Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22.053
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 183(3): 650-665.e15, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33031742

RESUMO

Endocannabinoids are host-derived lipid hormones that fundamentally impact gastrointestinal (GI) biology. The use of cannabis and other exocannabinoids as anecdotal treatments for various GI disorders inspired the search for mechanisms by which these compounds mediate their effects, which led to the discovery of the mammalian endocannabinoid system. Dysregulated endocannabinoid signaling was linked to inflammation and the gut microbiota. However, the effects of endocannabinoids on host susceptibility to infection has not been explored. Here, we show that mice with elevated levels of the endocannabinoid 2-arachidonoyl glycerol (2-AG) are protected from enteric infection by Enterobacteriaceae pathogens. 2-AG directly modulates pathogen function by inhibiting virulence programs essential for successful infection. Furthermore, 2-AG antagonizes the bacterial receptor QseC, a histidine kinase encoded within the core Enterobacteriaceae genome that promotes the activation of pathogen-associated type three secretion systems. Taken together, our findings establish that endocannabinoids are directly sensed by bacteria and can modulate bacterial function.


Assuntos
Endocanabinoides/metabolismo , Enterobacteriaceae/patogenicidade , Animais , Ácidos Araquidônicos/química , Ácidos Araquidônicos/metabolismo , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/metabolismo , Citrobacter rodentium/patogenicidade , Colo/microbiologia , Colo/patologia , Endocanabinoides/química , Infecções por Enterobacteriaceae/microbiologia , Feminino , Microbioma Gastrointestinal , Glicerídeos/química , Glicerídeos/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monoacilglicerol Lipases/metabolismo , Salmonella/patogenicidade , Virulência
2.
Cell ; 178(3): 552-566.e20, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31327526

RESUMO

Antibacterial autophagy (xenophagy) is an important host defense, but how it is initiated is unclear. Here, we performed a bacterial transposon screen and identified a T3SS effector SopF that potently blocked Salmonella autophagy. SopF was a general xenophagy inhibitor without affecting canonical autophagy. S. Typhimurium ΔsopF resembled S. flexneri ΔvirAΔicsB with the majority of intracellular bacteria targeted by autophagy, permitting a CRISPR screen that identified host V-ATPase as an essential factor. Upon bacteria-caused vacuolar damage, the V-ATPase recruited ATG16L1 onto bacteria-containing vacuole, which was blocked by SopF. Mammalian ATG16L1 bears a WD40 domain required for interacting with the V-ATPase. Inhibiting autophagy by SopF promoted S. Typhimurium proliferation in vivo. SopF targeted Gln124 of ATP6V0C in the V-ATPase for ADP-ribosylation. Mutation of Gln124 also blocked xenophagy, but not canonical autophagy. Thus, the discovery of SopF reveals the V-ATPase-ATG16L1 axis that critically mediates autophagic recognition of intracellular pathogen.


Assuntos
Proteínas Relacionadas à Autofagia/metabolismo , Proteínas de Bactérias/genética , Macroautofagia , Salmonella/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Fatores de Virulência/genética , ADP-Ribosilação , Proteínas Relacionadas à Autofagia/deficiência , Proteínas Relacionadas à Autofagia/genética , Proteínas de Bactérias/metabolismo , Sistemas CRISPR-Cas/genética , Edição de Genes , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Ligação Proteica , Salmonella/patogenicidade , Sistemas de Secreção Tipo III/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética , Fatores de Virulência/metabolismo
3.
Nat Immunol ; 21(1): 86-100, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31844327

RESUMO

By developing a high-density murine immunophenotyping platform compatible with high-throughput genetic screening, we have established profound contributions of genetics and structure to immune variation (http://www.immunophenotype.org). Specifically, high-throughput phenotyping of 530 unique mouse gene knockouts identified 140 monogenic 'hits', of which most had no previous immunologic association. Furthermore, hits were collectively enriched in genes for which humans show poor tolerance to loss of function. The immunophenotyping platform also exposed dense correlation networks linking immune parameters with each other and with specific physiologic traits. Such linkages limit freedom of movement for individual immune parameters, thereby imposing genetically regulated 'immunologic structures', the integrity of which was associated with immunocompetence. Hence, we provide an expanded genetic resource and structural perspective for understanding and monitoring immune variation in health and disease.


Assuntos
Infecções por Enterobacteriaceae/imunologia , Variação Genética/genética , Ensaios de Triagem em Larga Escala/métodos , Imunofenotipagem/métodos , Infecções por Salmonella/imunologia , Animais , Citrobacter/imunologia , Infecções por Enterobacteriaceae/microbiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Salmonella/imunologia , Infecções por Salmonella/microbiologia
4.
Immunity ; 55(3): 385-387, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35263564

RESUMO

Some bacteria and parasites, such as Salmonella, actively disrupt germinal centers and elicit only low affinity antibodies, but the mechanisms by which microbes alter these responses is poorly understood. In this issue of Immunity, Biram et al. (2022) uncover a mechanism by which Salmonella recruits Sca-1+ monocytes to germinal centers and impairs metabolic adaptation.


Assuntos
Infecções Bacterianas , Monócitos , Centro Germinativo/imunologia , Humanos , Salmonella
5.
Immunity ; 53(3): 471-473, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32937145

RESUMO

Although programmed cell death can control intracellular bacterial replication, the role of cell death during systemic Salmonella infection remained elusive. In this issue of Immunity, Doerflinger et al. discover a critical but overlapping role for cell death associated caspases during Salmonella infection.


Assuntos
Infecções por Salmonella , Apoptose , Caspases , Morte Celular , Humanos , Salmonella
6.
Immunity ; 53(3): 533-547.e7, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32735843

RESUMO

Programmed cell death contributes to host defense against pathogens. To investigate the relative importance of pyroptosis, necroptosis, and apoptosis during Salmonella infection, we infected mice and macrophages deficient for diverse combinations of caspases-1, -11, -12, and -8 and receptor interacting serine/threonine kinase 3 (RIPK3). Loss of pyroptosis, caspase-8-driven apoptosis, or necroptosis had minor impact on Salmonella control. However, combined deficiency of these cell death pathways caused loss of bacterial control in mice and their macrophages, demonstrating that host defense can employ varying components of several cell death pathways to limit intracellular infections. This flexible use of distinct cell death pathways involved extensive cross-talk between initiators and effectors of pyroptosis and apoptosis, where initiator caspases-1 and -8 also functioned as executioners when all known effectors of cell death were absent. These findings uncover a highly coordinated and flexible cell death system with in-built fail-safe processes that protect the host from intracellular infections.


Assuntos
Apoptose/imunologia , Macrófagos/imunologia , Necroptose/imunologia , Piroptose/imunologia , Infecções por Salmonella/imunologia , Salmonella/imunologia , Animais , Caspase 1/deficiência , Caspase 1/genética , Caspase 12/deficiência , Caspase 12/genética , Caspase 8/genética , Caspases Iniciadoras/deficiência , Caspases Iniciadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/genética
7.
Immunity ; 50(1): 166-180.e7, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30650375

RESUMO

Chronic inflammation drives the progression of colorectal cancer (CRC). Increased expression of interleukin (IL)-17A is associated with poor prognosis, and IL-17A blockade curbs tumor progression in preclinical models of CRC. Here we examined the impact of IL-1 signaling, a key regulator of the IL-17 pathway, in different cell types within the CRC microenvironment. Genetic deletion of the IL-1 receptor (IL-1R1) in epithelial cells alleviated tumorigenesis in the APC model of CRC, demonstrating a cell-autonomous role for IL-1 signaling in early tumor seed outgrowth. T cell specific ablation of IL-1R1 decreased tumor-elicited inflammation dependent on IL-17 and IL-22, thereby reducing CRC progression. The pro-tumorigenic roles of IL-1 were counteracted by its effects on myeloid cells, particularly neutrophils, where IL-1R1 ablation resulted in bacterial invasion into tumors, heightened inflammation and aggressive CRC progression. Thus, IL-1 signaling elicits cell-type-specific responses, which, in aggregate, set the inflammatory tone of the tumor microenvironment and determine the propensity for disease progression.


Assuntos
Neoplasias Colorretais/imunologia , Inflamação/metabolismo , Interleucina-17/metabolismo , Interleucina-1/metabolismo , Neutrófilos/imunologia , Salmonelose Animal/imunologia , Salmonella/imunologia , Animais , Carcinogênese , Células Cultivadas , Humanos , Interleucina-1/genética , Interleucina-1/imunologia , Interleucinas/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/ultraestrutura , Especificidade de Órgãos , Receptores de Interleucina-1/genética , Transdução de Sinais , Microambiente Tumoral , Interleucina 22
8.
PLoS Biol ; 22(4): e3002560, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38574172

RESUMO

In all domains of life, Hsp70 chaperones preserve protein homeostasis by promoting protein folding and degradation and preventing protein aggregation. We now report that the Hsp70 from the bacterial pathogen Salmonella enterica serovar Typhimurium-termed DnaK-independently reduces protein synthesis in vitro and in S. Typhimurium facing cytoplasmic Mg2+ starvation, a condition encountered during infection. This reduction reflects a 3-fold increase in ribosome association with DnaK and a 30-fold decrease in ribosome association with trigger factor, the chaperone normally associated with translating ribosomes. Surprisingly, this reduction does not involve J-domain cochaperones, unlike previously known functions of DnaK. Removing the 74 C-terminal amino acids of the 638-residue long DnaK impeded DnaK association with ribosomes and reduction of protein synthesis, rendering S. Typhimurium defective in protein homeostasis during cytoplasmic Mg2+ starvation. DnaK-dependent reduction in protein synthesis is critical for survival against Mg2+ starvation because inhibiting protein synthesis in a dnaK-independent manner overcame the 10,000-fold loss in viability resulting from DnaK truncation. Our results indicate that DnaK protects bacteria from infection-relevant stresses by coordinating protein synthesis with protein folding capacity.


Assuntos
Proteínas de Escherichia coli , Magnésio , Magnésio/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Chaperonas Moleculares/metabolismo , Dobramento de Proteína , Bactérias/metabolismo , Salmonella
9.
Nature ; 596(7871): 262-267, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34349263

RESUMO

Regulated cell death is an integral part of life, and has broad effects on organism development and homeostasis1. Malfunctions within the regulated cell death process, including the clearance of dying cells, can manifest in diverse pathologies throughout various tissues including the gastrointestinal tract2. A long appreciated, yet elusively defined relationship exists between cell death and gastrointestinal pathologies with an underlying microbial component3-6, but the direct effect of dying mammalian cells on bacterial growth is unclear. Here we advance a concept that several Enterobacteriaceae, including patient-derived clinical isolates, have an efficient growth strategy to exploit soluble factors that are released from dying gut epithelial cells. Mammalian nutrients released after caspase-3/7-dependent apoptosis boosts the growth of multiple Enterobacteriaceae and is observed using primary mouse colonic tissue, mouse and human cell lines, several apoptotic triggers, and in conventional as well as germ-free mice in vivo. The mammalian cell death nutrients induce a core transcriptional response in pathogenic Salmonella, and we identify the pyruvate formate-lyase-encoding pflB gene as a key driver of bacterial colonization in three contexts: a foodborne infection model, a TNF- and A20-dependent cell death model, and a chemotherapy-induced mucositis model. These findings introduce a new layer to the complex host-pathogen interaction, in which death-induced nutrient release acts as a source of fuel for intestinal bacteria, with implications for gut inflammation and cytotoxic chemotherapy treatment.


Assuntos
Apoptose , Enterobacteriaceae/crescimento & desenvolvimento , Enterobacteriaceae/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Intestinos/citologia , Intestinos/microbiologia , Acetiltransferases/genética , Acetiltransferases/metabolismo , Animais , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/patologia , Feminino , Doenças Transmitidas por Alimentos/microbiologia , Vida Livre de Germes , Interações Hospedeiro-Patógeno , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Masculino , Camundongos , Mucosite/induzido quimicamente , Salmonella/enzimologia , Salmonella/genética , Salmonella/crescimento & desenvolvimento , Salmonella/metabolismo , Transcriptoma , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Nature ; 594(7863): 413-417, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33981034

RESUMO

Humans and their microbiota have coevolved a mutually beneficial relationship in which the human host provides a hospitable environment for the microorganisms and the microbiota provides many advantages for the host, including nutritional benefits and protection from pathogen infection1. Maintaining this relationship requires a careful immune balance to contain commensal microorganisms within the lumen while limiting inflammatory anti-commensal responses1,2. Antigen-specific recognition of intestinal microorganisms by T cells has previously been described3,4. Although the local environment shapes the differentiation of effector cells3-5 it is unclear how microbiota-specific T cells are educated in the thymus. Here we show that intestinal colonization in early life leads to the trafficking of microbial antigens from the intestine to the thymus by intestinal dendritic cells, which then induce the expansion of microbiota-specific T cells. Once in the periphery, microbiota-specific T cells have pathogenic potential or can protect against related pathogens. In this way, the developing microbiota shapes and expands the thymic and peripheral T cell repertoire, allowing for enhanced recognition of intestinal microorganisms and pathogens.


Assuntos
Células Dendríticas/imunologia , Microbioma Gastrointestinal/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Envelhecimento/imunologia , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Receptor 1 de Quimiocina CX3C/metabolismo , DNA Bacteriano/análise , Células Dendríticas/metabolismo , Escherichia coli/imunologia , Feminino , Masculino , Camundongos , Especificidade de Órgãos , Salmonella/imunologia , Simbiose/imunologia , Timo/metabolismo
11.
Proc Natl Acad Sci U S A ; 121(14): e2308814121, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38527194

RESUMO

RNA decay is a crucial mechanism for regulating gene expression in response to environmental stresses. In bacteria, RNA-binding proteins (RBPs) are known to be involved in posttranscriptional regulation, but their global impact on RNA half-lives has not been extensively studied. To shed light on the role of the major RBPs ProQ and CspC/E in maintaining RNA stability, we performed RNA sequencing of Salmonella enterica over a time course following treatment with the transcription initiation inhibitor rifampicin (RIF-seq) in the presence and absence of these RBPs. We developed a hierarchical Bayesian model that corrects for confounding factors in rifampicin RNA stability assays and enables us to identify differentially decaying transcripts transcriptome-wide. Our analysis revealed that the median RNA half-life in Salmonella in early stationary phase is less than 1 min, a third of previous estimates. We found that over half of the 500 most long-lived transcripts are bound by at least one major RBP, suggesting a general role for RBPs in shaping the transcriptome. Integrating differential stability estimates with cross-linking and immunoprecipitation followed by RNA sequencing (CLIP-seq) revealed that approximately 30% of transcripts with ProQ binding sites and more than 40% with CspC/E binding sites in coding or 3' untranslated regions decay differentially in the absence of the respective RBP. Analysis of differentially destabilized transcripts identified a role for ProQ in the oxidative stress response. Our findings provide insights into posttranscriptional regulation by ProQ and CspC/E, and the importance of RBPs in regulating gene expression.


Assuntos
Perfilação da Expressão Gênica , Rifampina , Teorema de Bayes , Meia-Vida , Transcriptoma , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Salmonella/metabolismo , Estabilidade de RNA/genética
12.
Immunity ; 46(4): 522-524, 2017 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-28423331

RESUMO

Within the gut, Salmonella-infected enterocytes are expelled into the lumen, limiting pathogen replication. In this issue of Immunity, Rauch et al. (2017) expand our understanding of this cell-intrinsic response by characterizing the genetic determinants that control the expulsion and death of epithelial cells.


Assuntos
Infecções por Salmonella/imunologia , Salmonella/imunologia , Enterócitos/imunologia , Células Epiteliais , Imunidade
13.
Immunity ; 46(6): 977-979, 2017 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28636967

RESUMO

Whereas strong evidence supports the notion that the microbiota promotes immune system maturation in multiple tissues, the identity of the specific microbes that elicit protective immunity to different infections is less clear. In a recent issue of Cell Host & Microbe, Thiemann et al. (2017) report the identification of specific gut bacteria that protect from Salmonella infection by priming host IFN-γ responses.


Assuntos
Microbioma Gastrointestinal , Interferon gama/metabolismo , Infecções por Salmonella/imunologia , Salmonella/imunologia , Simbiose , Animais , Biodiversidade , Modelos Animais de Doenças , Suscetibilidade a Doenças , Exposição Ambiental , Humanos , Imunidade Inata , Interferon gama/genética , Camundongos , Probióticos , Especificidade da Espécie
14.
PLoS Biol ; 21(8): e3002198, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37594988

RESUMO

Pathogenic bacteria proliferating inside mammalian host cells need to rapidly adapt to the intracellular environment. How they achieve this and scavenge essential nutrients from the host has been an open question due to the difficulties in distinguishing between bacterial and host metabolites in situ. Here, we capitalized on the inability of mammalian cells to metabolize mannitol to develop a stable isotopic labeling approach to track Salmonella enterica metabolites during intracellular proliferation in host macrophage and epithelial cells. By measuring label incorporation into Salmonella metabolites with liquid chromatography-mass spectrometry (LC-MS), and combining it with metabolic modeling, we identify relevant carbon sources used by Salmonella, uncover routes of their metabolization, and quantify relative reaction rates in central carbon metabolism. Our results underline the importance of the Entner-Doudoroff pathway (EDP) and the phosphoenolpyruvate carboxylase for intracellularly proliferating Salmonella. More broadly, our metabolic labeling strategy opens novel avenues for understanding the metabolism of pathogens inside host cells.


Assuntos
Salmonella enterica , Salmonella , Animais , Carbono , Cromatografia Líquida , Isótopos , Mamíferos
15.
PLoS Biol ; 21(4): e3002051, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37014914

RESUMO

Detoxification, scavenging, and repair systems embody the archetypical antioxidant defenses of prokaryotic and eukaryotic cells. Metabolic rewiring also aids with the adaptation of bacteria to oxidative stress. Evolutionarily diverse bacteria combat the toxicity of reactive oxygen species (ROS) by actively engaging the stringent response, a stress program that controls many metabolic pathways at the level of transcription initiation via guanosine tetraphosphate and the α-helical DksA protein. Studies herein with Salmonella demonstrate that the interactions of structurally related, but functionally unique, α-helical Gre factors with the secondary channel of RNA polymerase elicit the expression of metabolic signatures that are associated with resistance to oxidative killing. Gre proteins both improve transcriptional fidelity of metabolic genes and resolve pauses in ternary elongation complexes of Embden-Meyerhof-Parnas (EMP) glycolysis and aerobic respiration genes. The Gre-directed utilization of glucose in overflow and aerobic metabolism satisfies the energetic and redox demands of Salmonella, while preventing the occurrence of amino acid bradytrophies. The resolution of transcriptional pauses in EMP glycolysis and aerobic respiration genes by Gre factors safeguards Salmonella from the cytotoxicity of phagocyte NADPH oxidase in the innate host response. In particular, the activation of cytochrome bd protects Salmonella from phagocyte NADPH oxidase-dependent killing by promoting glucose utilization, redox balancing, and energy production. Control of transcription fidelity and elongation by Gre factors represent important points in the regulation of metabolic programs supporting bacterial pathogenesis.


Assuntos
Estresse Oxidativo , Salmonella , Salmonella/genética , Estresse Oxidativo/genética , Oxirredução , NADPH Oxidases/metabolismo , Glucose/metabolismo
16.
Nucleic Acids Res ; 52(5): 2498-2518, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38300764

RESUMO

The SGI1 family integrative mobilizable elements, which are efficient agents in distribution of multidrug resistance in Gammaproteobacteria, have a complex, parasitic relationship with their IncC conjugative helper plasmids. Besides exploiting the transfer apparatus, SGI1 also hijacks IncC plasmid control mechanisms to time its own excision, replication and expression of self-encoded T4SS components, which provides advantages for SGI1 over its helpers in conjugal transfer and stable maintenance. Furthermore, SGI1 destabilizes its helpers in an unknown, replication-dependent way when they are concomitantly present in the same host. Here we report how SGI1 exploits the helper plasmid partitioning system to displace the plasmid and simultaneously increase its own stability. We show that SGI1 carries two copies of sequences mimicking the parS sites of IncC plasmids. These parS-like elements bind the ParB protein encoded by the plasmid and increase SGI1 stability by utilizing the parABS system of the plasmid for its own partitioning, through which SGI1 also destabilizes the helper plasmid. Furthermore, SGI1 expresses a small protein, Sci, which significantly strengthens this plasmid-destabilizing effect, as well as SGI1 maintenance. The plasmid-induced replication of SGI1 results in an increased copy-number of parS-like sequences and Sci expression leading to strong incompatibility with the helper plasmid.


Assuntos
Elementos de DNA Transponíveis , Salmonella , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Plasmídeos/genética , Salmonella/efeitos dos fármacos , Salmonella/genética , Farmacorresistência Bacteriana Múltipla
17.
Traffic ; 24(7): 270-283, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37114883

RESUMO

Intracellular membrane fusion is mediated by membrane-bridging complexes of soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs). SNARE proteins are one of the key players in vesicular transport. Several reports shed light on intracellular bacteria modulating host SNARE machinery to establish infection successfully. The critical SNAREs in macrophages responsible for phagosome maturation are Syntaxin 3 (STX3) and Syntaxin 4 (STX4). Reports also suggest that Salmonella actively modulates its vacuole membrane composition to escape lysosomal fusion. Salmonella containing vacuole (SCV) harbours recycling endosomal SNARE Syntaxin 12 (STX12). However, the role of host SNAREs in SCV biogenesis and pathogenesis remains unclear. Upon knockdown of STX3, we observed a reduction in bacterial proliferation, which is concomitantly restored upon the overexpression of STX3. Live-cell imaging of Salmonella-infected cells showed that STX3 localises to the SCV membranes and thus might help in the fusion of SCV with intracellular vesicles to acquire membrane for its division. We also found the interaction STX3-SCV was abrogated when we infected with SPI-2 encoded Type 3 secretion system (T3SS) apparatus mutant (STM ∆ssaV) but not with SPI-1 encoded T3SS apparatus mutant (STM ∆invC). These observations were also consistent in the mice model of Salmonella infection. Together, these results shed light on the effector molecules secreted through T3SS encoded by SPI-2, possibly involved in interaction with host SNARE STX3, which is essential to maintain the division of Salmonella in SCV and help to maintain a single bacterium per vacuole.


Assuntos
Salmonella , Vacúolos , Animais , Camundongos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Comunicação Celular , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Salmonella/metabolismo , Proteínas SNARE/metabolismo , Vacúolos/microbiologia
18.
J Biol Chem ; 300(3): 105710, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38309504

RESUMO

The bacterial envelope is an essential compartment involved in metabolism and metabolites transport, virulence, and stress defense. Its roles become more evident when homeostasis is challenged during host-pathogen interactions. In particular, the presence of free radical groups and excess copper in the periplasm causes noxious reactions, such as sulfhydryl group oxidation leading to enzymatic inactivation and protein denaturation. In response to this, canonical and accessory oxidoreductase systems are induced, performing quality control of thiol groups, and therefore contributing to restoring homeostasis and preserving survival under these conditions. Here, we examine recent advances in the characterization of the Dsb-like, Salmonella-specific Scs system. This system includes the ScsC/ScsB pair of Cu+-binding proteins with thiol-oxidoreductase activity, an alternative ScsB-partner, the membrane-linked ScsD, and a likely associated protein, ScsA, with a role in peroxide resistance. We discuss the acquisition of the scsABCD locus and its integration into a global regulatory pathway directing envelope response to Cu stress during the evolution of pathogens that also harbor the canonical Dsb systems. The evidence suggests that the canonical Dsb systems cannot satisfy the extra demands that the host-pathogen interface imposes to preserve functional thiol groups. This resulted in the acquisition of the Scs system by Salmonella. We propose that the ScsABCD complex evolved to connect Cu and redox stress responses in this pathogen as well as in other bacterial pathogens.


Assuntos
Proteínas de Bactérias , Proteínas de Transporte , Cobre , Salmonella , Proteínas de Bactérias/metabolismo , Cobre/metabolismo , Homeostase , Oxirredução , Oxirredutases/metabolismo , Salmonella/metabolismo , Compostos de Sulfidrila , Proteínas de Transporte/metabolismo
19.
PLoS Pathog ; 19(8): e1011537, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37535496

RESUMO

The bacterial flagellum is a rotary motor organelle and important virulence factor that propels motile pathogenic bacteria, such as Salmonella enterica, through their surroundings. Bacteriophages, or phages, are viruses that solely infect bacteria. As such, phages have myriad applications in the healthcare field, including phage therapy against antibiotic-resistant bacterial pathogens. Bacteriophage χ (Chi) is a flagellum-dependent (flagellotropic) bacteriophage, which begins its infection cycle by attaching its long tail fiber to the S. enterica flagellar filament as its primary receptor. The interactions between phage and flagellum are poorly understood, as are the reasons that χ only kills certain Salmonella serotypes while others entirely evade phage infection. In this study, we used molecular cloning, targeted mutagenesis, heterologous flagellin expression, and phage-host interaction assays to determine which domains within the flagellar filament protein flagellin mediate this complex interaction. We identified the antigenic N- and C-terminal D2 domains as essential for phage χ binding, with the hypervariable central D3 domain playing a less crucial role. Here, we report that the primary structure of the Salmonella flagellin D2 domains is the major determinant of χ adhesion. The phage susceptibility of a strain is directly tied to these domains. We additionally uncovered important information about flagellar function. The central and most variable domain, D3, is not required for motility in S. Typhimurium 14028s, as it can be deleted or its sequence composition can be significantly altered with minimal impacts on motility. Further knowledge about the complex interactions between flagellotropic phage χ and its primary bacterial receptor may allow genetic engineering of its host range for use as targeted antimicrobial therapy against motile pathogens of the χ-host genera Salmonella, Escherichia, or Serratia.


Assuntos
Bacteriófagos , Salmonella enterica , Bacteriófagos/fisiologia , Flagelina/genética , Flagelina/metabolismo , Salmonella enterica/genética , Salmonella enterica/metabolismo , Salmonella/metabolismo , Serratia
20.
Nature ; 566(7742): 110-114, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30675063

RESUMO

Small intestinal mononuclear cells that express CX3CR1 (CX3CR1+ cells) regulate immune responses1-5. CX3CR1+ cells take up luminal antigens by protruding their dendrites into the lumen1-4,6. However, it remains unclear how dendrite protrusion by CX3CR1+ cells is induced in the intestine. Here we show in mice that the bacterial metabolites pyruvic acid and lactic acid induce dendrite protrusion via GPR31 in CX3CR1+ cells. Mice that lack GPR31, which was highly and selectively expressed in intestinal CX3CR1+ cells, showed defective dendrite protrusions of CX3CR1+ cells in the small intestine. A methanol-soluble fraction of the small intestinal contents of specific-pathogen-free mice, but not germ-free mice, induced dendrite extension of intestinal CX3CR1+ cells in vitro. We purified a GPR31-activating fraction, and identified lactic acid. Both lactic acid and pyruvic acid induced dendrite extension of CX3CR1+ cells of wild-type mice, but not of Gpr31b-/- mice. Oral administration of lactate and pyruvate enhanced dendrite protrusion of CX3CR1+ cells in the small intestine of wild-type mice, but not in that of Gpr31b-/- mice. Furthermore, wild-type mice treated with lactate or pyruvate showed an enhanced immune response and high resistance to intestinal Salmonella infection. These findings demonstrate that lactate and pyruvate, which are produced in the intestinal lumen in a bacteria-dependent manner, contribute to enhanced immune responses by inducing GPR31-mediated dendrite protrusion of intestinal CX3CR1+ cells.


Assuntos
Bactérias/metabolismo , Receptor 1 de Quimiocina CX3C/metabolismo , Extensões da Superfície Celular/metabolismo , Intestino Delgado/citologia , Intestino Delgado/microbiologia , Ácido Láctico/metabolismo , Ácido Pirúvico/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Bactérias/imunologia , Receptor 1 de Quimiocina CX3C/deficiência , Receptor 1 de Quimiocina CX3C/genética , Extensões da Superfície Celular/efeitos dos fármacos , Extensões da Superfície Celular/imunologia , Feminino , Células HEK293 , Humanos , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/imunologia , Ácido Láctico/farmacologia , Lactobacillus helveticus/metabolismo , Masculino , Metanol , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Ácido Pirúvico/farmacologia , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Salmonella/imunologia , Salmonella/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA