Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
Nat Commun ; 15(1): 2452, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38503748

RESUMO

ADP-ribosylation is a reversible post-translational modification involved in various cellular activities. Removal of ADP-ribosylation requires (ADP-ribosyl)hydrolases, with macrodomain enzymes being a major family in this category. The pathogen Legionella pneumophila mediates atypical ubiquitination of host targets using the SidE effector family in a process that involves ubiquitin ADP-ribosylation on arginine 42 as an obligatory step. Here, we show that the Legionella macrodomain effector MavL regulates this pathway by reversing the arginine ADP-ribosylation, likely to minimize potential detrimental effects caused by the modified ubiquitin. We determine the crystal structure of ADP-ribose-bound MavL, providing structural insights into recognition of the ADP-ribosyl group and catalytic mechanism of its removal. Further analyses reveal DUF4804 as a class of MavL-like macrodomain enzymes whose representative members show unique selectivity for mono-ADP-ribosylated arginine residue in synthetic substrates. We find such enzymes are also present in eukaryotes, as exemplified by two previously uncharacterized (ADP-ribosyl)hydrolases in Drosophila melanogaster. Crystal structures of several proteins in this class provide insights into arginine specificity and a shared mode of ADP-ribose interaction distinct from previously characterized macrodomains. Collectively, our study reveals a new regulatory layer of SidE-catalyzed ubiquitination and expands the current understanding of macrodomain enzymes.


Assuntos
Legionella , Ubiquitina , Animais , Ubiquitina/metabolismo , Legionella/metabolismo , Drosophila melanogaster/metabolismo , ADP-Ribosilação , Adenosina Difosfato Ribose/metabolismo , Hidrolases/metabolismo
2.
Mol Microbiol ; 121(2): 243-259, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38153189

RESUMO

The intracellular pathogen Legionella pneumophila translocates more than 300 effector proteins into its host cells. The expression levels of the genes encoding these effectors are orchestrated by an intricate regulatory network. Here, we introduce LelA, the first L. pneumophila LysR-type transcriptional regulator of effectors. Through bioinformatic and experimental analyses, we identified the LelA target regulatory element and demonstrated that it directly activates the expression of three L. pneumophila effectors (legL7, legL6, and legU1). We further found that the gene encoding LelA is positively regulated by the RpoS sigma factor, thus linking it to the known effector regulatory network. Examination of other species throughout the Legionella genus revealed that this regulatory element is found upstream of 34 genes encoding validated effectors, putative effectors, and hypothetical proteins. Moreover, ten of these genes were examined and found to be activated by the L. pneumophila LelA as well as by their orthologs in the corresponding species. LelA represents a novel type of Legionella effector regulator, which coordinates the expression of both adjacently and distantly located effector-encoding genes, thus forming small groups of co-regulated effectors.


Assuntos
Legionella pneumophila , Legionella , Legionella/genética , Legionella/metabolismo , Proteínas de Bactérias/metabolismo , Legionella pneumophila/metabolismo , Fator sigma/genética , Fator sigma/metabolismo , Sequências Reguladoras de Ácido Nucleico
3.
Nat Cell Biol ; 25(11): 1600-1615, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37857833

RESUMO

A widespread strategy employed by pathogens to establish infection is to inhibit host-cell protein synthesis. Legionella pneumophila, an intracellular bacterial pathogen and the causative organism of Legionnaires' disease, secretes a subset of protein effectors into host cells that inhibit translation elongation. Mechanistic insights into how the bacterium targets translation elongation remain poorly defined. We report here that the Legionella effector SidI functions in an unprecedented way as a transfer-RNA mimic that directly binds to and glycosylates the ribosome. The 3.1 Å cryo-electron microscopy structure of SidI reveals an N-terminal domain with an 'inverted L' shape and surface-charge distribution characteristic of tRNA mimicry, and a C-terminal domain that adopts a glycosyl transferase fold that licenses SidI to utilize GDP-mannose as a sugar precursor. This coupling of tRNA mimicry and enzymatic action endows SidI with the ability to block protein synthesis with a potency comparable to ricin, one of the most powerful toxins known. In Legionella-infected cells, the translational pausing activated by SidI elicits a stress response signature mimicking the ribotoxic stress response, which is activated by elongation inhibitors that induce ribosome collisions. SidI-mediated effects on the ribosome activate the stress kinases ZAKα and p38, which in turn drive an accumulation of the protein activating transcription factor 3 (ATF3). Intriguingly, ATF3 escapes the translation block imposed by SidI, translocates to the nucleus and orchestrates the transcription of stress-inducible genes that promote cell death, revealing a major role for ATF3 in the response to collided ribosome stress. Together, our findings elucidate a novel mechanism by which a pathogenic bacterium employs tRNA mimicry to hijack a ribosome-to-nuclear signalling pathway that regulates cell fate.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Humanos , Legionella/metabolismo , Microscopia Crioeletrônica , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Doença dos Legionários/genética , Doença dos Legionários/microbiologia , Transferases/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia
4.
mBio ; 14(5): e0151023, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37819088

RESUMO

IMPORTANCE: Toxin-antitoxin (TA) systems are parasitic genetic elements found in almost all bacterial genomes. They are exchanged horizontally between cells and are typically poorly conserved across closely related strains and species. Here, we report the characterization of a tripartite TA system in the bacterial pathogen Legionella pneumophila that is highly conserved across Legionella species genomes. This system (denoted HipBSTLp) is a distant homolog of the recently discovered split-HipA system in Escherichia coli (HipBSTEc). We present bioinformatic, molecular, and structural analyses of the divergence between these two systems and the functionality of this newly described TA system family. Furthermore, we provide evidence to refute previous claims that the toxin in this system (HipTLp) possesses bifunctionality as an L. pneumophila virulence protein. Overall, this work expands our understanding of the split-HipA system architecture and illustrates the potential for undiscovered biology in these abundant genetic elements.


Assuntos
Proteínas de Escherichia coli , Legionella pneumophila , Legionella , Sistemas Toxina-Antitoxina , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Sistemas Toxina-Antitoxina/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Legionella/metabolismo , Proteínas de Bactérias/metabolismo
5.
mBio ; 14(5): e0170723, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37737612

RESUMO

IMPORTANCE: Inflammasomes are essential for host defense against intracellular bacterial pathogens like Legionella, as they activate caspases, which promote cytokine release and cell death to control infection. In mice, interferon (IFN) signaling promotes inflammasome responses against bacteria by inducing a family of IFN-inducible GTPases known as guanylate-binding proteins (GBPs). Within murine macrophages, IFN promotes the rupture of the Legionella-containing vacuole (LCV), while GBPs are dispensable for this process. Instead, GBPs facilitate the lysis of cytosol-exposed Legionella. In contrast, the functions of IFN and GBPs in human inflammasome responses to Legionella are poorly understood. We show that IFN-γ enhances inflammasome responses to Legionella in human macrophages. Human GBP1 is required for these IFN-γ-driven inflammasome responses. Furthermore, GBP1 co-localizes with Legionella and/or LCVs in a type IV secretion system (T4SS)-dependent manner and promotes damage to the LCV, which leads to increased exposure of the bacteria to the host cell cytosol. Thus, our findings reveal species- and pathogen-specific differences in how GBPs function to promote inflammasome responses.


Assuntos
Legionella pneumophila , Legionella , Humanos , Animais , Camundongos , Inflamassomos/metabolismo , Legionella/metabolismo , Vacúolos/metabolismo , Proteínas de Transporte/metabolismo , Transdução de Sinais , Legionella pneumophila/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo
6.
Chembiochem ; 24(21): e202300442, 2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37489700

RESUMO

Legionella pneumophila is the causative agent of Legionnaires' disease, a serious form of pneumonia. Its macrophage infectivity potentiator (Mip), a member of a highly conserved family of FK506-binding proteins (FKBPs), plays a major role in the proliferation of the gram-negative bacterium in host organisms. In this work, we test our library of >1000 FKBP-focused ligands for inhibition of LpMip. The [4.3.1]-bicyclic sulfonamide turned out as a highly preferred scaffold and provided the most potent LpMip inhibitors known so far. Selected compounds were non-toxic to human cells, displayed antibacterial activity and block bacterial proliferation in cellular infection-assays as well as infectivity in human lung tissue explants. The results confirm [4.3.1]-bicyclic sulfonamides as anti-legionellal agents, although their anti-infective properties cannot be explained by inhibition of LpMip alone.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Humanos , Doença dos Legionários/tratamento farmacológico , Doença dos Legionários/microbiologia , Proteínas de Ligação a Tacrolimo , Peptidilprolil Isomerase/química , Peptidilprolil Isomerase/metabolismo , Proteínas de Bactérias/metabolismo , Legionella pneumophila/metabolismo , Legionella/metabolismo
7.
Cell Rep ; 42(8): 112817, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37471226

RESUMO

Xenophagy is an evolutionarily conserved host defensive mechanism to eliminate invading microorganisms through autophagic machinery. The intracellular bacterial pathogen Legionella pneumophila can avoid clearance by the xenophagy pathway via the actions of multiple Dot/Icm effector proteins. Previous studies have shown that p62, an adaptor protein involved in xenophagy signaling, is excluded from Legionella-containing vacuoles (LCVs). Such defects are attributed to the multifunctional SidE family effectors (SidEs) that exhibit classic deubiquitinase (DUB) and phosphoribosyl ubiquitination (PR-ubiquitination) activities, yet the mechanism remains elusive. In the present study, we demonstrate that the host DUB USP14 is PR-ubiquitinated by SidEs at multiple serine residues, which impairs its DUB activity and its interactions with p62. The exclusion of p62 from the bacterial phagosome requires the ubiquitin ligase but not the DUB activity of SidEs. These results reveal that PR-ubiquitination of USP14 by SidEs contributes to the evasion of xenophagic clearance by L. pneumophila.


Assuntos
Legionella , Doença dos Legionários , Humanos , Legionella/metabolismo , Doença dos Legionários/metabolismo , Serina/metabolismo , Proteínas de Bactérias/metabolismo , Ubiquitinação , Ubiquitina/metabolismo , Fagossomos/metabolismo , Vacúolos/metabolismo , Ubiquitina Tiolesterase/metabolismo
8.
Elife ; 122023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37158597

RESUMO

The amoeba-resistant bacterium Legionella pneumophila causes Legionnaires' disease and employs a type IV secretion system (T4SS) to replicate in the unique, ER-associated Legionella-containing vacuole (LCV). The large fusion GTPase Sey1/atlastin is implicated in ER dynamics, ER-derived lipid droplet (LD) formation, and LCV maturation. Here, we employ cryo-electron tomography, confocal microscopy, proteomics, and isotopologue profiling to analyze LCV-LD interactions in the genetically tractable amoeba Dictyostelium discoideum. Dually fluorescence-labeled D. discoideum producing LCV and LD markers revealed that Sey1 as well as the L. pneumophila T4SS and the Ran GTPase activator LegG1 promote LCV-LD interactions. In vitro reconstitution using purified LCVs and LDs from parental or Δsey1 mutant D. discoideum indicated that Sey1 and GTP promote this process. Sey1 and the L. pneumophila fatty acid transporter FadL were implicated in palmitate catabolism and palmitate-dependent intracellular growth. Taken together, our results reveal that Sey1 and LegG1 mediate LD- and FadL-dependent fatty acid metabolism of intracellular L. pneumophila.


Assuntos
Dictyostelium , Legionella pneumophila , Legionella , Doença dos Legionários , Humanos , Legionella pneumophila/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Macrófagos/metabolismo , Dictyostelium/metabolismo , Gotículas Lipídicas/metabolismo , Vacúolos/metabolismo , Legionella/metabolismo , Doença dos Legionários/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
9.
Proc Natl Acad Sci U S A ; 120(23): e2119658120, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37252954

RESUMO

Peptidoglycan is a critical component of the bacteria cell envelope. Remodeling of the peptidoglycan is required for numerous essential cellular processes and has been linked to bacterial pathogenesis. Peptidoglycan deacetylases that remove the acetyl group of the N-acetylglucosamine (NAG) subunit protect bacterial pathogens from immune recognition and digestive enzymes secreted at the site of infection. However, the full extent of this modification on bacterial physiology and pathogenesis is not known. Here, we identify a polysaccharide deacetylase of the intracellular bacterial pathogen Legionella pneumophila and define a two-tiered role for this enzyme in Legionella pathogenesis. First, NAG deacetylation is important for the proper localization and function of the Type IVb secretion system, linking peptidoglycan editing to the modulation of host cellular processes through the action of secreted virulence factors. As a consequence, the Legionella vacuole mis-traffics along the endocytic pathway to the lysosome, preventing the formation of a replication permissive compartment. Second, within the lysosome, the inability to deacetylate the peptidoglycan renders the bacteria more sensitive to lysozyme-mediated degradation, resulting in increased bacterial death. Thus, the ability to deacetylate NAG is important for bacteria to persist within host cells and in turn, Legionella virulence. Collectively, these results expand the function of peptidoglycan deacetylases in bacteria, linking peptidoglycan editing, Type IV secretion, and the intracellular fate of a bacterial pathogen.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Humanos , Legionella pneumophila/metabolismo , Peptidoglicano/metabolismo , Vacúolos/metabolismo , Legionella/metabolismo , Lisossomos/metabolismo , Proteínas de Bactérias/metabolismo , Doença dos Legionários/microbiologia
10.
Life Sci Alliance ; 6(7)2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37100438

RESUMO

Although ubiquitin is found only in eukaryotes, several pathogenic bacteria and viruses possess proteins that hinder the host ubiquitin system. Legionella, a gram-negative intracellular bacterium, possesses an ovarian tumor (OTU) family of deubiquitinases (Lot DUBs). Herein, we describe the molecular characteristics of Lot DUBs. We elucidated the structure of the LotA OTU1 domain and revealed that entire Lot DUBs possess a characteristic extended helical lobe that is not found in other OTU-DUBs. The structural topology of an extended helical lobe is the same throughout the Lot family, and it provides an S1' ubiquitin-binding site. Moreover, the catalytic triads of Lot DUBs resemble those of the A20-type OTU-DUBs. Furthermore, we revealed a unique mechanism by which LotA OTU domains cooperate together to distinguish the length of the chain and preferentially cleave longer K48-linked polyubiquitin chains. The LotA OTU1 domain itself cleaves K6-linked ubiquitin chains, whereas it is also essential for assisting the cleavage of longer K48-linked polyubiquitin chains by the OTU2 domain. Thus, this study provides novel insights into the structure and mechanism of action of Lot DUBs.


Assuntos
Legionella , Neoplasias Ovarianas , Feminino , Humanos , Ubiquitina/metabolismo , Poliubiquitina/química , Poliubiquitina/metabolismo , Legionella/metabolismo , Enzimas Desubiquitinantes/genética , Enzimas Desubiquitinantes/metabolismo , Neoplasias Ovarianas/genética
11.
Nat Commun ; 14(1): 2245, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-37076474

RESUMO

Bacterial pathogens often make use of post-translational modifications to manipulate host cells. Legionella pneumophila, the causative agent of Legionnaires disease, secretes the enzyme AnkX that uses cytidine diphosphate-choline to post-translationally modify the human small G-Protein Rab1 with a phosphocholine moiety at Ser76. Later in the infection, the Legionella enzyme Lem3 acts as a dephosphocholinase, hydrolytically removing the phosphocholine. While the molecular mechanism for Rab1 phosphocholination by AnkX has recently been resolved, structural insights into the activity of Lem3 remained elusive. Here, we stabilise the transient Lem3:Rab1b complex by substrate mediated covalent capture. Through crystal structures of Lem3 in the apo form and in complex with Rab1b, we reveal Lem3's catalytic mechanism, showing that it acts on Rab1 by locally unfolding it. Since Lem3 shares high structural similarity with metal-dependent protein phosphatases, our Lem3:Rab1b complex structure also sheds light on how these phosphatases recognise protein substrates.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Humanos , Legionella/metabolismo , Fosforilcolina/metabolismo , Legionella pneumophila/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas de Bactérias/metabolismo , Proteínas rab1 de Ligação ao GTP/metabolismo
12.
Nat Commun ; 14(1): 2154, 2023 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-37059817

RESUMO

Legionella pneumophila replicates intracellularly by secreting effectors via a type IV secretion system. One of these effectors is a eukaryotic methyltransferase (RomA) that methylates K14 of histone H3 (H3K14me3) to counteract host immune responses. However, it is not known how L. pneumophila infection catalyses H3K14 methylation as this residue is usually acetylated. Here we show that L. pneumophila secretes a eukaryotic-like histone deacetylase (LphD) that specifically targets H3K14ac and works in synergy with RomA. Both effectors target host chromatin and bind the HBO1 histone acetyltransferase complex that acetylates H3K14. Full activity of RomA is dependent on the presence of LphD as H3K14 methylation levels are significantly decreased in a ∆lphD mutant. The dependency of these two chromatin-modifying effectors on each other is further substantiated by mutational and virulence assays revealing that the presence of only one of these two effectors impairs intracellular replication, while a double knockout (∆lphD∆romA) can restore intracellular replication. Uniquely, we present evidence for "para-effectors", an effector pair, that actively and coordinately modify host histones to hijack the host response. The identification of epigenetic marks modulated by pathogens has the potential to lead to the development of innovative therapeutic strategies to counteract bacterial infection and strengthening host defences.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Humanos , Legionella/metabolismo , Cromatina/metabolismo , Proteínas de Bactérias/metabolismo , Doença dos Legionários/genética , Histonas/metabolismo
13.
Infect Immun ; 91(4): e0044122, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-36912646

RESUMO

To replicate within host cells, bacterial pathogens must acquire host-derived nutrients while avoiding degradative antimicrobial pathways. Fundamental insights into bacterial pathogenicity have been revealed by bacteria of the genus Legionella, which naturally parasitize free-living protozoa by establishing a membrane-bound replicative niche termed the Legionella-containing vacuole (LCV). Biogenesis of the LCV and intracellular replication rely on rapid evasion of the endocytic pathway and acquisition of host-derived nutrients, much of which is mediated by bacterial effector proteins translocated into host cells by a Dot/Icm type IV secretion system. Billions of years of co-evolution with eukaryotic hosts and broad host tropism have resulted in expansion of the Legionella genome to accommodate a massive repertoire of effector proteins that promote LCV biogenesis, safeguard the LCV from endolysosomal maturation, and mediate the acquisition of host nutrients. This minireview is focused on the mechanisms by which an ancient intracellular pathogen leverages effector proteins and hijacks host cell biology to obtain essential host-derived nutrients and prevent lysosomal degradation.


Assuntos
Legionella pneumophila , Legionella , Legionella/genética , Legionella/metabolismo , Legionella pneumophila/metabolismo , Vacúolos/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Lisossomos/metabolismo , Nutrientes , Interações Hospedeiro-Patógeno
14.
BMC Neurol ; 23(1): 69, 2023 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-36782173

RESUMO

BACKGROUND: Autoimmune glial fibrillary acidic protein (GFAP) astrocytopathy is a recently identified recurrent meningoencephalomyelitis with GFAP immunoglobulin G presence in the serum or cerebrospinal fluid (CSF) as a specific biomarker. GFAP astrocytopathy is closely associated with the occurrence of some tumors and often coexists with other antibodies, such as the N-methyl-D-aspartate receptor and aquaporin-4 antibodies. However, GFAP astrocytopathy complicated by central nervous system infection is rare. CASE PRESENTATION: Here, we present the case of a patient admitted to a local hospital due to a prominent fever and cough. The patient had a 1-month history of headaches before admission that were not considered serious at the time. Metagenomic next-generation sequencing (mNGS) of bronchoalveolar lavage fluid revealed a high sequence number of Legionella pneumophila and a few mycobacteria. His cough and fever improved significantly after antibiotic treatment. Still, a slight headache remained. Subsequently, his condition worsened, and he visited our hospital with a disturbance of consciousness. Mycobacterium tuberculosis was detected with mNGS of the CSF, while the CSF and serum were also positive for GFAP antibodies. Following anti-tuberculosis and steroid therapy, the patient's symptoms improved, and he tested negative for the GFAP antibody. CONCLUSION: This is the first reported case of GFAP astrocytopathy complicated by tuberculous meningoencephalitis. Due to overlaps in the clinical manifestations of the two diseases, GFAP astrocytopathy is sometimes misdiagnosed as tuberculous meningoencephalitis. Therefore, in addition to ensuring careful identification of the two diseases, clinicians need to be aware of their possible co-existence.


Assuntos
Legionella , Meningoencefalite , Pneumonia , Tuberculose Meníngea , Masculino , Humanos , Proteína Glial Fibrilar Ácida , Tosse , Meningoencefalite/complicações , Meningoencefalite/diagnóstico , Autoanticorpos/líquido cefalorraquidiano , Febre , Legionella/metabolismo
15.
EMBO Rep ; 24(3): e56007, 2023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36588479

RESUMO

Legionella pneumophila replicates in macrophages and amoeba within a unique compartment, the Legionella-containing vacuole (LCV). Hallmarks of LCV formation are the phosphoinositide lipid conversion from PtdIns(3)P to PtdIns(4)P, fusion with ER-derived vesicles and a tight association with the ER. Proteomics of purified LCVs indicate the presence of membrane contact sites (MCS) proteins possibly implicated in lipid exchange. Using dually fluorescence-labeled Dictyostelium discoideum amoeba, we reveal that VAMP-associated protein (Vap) and the PtdIns(4)P 4-phosphatase Sac1 localize to the ER, and Vap also localizes to the LCV membrane. Furthermore, Vap as well as Sac1 promote intracellular replication of L. pneumophila and LCV remodeling. Oxysterol binding proteins (OSBPs) preferentially localize to the ER (OSBP8) or the LCV membrane (OSBP11), respectively, and restrict (OSBP8) or promote (OSBP11) bacterial replication and LCV expansion. The sterol probes GFP-D4H* and filipin indicate that sterols are rapidly depleted from LCVs, while PtdIns(4)P accumulates. In addition to Sac1, the PtdIns(4)P-subverting L. pneumophila effector proteins LepB and SidC also support LCV remodeling. Taken together, the Legionella- and host cell-driven PtdIns(4)P gradient at LCV-ER MCSs promotes Vap-, OSBP- and Sac1-dependent pathogen vacuole maturation.


Assuntos
Dictyostelium , Legionella pneumophila , Legionella , Vacúolos/metabolismo , Legionella/metabolismo , Dictyostelium/microbiologia , Fosfatidilinositóis/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Bactérias/metabolismo
16.
Sci Adv ; 8(50): eadd7945, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36525490

RESUMO

The intracellular bacterial pathogen Legionella pneumophila uses hundreds of effector proteins to manipulate multiple processes of the host cells to establish a replicative niche known as Legionella-containing vacuole (LCV). Biogenesis of the LCV has been known to depend on host small guanosine triphosphatases (GTPases), but whether bacterial effector GTPases are also involved remains unknown. Here, we show that an ankyrin repeat containing effector LegA15 localizes directly in host lipid droplets (LDs), leading to Golgi apparatus fragmentation of the host cells by hijacking the host vesicular transport factor p115. LegA15 is a GTPase with a unique catalytic mechanism, unlike any eukaryotic small GTPases. Moreover, the effector LegA15 co-opts p115 to modulate homeostasis of the host LDs in its GTPase-dependent manner. Together, our data reveal that an atypical GTPase effector regulates the host LDs through impeding the vesicle secretion system of the host cells for intracellular life cycle of Legionella.


Assuntos
Legionella , Legionella/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Gotículas Lipídicas/metabolismo , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno
17.
Sci Rep ; 12(1): 21782, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36526881

RESUMO

The pathogenic Legionella bacteria are notorious for delivering numerous effector proteins into the host cell with the aim of disturbing and hijacking cellular processes for their benefit. Despite intensive studies, many effectors remain uncharacterized. Motivated by the richness of Legionella effector repertoires and their oftentimes atypical biochemistry, also by several known atypical Legionella effector kinases and pseudokinases discovered recently, we undertook an in silico survey and exploration of the pan-kinome of the Legionella genus, i.e., the union of the kinomes of individual species. In this study, we discovered 13 novel (pseudo)kinase families (all are potential effectors) with the use of non-standard bioinformatic approaches. Together with 16 known families, we present a catalog of effector and non-effector protein kinase-like families within Legionella, available at http://bioinfo.sggw.edu.pl/kintaro/ . We analyze and discuss the likely functional roles of the novel predicted kinases. Notably, some of the kinase families are also present in other bacterial taxa, including other pathogens, often phylogenetically very distant from Legionella. This work highlights Nature's ingeniousness in the pathogen-host arms race and offers a useful resource for the study of infection mechanisms.


Assuntos
Legionella pneumophila , Legionella , Legionella/metabolismo , Proteínas Quinases/metabolismo , Biologia Computacional , Legionella pneumophila/metabolismo , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno
18.
Proc Natl Acad Sci U S A ; 119(48): e2209149119, 2022 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-36413498

RESUMO

Intravacuolar pathogens need to gradually expand their surrounding vacuole to accommodate the growing number of bacterial offspring during intracellular replication. Here we found that Legionella pneumophila controls vacuole expansion by fine-tuning the generation of lysophospholipids within the vacuolar membrane. Upon allosteric activation by binding to host ubiquitin, the type IVB (Dot/Icm) effector VpdC converts phospholipids into lysophospholipids which, at moderate concentrations, are known to promote membrane fusion but block it at elevated levels by generating excessive positive membrane curvature. Consequently, L. pneumophila overproducing VpdC were prevented from adequately expanding their surrounding membrane, trapping the replicating bacteria within spatially confined vacuoles and reducing their capability to proliferate intracellularly. Quantitative lipidomics confirmed a VpdC-dependent increase in several types of lysophospholipids during infection, and VpdC production in transiently transfected cells caused tubulation of organelle membranes as well as mitochondria fragmentation, processes that can be phenocopied by supplying cells with exogenous lysophospholipids. Together, these results demonstrate an important role for bacterial phospholipases in vacuolar expansion.


Assuntos
Legionella , Doença dos Legionários , Humanos , Legionella/metabolismo , Vacúolos/metabolismo , Doença dos Legionários/microbiologia , Fosfolipases/metabolismo , Ubiquitina/metabolismo , Proteínas de Bactérias/metabolismo , Lisofosfolipídeos/metabolismo
19.
Acta Crystallogr D Struct Biol ; 78(Pt 9): 1110-1119, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-36048151

RESUMO

The pathogen Legionella pneumophila, which is the causative agent of Legionnaires' disease, secrets hundreds of effectors into host cells via its Dot/Icm secretion system to subvert host-cell pathways during pathogenesis. VipF, a conserved core effector among Legionella species, is a putative acetyltransferase, but its structure and catalytic mechanism remain unknown. Here, three crystal structures of VipF in complex with its cofactor acetyl-CoA and/or a substrate are reported. The two GNAT-like domains of VipF are connected as two wings by two ß-strands to form a U-shape. Both domains bind acetyl-CoA or CoA, but only in the C-terminal domain does the molecule extend to the bottom of the U-shaped groove as required for an active transferase reaction; the molecule in the N-terminal domain folds back on itself. Interestingly, when chloramphenicol, a putative substrate, binds in the pocket of the central U-shaped groove adjacent to the N-terminal domain, VipF remains in an open conformation. Moreover, mutations in the central U-shaped groove, including Glu129 and Asp251, largely impaired the acetyltransferase activity of VipF, suggesting a unique enzymatic mechanism for the Legionella effector VipF.


Assuntos
Legionella pneumophila , Legionella , Doença dos Legionários , Acetilcoenzima A/metabolismo , Acetilação , Acetiltransferases , Proteínas de Bactérias/química , Legionella/metabolismo , Legionella pneumophila/química , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Doença dos Legionários/genética
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...