Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 22(11): 1367-1374, 2021 11.
Article in English | MEDLINE | ID: mdl-34686862

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) represent innate homologs of type 2 helper T cells (TH2) that participate in immune defense and tissue homeostasis through production of type 2 cytokines. While T lymphocytes metabolically adapt to microenvironmental changes, knowledge of human ILC2 metabolism is limited, and its key regulators are unknown. Here, we show that circulating 'naive' ILC2s have an unexpected metabolic profile with a higher level of oxidative phosphorylation (OXPHOS) than natural killer (NK) cells. Accordingly, ILC2s are severely reduced in individuals with mitochondrial disease (MD) and impaired OXPHOS. Metabolomic and nutrient receptor analysis revealed ILC2 uptake of amino acids to sustain OXPHOS at steady state. Following activation with interleukin-33 (IL-33), ILC2s became highly proliferative, relying on glycolysis and mammalian target of rapamycin (mTOR) to produce IL-13 while continuing to fuel OXPHOS with amino acids to maintain cellular fitness and proliferation. Our results suggest that proliferation and function are metabolically uncoupled in human ILC2s, offering new strategies to target ILC2s in disease settings.


Subject(s)
Cell Proliferation , Cytokines/metabolism , Energy Metabolism , Immunity, Innate , Lymphocyte Activation , Mitochondrial Diseases/metabolism , Th2 Cells/metabolism , Amino Acids, Branched-Chain/metabolism , Arginine/metabolism , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Energy Metabolism/drug effects , Humans , Immunity, Innate/drug effects , Interleukin-33/pharmacology , Lymphocyte Activation/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/immunology , Phenotype , Th2 Cells/drug effects , Th2 Cells/immunology
2.
Genome Res ; 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36109147

ABSTRACT

The unicellular amoeba Acanthamoeba castellanii is ubiquitous in aquatic environments, where it preys on bacteria. The organism also hosts bacterial endosymbionts, some of which are parasitic, including human pathogens such as Chlamydia and Legionella spp. Here we report complete, high-quality genome sequences for two extensively studied A. castellanii strains, Neff and C3. Combining long- and short-read data with Hi-C, we generated near chromosome-level assemblies for both strains with 90% of the genome contained in 29 scaffolds for the Neff strain and 31 for the C3 strain. Comparative genomics revealed strain-specific functional enrichment, most notably genes related to signal transduction in the C3 strain and to viral replication in Neff. Furthermore, we characterized the spatial organization of the A. castellanii genome and showed that it is reorganized during infection by Legionella pneumophila Infection-dependent chromatin loops were found to be enriched in genes for signal transduction and phosphorylation processes. In genomic regions where chromatin organization changed during Legionella infection, we found functional enrichment for genes associated with metabolism, organelle assembly, and cytoskeleton organization. Given Legionella infection is known to alter its host's cell cycle, to exploit the host's organelles, and to modulate the host's metabolism in its favor, these changes in chromatin organization may partly be related to mechanisms of host control during Legionella infection.

3.
PLoS Pathog ; 19(5): e1011375, 2023 05.
Article in English | MEDLINE | ID: mdl-37155695

ABSTRACT

The zebrafish has become a powerful model organism to study host-pathogen interactions. Here, we developed a zebrafish model to dissect the innate immune response to Legionella pneumophila during infection. We show that L. pneumophila cause zebrafish larvae death in a dose dependent manner. Additionally, we show that macrophages are the first line of defence and cooperate with neutrophils to clear the infection. Immunocompromised humans have an increased propensity to develop pneumonia, similarly, when either macrophages or neutrophils are depleted, these "immunocompromised" larvae become lethally sensitive to L. pneumophila. Also, as observed in human infections, the adaptor signalling molecule Myd88 is not required to control disease in the larvae. Furthermore, proinflammatory cytokine genes il1ß and tnf-α were upregulated during infection, recapitulating key immune responses seen in human infection. Strikingly, we uncovered a previously undescribed infection phenotype in zebrafish larvae, whereby bloodborne, wild type L. pneumophila invade and grow in the larval yolk region, a phenotype not observed with a type IV secretion system deficient mutant that cannot translocate effectors into its host cell. Thus, zebrafish larva represents an innovative L. pneumophila infection model that mimics important aspects of the human immune response to L. pneumophila infection and will allow the elucidation of mechanisms by which type IV secretion effectors allow L. pneumophila to cross host cell membranes and obtain nutrients from nutrient rich environments.


Subject(s)
Legionella pneumophila , Legionnaires' Disease , Animals , Humans , Zebrafish , Immunity, Innate , Macrophages , Larva
4.
Cell ; 139(4): 770-9, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19914169

ABSTRACT

Riboswitches are RNA elements acting in cis, controlling expression of their downstream genes through a metabolite-induced alteration of their secondary structure. Here, we demonstrate that two S-adenosylmethionine (SAM) riboswitches, SreA and SreB, can also function in trans and act as noncoding RNAs in Listeria monocytogenes. SreA and SreB control expression of the virulence regulator PrfA by binding to the 5'-untranslated region of its mRNA. Absence of the SAM riboswitches SreA and SreB increases the level of PrfA and virulence gene expression in L. monocytogenes. Thus, the impact of the SAM riboswitches on PrfA expression highlights a link between bacterial virulence and nutrient availability. Together, our results uncover an unexpected role for riboswitches and a distinct class of regulatory noncoding RNAs in bacteria.


Subject(s)
Bacterial Proteins/genetics , Listeria monocytogenes/genetics , Peptide Termination Factors/genetics , Regulatory Sequences, Ribonucleic Acid , 5' Untranslated Regions , Base Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Molecular Sequence Data , Temperature , Virulence
5.
Microbiology (Reading) ; 168(3)2022 03.
Article in English | MEDLINE | ID: mdl-35230931

ABSTRACT

Legionella pneumophila is an environmental bacterium that parasitizes aquatic protozoa and uses the same processes to infect humans. The facultative intracellular pathogen causes a life-threatening pneumonia with possible systemic complications. The co-evolution with protozoa is reflected in an armoury of bacterial effectors, and many of these type IV-secreted proteins have likely been acquired by interdomain horizontal gene transfer (HGT) from hosts. The unique features of L. pneumophila are the largest bacterial effector repertoire known to date, subversion of virtually all eukaryotic signalling pathways and acquisition of eukaryotic enzyme activities used to manipulate the host cell to the pathogen's advantage.


Subject(s)
Legionella pneumophila , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Eukaryota , Gene Transfer, Horizontal , Humans , Legionella pneumophila/genetics , Legionella pneumophila/metabolism
6.
Proc Natl Acad Sci U S A ; 116(6): 2265-2273, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30659146

ABSTRACT

The genus Legionella comprises 65 species, among which Legionella pneumophila is a human pathogen causing severe pneumonia. To understand the evolution of an environmental to an accidental human pathogen, we have functionally analyzed 80 Legionella genomes spanning 58 species. Uniquely, an immense repository of 18,000 secreted proteins encoding 137 different eukaryotic-like domains and over 200 eukaryotic-like proteins is paired with a highly conserved type IV secretion system (T4SS). Specifically, we show that eukaryotic Rho- and Rab-GTPase domains are found nearly exclusively in eukaryotes and Legionella Translocation assays for selected Rab-GTPase proteins revealed that they are indeed T4SS secreted substrates. Furthermore, F-box, U-box, and SET domains were present in >70% of all species, suggesting that manipulation of host signal transduction, protein turnover, and chromatin modification pathways are fundamental intracellular replication strategies for legionellae. In contrast, the Sec-7 domain was restricted to L. pneumophila and seven other species, indicating effector repertoire tailoring within different amoebae. Functional screening of 47 species revealed 60% were competent for intracellular replication in THP-1 cells, but interestingly, this phenotype was associated with diverse effector assemblages. These data, combined with evolutionary analysis, indicate that the capacity to infect eukaryotic cells has been acquired independently many times within the genus and that a highly conserved yet versatile T4SS secretes an exceptional number of different proteins shaped by interdomain gene transfer. Furthermore, we revealed the surprising extent to which legionellae have coopted genes and thus cellular functions from their eukaryotic hosts, providing an understanding of how dynamic reshuffling and gene acquisition have led to the emergence of major human pathogens.


Subject(s)
Genome, Bacterial , Legionella/physiology , Legionellosis/microbiology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Secretion Systems/genetics , Computational Biology/methods , Evolution, Molecular , Genomics/methods , Humans , Intracellular Space/microbiology , Legionella/classification , Phylogeny , Protein Domains
7.
Int Immunol ; 32(7): 475-483, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32441740

ABSTRACT

In this review, we propose that certain modifications in cellular metabolism might function as danger signals triggering inflammasome-mediated immune responses. We propose to call them danger-associated metabolic modifications (DAMMs). As intracellular bacteria can actively modulate macrophage metabolism for their benefit, infected host cells might sense bacteria-induced metabolic alterations and activate immune reactions. Here we report the known metabolic interactions that occur during infection of macrophages by intracellular bacteria and discuss the possible emergence of DAMMs upon bacteria-induced alterations of cellular metabolism.


Subject(s)
Bacterial Infections/metabolism , Macrophages/metabolism , Animals , Bacterial Infections/immunology , Humans , Macrophages/immunology
8.
Cell Microbiol ; 22(10): e13246, 2020 10.
Article in English | MEDLINE | ID: mdl-32720355

ABSTRACT

Intracellular bacterial pathogens harbour genes, the closest homologues of which are found in eukaryotes. Regulator of chromosome condensation 1 (RCC1) repeat proteins are phylogenetically widespread and implicated in protein-protein interactions, such as the activation of the small GTPase Ran by its cognate guanine nucleotide exchange factor, RCC1. Legionella pneumophila and Coxiella burnetii, the causative agents of Legionnaires' disease and Q fever, respectively, harbour RCC1 repeat coding genes. Legionella pneumophila secretes the RCC1 repeat 'effector' proteins LegG1, PpgA and PieG into eukaryotic host cells, where they promote the activation of the pleiotropic small GTPase Ran, microtubule stabilisation, pathogen vacuole motility and intracellular bacterial growth as well as host cell migration. The RCC1 repeat effectors localise to the pathogen vacuole or the host plasma membrane and target distinct components of the Ran GTPase cycle, including Ran modulators and the small GTPase itself. Coxiella burnetii translocates the RCC1 repeat effector NopA into host cells, where the protein localises to nucleoli. NopA binds to Ran GTPase and promotes the nuclear accumulation of Ran(GTP), thus pertubing the import of the transcription factor NF-κB and innate immune signalling. Hence, divergent evolution of bacterial RCC1 repeat effectors defines the range of Ran GTPase cycle targets and likely allows fine-tuning of Ran GTPase activation by the pathogens at different cellular sites.


Subject(s)
Biological Evolution , Coxiella burnetii/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Legionella pneumophila/metabolism , ran GTP-Binding Protein/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Nucleolus/enzymology , Coxiella burnetii/genetics , Coxiella burnetii/pathogenicity , Enzyme Activation , Genes, Bacterial , Host-Pathogen Interactions , Humans , Legionella/genetics , Legionella/metabolism , Legionella/pathogenicity , Legionella pneumophila/genetics , Legionnaires' Disease/microbiology , Protein Transport , Q Fever/microbiology , Vacuoles/metabolism , Vacuoles/microbiology
9.
PLoS Genet ; 13(2): e1006629, 2017 02.
Article in English | MEDLINE | ID: mdl-28212376

ABSTRACT

The carbon storage regulator protein CsrA regulates cellular processes post-transcriptionally by binding to target-RNAs altering translation efficiency and/or their stability. Here we identified and analyzed the direct targets of CsrA in the human pathogen Legionella pneumophila. Genome wide transcriptome, proteome and RNA co-immunoprecipitation followed by deep sequencing of a wild type and a csrA mutant strain identified 479 RNAs with potential CsrA interaction sites located in the untranslated and/or coding regions of mRNAs or of known non-coding sRNAs. Further analyses revealed that CsrA exhibits a dual regulatory role in virulence as it affects the expression of the regulators FleQ, LqsR, LetE and RpoS but it also directly regulates the timely expression of over 40 Dot/Icm substrates. CsrA controls its own expression and the stringent response through a regulatory feedback loop as evidenced by its binding to RelA-mRNA and links it to quorum sensing and motility. CsrA is a central player in the carbon, amino acid, fatty acid metabolism and energy transfer and directly affects the biosynthesis of cofactors, vitamins and secondary metabolites. We describe the first L. pneumophila riboswitch, a thiamine pyrophosphate riboswitch whose regulatory impact is fine-tuned by CsrA, and identified a unique regulatory mode of CsrA, the active stabilization of RNA anti-terminator conformations inside a coding sequence preventing Rho-dependent termination of the gap operon through transcriptional polarity effects. This allows L. pneumophila to regulate the pentose phosphate pathway and the glycolysis combined or individually although they share genes in a single operon. Thus the L. pneumophila genome has evolved to acclimate at least five different modes of regulation by CsrA giving it a truly unique position in its life cycle.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Genome, Bacterial/genetics , Legionella pneumophila/genetics , Repressor Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Blotting, Northern , Evolution, Molecular , Feedback, Physiological , Gene Expression Profiling/methods , Glycolysis/genetics , Host-Pathogen Interactions , Humans , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Legionnaires' Disease/microbiology , Mutation , Oligonucleotide Array Sequence Analysis , Operon/genetics , Pentose Phosphate Pathway/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Riboswitch/genetics , Tandem Mass Spectrometry , Virulence/genetics
10.
11.
PLoS Genet ; 13(6): e1006855, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28650958

ABSTRACT

Legionella pneumophila is an environmental bacterium and the causative agent of Legionnaires' disease. Previous genomic studies have shown that recombination accounts for a high proportion (>96%) of diversity within several major disease-associated sequence types (STs) of L. pneumophila. This suggests that recombination represents a potentially important force shaping adaptation and virulence. Despite this, little is known about the biological effects of recombination in L. pneumophila, particularly with regards to homologous recombination (whereby genes are replaced with alternative allelic variants). Using newly available population genomic data, we have disentangled events arising from homologous and non-homologous recombination in six major disease-associated STs of L. pneumophila (subsp. pneumophila), and subsequently performed a detailed characterisation of the dynamics and impact of homologous recombination. We identified genomic "hotspots" of homologous recombination that include regions containing outer membrane proteins, the lipopolysaccharide (LPS) region and Dot/Icm effectors, which provide interesting clues to the selection pressures faced by L. pneumophila. Inference of the origin of the recombined regions showed that isolates have most frequently imported DNA from isolates belonging to their own clade, but also occasionally from other major clades of the same subspecies. This supports the hypothesis that the possibility for horizontal exchange of new adaptations between major clades of the subspecies may have been a critical factor in the recent emergence of several clinically important STs from diverse genomic backgrounds. However, acquisition of recombined regions from another subspecies, L. pneumophila subsp. fraseri, was rarely observed, suggesting the existence of a recombination barrier and/or the possibility of ongoing speciation between the two subspecies. Finally, we suggest that multi-fragment recombination may occur in L. pneumophila, whereby multiple non-contiguous segments that originate from the same molecule of donor DNA are imported into a recipient genome during a single episode of recombination.


Subject(s)
Evolution, Molecular , Homologous Recombination/genetics , Legionella pneumophila/genetics , Legionnaires' Disease/genetics , Bacterial Outer Membrane Proteins/genetics , Genome, Bacterial , Legionnaires' Disease/microbiology , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/genetics , Phylogeny , Recombinant Proteins/genetics
12.
Genes Immun ; 20(5): 394-402, 2019 05.
Article in English | MEDLINE | ID: mdl-31053752

ABSTRACT

Legionella pneumophila is an intracellular pathogen that causes a severe pneumonia called Legionnaires' disease that is often fatal when not promptly diagnosed and treated. However, L. pneumophila is mainly an environmental pathogen of protozoa. This bacterium parasitizes free-living amoeba and other aquatic protozoa with which it co-evolved over an evolutionary long time. Due to the close relationship between hosts and pathogens, their co-evolution leads to molecular interactions such as the exchange of genetic material through horizontal gene transfer (HGT). Those genes that confer an advantage to the bacteria were fixed in their genomes and help these pathogens to subvert host functions to their advantage. Genome sequencing of L. pneumophila and recently of the entire genus Legionella that comprises over 60 species revealed that Legionellae have co-opted genes and thus cellular functions from their eukaryotic hosts to a surprisingly high extent never observed before for an prokaryotic organism. Acquisition and loss of these eukaryotic-like genes and eukaryotic domains is an ongoing process underlining the highly dynamic nature of the Legionella genomes. Although the large amount and diversity of HGT that occurred between Legionella and their protozoan hosts seems to be unique in the prokaryotic world, the analyses of more and more genomes from environmental organisms and symbionts of amoeba revealed that such genetic exchanges occur among all amoeba-associated bacteria and also among the different microorganisms that infect amoeba such as viruses. This dynamic reshuffling and gene-acquisition has led to the emergence of major human pathogens such as Legionella and may lead to the emergence of new human pathogens from the environment.


Subject(s)
Evolution, Molecular , Host-Pathogen Interactions/genetics , Legionella pneumophila/pathogenicity , Amoeba/genetics , Amoeba/microbiology , Gene Transfer, Horizontal , Legionella pneumophila/genetics
13.
Environ Microbiol ; 21(3): 1035-1053, 2019 03.
Article in English | MEDLINE | ID: mdl-30623561

ABSTRACT

The causative agent of Legionnaires' disease, Legionella pneumophila, colonizes amoebae and biofilms in the environment. The opportunistic pathogen employs the Lqs (Legionella quorum sensing) system and the signalling molecule LAI-1 (Legionella autoinducer-1) to regulate virulence, motility, natural competence and expression of a 133 kb genomic "fitness island", including a putative novel regulator. Here, we show that the regulator termed LvbR is an LqsS-regulated transcription factor that binds to the promoter of lpg1056/hnox1 (encoding an inhibitor of the diguanylate cyclase Lpg1057), and thus, regulates proteins involved in c-di-GMP metabolism. LvbR determines biofilm architecture, since L. pneumophila lacking lvbR accumulates less sessile biomass and forms homogeneous mat-like structures, while the parental strain develops more compact bacterial aggregates. Comparative transcriptomics of sessile and planktonic ΔlvbR or ΔlqsR mutant strains revealed concerted (virulence, fitness island, metabolism) and reciprocally (motility) regulated genes in biofilm and broth respectively. Moreover, ΔlvbR is hyper-competent for DNA uptake, defective for phagocyte infection, outcompeted by the parental strain in amoebae co-infections and impaired for cell migration inhibition. Taken together, our results indicate that L. pneumophila LvbR is a novel pleiotropic transcription factor, which links the Lqs and c-di-GMP regulatory networks to control biofilm architecture and pathogen-host cell interactions.


Subject(s)
Bacterial Proteins/metabolism , Biofilms , Cyclic GMP/analogs & derivatives , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Legionella pneumophila/genetics , Transcription Factors/metabolism , 4-Butyrolactone/analogs & derivatives , Bacterial Proteins/genetics , Cyclic GMP/metabolism , Legionella pneumophila/pathogenicity , Legionnaires' Disease/microbiology , Quorum Sensing , Virulence
14.
Genome Res ; 26(11): 1555-1564, 2016 11.
Article in English | MEDLINE | ID: mdl-27662900

ABSTRACT

Legionella pneumophila is an environmental bacterium and the leading cause of Legionnaires' disease. Just five sequence types (ST), from more than 2000 currently described, cause nearly half of disease cases in northwest Europe. Here, we report the sequence and analyses of 364 L. pneumophila genomes, including 337 from the five disease-associated STs and 27 representative of the species diversity. Phylogenetic analyses revealed that the five STs have independent origins within a highly diverse species. The number of de novo mutations is extremely low with maximum pairwise single-nucleotide polymorphisms (SNPs) ranging from 19 (ST47) to 127 (ST1), which suggests emergences within the last century. Isolates sampled geographically far apart differ by only a few SNPs, demonstrating rapid dissemination. These five STs have been recombining recently, leading to a shared pool of allelic variants potentially contributing to their increased disease propensity. The oldest clone, ST1, has spread globally; between 1940 and 2000, four new clones have emerged in Europe, which show long-distance, rapid dispersal. That a large proportion of clinical cases is caused by recently emerged and internationally dispersed clones, linked by convergent evolution, is surprising for an environmental bacterium traditionally considered to be an opportunistic pathogen. To simultaneously explain recent emergence, rapid spread and increased disease association, we hypothesize that these STs have adapted to new man-made environmental niches, which may be linked by human infection and transmission.


Subject(s)
Evolution, Molecular , Legionella pneumophila/genetics , Legionnaires' Disease/microbiology , Humans , Legionella pneumophila/classification , Legionella pneumophila/isolation & purification , Legionella pneumophila/pathogenicity , Mutation , Phylogeny , Polymorphism, Single Nucleotide , Selection, Genetic , Virulence/genetics
15.
Cell Microbiol ; 20(9): e12852, 2018 09.
Article in English | MEDLINE | ID: mdl-29691989

ABSTRACT

The intracellular pathogen Legionella pneumophila influences numerous eukaryotic cellular processes through the Dot/Icm-dependent translocation of more than 300 effector proteins into the host cell. Although many translocated effectors localise to the Legionella replicative vacuole, other effectors can affect remote intracellular sites. Following infection, a subset of effector proteins localises to the nucleus where they subvert host cell transcriptional responses to infection. Here, we identified Lpw27461 (Lpp2587), Lpg2519 as a new nuclear-localised effector that we have termed SnpL. Upon ectopic expression or during L. pneumophila infection, SnpL showed strong nuclear localisation by immunofluorescence microscopy but was excluded from nucleoli. Using immunoprecipitation and mass spectrometry, we determined the host-binding partner of SnpL as the eukaryotic transcription elongation factor, Suppressor of Ty5 (SUPT5H)/Spt5. SUPT5H is an evolutionarily conserved component of the DRB sensitivity-inducing factor complex that regulates RNA Polymerase II dependent mRNA processing and transcription elongation. Protein interaction studies showed that SnpL bound to the central Kyprides, Ouzounis, Woese motif region of SUPT5H. Ectopic expression of SnpL led to massive upregulation of host gene expression and macrophage cell death. The activity of SnpL further highlights the ability of L. pneumophila to control fundamental eukaryotic processes such as transcription that, in the case of SnpL, leads to global upregulation of host gene expression.


Subject(s)
Host-Pathogen Interactions , Legionella pneumophila/pathogenicity , Membrane Transport Proteins/metabolism , Nuclear Proteins/metabolism , RNA Polymerase II/metabolism , Transcriptional Elongation Factors/metabolism , Virulence Factors/metabolism , Animals , Cell Death , Cell Line , Cell Nucleus/chemistry , Humans , Immunoprecipitation , Macrophages/microbiology , Macrophages/physiology , Mass Spectrometry , Microscopy, Fluorescence , Protein Binding , Protein Transport
16.
Bioessays ; 39(2)2017 02.
Article in English | MEDLINE | ID: mdl-28026026

ABSTRACT

Pathogenic bacteria frequently target the endoplasmic reticulum (ER) and mitochondria in order to exploit host functions. ER-mitochondria inter-organelle communication is topologically sub-compartmentalized at mitochondria-associated ER membranes (MAMs). MAMs are specific membranous microdomains with unique regulatory functions such as lipid synthesis and trafficking, calcium homeostasis, mitochondrial morphology, inflammasome activation, autophagosome formation, and apoptosis. These important cellular processes are all modulated by pathogens to subvert host functions and promote infection, thus it is tempting to assume that pathogenic bacteria target MAMs to subvert these different pathways in their hosts. First lines of evidence that support this hypothesis come from Legionella pneumophila. This intracellular bacterium secretes an effector that exhibits sphingosine-1 phosphate lyase activity (LpSpl) that seems to target MAMs to modulate the autophagy response to infection. Here we thus propose the concept that MAMs could be targeted by pathogenic bacteria to undermine key host cellular processes.


Subject(s)
Aldehyde-Lyases/metabolism , Endoplasmic Reticulum/microbiology , Legionella pneumophila/enzymology , Membrane Microdomains/microbiology , Mitochondria/microbiology , Animals , Autophagy , Humans , Legionella pneumophila/pathogenicity , Legionnaires' Disease/microbiology , Membrane Microdomains/metabolism
17.
Proc Natl Acad Sci U S A ; 113(7): 1901-6, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26831115

ABSTRACT

Autophagy is an essential component of innate immunity, enabling the detection and elimination of intracellular pathogens. Legionella pneumophila, an intracellular pathogen that can cause a severe pneumonia in humans, is able to modulate autophagy through the action of effector proteins that are translocated into the host cell by the pathogen's Dot/Icm type IV secretion system. Many of these effectors share structural and sequence similarity with eukaryotic proteins. Indeed, phylogenetic analyses have indicated their acquisition by horizontal gene transfer from a eukaryotic host. Here we report that L. pneumophila translocates the effector protein sphingosine-1 phosphate lyase (LpSpl) to target the host sphingosine biosynthesis and to curtail autophagy. Our structural characterization of LpSpl and its comparison with human SPL reveals high structural conservation, thus supporting prior phylogenetic analysis. We show that LpSpl possesses S1P lyase activity that was abrogated by mutation of the catalytic site residues. L. pneumophila triggers the reduction of several sphingolipids critical for macrophage function in an LpSpl-dependent and -independent manner. LpSpl activity alone was sufficient to prevent an increase in sphingosine levels in infected host cells and to inhibit autophagy during macrophage infection. LpSpl was required for efficient infection of A/J mice, highlighting an important virulence role for this effector. Thus, we have uncovered a previously unidentified mechanism used by intracellular pathogens to inhibit autophagy, namely the disruption of host sphingolipid biosynthesis.


Subject(s)
Aldehyde-Lyases/metabolism , Autophagy , Legionella pneumophila/enzymology , Sphingolipids/metabolism , Aldehyde-Lyases/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Legionnaires' Disease/immunology , Mice , Protein Conformation
18.
Curr Issues Mol Biol ; 25: 81-98, 2018.
Article in English | MEDLINE | ID: mdl-28875941

ABSTRACT

Autophagy is a highly conserved catabolic process, degrading unnecessary or damaged components in the eukaryotic cell to maintain cellular homeostasis, but it is also an intrinsic cellular defence mechanism to remove invading pathogens. A crosstalk between autophagy and innate or adaptive immune responses has been recently reported, whereby autophagy influences both, innate and adaptive immunity like the production and secretion of pro-inflammatory cytokines or MHC class II antigen presentation to T cells. Pathogenic bacteria have evolved diverse strategies to manipulate autophagy, mechanisms that also impact host immune responses at different levels. Here we discuss the influence of autophagy on self-autonomous, innate and adaptive immunity and then focus on how bacterial mechanisms that shape autophagy may impact the host immune system.


Subject(s)
Autophagy/genetics , Gram-Negative Bacteria/immunology , Gram-Positive Bacteria/immunology , Host-Pathogen Interactions , Immunity, Innate , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Autophagosomes/immunology , Autophagosomes/microbiology , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Gram-Negative Bacteria/growth & development , Gram-Positive Bacteria/growth & development , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , NLR Proteins/genetics , NLR Proteins/immunology , Signal Transduction , T-Lymphocytes/microbiology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
19.
Curr Top Microbiol Immunol ; 413: 1-30, 2017.
Article in English | MEDLINE | ID: mdl-29536353

ABSTRACT

The bacterial type IV secretion systems (T4SSs) are a highly functionally and structurally diverse superfamily of secretion systems found in many species of Gram-negative and -positive bacteria. Collectively, the T4SSs can translocate DNA and monomeric and multimeric protein substrates to a variety of bacterial and eukaryotic cell types. Detailed phylogenomics analyses have established that the T4SSs evolved from ancient conjugation machines whose original functions were to disseminate mobile DNA elements within and between bacterial species. How members of the T4SS superfamily evolved to recognize and translocate specific substrate repertoires to prokaryotic or eukaryotic target cells is a fascinating question from evolutionary, biological, and structural perspectives. In this chapter, we will summarize recent findings that have shaped our current view of the biological diversity of the T4SSs. We focus mainly on two subtypes, designated as the types IVA (T4ASS) and IVB (T4BSS) systems that respectively are represented by the paradigmatic Agrobacterium tumefaciens VirB/VirD4 and Legionella pneumophila Dot/Icm T4SSs. We present current information about the composition and architectures of these representative systems. We also describe how these and a few related T4ASS and T4BSS members evolved as specialized nanomachines through acquisition of novel domains or subunits, a process that ultimately generated extensive genetic and structural mosaicism among this secretion superfamily. Finally, we present new phylogenomics information establishing that the T4BSSs are much more broadly distributed than initially envisioned.


Subject(s)
Biological Evolution , Legionella pneumophila , Agrobacterium tumefaciens , Bacterial Proteins , Bacterial Secretion Systems , Biodiversity , Type IV Secretion Systems
20.
J Infect Dis ; 215(3): 440-451, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27932612

ABSTRACT

BACKGROUND: Legionella longbeachae (Llo) and Legionella pneumophila (Lpn) are the most common pneumonia-causing agents of the genus. Although both species can be lethal to humans and are highly prevalent, little is known about the molecular pathogenesis of Llo infections. In murine models of infection, Lpn infection is self-limited, whereas Llo infection is lethal. METHODS: We used mouse macrophages, human macrophages, human epithelial cells, and mouse infections in vivo to evaluate multiple parameters of the infection. RESULTS: We determined that the Llo Dot/Icm secretion system is critical for virulence. Different than Lpn, Llo disseminates and the animals develop a severe pulmonary failure, as demonstrated by lung mechanics and blood oxygenation assays. As compared to Lpn, Llo is immunologically silent and fails to trigger the production of cytokines in human pulmonary epithelial cells and in mouse and human macrophages. Infections in Tnfr1-/-, Ifng-/-, and Il12p40-/- mice supported the participation of cytokines for the resistance phenotype. CONCLUSIONS: Both Lpn and Llo require the Dot/Icm system for pathogenesis, but the infection outcome is strikingly different. Llo is immunologically silent, highly virulent, and lethal. The differences reported herein may reflect unappreciated clinical differences in patients infected with Lpn or Llo.


Subject(s)
Legionella longbeachae/immunology , Legionella longbeachae/pathogenicity , Legionellosis/immunology , Animals , Cytokines/metabolism , Disease Resistance/immunology , Female , Humans , Legionella pneumophila/immunology , Legionellosis/microbiology , Legionellosis/pathology , Legionellosis/physiopathology , Leukocytes, Mononuclear , Lung/physiopathology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Species Specificity , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL