Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
Mucosal Immunol ; 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38750967

RESUMO

The immune response against Legionella longbeachae, a causative agent of the often-fatal Legionnaires' pneumonia, is poorly understood. Here we investigated the specific roles of tissue-resident alveolar macrophages (AM) and infiltrating phagocytes during infection with this pathogen. AM were the predominant cell type that internalized bacteria one day after infection. Three and five days after infection, AM numbers were greatly reduced while there was an influx of neutrophils and later monocyte-derived cells (MC) into lung tissue. AM carried greater numbers of viable L.longbeachae than neutrophils and MC, which correlated with a higher capacity of L.longbeachae to translocate bacterial effector proteins required for bacterial replication into the AM cytosol. Cell ablation experiments demonstrated that AM promoted infection whereas neutrophils and MC were required for efficient bacterial clearance. IL-18 was important for IFN-γ production by IL-18R+ NK cells and T cells which, in turn, stimulated ROS-mediated bactericidal activity in neutrophils resulting in restriction of L.longbeachae infection. Ciliated bronchiolar epithelial cells also expressed IL-18R but did not play a role in IL-18-mediated L.longbeachae clearance. Our results have identified opposing innate functions of tissue-resident and infiltrating immune cells during L.longbeachae infection that may be manipulated to improve protective responses.

3.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L373-L384, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36719079

RESUMO

Legionella pneumophila is the main etiological agent of Legionnaires' disease, a severe bacterial pneumonia. L. pneumophila is initially engulfed by alveolar macrophages (AMs) and subvert normal cellular functions to establish a replicative vacuole. Cigarette smokers are particularly susceptible to developing Legionnaires' disease and other pulmonary infections; however, little is known about the cellular mechanisms underlying this susceptibility. To investigate this, we used a mouse model of acute cigarette smoke exposure to examine the immune response to cigarette smoke and subsequent L. pneumophila infection. Contrary to previous reports, we show that cigarette smoke exposure alone causes a significant depletion of AMs using enzymatic digestion to extract cells, or via imaging intact lung lobes by light-sheet microscopy. Furthermore, treatment of mice deficient in specific types of cell death with smoke suggests that NLRP3-driven pyroptosis is a contributor to smoke-induced death of AMs. After infection, smoke-exposed mice displayed increased pulmonary L. pneumophila loads and developed more severe disease compared with air-exposed controls. We tested if depletion of AMs was related to this phenotype by directly depleting them with clodronate liposomes and found that this also resulted in increased L. pneumophila loads. In summary, our results showed that cigarette smoke depleted AMs from the lung and that this likely contributed to more severe Legionnaires' disease. Furthermore, the role of AMs in L. pneumophila infection is more nuanced than simply providing a replicative niche, and our studies suggest they play a major role in bacterial clearance.


Assuntos
Fumar Cigarros , Legionella pneumophila , Doença dos Legionários , Camundongos , Animais , Macrófagos Alveolares/metabolismo , Doença dos Legionários/metabolismo , Doença dos Legionários/microbiologia , Pulmão/microbiologia
4.
Curr Opin Cell Biol ; 79: 102131, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36215855

RESUMO

Subversion of the host cell cytoskeleton is a virulence attribute common to many bacterial pathogens. On mucosal surfaces, bacteria have evolved distinct ways of interacting with the polarised epithelium and manipulating host cell structure to propagate infection. For example, Shigella and Listeria induce cytoskeletal changes to induce their own uptake into enterocytes in order to replicate within an intracellular environment and then spread from cell-to-cell by harnessing the host actin cytoskeleton. In this review, we highlight some recent studies that advance our understanding of the role of the host cell cytoskeleton in the mechanical and molecular processes of pathogen invasion, cell-to-cell spread and the impact of infection on epithelial intercellular tension and innate mucosal defence.


Assuntos
Listeria , Shigella , Citoesqueleto/metabolismo , Células Epiteliais , Bactérias , Interações Hospedeiro-Patógeno
5.
Proc Natl Acad Sci U S A ; 119(34): e2204332119, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35976880

RESUMO

Attaching and effacing (AE) lesion formation on enterocytes by enteropathogenic Escherichia coli (EPEC) requires the EPEC type III secretion system (T3SS). Two T3SS effectors injected into the host cell during infection are the atypical kinases, NleH1 and NleH2. However, the host targets of NleH1 and NleH2 kinase activity during infection have not been reported. Here phosphoproteomics identified Ser775 in the microvillus protein Eps8 as a bona fide target of NleH1 and NleH2 phosphorylation. Both kinases interacted with Eps8 through previously unrecognized, noncanonical "proline-rich" motifs, PxxDY, that bound the Src Homology 3 (SH3) domain of Eps8. Structural analysis of the Eps8 SH3 domain bound to a peptide containing one of the proline-rich motifs from NleH showed that the N-terminal part of the peptide adopts a type II polyproline helix, and its C-terminal "DY" segment makes multiple contacts with the SH3 domain. Ser775 phosphorylation by NleH1 or NleH2 hindered Eps8 bundling activity and drove dispersal of Eps8 from the AE lesion during EPEC infection. This finding suggested that NleH1 and NleH2 altered the cellular localization of Eps8 and the cytoskeletal composition of AE lesions during EPEC infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Escherichia coli Enteropatogênica , Infecções por Escherichia coli , Proteínas de Escherichia coli , Fosfotransferases , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Núcleo Celular/metabolismo , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Humanos , Microvilosidades/metabolismo , Fosforilação , Fosfotransferases/metabolismo
6.
Methods Mol Biol ; 2523: 23-41, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35759189

RESUMO

Legionella pneumophila is an intracellular bacterial pathogen that uses a type IV secretion system (T4SS), termed Dot/Icm, to secrete more than 330 virulence effector proteins into the infected host cell. Many Dot/Icm effectors are involved in biogenesis of the Legionella-containing vacuole (LCV), which allows intracellular bacterial replication in environmental amoebae and alveolar macrophages. Through their activity, some effectors trigger the mammalian host immune response in a phenomenon termed effector-triggered immunity (ETI). Here, we describe a protocol to create and use L. pneumophila genome deletion mutants to identify effector(s) that alter pro-inflammatory cytokine production and bacterial clearance in the lungs of mice.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Animais , Genômica , Legionella/metabolismo , Legionella pneumophila/metabolismo , Doença dos Legionários/microbiologia , Mamíferos/metabolismo , Camundongos , Sistemas de Secreção Tipo IV/genética
7.
Mol Microbiol ; 117(3): 551-552, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35303397
8.
PLoS Pathog ; 18(1): e1010166, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35007292

RESUMO

A hallmark of Listeria (L.) monocytogenes pathogenesis is bacterial escape from maturing entry vacuoles, which is required for rapid bacterial replication in the host cell cytoplasm and cell-to-cell spread. The bacterial transcriptional activator PrfA controls expression of key virulence factors that enable exploitation of this intracellular niche. The transcriptional activity of PrfA within infected host cells is controlled by allosteric coactivation. Inhibitory occupation of the coactivator site has been shown to impair PrfA functions, but consequences of PrfA inhibition for L. monocytogenes infection and pathogenesis are unknown. Here we report the crystal structure of PrfA with a small molecule inhibitor occupying the coactivator site at 2.0 Å resolution. Using molecular imaging and infection studies in macrophages, we demonstrate that PrfA inhibition prevents the vacuolar escape of L. monocytogenes and enables extensive bacterial replication inside spacious vacuoles. In contrast to previously described spacious Listeria-containing vacuoles, which have been implicated in supporting chronic infection, PrfA inhibition facilitated progressive clearance of intracellular L. monocytogenes from spacious vacuoles through lysosomal degradation. Thus, inhibitory occupation of the PrfA coactivator site facilitates formation of a transient intravacuolar L. monocytogenes replication niche that licenses macrophages to effectively eliminate intracellular bacteria. Our findings encourage further exploration of PrfA as a potential target for antimicrobials and highlight that intra-vacuolar residence of L. monocytogenes in macrophages is not inevitably tied to bacterial persistence.


Assuntos
Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Macrófagos/microbiologia , Vacúolos/microbiologia , Virulência/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
Biomolecules ; 11(12)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34944446

RESUMO

Legionella pneumophila is a Gram-negative intracellular pathogen that causes Legionnaires' disease in elderly or immunocompromised individuals. This bacterium relies on the Dot/Icm (Defective in organelle trafficking/Intracellular multiplication) Type IV Secretion System (T4SS) and a large (>330) set of effector proteins to colonize the host cell. The structural variability of these effectors allows them to disrupt many host processes. Herein, we report the crystal structure of MavL to 2.65 Å resolution. MavL adopts an ADP-ribosyltransferase (ART) fold and contains the distinctive ligand-binding cleft of ART proteins. Indeed, MavL binds ADP-ribose with Kd of 13 µM. Structural overlay of MavL with poly-(ADP-ribose) glycohydrolases (PARGs) revealed a pair of aspartate residues in MavL that align with the catalytic glutamates in PARGs. MavL also aligns with ADP-ribose "reader" proteins (proteins that recognize ADP-ribose). Since no glycohydrolase activity was observed when incubated in the presence of ADP-ribosylated PARP1, MavL may play a role as a signaling protein that binds ADP-ribose. An interaction between MavL and the mammalian ubiquitin-conjugating enzyme UBE2Q1 was revealed by yeast two-hybrid and co-immunoprecipitation experiments. This work provides structural and molecular insights to guide biochemical studies aimed at elucidating the function of MavL. Our findings support the notion that ubiquitination and ADP-ribosylation are global modifications exploited by L. pneumophila.


Assuntos
Legionella pneumophila/crescimento & desenvolvimento , Sistemas de Secreção Tipo IV/química , Sistemas de Secreção Tipo IV/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Adenosina Difosfato Ribose/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Células HEK293 , Células HeLa , Humanos , Legionella pneumophila/enzimologia , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Células THP-1 , Ubiquitinação
10.
PLoS Pathog ; 17(6): e1009658, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34133469

RESUMO

During infection, enteropathogenic Escherichia coli (EPEC) and enterohaemorrhagic E. coli (EHEC) directly manipulate various aspects of host cell function through the translocation of type III secretion system (T3SS) effector proteins directly into the host cell. Many T3SS effector proteins are enzymes that mediate post-translational modifications of host proteins, such as the glycosyltransferase NleB1, which transfers a single N-acetylglucosamine (GlcNAc) to arginine residues, creating an Arg-GlcNAc linkage. NleB1 glycosylates death-domain containing proteins including FADD, TRADD and RIPK1 to block host cell death. The NleB1 paralogue, NleB2, is found in many EPEC and EHEC strains but to date its enzymatic activity has not been described. Using in vitro glycosylation assays combined with mass spectrometry, we found that NleB2 can utilize multiple sugar donors including UDP-glucose, UDP-GlcNAc and UDP-galactose during glycosylation of the death domain protein, RIPK1. Sugar donor competition assays demonstrated that UDP-glucose was the preferred substrate of NleB2 and peptide sequencing identified the glycosylation site within RIPK1 as Arg603, indicating that NleB2 catalyses arginine glucosylation. We also confirmed that NleB2 catalysed arginine-hexose modification of Flag-RIPK1 during infection of HEK293T cells with EPEC E2348/69. Using site-directed mutagenesis and in vitro glycosylation assays, we identified that residue Ser252 in NleB2 contributes to the specificity of this distinct catalytic activity. Substitution of Ser252 in NleB2 to Gly, or substitution of the corresponding Gly255 in NleB1 to Ser switches sugar donor preference between UDP-GlcNAc and UDP-glucose. However, this switch did not affect the ability of the NleB variants to inhibit inflammatory or cell death signalling during HeLa cell transfection or EPEC infection. NleB2 is thus the first identified bacterial Arg-glucose transferase that, similar to the NleB1 Arg-GlcNAc transferase, inhibits host protein function by arginine glycosylation.


Assuntos
Arginina/metabolismo , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/metabolismo , Glucose/metabolismo , Glicosiltransferases/metabolismo , Fatores de Virulência/metabolismo , Linhagem Celular , Humanos
12.
Cell Microbiol ; 23(10): e13368, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34041837

RESUMO

The Dot/Icm system of Legionella pneumophila is essential for virulence and delivers a large repertoire of effectors into infected host cells to create the Legionella containing vacuole. Since the secretion of effectors via the Dot/Icm system does not occur in the absence of host cells, we hypothesised that host factors actively participate in Dot/Icm effector translocation. Here we employed a high-throughput, genome-wide siRNA screen to systematically test the effect of silencing 18,120 human genes on translocation of the Dot/Icm effector, RalF, into HeLa cells. For the primary screen, we found that silencing of 119 genes led to increased translocation of RalF, while silencing of 321 genes resulted in decreased translocation. Following secondary screening, 70 genes were successfully validated as 'high confidence' targets. Gene set enrichment analysis of siRNAs leading to decreased RalF translocation, showed that ubiquitination was the most highly overrepresented category in the pathway analysis. We further showed that two host factors, the E2 ubiquitin-conjugating enzyme, UBE2E1, and the E3 ubiquitin ligase, CUL7, were important for supporting Dot/Icm translocation and L. pneumophila intracellular replication. In summary, we identified host ubiquitin pathways as important for the efficiency of Dot/Icm effector translocation by L. pneumophila, suggesting that host-derived ubiquitin-conjugating enzymes and ubiquitin ligases participate in the translocation of Legionella effector proteins and influence intracellular persistence and survival.


Assuntos
Legionella pneumophila , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células HeLa , Humanos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Ubiquitinação , Vacúolos/metabolismo
13.
Biochem Soc Trans ; 49(3): 1287-1297, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34003245

RESUMO

Interferon (IFN)-induced guanosine triphosphate hydrolysing enzymes (GTPases) have been identified as cornerstones of IFN-mediated cell-autonomous defence. Upon IFN stimulation, these GTPases are highly expressed in various host cells, where they orchestrate anti-microbial activities against a diverse range of pathogens such as bacteria, protozoan and viruses. IFN-induced GTPases have been shown to interact with various host pathways and proteins mediating pathogen control via inflammasome activation, destabilising pathogen compartments and membranes, orchestrating destruction via autophagy and the production of reactive oxygen species as well as inhibiting pathogen mobility. In this mini-review, we provide an update on how the IFN-induced GTPases target pathogens and mediate host defence, emphasising findings on protection against bacterial pathogens.


Assuntos
Bactérias/imunologia , Infecções Bacterianas/imunologia , GTP Fosfo-Hidrolases/imunologia , Imunidade Inata/imunologia , Interferons/imunologia , Animais , Bactérias/patogenicidade , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , GTP Fosfo-Hidrolases/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferons/metabolismo , Transdução de Sinais/imunologia , Virulência/imunologia
14.
Microorganisms ; 9(4)2021 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-33805575

RESUMO

The unfolded protein response (UPR) is a homeostatic response to endoplasmic reticulum (ER) stress within eukaryotic cells. The UPR initiates transcriptional and post-transcriptional programs to resolve ER stress; or, if ER stress is severe or prolonged, initiates apoptosis. ER stress is a common feature of bacterial infection although the role of the UPR in host defense is only beginning to be understood. While the UPR is important for host defense against pore-forming toxins produced by some bacteria, other bacterial effector proteins hijack the UPR through the activity of translocated effector proteins that facilitate intracellular survival and proliferation. UPR-mediated apoptosis can limit bacterial replication but also often contributes to tissue damage and disease. Here, we discuss the dual nature of the UPR during infection and the implications of UPR activation or inhibition for inflammation and immunity as illustrated by different bacterial pathogens.

15.
Methods Mol Biol ; 2291: 317-332, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33704761

RESUMO

Shiga toxin-producing Escherichia coli (STEC) and the related pathogen enteropathogenic Escherichia coli (EPEC) use a type III secretion system to translocate effector proteins into host cells to modulate inflammatory signaling pathways during infection. Here we describe the procedures to investigate effector-driven modulation of host inflammatory signaling pathways in mammalian cells where bacterial effectors are ectopically expressed or in cell lines infected with STEC or EPEC. We focus on the TNF-induced NF-κB response by examining IκBα degradation by immunoblot and p65 nuclear localization in addition to using an NF-κB-dependent luciferase reporter and cytokine secretion assays. These methods can be adapted for examining effector-mediated modulation of other inflammatory stimuli and host signaling pathways.


Assuntos
Escherichia coli Enteropatogênica/metabolismo , Infecções por Escherichia coli/metabolismo , Escherichia coli Shiga Toxigênica/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , Animais , Linhagem Celular , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/patologia , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Inibidor de NF-kappaB alfa/metabolismo , Escherichia coli Shiga Toxigênica/patogenicidade
16.
Protein Sci ; 30(5): 940-955, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33660322

RESUMO

Legionella pneumophila is an intracellular pathogen that causes Legionnaire's disease in humans. This bacterium can be found in freshwater environments as a free-living organism, but it is also an intracellular parasite of protozoa. Human infection occurs when inhaled aerosolized pathogen comes into contact with the alveolar mucosa and replicates in alveolar macrophages. Legionella enters the host cell by phagocytosis and redirects the Legionella-containing phagosomes from the phagocytic maturation pathway. These nascent phagosomes fuse with ER-derived secretory vesicles and membranes forming the Legionella-containing vacuole. Legionella subverts many host cellular processes by secreting over 300 effector proteins into the host cell via the Dot/Icm type IV secretion system. The cellular function for many Dot/Icm effectors is still unknown. Here, we present a structural and functional study of L. pneumophila effector RavA (Lpg0008). Structural analysis revealed that the RavA consists of four ~85 residue long α-helical domains with similar folds, which show only a low level of structural similarity to other protein domains. The ~90 residues long C-terminal segment is predicted to be natively unfolded. We show that during L. pneumophila infection of human cells, RavA localizes to the Golgi apparatus and to the plasma membrane. The same localization is observed when RavA is expressed in human cells. The localization signal resides within the C-terminal sequence C409 WTSFCGLF417 . Yeast-two-hybrid screen using RavA as bait identified RAB11A as a potential binding partner. RavA is present in L. pneumophila strains but only distant homologs are found in other Legionella species, where the number of repeats varies.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Legionella pneumophila/enzimologia , Adenosina Trifosfatases/genética , Proteínas de Bactérias/genética , Células HEK293 , Humanos , Legionella pneumophila/genética , Conformação Proteica em alfa-Hélice , Domínios Proteicos , Proteínas rab de Ligação ao GTP/química , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
17.
Curr Opin Microbiol ; 59: 58-64, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32862049

RESUMO

Many Gram-negative enteric pathogens, including enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC), Salmonella, Shigella, and Yersinia species have evolved strategies to combat host defence mechanisms. Critical bacterial virulence factors, which often include but are not limited to type III secreted effector proteins, are deployed to cooperatively interfere with key host defence pathways. Recent studies in this area have not only contributed to our knowledge of bacterial pathogenesis, but have also shed light on the host pathways that are critical for controlling bacterial infection. In this review, we summarise recent breakthroughs in our understanding of the mechanisms utilised by enteric bacterial pathogens to rewire critical host innate immune responses, including cell death and inflammatory signaling and cell-intrinsic anti-microbial responses such as xenophagy.


Assuntos
Microbioma Gastrointestinal , Interações Hospedeiro-Patógeno , Imunidade Inata , Animais , Morte Celular , Microbioma Gastrointestinal/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/imunologia , Transdução de Sinais , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
18.
Artigo em Inglês | MEDLINE | ID: mdl-32974215

RESUMO

During infection, Salmonella species inject multiple type III secretion system (T3SS) effector proteins into host cells that mediate invasion and subsequent intracellular replication. At early stages of infection, Salmonella exploits key regulators of host intracellular vesicle transport, including the small GTPases Rab5 and Rab7, to subvert host endocytic vesicle trafficking and establish the Salmonella-containing vacuole (SCV). At later stages of intracellular replication, interactions of the SCV with Rab GTPases are less well defined. Here we report that Rab1, Rab5, and Rab11 are modified at later stages of Salmonella infection by SseK3, an arginine N-acetylglucosamine (GlcNAc) transferase effector translocated via the Salmonella pathogenicity island 2 (SPI-2) type III secretion system. SseK3 modified arginines at positions 74, 82, and 111 within Rab1 and this modification occurred independently of Rab1 nucleotide binding. SseK3 exhibited Golgi localization that was independent of its glycosyltransferase activity but Arg-GlcNAc transferase activity was required for inhibition of alkaline phosphatase secretion in transfected cells. While SseK3 had a modest effect on SEAP secretion during infection of HeLa229 cells, inhibition of IL-1 and GM-CSF cytokine secretion was only observed upon over-expression of SseK3 during infection of RAW264.7 cells. Our results suggest that, in addition to targeting death receptor signaling, SseK3 may contribute to Salmonella infection by interfering with the activity of key Rab GTPases.


Assuntos
Infecções por Salmonella , Proteínas rab de Ligação ao GTP , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Células HeLa , Humanos , Salmonella/metabolismo
19.
Artigo em Inglês | MEDLINE | ID: mdl-32974218

RESUMO

Legionella pneumophila is an environmental bacterium that has evolved to survive predation by soil and water amoebae such as Acanthamoeba castellanii, and this has inadvertently led to the ability of L. pneumophila to survive and replicate in human cells. L. pneumophila causes Legionnaire's Disease, with human exposure occurring via the inhalation of water aerosols containing both amoebae and the bacteria. These aerosols originate from aquatic biofilms found in artifical water sources, such as air-conditioning cooling towers and humidifiers. In these man-made environments, A. castellanii supports L. pneumophila intracellular replication, thereby promoting persistence and dissemination of the bacteria and providing protection from external stress. Despite this close evolutionary relationship, very little is known about how A. castellanii responds to L. pneumophila infection. In this study, we examined the global transcriptional response of A. castellanii to L. pneumophila infection. We compared A. castellanii infected with wild type L. pneumophila to A. castellanii infected with an isogenic ΔdotA mutant strain, which is unable to replicate intracellularly. We showed that A. castellanii underwent clear morphological and transcriptional rewiring over the course of L. pneumophila infection. Through improved annotation of the A. castellanii genome, we determined that these transcriptional changes primarily involved biological processes utilizing small GTPases, including cellular transport, signaling, metabolism and replication. In addition, a number of sirtuin-encoding genes in A. castellanii were found to be conserved and upregulated during L. pneumophila infection. Silencing of sirtuin gene, sir6f (ACA1_153540) resulted in the inhibition of A. castellanii cell proliferation during infection and reduced L. pneumophila replication. Overall our findings identified several biological pathways in amoebae that may support L. pneumophila replication and A. castellanii proliferation in environmental conditions.


Assuntos
Acanthamoeba castellanii , Legionella pneumophila , Doença dos Legionários , Sirtuínas , Proteínas de Bactérias/genética , Humanos , Legionella pneumophila/genética , Transcriptoma
20.
J Biol Chem ; 295(11): 3401-3402, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32169854

RESUMO

Chronic recurrent multifocal osteomyelitis (CRMO) is an autoinflammatory bone disease mediated by the inflammatory cytokine, IL-1ß. Although IL-1ß is known as the key driver of bone lesions in CRMO, the signaling events leading to pathogenic levels of the cytokine are not fully understood. Using a genetic mouse model of CRMO, Dasari et al. find a role for the nonreceptor spleen tyrosine kinase (SYK) in upstream signaling leading to IL-1ß up-regulation. Their findings suggest that SYK may constitute a new therapeutic target for CRMO.


Assuntos
Inflamassomos , Osteomielite , Animais , Caspase 8 , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Tirosina Quinases , Quinase Syk/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA