Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 663
Filter
2.
BMC Microbiol ; 24(1): 266, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39026145

ABSTRACT

BACKGROUND: Legionella pneumophila is a Gram-negative intracellular bacillus and is the causative agent of a severe form of pneumonia called Legionnaires' disease which accounts for 2-9% of cases of community acquired pneumonia. It produces an extremely large protein belonging to the RTX (Repeats in ToXin) family, called RtxA, and we previously reported that RtxA is transported by a dedicated type 1 secretion system (T1SS) to the cell surface. RTX proteins have been shown to participate in the virulence or biofilm formation of various bacteria, the most studied models being the pore forming hemolysin A (HlyA) of Escherichia coli and the biofilm associated protein LapA of P. fluorescens. LapA localization depends on the enzymatic release by LapD/LapG complex activity. This study aimed to elucidate the dual localization (cell surface associated or released state) of L. pneumophila RTX protein (RtxA) and whether this released versus sequestered state of RtxA plays a role in L. pneumophila virulence. RESULTS: The hereby work reveals that, in vitro, LapG periplasmic protease cleaves RtxA N-terminus in the middle of a di-alanine motif (position 108-109). Consistently, a strain lacking LapG protease maintains RtxA on the cell surface, whereas a strain lacking the c-di-GMP receptor LapD does not exhibit cell surface RtxA because of its continuous cleavage and release, as in the LapA-D-G model of Pseudomonas fluorescens. Interestingly, our data point out a key role of RtxA in enhancing the infection process of amoeba cells, regardless of its location (embedded or released); therefore, this may be the result of a secondary role of this surface protein. CONCLUSIONS: This is the first experimental identification of the cleavage site within the RTX protein family. The primary role of RtxA in Legionella is still questionable as in many other bacterial species, hence it sounds reasonable to propose a major function in biofilm formation, promoting cell aggregation when RtxA is embedded in the outer membrane and facilitating biofilm dispersion in case of RtxA release. The role of RtxA in enhancing the infection process may be a result of its action on host cells (i.e., PDI interaction or pore-formation), and independently of its status (embedded or released).


Subject(s)
Bacterial Proteins , Legionella pneumophila , Legionella pneumophila/pathogenicity , Legionella pneumophila/metabolism , Legionella pneumophila/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Virulence , Bacterial Toxins/metabolism , Biofilms/growth & development , Legionnaires' Disease/microbiology , Type I Secretion Systems/metabolism , Type I Secretion Systems/genetics , Cell Membrane/metabolism
3.
Nat Commun ; 15(1): 5953, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009586

ABSTRACT

The intracellular bacterial pathogen Legionella pneumophila modulates host cell functions by secreting multiple effectors with diverse biochemical activities. In particular, effectors of the SidE family interfere with host protein ubiquitination in a process that involves production of phosphoribosyl ubiquitin (PR-Ub). Here, we show that effector LnaB converts PR-Ub into ADP-ribosylated ubiquitin, which is further processed to ADP-ribose and functional ubiquitin by the (ADP-ribosyl)hydrolase MavL, thus maintaining ubiquitin homeostasis in infected cells. Upon being activated by actin, LnaB also undergoes self-AMPylation on tyrosine residues. The activity of LnaB requires a motif consisting of Ser, His and Glu (SHxxxE) present in a large family of toxins from diverse bacterial pathogens. Thus, our study sheds light on the mechanisms by which a pathogen maintains ubiquitin homeostasis and identifies a family of enzymes capable of protein AMPylation.


Subject(s)
Bacterial Proteins , Homeostasis , Legionella pneumophila , Ubiquitin , Ubiquitination , Ubiquitin/metabolism , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , ADP-Ribosylation , Host-Pathogen Interactions , Adenosine Diphosphate Ribose/metabolism , Legionnaires' Disease/metabolism , Legionnaires' Disease/microbiology , HEK293 Cells , Actins/metabolism , HeLa Cells
4.
Curr Opin Microbiol ; 80: 102495, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908045

ABSTRACT

Legionella species are Gram-negative intracellular bacteria that evolved in soil and freshwater environments, where they infect and replicate within various unicellular protozoa. The primary virulence factor of Legionella is the expression of a type IV secretion system (T4SS), which contributes to the translocation of effector proteins that subvert biological processes of the host cells. Because of its evolution in unicellular organisms, T4SS effector proteins are not adapted to subvert specific mammalian signaling pathways and immunity. Consequently, Legionella pneumophila has emerged as an interesting infection model for investigating immune responses against pathogenic bacteria in multicellular organisms. This review highlights recent advances in our understanding of mammalian innate immunity derived from studies involving L. pneumophila. This includes recent insights into inflammasome-mediated mechanisms restricting bacterial replication in macrophages, mechanisms inducing cell death in response to infection, induction of effector-triggered immunity, activation of specific pulmonary cell types in mammalian lungs, and the protective role of recruiting monocyte-derived cells to infected lungs.


Subject(s)
Immunity, Innate , Legionella pneumophila , Legionnaires' Disease , Legionella pneumophila/immunology , Legionella pneumophila/pathogenicity , Humans , Animals , Legionnaires' Disease/immunology , Legionnaires' Disease/microbiology , Phagocytes/immunology , Phagocytes/microbiology , Type IV Secretion Systems/immunology , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Monocytes/immunology , Monocytes/microbiology , Virulence Factors/immunology , Virulence Factors/metabolism , Macrophages/immunology , Macrophages/microbiology , Host-Pathogen Interactions/immunology
5.
Nature ; 631(8020): 393-401, 2024 07.
Article in English | MEDLINE | ID: mdl-38776962

ABSTRACT

AMPylation is a post-translational modification in which AMP is added to the amino acid side chains of proteins1,2. Here we show that, with ATP as the ligand and actin as the host activator, the effector protein LnaB of Legionella pneumophila exhibits AMPylase activity towards the phosphoryl group of phosphoribose on PRR42-Ub that is generated by the SidE family of effectors, and deubiquitinases DupA and DupB in an E1- and E2-independent ubiquitination process3-7. The product of LnaB is further hydrolysed by an ADP-ribosylhydrolase, MavL, to Ub, thereby preventing the accumulation of PRR42-Ub and ADPRR42-Ub and protecting canonical ubiquitination in host cells. LnaB represents a large family of AMPylases that adopt a common structural fold, distinct from those of the previously known AMPylases, and LnaB homologues are found in more than 20 species of bacterial pathogens. Moreover, LnaB also exhibits robust phosphoryl AMPylase activity towards phosphorylated residues and produces unique ADPylation modifications in proteins. During infection, LnaB AMPylates the conserved phosphorylated tyrosine residues in the activation loop of the Src family of kinases8,9, which dampens downstream phosphorylation signalling in the host. Structural studies reveal the actin-dependent activation and catalytic mechanisms of the LnaB family of AMPylases. This study identifies, to our knowledge, an unprecedented molecular regulation mechanism in bacterial pathogenesis and protein phosphorylation.


Subject(s)
Adenosine Monophosphate , Bacterial Proteins , Legionella pneumophila , Phosphotyrosine , Signal Transduction , Humans , Actins/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , ADP-Ribosylation , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Hydrolysis , Legionella pneumophila/enzymology , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Ligands , Models, Molecular , N-Glycosyl Hydrolases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Tyrosine/metabolism , Tyrosine/chemistry , Ubiquitin/metabolism , Ubiquitination , Deubiquitinating Enzymes/metabolism , Protein Folding , Phosphotyrosine/chemistry , Phosphotyrosine/metabolism
6.
mBio ; 15(6): e0071024, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38682908

ABSTRACT

The causative agent of Legionnaires' disease, Legionella pneumophila, is an environmental bacterium, that replicates in macrophages, parasitizes amoeba, and forms biofilms. L. pneumophila employs the Legionella quorum sensing (Lqs) system and the transcription factor LvbR to control various bacterial traits, including virulence and biofilm architecture. LvbR negatively regulates the nitric oxide (NO) receptor Hnox1, linking quorum sensing to NO signaling. Here, we assessed the response of L. pneumophila to NO and investigated bacterial receptors underlying this process. Chemical NO donors, such as dipropylenetriamine (DPTA) NONOate and sodium nitroprusside (SNP), delayed and reduced the expression of the promoters for flagellin (PflaA) and the 6S small regulatory RNA (P6SRNA). Marker-less L. pneumophila mutant strains lacking individual (Hnox1, Hnox2, or NosP) or all three NO receptors (triple knockout, TKO) grew like the parental strain in media. However, in the TKO strain, the reduction of PflaA expression by DPTA NONOate was less pronounced, suggesting that the NO receptors are implicated in NO signaling. In the ΔnosP mutant, the lvbR promoter was upregulated, indicating that NosP negatively regulates LvbR. The single and triple NO receptor mutant strains were impaired for growth in phagocytes, and phenotypic heterogeneity of non-growing/growing bacteria in amoebae was regulated by the NO receptors. The single NO receptor and TKO mutant strains showed altered biofilm architecture and lack of response of biofilms to NO. In summary, we provide evidence that L. pneumophila regulates virulence, intracellular phenotypic heterogeneity, and biofilm formation through NO and three functionally non-redundant NO receptors, Hnox1, Hnox2, and NosP. IMPORTANCE: The highly reactive diatomic gas molecule nitric oxide (NO) is produced by eukaryotes and bacteria to promote short-range and transient signaling within and between neighboring cells. Despite its importance as an inter-kingdom and intra-bacterial signaling molecule, the bacterial response and the underlying components of the signaling pathways are poorly characterized. The environmental bacterium Legionella pneumophila forms biofilms and replicates in protozoan and mammalian phagocytes. L. pneumophila harbors three putative NO receptors, one of which crosstalks with the Legionella quorum sensing (Lqs)-LvbR network to regulate various bacterial traits, including virulence and biofilm architecture. In this study, we used pharmacological, genetic, and cell biological approaches to assess the response of L. pneumophila to NO and to demonstrate that the putative NO receptors are implicated in NO detection, bacterial replication in phagocytes, intracellular phenotypic heterogeneity, and biofilm formation.


Subject(s)
Bacterial Proteins , Biofilms , Gene Expression Regulation, Bacterial , Legionella pneumophila , Nitric Oxide , Signal Transduction , Biofilms/growth & development , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Legionella pneumophila/physiology , Legionella pneumophila/metabolism , Nitric Oxide/metabolism , Virulence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Phenotype , Macrophages/microbiology , Quorum Sensing
7.
Cell Rep ; 43(4): 114033, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38568811

ABSTRACT

Small GTPases of the Ras subfamily are best known for their role as proto-oncoproteins, while their function during microbial infection has remained elusive. Here, we show that Legionella pneumophila hijacks the small GTPase NRas to the Legionella-containing vacuole (LCV) surface. A CRISPR interference screen identifies a single L. pneumophila effector, DenR (Lpg1909), required for this process. Recruitment is specific for NRas, while its homologs KRas and HRas are excluded from LCVs. The C-terminal hypervariable tail of NRas is sufficient for recruitment, and interference with either NRas farnesylation or S-acylation sites abrogates recruitment. Intriguingly, we detect markers of active NRas signaling on the LCV, suggesting it acts as a signaling platform. Subsequent phosphoproteomics analyses show that DenR rewires the host NRas signaling landscape, including dampening of the canonical mitogen-activated protein kinase pathway. These results provide evidence for L. pneumophila targeting NRas and suggest a link between NRas GTPase signaling and microbial infection.


Subject(s)
Bacterial Proteins , GTP Phosphohydrolases , Legionella pneumophila , MAP Kinase Signaling System , Membrane Proteins , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , GTP Phosphohydrolases/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Down-Regulation , HEK293 Cells , Legionnaires' Disease/microbiology , Legionnaires' Disease/metabolism , Vacuoles/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/genetics
8.
Elife ; 112022 01 27.
Article in English | MEDLINE | ID: mdl-35084332

ABSTRACT

The intracellular pathogen Legionella pneumophila delivers more than 330 effectors into host cells by its Dot/Icm secretion system. Those effectors direct the biogenesis of the Legionella-containing vacuole (LCV) that permits its intracellular survival and replication. It has long been documented that the LCV is associated with mitochondria and a number of Dot/Icm effectors have been shown to target to this organelle. Yet, the biochemical function and host cell target of most of these effectors remain unknown. Here, we found that the Dot/Icm substrate Ceg3 (Lpg0080) is a mono-ADP-ribosyltransferase that localizes to the mitochondria in host cells where it attacks ADP/ATP translocases by ADP-ribosylation, and blunts their ADP/ATP exchange activity. The modification occurs on the second arginine residue in the -RRRMMM- element, which is conserved among all known ADP/ATP carriers from different organisms. Our results reveal modulation of host energy metabolism as a virulence mechanism for L. pneumophila.


Subject(s)
Energy Metabolism/physiology , Legionella pneumophila/pathogenicity , Mitochondrial ADP, ATP Translocases/metabolism , Vacuoles/microbiology , ADP-Ribosylation/physiology , HEK293 Cells , HeLa Cells , Humans , Legionnaires' Disease/metabolism , Legionnaires' Disease/microbiology , Vacuoles/physiology , Virulence
9.
J Leukoc Biol ; 112(2): 257-271, 2022 08.
Article in English | MEDLINE | ID: mdl-34826345

ABSTRACT

Macrophages use an array of innate immune sensors to detect intracellular pathogens and to tailor effective antimicrobial responses. In addition, extrinsic activation with the cytokine IFN-γ is often required as well to tip the scales of the host-pathogen balance toward pathogen restriction. However, little is known about how host-pathogen sensing impacts the antimicrobial IFN-γ-activated state. It was observed that in the absence of IRF3, a key downstream component of pathogen sensing pathways, IFN-γ-primed macrophages more efficiently restricted the intracellular bacterium Legionella pneumophila and the intracellular protozoan parasite Trypanosoma cruzi. This effect did not require IFNAR, the receptor for Type I IFNs known to be induced by IRF3, nor the sensing adaptors MyD88/TRIF, MAVS, or STING. This effect also did not involve differential activation of STAT1, the major signaling protein downstream of both Type 1 and Type 2 IFN receptors. IRF3-deficient macrophages displayed a significantly altered IFN-γ-induced gene expression program, with up-regulation of microbial restriction factors such as Nos2. Finally, we found that IFN-γ-primed but not unprimed macrophages largely excluded the activated form of IRF3 from the nucleus following bacterial infection. These data are consistent with a relationship of mutual inhibition between IRF3 and IFN-γ-activated programs, possibly as a component of a partially reversible mechanism for modulating the activity of potent innate immune effectors (such as Nos2) in the context of intracellular infection.


Subject(s)
Interferon Regulatory Factor-3 , Interferon-gamma , Legionella pneumophila , Macrophages , Trypanosoma cruzi , Adaptor Proteins, Signal Transducing/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-gamma/metabolism , Legionella pneumophila/pathogenicity , Macrophages/metabolism , Nitric Oxide Synthase Type II/metabolism , Trypanosoma cruzi/pathogenicity
10.
Eur J Histochem ; 65(3)2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34468107

ABSTRACT

In this study, we studied the long-term proliferation trajectory of myeloid-derived suppressor cells (MDSCs) in murine sepsis model and investigated whether swertianolin could modulate the immunosuppressive function of MDSCs. A murine sepsis model was established by cecal ligation and perforation (CLP), according to the Minimum Quality Threshold in Pre-Clinical Sepsis Studies (MQTiPSS) guidelines. The bone marrow and spleen of the mice were collected at 24 h, 72 h, 7 and 15 d after sepsis induction. The proportions of monocytic-MDSCs (M-MDSCs; CD11b+LY6G-LY6Chi) and granulocytic-MDSCs (G-MDSC, CD11b+ Ly6G+ Ly6Clow) were analyzed by flow cytometry. Then, we have investigated whether swertianolin could modulate the immunosuppressive function of MDSCs in in vitro experiments. G-MDSCs and M-MDSCs increased acutely after sepsis with high levels sustained over a long period of time. G-MDSCs were the main subtype identified in the murine model of sepsis with polymicrobial peritonitis. Furthermore, it was found that swertianolin reduced significantly interleukin-10 (IL-10), nitric oxide (NO), reactive oxygen species (ROS), and arginase production in MDSCs, while reducing MDSC proliferation and promoting MDSC differentiation into dendritic cells. Swertianolin also improved T-cell activity by blocking the immunosuppressive effect of MDSCs. Both subsets of MDSCs significantly increased in the bone marrow and spleen of the mice with sepsis, with G-MDSCs being the main subtype identified. Swertianolin effectively regulated the functions of MDSCs and reduced immune suppression.


Subject(s)
Immune Tolerance/drug effects , Iridoid Glucosides/pharmacology , Myeloid-Derived Suppressor Cells/drug effects , Sepsis/metabolism , Xanthones/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dendritic Cells/metabolism , Legionella pneumophila/pathogenicity , Lung/drug effects , Lung/pathology , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/classification , Myeloid-Derived Suppressor Cells/metabolism , Peritonitis/metabolism , Peritonitis/pathology , Sepsis/pathology
11.
PLoS One ; 16(4): e0250364, 2021.
Article in English | MEDLINE | ID: mdl-33886659

ABSTRACT

Legionellosis is an infection acquired through inhalation of aerosols that are contaminated with environmental bacteria Legionella spp. The bacteria require warm temperature for proliferation in bodies of water and moist soil. The legionellosis incidence in the United States has been rising rapidly in the past two decades without a clear explanation. In the meantime, the US has recorded consecutive years of above-norm temperature since 1997 and precipitation surplus since 2008. The present study analyzed the legionellosis incidence in the US during the 20-year period of 1999 to 2018 and correlated with concurrent temperature, precipitation, solar ultraviolet B (UVB) radiation, and vehicle mileage data. The age-adjusted legionellosis incidence rates rose exponentially from 0.40/100,000 in 1999 (with 1108 cases) to 2.69/100,000 in 2018 (with 9933 cases) at a calculated annual increase of 110%. In regression analyses, the rise correlated with an increase in vehicle miles driven and with temperature and precipitation levels that have been above the 1901-2000 mean since 1997 and 2008, respectively, suggesting more road exposure to traffic-generated aerosols and promotive effects of anomalous climate. Remarkably, the regressions with cumulative anomalies of temperature and precipitation were robust (R2 ≥ 0.9145, P ≤ 4.7E-11), implying possible changes to microbial ecology in the terrestrial and aquatic environments. An interactive synergy between annual precipitation and vehicle miles was also found in multiple regressions. Meanwhile, the bactericidal UVB radiation has been decreasing, which also contributed to the rising incidence in an inverse correlation. The 2018 legionellosis incidence peak corresponded to cumulative effects of the climate anomalies, vast vehicle miles (3,240 billion miles, 15904 km per capita), record high precipitation (880.1 mm), near record low UVB radiation (7488 kJ/m2), and continued above-norm temperature (11.96°C). These effects were examined and demonstrated in California, Florida, New Jersey, Ohio, and Wisconsin, states that represent diverse incidence rates and climates. The incidence and above-norm temperature both rose most in cold Wisconsin. These results suggest that warming temperature and precipitation surplus have likely elevated the density of Legionella bacteria in the environment, and together with road exposure explain the rapidly rising incidence of legionellosis in the United States. These trends are expected to continue, warranting further research and efforts to prevent infection.


Subject(s)
Global Warming , Hot Temperature , Legionella pneumophila/pathogenicity , Legionellosis/epidemiology , Sunlight , Ultraviolet Rays , Adolescent , Adult , Aged , Centers for Disease Control and Prevention, U.S. , Child , Child, Preschool , Female , Humans , Incidence , Infant , Infant, Newborn , Legionellosis/microbiology , Male , Middle Aged , Rural Population , United States/epidemiology , Urban Population , Young Adult
12.
PLoS Pathog ; 17(3): e1009437, 2021 03.
Article in English | MEDLINE | ID: mdl-33760868

ABSTRACT

Legionella pneumophila (L. pneumophila) is a gram-negative bacterium that replicates in a compartment that resembles the host endoplasmic reticulum (ER). To create its replicative niche, L. pneumophila manipulates host membrane traffic and fusion machineries. Bacterial proteins called Legionella effectors are translocated into the host cytosol and play a crucial role in these processes. In an early stage of infection, Legionella subverts ER-derived vesicles (ERDVs) by manipulating GTPase Rab1 to facilitate remodeling of the Legionella-containing vacuole (LCV). Subsequently, the LCV associates with the ER in a mechanism that remains elusive. In this study, we show that L. pneumophila recruits GTPases Rab33B and Rab6A, which regulate vesicle trafficking from the Golgi to the ER, to the LCV to promote the association of LCV with the ER. We found that recruitment of Rab6A to the LCV depends on Rab33B. Legionella effector SidE family proteins, which phosphoribosyl-ubiquitinate Rab33B, were found to be necessary for the recruitment of Rab33B to the LCV. Immunoprecipitation experiments revealed that L. pneumophila facilitates the interaction of Rab6 with ER-resident SNAREs comprising syntaxin 18, p31, and BNIP1, but not tethering factors including NAG, RINT-1, and ZW10, which are normally required for syntaxin 18-mediated fusion of Golgi-derived vesicles with the ER. Our results identified a Rab33B-Rab6A cascade on the LCV and the interaction of Rab6 with ER-resident SNARE proteins for the association of LCV with the ER and disclosed the unidentified physiological role of SidE family proteins.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Legionella pneumophila/pathogenicity , Legionnaires' Disease/metabolism , Vacuoles/microbiology , Bacterial Proteins/metabolism , Endoplasmic Reticulum/microbiology , Golgi Apparatus/microbiology , HEK293 Cells , HeLa Cells , Humans , Legionella pneumophila/metabolism , Protein Transport/physiology , Vacuoles/metabolism
13.
Pathog Dis ; 79(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33734371

ABSTRACT

The human pulmonary environment is complex, containing a matrix of cells, including fibroblasts, epithelial cells, interstitial macrophages, alveolar macrophages and neutrophils. When confronted with foreign material or invading pathogens, these cells mount a robust response. Nevertheless, many bacterial pathogens with an intracellular lifecycle stage exploit this environment for replication and survival. These include, but are not limited to, Coxiella burnetii, Legionella pneumophila, Yersinia pestis, Mycobacterium tuberculosis and Staphylococcus aureus. Currently, few human disease-relevant model systems exist for studying host-pathogen interactions during these bacterial infections in the lung. Here, we present two novel infection platforms, human alveolar macrophages (hAMs) and human precision-cut lung slices (hPCLS), along with an up-to-date synopsis of research using said models. Additionally, alternative uses for these systems in the absence of pathogen involvement are presented, such as tissue banking and further characterization of the human lung environment. Overall, hAMs and hPCLS allow novel human disease-relevant investigations that other models, such as cell lines and animal models, cannot completely provide.


Subject(s)
Bacterial Infections/microbiology , Host-Pathogen Interactions/immunology , Lung Diseases/microbiology , Lung/microbiology , Macrophages, Alveolar/microbiology , Models, Biological , Bacterial Infections/immunology , Bacterial Infections/pathology , Coxiella burnetii/growth & development , Coxiella burnetii/immunology , Coxiella burnetii/pathogenicity , Humans , Legionella pneumophila/growth & development , Legionella pneumophila/immunology , Legionella pneumophila/pathogenicity , Lung/immunology , Lung/pathology , Lung Diseases/immunology , Lung Diseases/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Microtomy , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Primary Cell Culture , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Tissue Banks , Tissue Culture Techniques , Yersinia pestis/growth & development , Yersinia pestis/immunology , Yersinia pestis/pathogenicity
14.
Korean J Parasitol ; 59(1): 67-75, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33684989

ABSTRACT

Legionella pneumophila is an opportunistic pathogen that survives and proliferates within protists such as Acanthamoeba spp. in environment. However, intracellular pathogenic endosymbiosis and its implications within Acanthamoeba spp. remain poorly understood. In this study, RNA sequencing analysis was used to investigate transcriptional changes in A. castellanii in response to L. pneumophila infection. Based on RNA sequencing data, we identified 1,211 upregulated genes and 1,131 downregulated genes in A. castellanii infected with L. pneumophila for 12 hr. After 24 hr, 1,321 upregulated genes and 1,379 downregulated genes were identified. Gene ontology (GO) analysis revealed that L. pneumophila endosymbiosis enhanced hydrolase activity, catalytic activity, and DNA binding while reducing oxidoreductase activity in the molecular function (MF) domain. In particular, multiple genes associated with the GO term 'integral component of membrane' were downregulated during endosymbiosis. The endosymbiont also induced differential expression of various methyltransferases and acetyltransferases in A. castellanii. Findings herein are may significantly contribute to understanding endosymbiosis of L. pneumophila within A. castellanii.


Subject(s)
Acanthamoeba castellanii/genetics , Acanthamoeba castellanii/microbiology , Genes, Protozoan/genetics , Legionella pneumophila/physiology , Symbiosis/genetics , Transcriptome/genetics , Acanthamoeba castellanii/enzymology , Acetyltransferases/genetics , Acetyltransferases/metabolism , Catalysis , Gene Ontology , Hydrolases/metabolism , Legionella pneumophila/pathogenicity , Methyltransferases/genetics , Methyltransferases/metabolism , Oxidoreductases/metabolism , Sequence Analysis, RNA , Transcription, Genetic
15.
mBio ; 12(1)2021 02 09.
Article in English | MEDLINE | ID: mdl-33563829

ABSTRACT

Diversion of the Legionella pneumophila-containing vacuole (LCV) from the host endosomal-lysosomal degradation pathway is one of the main virulence features essential for manifestation of Legionnaires' pneumonia. Many of the ∼350 Dot/Icm-injected effectors identified in L. pneumophila have been shown to interfere with various host pathways and processes, but no L. pneumophila effector has ever been identified to be indispensable for lysosomal evasion. While most single effector mutants of L. pneumophila do not exhibit a defective phenotype within macrophages, we show that the MavE effector is essential for intracellular growth of L. pneumophila in human monocyte-derived macrophages (hMDMs) and amoebae and for intrapulmonary proliferation in mice. The mavE null mutant fails to remodel the LCV with endoplasmic reticulum (ER)-derived vesicles and is trafficked to the lysosomes where it is degraded, similar to formalin-killed bacteria. During infection of hMDMs, the MavE effector localizes to the poles of the LCV membrane. The crystal structure of MavE, resolved to 1.8 Å, reveals a C-terminal transmembrane helix, three copies of tyrosine-based sorting motifs, and an NPxY eukaryotic motif, which binds phosphotyrosine-binding domains present on signaling and adaptor eukaryotic proteins. Two point mutations within the NPxY motif result in attenuation of L. pneumophila in both hMDMs and amoeba. The substitution defects of P78 and D64 are associated with failure of vacuoles harboring the mutant to be remodeled by the ER and results in fusion of the vacuole to the lysosomes leading to bacterial degradation. Therefore, the MavE effector of L. pneumophila is indispensable for phagosome biogenesis and lysosomal evasion.IMPORTANCE Intracellular proliferation of Legionella pneumophila within a vacuole in human alveolar macrophages is essential for manifestation of Legionnaires' pneumonia. Intravacuolar growth of the pathogen is totally dependent on remodeling the L. pneumophila-containing vacuole (LCV) by the ER and on its evasion of the endosomal-lysosomal degradation pathway. The pathogen has evolved to inject ∼350 protein effectors into the host cell where they modulate various host processes, but no L. pneumophila effector has ever been identified to be indispensable for lysosomal evasion. We show that the MavE effector localizes to the poles of the LCV membrane and is essential for lysosomal evasion and intracellular growth of L. pneumophila and for intrapulmonary proliferation in mice. The crystal structure of MavE shows an NPxY eukaryotic motif essential for ER-mediated remodeling and lysosomal evasion by the LCV. Therefore, the MavE effector of L. pneumophila is indispensable for phagosome biogenesis and lysosomal evasion.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Lysosomes/microbiology , Macrophages/microbiology , Animals , Bacterial Proteins/chemistry , Cells, Cultured , Crystallization , Host-Pathogen Interactions , Humans , Mice , Protein Transport , Vacuoles/microbiology , Virulence
16.
Commun Biol ; 4(1): 157, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542442

ABSTRACT

Catalytically inactive dCas9 imposes transcriptional gene repression by sterically precluding RNA polymerase activity at a given gene to which it was directed by CRISPR (cr)RNAs. This gene silencing technology, known as CRISPR interference (CRISPRi), has been employed in various bacterial species to interrogate genes, mostly individually or in pairs. Here, we developed a multiplex CRISPRi platform in the pathogen Legionella pneumophila capable of silencing up to ten genes simultaneously. Constraints on precursor-crRNA expression were overcome by combining a strong promoter with a boxA element upstream of a CRISPR array. Using crRNAs directed against virulence protein-encoding genes, we demonstrated that CRISPRi is fully functional not only during growth in axenic media, but also during macrophage infection, and that gene depletion by CRISPRi recapitulated the growth defect of deletion strains. By altering the position of crRNA-encoding spacers within the CRISPR array, our platform achieved the gradual depletion of targets that was mirrored by the severity in phenotypes. Multiplex CRISPRi thus holds great promise for probing large sets of genes in bulk in order to decipher virulence strategies of L. pneumophila and other bacterial pathogens.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Silencing , Legionella pneumophila/genetics , Virulence Factors/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9/metabolism , Gene Expression Regulation, Bacterial , Humans , Legionella pneumophila/growth & development , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Proof of Concept Study , U937 Cells , Virulence/genetics , Virulence Factors/metabolism
17.
mSphere ; 6(1)2021 01 06.
Article in English | MEDLINE | ID: mdl-33408238

ABSTRACT

Bryan D. Bryson works in the field of biological engineering with a specific interest in host-mycobacterium interactions. In this mSphere of Influence article, he reflects on how "IRG1 and inducible nitric oxide synthase act redundantly with other interferon-gamma-induced factors to restrict intracellular replication of Legionella pneumophila" by Price and colleagues (J. V. Price, D. Russo, D. X. Ji, R. A. Chavez, et al., mBio 10:e02629-19, 2019, https://doi.org/10.1128/mBio.02629-19) made an impact on him by reinforcing the complexity of intracellular pathogen control.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate/genetics , Interferon-gamma/immunology , Legionella pneumophila/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Legionella pneumophila/pathogenicity , Narration , Nitric Oxide Synthase Type II , Phagosomes/immunology , Phagosomes/microbiology , Protein Transport
18.
BMC Infect Dis ; 21(1): 32, 2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33413170

ABSTRACT

BACKGROUND: Legionnaire's disease is one of the major causes of community-acquired pneumonia and is occasionally complicated by neurological symptoms. However, reports of ocular lesions due to Legionnaire's disease are limited. CASE PRESENTATION: We report the case of a patient with Legionnaire's disease presenting as bilateral central scotomata due to retinal lesions. The patient consulted due to fever and bilateral central scotomata, as well as other extrapulmonary symptoms. Optical coherence tomography (OCT) showed bilateral accumulations of fluid under the retina, and the patient was diagnosed with bilateral exudative retinal detachment. Later, Legionnaire's disease was confirmed by pulmonary infiltrates on chest imaging and positive urinary antigen for Legionella pneumophila. After administration of antibiotics, the bilateral central scotomata and bilateral subretinal fluid accumulations completely resolved, as did the other extrapulmonary symptoms and the pulmonary infiltrates. Thus, the bilateral central scotomata due to exudative retinal detachment were thought to be caused by Legionnaire's disease. CONCLUSIONS: This case demonstrates that Legionnaire's disease can present as bilateral central scotomata. We may consider the possibility of extrapulmonary involvement complicating Legionnaire's disease when we encounter bilateral ocular lesions in patients with fever and pneumonia.


Subject(s)
Legionnaires' Disease/diagnosis , Legionnaires' Disease/physiopathology , Scotoma/etiology , Anti-Bacterial Agents/therapeutic use , Humans , Legionella pneumophila/immunology , Legionella pneumophila/pathogenicity , Legionnaires' Disease/drug therapy , Legionnaires' Disease/etiology , Male , Middle Aged , Pneumonia, Bacterial/diagnosis , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/physiopathology , Scotoma/diagnosis , Scotoma/pathology , Tomography, Optical Coherence
19.
Cell Microbiol ; 23(5): e13313, 2021 05.
Article in English | MEDLINE | ID: mdl-33491325

ABSTRACT

ProA is a secreted zinc metalloprotease of Legionella pneumophila causing lung damage in animal models of Legionnaires' disease. Here we demonstrate that ProA promotes infection of human lung tissue explants (HLTEs) and dissect the contribution to cell type specific replication and extracellular virulence mechanisms. For the first time, we reveal that co-incubation of HLTEs with purified ProA causes a significant increase of the alveolar septal thickness. This destruction of connective tissue fibres was further substantiated by collagen IV degradation assays. The moderate attenuation of a proA-negative mutant in A549 epithelial cells and THP-1 macrophages suggests that effects of ProA in tissue mainly result from extracellular activity. Correspondingly, ProA contributes to dissemination and serum resistance of the pathogen, which further expands the versatile substrate spectrum of this thermolysin-like protease. The crystal structure of ProA at 1.48 Å resolution showed high congruence to pseudolysin of Pseudomonas aeruginosa, but revealed deviations in flexible loops, the substrate binding pocket S1 ' and the repertoire of cofactors, by which ProA can be distinguished from respective homologues. In sum, this work specified virulence features of ProA at different organisational levels by zooming in from histopathological effects in human lung tissue to atomic details of the protease substrate determination.


Subject(s)
Bacterial Proteins/metabolism , Collagen Type IV/metabolism , Legionella pneumophila/enzymology , Legionella pneumophila/pathogenicity , Lung/microbiology , Metalloendopeptidases/metabolism , Pulmonary Alveoli/pathology , Virulence Factors/metabolism , A549 Cells , Bacterial Proteins/chemistry , Blood Bactericidal Activity , Humans , Legionella pneumophila/growth & development , Lung/pathology , Metalloendopeptidases/chemistry , Proteolysis , Pulmonary Alveoli/metabolism , THP-1 Cells , Virulence , Virulence Factors/chemistry
20.
J Biol Chem ; 296: 100301, 2021.
Article in English | MEDLINE | ID: mdl-33476647

ABSTRACT

ADP-ribosyltransferases (ARTs) are a widespread superfamily of enzymes frequently employed in pathogenic strategies of bacteria. Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaire's disease, has acquired over 330 translocated effectors that showcase remarkable biochemical and structural diversity. However, the ART effectors that influence L. pneumophila have not been well defined. Here, we took a bioinformatic approach to search the Legionella effector repertoire for additional divergent members of the ART superfamily and identified an ART domain in Legionella pneumophila gene0181, which we hereafter refer to as Legionella ADP-Ribosyltransferase 1 (Lart1) (Legionella ART 1). We show that L. pneumophila Lart1 targets a specific class of 120-kDa NAD+-dependent glutamate dehydrogenase (GDH) enzymes found in fungi and protists, including many natural hosts of Legionella. Lart1 targets a conserved arginine residue in the NAD+-binding pocket of GDH, thereby blocking oxidative deamination of glutamate. Therefore, Lart1 could be the first example of a Legionella effector which directly targets a host metabolic enzyme during infection.


Subject(s)
ADP Ribose Transferases/chemistry , Bacterial Proteins/chemistry , Glutamate Dehydrogenase/chemistry , Legionella pneumophila/genetics , ADP Ribose Transferases/genetics , ADP Ribose Transferases/metabolism , ADP-Ribosylation , Amino Acid Sequence , Amoeba/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Deamination , Escherichia coli/genetics , Escherichia coli/metabolism , Fungi , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutamate Dehydrogenase/genetics , Glutamate Dehydrogenase/metabolism , Host-Pathogen Interactions , Kinetics , Legionella pneumophila/enzymology , Legionella pneumophila/pathogenicity , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL