Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Infect Immun ; 91(1): e0019922, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36537791

ABSTRACT

MicroRNAs (miRNAs), a class of small noncoding RNAs, are critical to gene regulation in eukaryotes. They are involved in modulating a variety of physiological processes, including the host response to intracellular infections. Little is known about miRNA functions during infection by Coxiella burnetii, the causative agent of human Q fever. This bacterial pathogen establishes a large replicative vacuole within macrophages by manipulating host processes such as apoptosis and autophagy. We investigated miRNA expression in C. burnetii-infected macrophages and identified several miRNAs that were down- or upregulated during infection. We further explored the functions of miR-143-3p, an miRNA whose expression is downregulated in macrophages infected with C. burnetii, and show that increasing the abundance of this miRNA in human cells results in increased apoptosis and reduced autophagy-conditions that are unfavorable to C. burnetii intracellular growth. In sum, this study demonstrates that C. burnetii infection elicits a robust miRNA-based host response, and because miR-143-3p promotes apoptosis and inhibits autophagy, downregulation of miR-143-3p expression during C. burnetii infection likely benefits the pathogen.


Subject(s)
Coxiella burnetii , MicroRNAs , Q Fever , Humans , Coxiella burnetii/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Host-Pathogen Interactions/genetics , Q Fever/genetics , Q Fever/metabolism , Macrophages/microbiology , Vacuoles/microbiology
2.
Infect Immun ; 88(7)2020 06 22.
Article in English | MEDLINE | ID: mdl-32284364

ABSTRACT

Coxiella burnetii is the causative agent of human Q fever, eliciting symptoms that range from acute fever and fatigue to chronic fatal endocarditis. C. burnetii is a Gram-negative intracellular bacterium that replicates within an acidic lysosome-like parasitophorous vacuole (PV) in human macrophages. During intracellular growth, C. burnetii delivers bacterial proteins directly into the host cytoplasm using a Dot/Icm type IV secretion system (T4SS). Multiple T4SS effectors localize to and/or disrupt the endoplasmic reticulum (ER) and secretory transport, but their role in infection is unknown. During microbial infection, unfolded nascent proteins may exceed the folding capacity of the ER, activating the unfolded protein response (UPR) and restoring the ER to its normal physiological state. A subset of intracellular pathogens manipulates the UPR to promote survival and replication in host cells. In this study, we investigated the impact of C. burnetii infection on activation of the three arms of the UPR. An inhibitor of the UPR antagonized PV expansion in macrophages, indicating this process is needed for bacterial replication niche formation. Protein kinase RNA-like ER kinase (PERK) signaling was activated during infection, leading to increased levels of phosphorylated eukaryotic initiation factor α, which was required for C. burnetii growth. Increased production and nuclear translocation of the transcription factor ATF4 also occurred, which normally drives expression of the proapoptotic C/EBP homologous protein (CHOP). CHOP protein production increased during infection; however, C. burnetii actively prevented CHOP nuclear translocation and downstream apoptosis in a T4SS-dependent manner. The results collectively demonstrate interplay between C. burnetii and specific components of the eIF2α signaling cascade to parasitize human macrophages.


Subject(s)
Coxiella burnetii/physiology , Eukaryotic Initiation Factor-2/metabolism , Host-Pathogen Interactions , Q Fever/metabolism , Q Fever/microbiology , Activating Transcription Factor 6/metabolism , Bacterial Secretion Systems , Histones/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Protein Transport , Transcription Factor CHOP/metabolism
3.
Infect Immun ; 87(5)2019 03.
Article in English | MEDLINE | ID: mdl-30833339

ABSTRACT

Human Q fever is caused by the intracellular bacterial pathogen Coxiella burnetii Q fever presents with acute flu-like and pulmonary symptoms or can progress to chronic, severe endocarditis. After human inhalation, C. burnetii is engulfed by alveolar macrophages and transits through the phagolysosomal maturation pathway, resisting the acidic pH of lysosomes to form a parasitophorous vacuole (PV) in which to replicate. Previous studies showed that C. burnetii replicates efficiently in primary human alveolar macrophages (hAMs) in ex vivo human lung tissue. Although C. burnetii replicates in most cell types in vitro, the pathogen does not grow in non-hAM cells of human lung tissue. In this study, we investigated the interaction between C. burnetii and other pulmonary cell types apart from the lung environment. C. burnetii formed a prototypical PV and replicated efficiently in human pulmonary fibroblasts and in airway, but not alveolar, epithelial cells. Atypical PV expansion in alveolar epithelial cells was attributed in part to defective recruitment of autophagy-related proteins. Further assessment of the C. burnetii growth niche showed that macrophages mounted a robust interleukin 8 (IL-8), neutrophil-attracting response to C. burnetii and ultimately shifted to an M2-polarized phenotype characteristic of anti-inflammatory macrophages. Considering our findings together, this study provides further clarity on the unique C. burnetii-lung dynamic during early stages of human acute Q fever.


Subject(s)
Coxiella burnetii/pathogenicity , Host-Pathogen Interactions/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Q Fever/immunology , Q Fever/physiopathology , Humans , Macrophages, Alveolar/microbiology , Q Fever/microbiology
4.
Infect Immun ; 87(7)2019 07.
Article in English | MEDLINE | ID: mdl-31010814

ABSTRACT

Pulmonary pathogens encounter numerous insults, including phagocytic cells designed to degrade bacteria, while establishing infection in the human lung. Staphylococcus aureus is a versatile, opportunistic pathogen that can cause severe pneumonia, and methicillin-resistant isolates are of particular concern. Recent reports present conflicting data regarding the ability of S. aureus to survive and replicate within macrophages. However, due to use of multiple strains and macrophage sources, making comparisons between reports remains difficult. Here, we established a disease-relevant platform to study innate interactions between S. aureus and human lungs. Human precision-cut lung slices (hPCLS) were subjected to infection by S. aureus LAC (methicillin-resistant) or UAMS-1 (methicillin-sensitive) isolates. Additionally, primary human alveolar macrophages (hAMs) were infected with S. aureus, and antibacterial activity was assessed. Although both S. aureus isolates survived within hAM phagosomes, neither strain replicated efficiently in these cells. S. aureus was prevalent within the epithelial and interstitial regions of hPCLS, with limited numbers present in a subset of hAMs, suggesting that the pathogen may not target phagocytic cells for intracellular growth during natural pulmonary infection. S. aureus-infected hAMs mounted a robust inflammatory response that reflected natural human disease. S. aureus LAC was significantly more cytotoxic to hAMs than UAMS-1, potentially due to isolate-specific virulence factors. The bicomponent toxin Panton-Valentine leukocidin was not produced during intracellular infection, while alpha-hemolysin was produced but was not hemolytic, suggesting that hAMs alter toxin activity. Overall, this study defined a new disease-relevant infection platform to study S. aureus interaction with human lungs and to define virulence factors that incapacitate pulmonary cells.


Subject(s)
Bacterial Toxins/metabolism , Exotoxins/metabolism , Leukocidins/metabolism , Macrophages, Alveolar/microbiology , Phagosomes/microbiology , Staphylococcal Infections , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Virulence Factors/metabolism , Anti-Bacterial Agents/pharmacology , Humans , Lung/metabolism , Lung/microbiology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology
5.
Microbiology (Reading) ; 165(1): 1-3, 2019 01.
Article in English | MEDLINE | ID: mdl-30422108

ABSTRACT

Coxiella burnetii is an obligate intracellular pathogen that causes acute and chronic Q fever. C. burnetii grows within a eukaryotic host cell in a vacuole highly similar to a phagolysosome. Found worldwide, this environmentally stable pathogen is maintained in nature via chronic infection of ruminants. Aerosol-mediated infection of humans results in infection and usurpation of alveolar macrophages through mechanisms using a bacterial Type 4B Secretion System and secreted effector proteins. Advances in axenic culture and genetic systems are changing our understanding of the pathogen's physiology and intimate molecular manipulations of host cells during infection.


Subject(s)
Coxiella burnetii/metabolism , Q Fever/microbiology , Acids/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/metabolism , Coxiella burnetii/classification , Coxiella burnetii/genetics , Coxiella burnetii/isolation & purification , Genome, Bacterial , Humans , Hydrogen-Ion Concentration , Phylogeny , Vacuoles/chemistry , Vacuoles/microbiology
6.
Infect Immun ; 86(5)2018 05.
Article in English | MEDLINE | ID: mdl-29483292

ABSTRACT

Coxiella burnetii is the causative agent of human Q fever, a debilitating flu-like illness that can progress to chronic disease presenting as endocarditis. Following inhalation, C. burnetii is phagocytosed by alveolar macrophages and generates a lysosome-like replication compartment termed the parasitophorous vacuole (PV). A type IV secretion system (T4SS) is required for PV generation and is one of the pathogen's few known virulence factors. We previously showed that C. burnetii actively recruits autophagosomes to the PV using the T4SS but does not alter macroautophagy. In the current study, we confirmed that the cargo receptor p62/sequestosome 1 (SQSTM-1) localizes near the PV in primary human alveolar macrophages infected with virulent C. burnetii p62 and LC3 typically interact to select cargo for autophagy-mediated degradation, resulting in p62 degradation and LC3 recycling. However, in C. burnetii-infected macrophages, p62 was not degraded when cells were starved, suggesting that the pathogen stabilizes the protein. In addition, phosphorylated p62 levels increased, indicative of activation, during infection. Small interfering RNA experiments indicated that p62 is not absolutely required for intracellular growth, suggesting that the protein serves a signaling role during infection. Indeed, the Nrf2-Keap1 cytoprotective pathway was activated during infection, as evidenced by sustained maintenance of Nrf2 levels and translocation of the protein to the nucleus in C. burnetii-infected cells. Collectively, our studies identify a new p62-regulated host signaling pathway exploited by C. burnetii during intramacrophage growth.


Subject(s)
Coxiella burnetii/pathogenicity , Host-Pathogen Interactions/physiology , Macrophages/metabolism , Macrophages/pathology , NF-E2-Related Factor 2/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction/physiology , Humans
7.
PLoS Pathog ; 12(10): e1005915, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27711191

ABSTRACT

Coxiella burnetii is an intracellular bacterial pathogen that causes human Q fever, an acute flu-like illness that can progress to chronic endocarditis and liver and bone infections. Humans are typically infected by aerosol-mediated transmission, and C. burnetii initially targets alveolar macrophages wherein the pathogen replicates in a phagolysosome-like niche known as the parasitophorous vacuole (PV). C. burnetii manipulates host cAMP-dependent protein kinase (PKA) signaling to promote PV formation, cell survival, and bacterial replication. In this study, we identified the actin regulatory protein vasodilator-stimulated phosphoprotein (VASP) as a PKA substrate that is increasingly phosphorylated at S157 and S239 during C. burnetii infection. Avirulent and virulent C. burnetii triggered increased levels of phosphorylated VASP in macrophage-like THP-1 cells and primary human alveolar macrophages, and this event required the Cα subunit of PKA. VASP phosphorylation also required bacterial protein synthesis and secretion of effector proteins via a type IV secretion system, indicating the pathogen actively triggers prolonged VASP phosphorylation. Optimal PV formation and intracellular bacterial replication required VASP activity, as siRNA-mediated depletion of VASP reduced PV size and bacterial growth. Interestingly, ectopic expression of a phospho-mimetic VASP (S239E) mutant protein prevented optimal PV formation, whereas VASP (S157E) mutant expression had no effect. VASP (S239E) expression also prevented trafficking of bead-containing phagosomes to the PV, indicating proper VASP activity is critical for heterotypic fusion events that control PV expansion in macrophages. Finally, expression of dominant negative VASP (S157A) in C. burnetii-infected cells impaired PV formation, confirming importance of the protein for proper infection. This study provides the first evidence of VASP manipulation by an intravacuolar bacterial pathogen via activation of PKA in human macrophages.


Subject(s)
Cell Adhesion Molecules/metabolism , Host-Pathogen Interactions/physiology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Q Fever/metabolism , Coxiella burnetii , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/physiology , Gene Knockdown Techniques , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Confocal
8.
Infect Immun ; 84(5): 1438-1445, 2016 05.
Article in English | MEDLINE | ID: mdl-26902725

ABSTRACT

Coxiella burnetii is an intracellular bacterial pathogen that causes human Q fever, an acute debilitating flu-like illness that can also present as chronic endocarditis. Disease typically occurs following inhalation of contaminated aerosols, resulting in an initial pulmonary infection. In human cells, C. burnetii generates a replication niche termed the parasitophorous vacuole (PV) by directing fusion with autophagosomes and lysosomes. C. burnetii requires this lysosomal environment for replication and uses a Dot/Icm type IV secretion system to generate the large PV. However, we do not understand how C. burnetii evades the intracellular immune surveillance that triggers an inflammatory response. We recently characterized human alveolar macrophage (hAM) infection in vitro and found that avirulent C. burnetii triggers sustained interleukin-1ß (IL-1ß) production. Here, we evaluated infection of ex vivo human lung tissue, defining a valuable approach for characterizing C. burnetii interactions with a human host. Within whole lung tissue, C. burnetii preferentially replicated in hAMs. Additionally, IL-1ß production correlated with formation of an apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC)-dependent inflammasome in response to infection. We also assessed potential activation of a human-specific noncanonical inflammasome and found that caspase-4 and caspase-5 are processed during infection. Interestingly, although inflammasome activation is closely linked to pyroptosis, lytic cell death did not occur following C. burnetii-triggered inflammasome activation, indicating an atypical response after intracellular detection. Together, these studies provide a novel platform for studying the human innate immune response to C. burnetii.


Subject(s)
Coxiella burnetii/pathogenicity , Lung/microbiology , Lung/pathology , Models, Biological , Organ Culture Techniques/methods , Q Fever/microbiology , Q Fever/pathology , Humans , Models, Theoretical
9.
Infect Immun ; 83(3): 1190-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25605765

ABSTRACT

Coxiella burnetii causes human Q fever, a zoonotic disease that presents with acute flu-like symptoms and can result in chronic life-threatening endocarditis. In human alveolar macrophages, C. burnetii uses a Dot/Icm type IV secretion system (T4SS) to generate a phagolysosome-like parasitophorous vacuole (PV) in which to replicate. The T4SS translocates effector proteins, or substrates, into the host cytosol, where they mediate critical cellular events, including interaction with autophagosomes, PV formation, and prevention of apoptosis. Over 100 C. burnetii Dot/Icm substrates have been identified, but the function of most remains undefined. Here, we identified a novel Dot/Icm substrate-encoding open reading frame (CbuD1884) present in all C. burnetii isolates except the Nine Mile reference isolate, where the gene is disrupted by a frameshift mutation, resulting in a pseudogene. The CbuD1884 protein contains two transmembrane helices (TMHs) and a coiled-coil domain predicted to mediate protein-protein interactions. The C-terminal region of the protein contains a predicted Dot/Icm translocation signal and was secreted by the T4SS, while the N-terminal portion of the protein was not secreted. When ectopically expressed in eukaryotic cells, the TMH-containing N-terminal region of the CbuD1884 protein trafficked to the endoplasmic reticulum (ER), with the C terminus dispersed nonspecifically in the host cytoplasm. This new Dot/Icm substrate is now termed ElpA (ER-localizing protein A). Full-length ElpA triggered substantial disruption of ER structure and host cell secretory transport. These results suggest that ElpA is a pathotype-specific T4SS effector that influences ER function during C. burnetii infection.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Secretion Systems/genetics , Carrier Proteins/chemistry , Coxiella burnetii/chemistry , Endoplasmic Reticulum/chemistry , Recombinant Proteins/chemistry , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Coxiella burnetii/genetics , Coxiella burnetii/metabolism , Coxiella burnetii/pathogenicity , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Monocytes/cytology , Monocytes/metabolism , Mutation , Open Reading Frames , Plasmids , Protein Sorting Signals , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transgenes
10.
Infect Immun ; 83(2): 661-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25422265

ABSTRACT

The intracellular bacterial pathogen Coxiella burnetii directs biogenesis of a parasitophorous vacuole (PV) that acquires host endolysosomal components. Formation of a PV that supports C. burnetii replication requires a Dot/Icm type 4B secretion system (T4BSS) that delivers bacterial effector proteins into the host cell cytosol. Thus, a subset of T4BSS effectors are presumed to direct PV biogenesis. Recently, the PV-localized effector protein CvpA was found to promote C. burnetii intracellular growth and PV expansion. We predict additional C. burnetii effectors localize to the PV membrane and regulate eukaryotic vesicle trafficking events that promote pathogen growth. To identify these vacuolar effector proteins, a list of predicted C. burnetii T4BSS substrates was compiled using bioinformatic criteria, such as the presence of eukaryote-like coiled-coil domains. Adenylate cyclase translocation assays revealed 13 proteins were secreted in a Dot/Icm-dependent fashion by C. burnetii during infection of human THP-1 macrophages. Four of the Dot/Icm substrates, termed Coxiella vacuolar protein B (CvpB), CvpC, CvpD, and CvpE, labeled the PV membrane and LAMP1-positive vesicles when ectopically expressed as fluorescently tagged fusion proteins. C. burnetii ΔcvpB, ΔcvpC, ΔcvpD, and ΔcvpE mutants exhibited significant defects in intracellular replication and PV formation. Genetic complementation of the ΔcvpD and ΔcvpE mutants rescued intracellular growth and PV generation, whereas the growth of C. burnetii ΔcvpB and ΔcvpC was rescued upon cohabitation with wild-type bacteria in a common PV. Collectively, these data indicate C. burnetii encodes multiple effector proteins that target the PV membrane and benefit pathogen replication in human macrophages.


Subject(s)
Bacterial Proteins/metabolism , Coxiella burnetii/metabolism , Macrophages/microbiology , Protein Transport/genetics , Vacuoles/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Secretion Systems/physiology , Cell Line, Tumor , Cell Membrane , Coxiella burnetii/genetics , Cytosol/metabolism , Gene Deletion , HeLa Cells , Host-Pathogen Interactions , Humans , Q Fever/microbiology , Q Fever/pathology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vacuoles/genetics , Vacuoles/microbiology
11.
PLoS Pathog ; 9(9): e1003583, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068923

ABSTRACT

Lytic gammaherpesvirus (GHV) replication facilitates the establishment of lifelong latent infection, which places the infected host at risk for numerous cancers. As obligate intracellular parasites, GHVs must control and usurp cellular signaling pathways in order to successfully replicate, disseminate to stable latency reservoirs in the host, and prevent immune-mediated clearance. To facilitate a systems-level understanding of phosphorylation-dependent signaling events directed by GHVs during lytic replication, we utilized label-free quantitative mass spectrometry to interrogate the lytic replication cycle of murine gammaherpesvirus-68 (MHV68). Compared to controls, MHV68 infection regulated by 2-fold or greater ca. 86% of identified phosphopeptides - a regulatory scale not previously observed in phosphoproteomic evaluations of discrete signal-inducing stimuli. Network analyses demonstrated that the infection-associated induction or repression of specific cellular proteins globally altered the flow of information through the host phosphoprotein network, yielding major changes to functional protein clusters and ontologically associated proteins. A series of orthogonal bioinformatics analyses revealed that MAPK and CDK-related signaling events were overrepresented in the infection-associated phosphoproteome and identified 155 host proteins, such as the transcription factor c-Jun, as putative downstream targets. Importantly, functional tests of bioinformatics-based predictions confirmed ERK1/2 and CDK1/2 as kinases that facilitate MHV68 replication and also demonstrated the importance of c-Jun. Finally, a transposon-mutant virus screen identified the MHV68 cyclin D ortholog as a viral protein that contributes to the prominent MAPK/CDK signature of the infection-associated phosphoproteome. Together, these analyses enhance an understanding of how GHVs reorganize and usurp intracellular signaling networks to facilitate infection and replication.


Subject(s)
Gammaherpesvirinae/physiology , Host-Pathogen Interactions , Models, Biological , Phosphoproteins/metabolism , Signal Transduction , Viral Proteins/metabolism , Virus Replication , 3T3 Cells , Animals , Chromatography, High Pressure Liquid , Computational Biology , Cyclin D/chemistry , Cyclin D/genetics , Cyclin D/metabolism , Gammaherpesvirinae/genetics , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , MAP Kinase Signaling System , Mice , Mutation , Phosphoproteins/chemistry , Phosphoproteins/genetics , Proteome/chemistry , Proteome/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-jun/chemistry , Proto-Oncogene Proteins c-jun/metabolism , Tandem Mass Spectrometry , Viral Proteins/chemistry , Viral Proteins/genetics
12.
Cell Microbiol ; 16(1): 146-59, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24028560

ABSTRACT

Intracellular bacterial pathogens often subvert apoptosis signalling to regulate survival of their host cell, allowing propagation of the bacterial population. Coxiella burnetii, the intracellular agent of human Q fever, inhibits host cell apoptosis through several mechanisms, including prevention of mitochondrial cytochrome c release, triggering of an anti-apoptotic transcriptional programme, and activation of pro-survival kinases. To control host cell survival, C. burnetii delivers effector proteins to the eukaryotic cytosol using a specialized Dot/Icm type IV secretion system (T4SS). Effectors are predicted to regulate activity of pro-survival host signalling proteins, such as Akt and cAMP-dependent protein kinase (PKA), to control infection. Here, we show that host PKA activity is required for C. burnetii inhibition of macrophage apoptosis. PKA is activated during infection and inhibits activity of the pro-apoptotic protein Bad via phosphorylation. Bad is also phosphorylated at an Akt-specific residue, indicating C. burnetii uses two kinases to fully inactivate Bad. Additionally, Bad and the tethering protein 14-3-3ß colocalize at the C. burnetii parasitophorous vacuole (PV) membrane during infection, an event predicted to alter Bad promotion of apoptosis. Inhibiting PKA activity prevents Bad recruitment to the PV, but the protein is retained at the membrane during induction of apoptosis. Finally, PKA regulatory subunit I (RI) traffics to the PV membrane in a T4SS-dependent manner, suggesting a C. burnetii effector(s) regulates PKA-dependent activities. This study is the first to demonstrate subversion of host PKA activity by an intracellular bacterial pathogen to prevent apoptosis and survive within macrophages.


Subject(s)
Coxiella burnetii/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Host-Pathogen Interactions , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/physiology , Cell Survival , Cells, Cultured , Humans
13.
Infect Immun ; 82(6): 2229-38, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24643534

ABSTRACT

Coxiella burnetii is an intracellular Gram-negative bacterium that causes human Q fever, a flu-like disease that can progress to chronic, life-threatening endocarditis. In humans, C. burnetii infects alveolar macrophages and promotes phagosomal fusion with autophagosomes and lysosomes, establishing a unique parasitophorous vacuole (PV) in which to replicate. The pathogen uses a Dot/Icm type IV secretion system (T4SS) to deliver effector proteins to the host cytoplasm, where they alter cellular processes to benefit the pathogen. The T4SS is required for PV expansion and prevention of apoptosis, but little else is known about the role of the system during intracellular growth. Recent reports suggest that C. burnetii actively recruits autophagosomes to the PV to deliver nutrients to the pathogen and provide membrane for the expanding vacuole. In this study, we examined the role of the T4SS in mediating PV interactions with autophagosomes. We found that the autophagy-related proteins LC3 and p62 localized to wild-type PV but not to T4SS mutant organism-containing phagosomes in human macrophage-like cells, primary human alveolar macrophages, and Chinese hamster ovary cells. However, while lipidated LC3 levels were elevated regardless of T4SS activity, no p62 turnover was observed during C. burnetii growth in macrophages, suggesting that the pathogen recruits preformed autophagosomes. When the T4SS was activated 24 h after infection, autophagosome recruitment ensued, indicating that autophagosome interactions are dispensable for initial PV maturation to a phagolysosome-like compartment but are involved in vacuole expansion. Together, these results demonstrate that C. burnetii actively directs PV-autophagosome interactions by using the Dot/Icm T4SS.


Subject(s)
Autophagy/physiology , Bacterial Secretion Systems/physiology , Coxiella burnetii/pathogenicity , Host-Pathogen Interactions/physiology , Macrophages/microbiology , Phagosomes/microbiology , Humans , Macrophages/metabolism , Phagosomes/metabolism , Vacuoles/metabolism , Vacuoles/microbiology
14.
Cell Microbiol ; 15(6): 1012-25, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23279051

ABSTRACT

The intracellular bacterial pathogen Coxiella burnetii is a category B select agent that causes human Q fever. In vivo, C. burnetii targets alveolar macrophages wherein the pathogen replicates in a lysosome-like parasitophorous vacuole (PV). In vitro, C. burnetii infects a variety of cultured cell lines that have collectively been used to model the pathogen's infectious cycle. However, differences in the cellular response to infection have been observed, and virulent C. burnetii isolate infection of host cells has not been well defined. Because alveolar macrophages are routinely implicated in disease, we established primary human alveolar macrophages (hAMs) as an in vitro model of C. burnetii-host cell interactions. C. burnetii pathotypes, including acute disease and endocarditis isolates, replicated in hAMs, albeit with unique PV properties. Each isolate replicated in large, typical PV and small, non-fused vacuoles, and lipid droplets were present in avirulent C. burnetii PV. Interestingly, a subset of small vacuoles harboured single organisms undergoing degradation. Prototypical PV formation and bacterial growth in hAMs required a functional type IV secretion system, indicating C. burnetii secretes effector proteins that control macrophage functions. Avirulent C. burnetii promoted sustained activation of Akt and Erk1/2 pro-survival kinases and short-termphosphorylation of stress-related p38. Avirulent organisms also triggered a robust, early pro-inflammatory response characterized by increased secretion of TNF-α and IL-6, while virulent isolates elicited substantially reduced secretion of these cytokines. A corresponding increase in pro- and mature IL-1ß occurred in hAMs infected with avirulent C. burnetii, while little accumulation was observed following infection with virulent isolates. Finally, treatment of hAMs with IFN-γ controlled intracellular replication, supporting a role for this antibacterial insult in the host response to C. burnetii. Collectively, the current results demonstrate the hAM model is a human disease-relevant platform for defining novel innate immune responses to C. burnetii.


Subject(s)
Coxiella burnetii/pathogenicity , Host-Pathogen Interactions/physiology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Q Fever/pathology , Cells, Cultured , Coxiella burnetii/growth & development , Humans , In Vitro Techniques , Interleukin-1beta/physiology , Interleukin-6/physiology , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-akt/physiology , Q Fever/physiopathology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Virulence
15.
PLoS Negl Trop Dis ; 18(7): e0012348, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39038047

ABSTRACT

Relapsing fever (RF), a vector-borne disease caused by Borrelia spp., is characterized by recurring febrile episodes due to repeated bouts of bacteremia. RF spirochetes can be geographically and phylogenetically divided into two distinct groups; Old World RF Borrelia (found in Africa, Asia, and Europe) and New World RF Borrelia (found in the Americas). While RF is a rarely reported disease in the Americas, RF is prevalent in endemic parts of Africa. Despite phylogenetic differences between Old World and New World RF Borrelia and higher incidence of disease associated with Old World RF spirochete infection, genetic manipulation has only been described in New World RF bacteria. Herein, we report the generation of genetic tools for use in the Old World RF spirochete, Borrelia duttonii. We describe methods for transformation and establish shuttle vector- and integration-based approaches for genetic complementation, creating green fluorescent protein (gfp)-expressing B. duttonii strains as a proof of principle. Allelic exchange mutagenesis was also used to inactivate a homolog of the Borrelia burgdorferi p66 gene, which encodes an important virulence factor, in B. duttonii and demonstrate that this mutant was attenuated in a murine model of RF. Finally, the B. duttonii p66 mutant was complemented using shuttle vector- and cis integration-based approaches. As expected, complemented p66 mutant strains were fully infectious, confirming that P66 is required for optimal mammalian infection. The genetic tools and techniques reported herein represent an important advancement in the study of RF Borrelia that allows for future characterization of virulence determinants and colonization factors important for the enzootic cycle of Old World RF spirochetes.


Subject(s)
Borrelia , Relapsing Fever , Animals , Relapsing Fever/microbiology , Borrelia/genetics , Borrelia/classification , Mice , Female , Genetic Complementation Test , Green Fluorescent Proteins/genetics , Humans
16.
J Bacteriol ; 195(14): 3269-76, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23687269

ABSTRACT

The intracellular bacterial agent of Q fever, Coxiella burnetii, translocates effector proteins into its host cell cytosol via a Dot/Icm type IV secretion system (T4SS). The T4SS is essential for parasitophorous vacuole formation, intracellular replication, and inhibition of host cell death, but the effectors mediating these events remain largely undefined. Six Dot/Icm substrate-encoding genes were recently discovered on the C. burnetii cryptic QpH1 plasmid, three of which are conserved among all C. burnetii isolates, suggesting that they are critical for conserved pathogen functions. However, the remaining hypothetical proteins encoded by plasmid genes have not been assessed for their potential as T4SS substrates. In the current study, we further defined the T4SS effector repertoire encoded by the C. burnetii QpH1, QpRS, and QpDG plasmids that were originally isolated from acute-disease, chronic-disease, and severely attenuated isolates, respectively. Hypothetical proteins, including those specific to QpRS or QpDG, were screened for translocation using the well-established Legionella pneumophila T4SS secretion model. In total, six novel plasmid-encoded proteins were translocated into macrophage-like cells by the Dot/Icm T4SS. Four newly identified effectors are encoded by genes present only on the QpDG plasmid from severely attenuated Dugway isolates, suggesting that the presence of specific effectors correlates with decreased virulence. These results further support the idea of a critical role for extrachromosomal elements in C. burnetii pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems , Coxiella burnetii/metabolism , Plasmids , Virulence Factors/metabolism , Bacterial Proteins/genetics , Cell Line , Coxiella burnetii/genetics , Humans , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Macrophages/microbiology , Substrate Specificity , Virulence Factors/genetics
17.
Infect Immun ; 80(6): 1980-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22473604

ABSTRACT

Coxiella burnetii is the bacterial agent of human Q fever, an acute, flu-like illness that can present as chronic endocarditis in immunocompromised individuals. Following aerosol-mediated transmission, C. burnetii replicates in alveolar macrophages in a unique phagolysosome-like parasitophorous vacuole (PV) required for survival. The mechanisms of C. burnetii intracellular survival are poorly defined and a recent Q fever outbreak in the Netherlands emphasizes the need for better understanding this unique host-pathogen interaction. We recently demonstrated that inhibition of host cyclic AMP-dependent protein kinase (PKA) activity negatively impacts PV formation. In the current study, we confirmed PKA involvement in PV biogenesis and probed the role of PKA signaling during C. burnetii infection of macrophages. Using PKA-specific inhibitors, we found the kinase was needed for biogenesis of prototypical PV and C. burnetii replication. PKA and downstream targets were differentially phosphorylated throughout infection, suggesting prolonged regulation of the pathway. Importantly, the pathogen actively triggered PKA activation, which was also required for PV formation by virulent C. burnetii isolates during infection of primary human alveolar macrophages. A subset of PKA-specific substrates were differentially phosphorylated during C. burnetii infection, suggesting the pathogen uses PKA signaling to control distinct host cell responses. Collectively, the current results suggest a versatile role for PKA in C. burnetii infection and indicate virulent organisms usurp host kinase cascades for efficient intracellular growth.


Subject(s)
Bacterial Proteins/metabolism , Coxiella burnetii/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Macrophages, Alveolar/microbiology , Signal Transduction/physiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Cells, Cultured , Chloramphenicol/pharmacology , Coxiella burnetii/drug effects , Coxiella burnetii/genetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Gene Expression Regulation, Bacterial/physiology , Humans , Phosphorylation
18.
Adv Exp Med Biol ; 984: 131-40, 2012.
Article in English | MEDLINE | ID: mdl-22711630

ABSTRACT

Coxiella burnetii is a highly infectious bacterial pathogen that replicates in a specialized vacuole inside eukaryotic cells. Due to a prolonged growth cycle, Coxiella continuously manipulates cellular processes to generate this parasitophorous vacuole (PV) and promote host cell viability. Here, we discuss recent findings that indicate Coxiella modulates several host signaling pathways to influence survival and ensure intracellular replication. The pathogen actively inhibits apoptotic cell death and activates the pro-survival kinases Akt and Erk1/2 to promote host viability. Coxiella's anti-apoptotic activity also involves the interface between autophagy and apoptosis, which is regulated by the interaction of autophagy-related Beclin-1 and anti-apoptotic Bcl-2. Additionally, Coxiella requires host kinase activity for PV biogenesis and maintenance. Thus, signaling modulation by Coxiella is critical for multiple aspects of host cell parasitism. Collectively, recent signaling studies have enhanced our understanding of the unique Coxiella-host cell interaction. Identification of bacterial factors that regulate signaling events will further our ability to model this intriguing infectious process.


Subject(s)
Coxiella burnetii/metabolism , Vacuoles/metabolism , Vacuoles/microbiology , Animals , Apoptosis/physiology , Autophagy/physiology , Cell Survival/physiology , Humans , Signal Transduction
19.
Front Cell Infect Microbiol ; 12: 934460, 2022.
Article in English | MEDLINE | ID: mdl-35899042

ABSTRACT

Lung macrophages are substantially distinct from other tissue-resident macrophages. They act as frontier sentinels of the alveolar-blood interface and are constantly exposed to various pathogens. Additionally, they precisely regulate immune responses under homeostatic and pathological conditions to curtail tissue damage while containing respiratory infections. As a highly heterogeneous population, the phenotypes and functions of lung macrophages with differing developmental ontogenies are linked to both intrinsic and extrinsic metabolic processes. Importantly, targeting these metabolic pathways greatly impacts macrophage functions, which in turn leads to different disease outcomes in the lung. In this review, we will discuss underlying metabolic regulation of lung macrophage subsets and how metabolic circuits, together with epigenetic modifications, dictate lung macrophage function during bacterial infection.


Subject(s)
Bacterial Infections , Macrophages, Alveolar , Bacterial Infections/pathology , Humans , Immunity , Lung/microbiology , Macrophages
20.
J Bacteriol ; 193(7): 1493-503, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21216993

ABSTRACT

The intracellular bacterial pathogen Coxiella burnetii directs biogenesis of a phagolysosome-like parasitophorous vacuole (PV), in which it replicates. The organism encodes a Dot/Icm type IV secretion system (T4SS) predicted to deliver to the host cytosol effector proteins that mediate PV formation and other cellular events. All C. burnetii isolates carry a large, autonomously replicating plasmid or have chromosomally integrated plasmid-like sequences (IPS), suggesting that plasmid and IPS genes are critical for infection. Bioinformatic analyses revealed two candidate Dot/Icm substrates with eukaryotic-like motifs uniquely encoded by the QpH1 plasmid from the Nine Mile reference isolate. CpeC, containing an F-box domain, and CpeD, possessing kinesin-related and coiled-coil regions, were secreted by the closely related Legionella pneumophila Dot/Icm T4SS. An additional QpH1-specific gene, cpeE, situated in a predicted operon with cpeD, also encoded a secreted effector. Further screening revealed that three hypothetical proteins (CpeA, CpeB, and CpeF) encoded by all C. burnetii plasmids and IPS are Dot/Icm substrates. By use of new genetic tools, secretion of plasmid effectors by C. burnetii during host cell infection was confirmed using ß-lactamase and adenylate cyclase translocation assays, and a C-terminal secretion signal was identified. When ectopically expressed in HeLa cells, plasmid effectors trafficked to different subcellular sites, including autophagosomes (CpeB), ubiquitin-rich compartments (CpeC), and the endoplasmic reticulum (CpeD). Collectively, these results suggest that C. burnetii plasmid-encoded T4SS substrates play important roles in subversion of host cell functions, providing a plausible explanation for the absolute maintenance of plasmid genes by this pathogen.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/physiology , Coxiella burnetii/metabolism , Gene Expression Regulation, Bacterial/physiology , Plasmids/genetics , Bacterial Proteins/genetics , Bacterial Secretion Systems/genetics , Base Sequence , Cell Line , Coxiella burnetii/genetics , DNA, Bacterial/genetics , HeLa Cells , Humans , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL