/ultraestructura , Salmonella typhimurium/ultraestructura , Vías Secretoras , Sistemas de Secreción Tipo III/ultraestructura , Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Simulación del Acoplamiento Molecular , Salmonella typhimurium/metabolismo , Sistemas de Secreción Tipo III/metabolismo
15.
Nat Commun ; 10(1): 3684, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31417089

RESUMEN

Bacterial toxins with an AB5 architecture consist of an active (A) subunit inserted into a ring-like platform comprised of five delivery (B) subunits. Salmonella Typhi, the cause of typhoid fever, produces an unusual A2B5 toxin known as typhoid toxin. Here, we report that upon infection of human cells, S. Typhi produces two forms of typhoid toxin that have distinct delivery components but share common active subunits. The two typhoid toxins exhibit different trafficking properties, elicit different effects when administered to laboratory animals, and are expressed using different regulatory mechanisms and in response to distinct metabolic cues. Collectively, these results indicate that the evolution of two typhoid toxin variants has conferred functional versatility to this virulence factor. More broadly, this study reveals a new paradigm in toxin biology and suggests that the evolutionary expansion of AB5 toxins was likely fueled by the plasticity inherent to their structural design coupled to the functional versatility afforded by the combination of homologous toxin components.


Asunto(s)
Toxinas Bacterianas/genética , Multimerización de Proteína/genética , Salmonella typhi/genética , Factores de Virulencia/genética , Animales , Línea Celular Tumoral , Humanos , Ratones , Subunidades de Proteína/genética , Homología de Secuencia de Ácido Nucleico
16.
PLoS Biol ; 17(7): e3000351, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31260457

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/metabolismo , Complejos Multiproteicos/metabolismo , Salmonella typhimurium/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos/química , Sistemas de Secreción Bacterianos/genética , Microscopía por Crioelectrón , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/ultraestructura , Mutación , Conformación Proteica , Transporte de Proteínas/genética , Salmonella typhimurium/genética , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/ultraestructura
17.
Nature ; 569(7754): 44-45, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31036968
18.
PLoS Pathog ; 15(4): e1007704, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30951565

RESUMEN

Typhoid toxin is a virulence factor for Salmonella Typhi and Paratyphi, the cause of typhoid fever in humans. This toxin has a unique architecture in that its pentameric B subunit, made of PltB, is linked to two enzymatic A subunits, the ADP ribosyl transferase PltA and the deoxyribonuclease CdtB. Typhoid toxin is uniquely adapted to humans, recognizing surface glycoprotein sialoglycans terminated in acetyl neuraminic acid, which are preferentially expressed by human cells. The transport pathway to its cellular targets followed by typhoid toxin after receptor binding is currently unknown. Through a genome-wide CRISPR/Cas9-mediated screen we have characterized the mechanisms by which typhoid toxin is transported within human cells. We found that typhoid toxin hijacks specific elements of the retrograde transport and endoplasmic reticulum-associated degradation machineries to reach its subcellular destination within target cells. Our study reveals unique and common features in the transport mechanisms of bacterial toxins that could serve as the bases for the development of novel anti-toxin therapeutic strategies.


Asunto(s)
Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Salmonella typhi/patogenicidad , Fiebre Tifoidea/microbiología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Transporte Biológico , Sistemas CRISPR-Cas , Células HeLa , Humanos , Espacio Intracelular/metabolismo , Unión Proteica , Salmonella typhi/genética , Fiebre Tifoidea/genética , Fiebre Tifoidea/metabolismo
19.
EcoSal Plus ; 8(2)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30942149

RESUMEN

Type III protein secretion systems (T3SSs), or injectisomes, are multiprotein nanomachines present in many Gram-negative bacteria that have a sustained long-standing close relationship with a eukaryotic host. These secretion systems have evolved to modulate host cellular functions through the activity of the effector proteins they deliver. To reach their destination, T3SS effectors must cross the multibarrier bacterial envelope and the eukaryotic cell membrane. Passage through the bacterial envelope is mediated by the needle complex, a central component of T3SSs that expands both the inner and outer membranes of Gram-negative bacteria. A set of T3SS secreted proteins, known as translocators, form a channel in the eukaryotic plasma membrane through which the effector proteins are delivered to reach the host cell cytosol. While the effector proteins are tailored to the specific lifestyle of the bacterium that encodes them, the injectisome is conserved among the different T3SSs. The central role of T3SSs in pathogenesis and their high degree of conservation make them a desirable target for the development of antimicrobial therapies against several important bacterial pathogens.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Eucariotas/microbiología , Bacterias Gramnegativas/patogenicidad , Interacciones Huésped-Patógeno , Sistemas de Secreción Tipo III/metabolismo , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Transporte de Proteínas , Sistemas de Secreción Tipo III/genética , Factores de Virulencia/genética
20.
J Bacteriol ; 201(13)2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31010901

RESUMEN

The bacterial flagellum is a sophisticated self-assembling nanomachine responsible for motility in many bacterial pathogens, including Pseudomonas aeruginosa, Vibrio spp., and Salmonella enterica The bacterial flagellum has been studied extensively in the model systems Escherichia coli and Salmonella enterica serovar Typhimurium, yet the range of variation in flagellar structure and assembly remains incompletely understood. Here, we used cryo-electron tomography and subtomogram averaging to determine in situ structures of polar flagella in P. aeruginosa and peritrichous flagella in S Typhimurium, revealing notable differences between these two flagellar systems. Furthermore, we observed flagellar outer membrane complexes as well as many incomplete flagellar subassemblies, which provide additional insight into mechanisms underlying flagellar assembly and loss in both P. aeruginosa and S Typhimurium.IMPORTANCE The bacterial flagellum has evolved as one of the most sophisticated self-assembled molecular machines, which confers locomotion and is often associated with virulence of bacterial pathogens. Variation in species-specific features of the flagellum, as well as in flagellar number and placement, results in structurally distinct flagella that appear to be adapted to the specific environments that bacteria encounter. Here, we used cutting-edge imaging techniques to determine high-resolution in situ structures of polar flagella in Pseudomonas aeruginosa and peritrichous flagella in Salmonella enterica serovar Typhimurium, demonstrating substantial variation between flagella in these organisms. Importantly, we observed novel flagellar subassemblies and provided additional insight into the structural basis of flagellar assembly and loss in both P. aeruginosa and S Typhimurium.


Asunto(s)
Microscopía por Crioelectrón , Tomografía con Microscopio Electrónico , Flagelos/ultraestructura , Pseudomonas aeruginosa/citología , Salmonella typhimurium/citología , Proteínas Bacterianas/metabolismo , Flagelos/genética , Regulación Bacteriana de la Expresión Génica , Pseudomonas aeruginosa/genética , Salmonella typhimurium/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA