Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Cell ; 169(2): 273-285.e17, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388411

ABSTRACT

How phospholipids are trafficked between the bacterial inner and outer membranes through the hydrophilic space of the periplasm is not known. We report that members of the mammalian cell entry (MCE) protein family form hexameric assemblies with a central channel capable of mediating lipid transport. The E. coli MCE protein, MlaD, forms a ring associated with an ABC transporter complex in the inner membrane. A soluble lipid-binding protein, MlaC, ferries lipids between MlaD and an outer membrane protein complex. In contrast, EM structures of two other E. coli MCE proteins show that YebT forms an elongated tube consisting of seven stacked MCE rings, and PqiB adopts a syringe-like architecture. Both YebT and PqiB create channels of sufficient length to span the periplasmic space. This work reveals diverse architectures of highly conserved protein-based channels implicated in the transport of lipids between the membranes of bacteria and some eukaryotic organelles.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Membrane Proteins/chemistry , Cell Membrane/chemistry , Crystallography, X-Ray , Microscopy, Electron , Models, Molecular , Multiprotein Complexes/chemistry
2.
Cell ; 161(3): 501-512, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25865481

ABSTRACT

Mycobacterium tuberculosis and Staphylococcus aureus secrete virulence factors via type VII protein secretion (T7S), a system that intriguingly requires all of its secretion substrates for activity. To gain insights into T7S function, we used structural approaches to guide studies of the putative translocase EccC, a unique enzyme with three ATPase domains, and its secretion substrate EsxB. The crystal structure of EccC revealed that the ATPase domains are joined by linker/pocket interactions that modulate its enzymatic activity. EsxB binds via its signal sequence to an empty pocket on the C-terminal ATPase domain, which is accompanied by an increase in ATPase activity. Surprisingly, substrate binding does not activate EccC allosterically but, rather, by stimulating its multimerization. Thus, the EsxB substrate is also an integral T7S component, illuminating a mechanism that helps to explain interdependence of substrates, and suggests a model in which binding of substrates modulates their coordinate release from the bacterium.


Subject(s)
Actinobacteria/enzymology , Bacterial Secretion Systems , Actinobacteria/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Crystallography, X-Ray , Models, Molecular , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Staphylococcus aureus/enzymology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Virulence Factors/chemistry
3.
Cell ; 150(4): 803-15, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22901810

ABSTRACT

Eukaryotic cells sterilize the cytosol by using autophagy to route invading bacterial pathogens to the lysosome. During macrophage infection with Mycobacterium tuberculosis, a vacuolar pathogen, exogenous induction of autophagy can limit replication, but the mechanism of autophagy targeting and its role in natural infection remain unclear. Here we show that phagosomal permeabilization mediated by the bacterial ESX-1 secretion system allows cytosolic components of the ubiquitin-mediated autophagy pathway access to phagosomal M. tuberculosis. Recognition of extracelluar bacterial DNA by the STING-dependent cytosolic pathway is required for marking bacteria with ubiquitin, and delivery of bacilli to autophagosomes requires the ubiquitin-autophagy receptors p62 and NDP52 and the DNA-responsive kinase TBK1. Remarkably, mice with monocytes incapable of delivering bacilli to the autophagy pathway are extremely susceptible to infection. Our results reveal an unexpected link between DNA sensing, innate immunity, and autophagy and indicate a major role for this autophagy pathway in resistance to M. tuberculosis infection.


Subject(s)
Autophagy , DNA, Bacterial/immunology , Immunity, Innate , Macrophages/immunology , Macrophages/microbiology , Mycobacterium tuberculosis/physiology , Animals , Autophagy-Related Protein 5 , Cytosol/microbiology , Deoxyribonucleases/metabolism , Lysosomes/microbiology , Macrophages/cytology , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mycobacterium tuberculosis/genetics , Phagosomes/microbiology , Ubiquitin/metabolism , Ubiquitination
4.
Mol Cell ; 71(4): 637-648.e5, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30118682

ABSTRACT

Although macrophages are armed with potent antibacterial functions, Mycobacterium tuberculosis (Mtb) replicates inside these innate immune cells. Determinants of macrophage intrinsic bacterial control, and the Mtb strategies to overcome them, are poorly understood. To further study these processes, we used an affinity tag purification mass spectrometry (AP-MS) approach to identify 187 Mtb-human protein-protein interactions (PPIs) involving 34 secreted Mtb proteins. This interaction map revealed two factors involved in Mtb pathogenesis-the secreted Mtb protein, LpqN, and its binding partner, the human ubiquitin ligase CBL. We discovered that an lpqN Mtb mutant is attenuated in macrophages, but growth is restored when CBL is removed. Conversely, Cbl-/- macrophages are resistant to viral infection, indicating that CBL regulates cell-intrinsic polarization between antibacterial and antiviral immunity. Collectively, these findings illustrate the utility of this Mtb-human PPI map for developing a deeper understanding of the intricate interactions between Mtb and its host.


Subject(s)
Bacterial Proteins/genetics , HIV/genetics , Host-Pathogen Interactions , Mycobacterium tuberculosis/genetics , Proto-Oncogene Proteins c-cbl/genetics , Virulence Factors/genetics , Animals , Bacterial Proteins/immunology , Cell Line, Tumor , Chlamydia trachomatis/genetics , Chlamydia trachomatis/immunology , Gene Expression Regulation , HIV/immunology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Humans , Lymphocytes/microbiology , Lymphocytes/virology , Macrophages/microbiology , Macrophages/virology , Mice , Mycobacterium tuberculosis/immunology , Primary Cell Culture , Protein Binding , Protein Interaction Mapping , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/immunology , RAW 264.7 Cells , Signal Transduction , Virulence Factors/immunology
5.
PLoS Pathog ; 19(6): e1011088, 2023 06.
Article in English | MEDLINE | ID: mdl-37352334

ABSTRACT

Macrophages employ an array of pattern recognition receptors to detect and eliminate intracellular pathogens that access the cytosol. The cytosolic carbohydrate sensors Galectin-3, -8, and -9 (Gal-3, Gal-8, and Gal-9) recognize damaged pathogen-containing phagosomes, and Gal-3 and Gal-8 are reported to restrict bacterial growth via autophagy in cultured cells. However, the contribution of these galectins to host resistance during bacterial infection in vivo remains unclear. We found that Gal-9 binds directly to Mycobacterium tuberculosis (Mtb) and Salmonella enterica serovar Typhimurium (Stm) and localizes to Mtb in macrophages. To determine the combined contribution of membrane damage-sensing galectins to immunity, we generated Gal-3, -8, and -9 triple knockout (TKO) mice. Mtb infection of primary macrophages from TKO mice resulted in defective autophagic flux but normal bacterial replication. Surprisingly, these mice had no discernable defect in resistance to acute infection with Mtb, Stm or Listeria monocytogenes, and had only modest impairments in bacterial growth restriction and CD4 T cell activation during chronic Mtb infection. Collectively, these findings indicate that while Gal-3, -8, and -9 respond to an array of intracellular pathogens, together these membrane damage-sensing galectins play a limited role in host resistance to bacterial infection.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Mice , Animals , Galectin 3/genetics , Tuberculosis/metabolism , Galectins/genetics , Galectins/metabolism , Macrophages , Salmonella typhimurium , Mice, Knockout
6.
PLoS Pathog ; 18(7): e1010721, 2022 07.
Article in English | MEDLINE | ID: mdl-35877763

ABSTRACT

The prevailing model of protective immunity to tuberculosis is that CD4 T cells produce the cytokine IFN-γ to activate bactericidal mechanisms in infected macrophages. Although IFN-γ-independent CD4 T cell based control of M. tuberculosis infection has been demonstrated in vivo it is unclear whether CD4 T cells are capable of directly activating macrophages to control infection in the absence of IFN-γ. We developed a co-culture model using CD4 T cells isolated from the lungs of infected mice and M. tuberculosis-infected murine bone marrow-derived macrophages (BMDMs) to investigate mechanisms of CD4 dependent control of infection. We found that even in the absence of IFN-γ signaling, CD4 T cells drive macrophage activation, M1 polarization, and control of infection. This IFN-γ-independent control of infection requires activation of the transcription factor HIF-1α and a shift to aerobic glycolysis in infected macrophages. While HIF-1α activation following IFN-γ stimulation requires nitric oxide, HIF-1α-mediated control in the absence of IFN-γ is nitric oxide-independent, indicating that distinct pathways can activate HIF-1α during infection. We show that CD4 T cell-derived GM-CSF is required for IFN-γ-independent control in BMDMs, but that recombinant GM-CSF is insufficient to control infection in BMDMs or alveolar macrophages and does not rescue the absence of control by GM-CSF-deficient T cells. In contrast, recombinant GM-CSF controls infection in peritoneal macrophages, induces lipid droplet biogenesis, and also requires HIF-1α for control. These results advance our understanding of CD4 T cell-mediated immunity to M. tuberculosis, reveal important differences in immune activation of distinct macrophage types, and outline a novel mechanism for the activation of HIF-1α. We establish a previously unknown functional link between GM-CSF and HIF-1α and provide evidence that CD4 T cell-derived GM-CSF is a potent bactericidal effector.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , CD4-Positive T-Lymphocytes , Granulocyte-Macrophage Colony-Stimulating Factor , Hypoxia-Inducible Factor 1, alpha Subunit , Interferon-gamma , Mice , Nitric Oxide
7.
J Immunol ; 205(1): 153-167, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32404352

ABSTRACT

Tripartite motif-containing proteins (TRIMs) play a variety of recently described roles in innate immunity. Although many TRIMs regulate type I IFN expression following cytosolic nucleic acid sensing of viruses, their contribution to innate immune signaling and gene expression during bacterial infection remains largely unknown. Because Mycobacterium tuberculosis is an activator of cGAS-dependent cytosolic DNA sensing, we set out to investigate a role for TRIM proteins in regulating macrophage responses to M. tuberculosis In this study, we demonstrate that TRIM14, a noncanonical TRIM that lacks an E3 ubiquitin ligase RING domain, is a critical negative regulator of the type I IFN response in Mus musculus macrophages. We show that TRIM14 interacts with both cGAS and TBK1 and that macrophages lacking TRIM14 dramatically hyperinduce IFN stimulated gene (ISG) expression following M. tuberculosis infection, cytosolic nucleic acid transfection, and IFN-ß treatment. Consistent with a defect in resolution of the type I IFN response, Trim14 knockout macrophages have more phospho-Ser754 STAT3 relative to phospho-Ser727 and fail to upregulate the STAT3 target Socs3, which is required to turn off IFNAR signaling. These data support a model whereby TRIM14 acts as a scaffold between TBK1 and STAT3 to promote phosphorylation of STAT3 at Ser727 and resolve ISG expression. Remarkably, Trim14 knockout macrophages hyperinduce expression of antimicrobial genes like Nos2 and are significantly better than control cells at limiting M. tuberculosis replication. Collectively, these data reveal an unappreciated role for TRIM14 in resolving type I IFN responses and controlling M. tuberculosis infection.


Subject(s)
Interferon Type I/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mycobacterium tuberculosis/immunology , Signal Transduction/immunology , Tripartite Motif Proteins/metabolism , Tuberculosis/immunology , Animals , Disease Models, Animal , Gene Expression Regulation/immunology , Gene Knockout Techniques , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/isolation & purification , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/isolation & purification , Nucleotidyltransferases/metabolism , Phosphorylation/immunology , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/isolation & purification , Protein Serine-Threonine Kinases/metabolism , RAW 264.7 Cells , Receptor, Interferon alpha-beta/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , STAT3 Transcription Factor/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/isolation & purification , Tuberculosis/microbiology
8.
Proc Natl Acad Sci U S A ; 115(2): E210-E217, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29279409

ABSTRACT

Xenophagy is a selective macroautophagic process that protects the host cytosol by entrapping and delivering microbes to a degradative compartment. Both noncanonical autophagic pathways and xenophagy are activated by microbes during infection, but the relative importance and function of these distinct processes are not clear. In this study, we used bacterial and host mutants to dissect the contribution of autophagic processes responsible for bacterial growth restriction of Listeria monocytogenesL. monocytogenes is a facultative intracellular pathogen that escapes from phagosomes, grows in the host cytosol, and avoids autophagy by expressing three determinants of pathogenesis: two secreted phospholipases C (PLCs; PlcA and PlcB) and a surface protein (ActA). We found that shortly after phagocytosis, wild-type (WT) L. monocytogenes escaped from a noncanonical autophagic process that targets damaged vacuoles. During this process, the autophagy marker LC3 localized to single-membrane phagosomes independently of the ULK complex, which is required for initiation of macroautophagy. However, growth restriction of bacteria lacking PlcA, PlcB, and ActA required FIP200 and TBK1, both involved in the engulfment of microbes by xenophagy. Time-lapse video microscopy revealed that deposition of LC3 on L. monocytogenes-containing vacuoles via noncanonical autophagy had no apparent role in restricting bacterial growth and that, upon access to the host cytosol, WT L. monocytogenes utilized PLCs and ActA to avoid subsequent xenophagy. In conclusion, although noncanonical autophagy targets phagosomes, xenophagy was required to restrict the growth of L. monocytogenes, an intracellular pathogen that damages the entry vacuole.


Subject(s)
Autophagy , Listeria monocytogenes/physiology , Macrophages/microbiology , Phagocytosis , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Cytosol/metabolism , Cytosol/microbiology , Host-Pathogen Interactions , Listeria monocytogenes/genetics , Macrophages/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Mutation , Phagosomes/metabolism , Phagosomes/microbiology , Time-Lapse Imaging/methods , Type C Phospholipases/genetics , Type C Phospholipases/metabolism
9.
Nature ; 501(7468): 512-6, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24005326

ABSTRACT

Ubiquitin-mediated targeting of intracellular bacteria to the autophagy pathway is a key innate defence mechanism against invading microbes, including the important human pathogen Mycobacterium tuberculosis. However, the ubiquitin ligases responsible for catalysing ubiquitin chains that surround intracellular bacteria are poorly understood. The parkin protein is a ubiquitin ligase with a well-established role in mitophagy, and mutations in the parkin gene (PARK2) lead to increased susceptibility to Parkinson's disease. Surprisingly, genetic polymorphisms in the PARK2 regulatory region are also associated with increased susceptibility to intracellular bacterial pathogens in humans, including Mycobacterium leprae and Salmonella enterica serovar Typhi, but the function of parkin in immunity has remained unexplored. Here we show that parkin has a role in ubiquitin-mediated autophagy of M. tuberculosis. Both parkin-deficient mice and flies are sensitive to various intracellular bacterial infections, indicating parkin has a conserved role in metazoan innate defence. Moreover, our work reveals an unexpected functional link between mitophagy and infectious disease.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/microbiology , Immunity, Innate/immunology , Mycobacterium marinum/immunology , Mycobacterium tuberculosis/immunology , Salmonella typhimurium/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Autophagy/immunology , Bone Marrow Cells/microbiology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Lysine/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Mitophagy , Models, Immunological , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Polyubiquitin/chemistry , Polyubiquitin/metabolism , Symbiosis/immunology , Tuberculosis/enzymology , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology , Ubiquitin/analysis , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
10.
Am J Respir Crit Care Med ; 196(4): 502-511, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28463648

ABSTRACT

RATIONALE: The molecular mechanisms that regulate tuberculosis susceptibility and bacillus Calmette-Guérin (BCG)-induced immunity are mostly unknown. However, induction of the adaptive immune response is a critical step in host control of Mycobacterium tuberculosis. Toll-interacting protein (TOLLIP) is a ubiquitin-binding protein that regulates innate immune responses, including Toll-like receptor signaling, which initiate adaptive immunity. TOLLIP variation is associated with susceptibility to tuberculosis, but the mechanism by which it regulates tuberculosis immunity is poorly understood. OBJECTIVES: To identify functional TOLLIP variants and evaluate the role of TOLLIP variation on innate and adaptive immune responses to mycobacteria and susceptibility to tuberculosis. METHODS: We used human cellular immunology approaches to characterize the role of a functional TOLLIP variant on monocyte mRNA expression and M. tuberculosis-induced monocyte immune functions. We also examined the association of TOLLIP variation with BCG-induced T-cell responses and susceptibility to latent tuberculosis infection. MEASUREMENTS AND MAIN RESULTS: We identified a functional TOLLIP promoter region single-nucleotide polymorphism, rs5743854, which was associated with decreased TOLLIP mRNA expression in infant monocytes. After M. tuberculosis infection, TOLLIP-deficient monocytes demonstrated increased IL-6, increased nitrite, and decreased bacterial replication. The TOLLIP-deficiency G/G genotype was associated with decreased BCG-specific IL-2+ CD4+ T-cell frequency and proliferation. This genotype was also associated with increased susceptibility to latent tuberculosis infection. CONCLUSIONS: TOLLIP deficiency is associated with decreased BCG-specific T-cell responses and increased susceptibility to tuberculosis. We hypothesize that the heightened antibacterial monocyte responses after vaccination of TOLLIP-deficient infants are responsible for decreased BCG-specific T-cell responses. Activating TOLLIP may provide a novel adjuvant strategy for BCG vaccination.


Subject(s)
Immunity, Innate/immunology , Intracellular Signaling Peptides and Proteins/immunology , Mycobacterium bovis/immunology , Tuberculosis/immunology , Humans , Immunity, Innate/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mycobacterium bovis/genetics , Polymorphism, Single Nucleotide/genetics , Polymorphism, Single Nucleotide/immunology , Prospective Studies , Tuberculosis/genetics
12.
Proc Natl Acad Sci U S A ; 111(41): 14758-63, 2014 Oct 14.
Article in English | MEDLINE | ID: mdl-25275011

ABSTRACT

Nearly 10% of the coding capacity of the Mycobacterium tuberculosis genome is devoted to two highly expanded and enigmatic protein families called PE and PPE, some of which are important virulence/immunogenicity factors and are secreted during infection via a unique alternative secretory system termed "type VII." How PE-PPE proteins function during infection and how they are translocated to the bacterial surface through the five distinct type VII secretion systems [ESAT-6 secretion system (ESX)] of M. tuberculosis is poorly understood. Here, we report the crystal structure of a PE-PPE heterodimer bound to ESX secretion-associated protein G (EspG), which adopts a novel fold. This PE-PPE-EspG complex, along with structures of two additional EspGs, suggests that EspG acts as an adaptor that recognizes specific PE-PPE protein complexes via extensive interactions with PPE domains, and delivers them to ESX machinery for secretion. Surprisingly, secretion of most PE-PPE proteins in M. tuberculosis is likely mediated by EspG from the ESX-5 system, underscoring the importance of ESX-5 in mycobacterial pathogenesis. Moreover, our results indicate that PE-PPE domains function as cis-acting targeting sequences that are read out by EspGs, revealing the molecular specificity for secretion through distinct ESX pathways.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Secretion Systems , Mycobacterium tuberculosis/metabolism , Amino Acid Motifs , Bacterial Proteins/genetics , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Models, Biological , Models, Molecular , Multigene Family , Protein Binding , Protein Subunits/metabolism , Sequence Homology, Amino Acid
13.
Proc Natl Acad Sci U S A ; 111(29): 10404-10, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-24994900

ABSTRACT

WOPR-domain proteins are found throughout the fungal kingdom where they function as master regulators of cell morphology and pathogenesis. Genetic and biochemical experiments previously demonstrated that these proteins bind to specific DNA sequences and thereby regulate transcription. However, their primary sequence showed no relationship to any known DNA-binding domain, and the basis for their ability to recognize DNA sequences remained unknown. Here, we describe the 2.6-Å crystal structure of a WOPR domain in complex with its preferred DNA sequence. The structure reveals that two highly conserved regions, separated by an unconserved linker, form an interdigitated ß-sheet that is tilted into the major groove of DNA. Although the main interaction surface is in the major groove, the highest-affinity interactions occur in the minor groove, primarily through a deeply penetrating arginine residue. The structure reveals a new, unanticipated mechanism by which proteins can recognize specific sequences of DNA.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Fungi/pathogenicity , Amino Acids/metabolism , Base Sequence , Conserved Sequence/genetics , Crystallography, X-Ray , DNA, Fungal/chemistry , DNA, Fungal/metabolism , Evolution, Molecular , Fungi/metabolism , Gene Expression Regulation, Fungal , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Phylogeny , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcriptional Activation/genetics
14.
Curr Top Microbiol Immunol ; 374: 211-41, 2013.
Article in English | MEDLINE | ID: mdl-23881288

ABSTRACT

The intimate and persistent connection between Mycobacterium tuberculosis and its human host suggests that the pathogen has evolved extensive mechanisms to evade eradication by the immune system. In particular, the organism has adapted to replicate within phagocytic cells, especially macrophages, which are specialized to kill microbes. Over the past decade of M. tuberculosis research, the means to manipulate both the organism and the host has ushered in an exciting time that has uncovered some of the mechanisms of the innate macrophage-pathogen interactions that lie at the heart of M. tuberculosis pathogenesis, though many interactions likely still await discovery. In this chapter, we will delve into some of these advances, with an emphasis on the interactions that occur on the cellular level when M. tuberculosis cells encounter macrophages. In particular, we focus on two major aspects of M. tuberculosis biology regarding the proximal physical interface between the bacterium and host, namely the interactions with the phagosomal membrane as well as the distinctive mycobacterial cell wall. Importantly, some of the emerging paradigms in M. tuberculosis pathogenesis and host response represent common themes in bacterial pathogenesis, such as the role of host cell membrane perforation in intracellular survival and host response. However, the array of unique bacterial lipid mediators and their interaction with host cells highlights the unique biology of this persistent pathogen.


Subject(s)
Immune Evasion , Macrophages/microbiology , Mycobacterium tuberculosis/metabolism , Phagosomes/microbiology , Tuberculosis, Pulmonary/microbiology , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Autophagy , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Wall/chemistry , Cell Wall/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Host-Pathogen Interactions , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Lipid Metabolism , Lipids/biosynthesis , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Mycolic Acids/metabolism , Phagosomes/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology
15.
Nature ; 454(7205): 717-21, 2008 Aug 07.
Article in English | MEDLINE | ID: mdl-18685700

ABSTRACT

Bacterial pathogens trigger specialized virulence factor secretion systems on encountering host cells. The ESX-1 protein secretion system of Mycobacterium tuberculosis-the causative agent of the human disease tuberculosis-delivers bacterial proteins into host cells during infection and is critical for virulence, but how it is regulated is unknown. Here we show that EspR (also known as Rv3849) is a key regulator of ESX-1 that is required for secretion and virulence in mice. EspR activates transcription of an operon that includes three ESX-1 components, Rv3616c-Rv3614c, whose expression in turn promotes secretion of ESX-1 substrates. EspR directly binds to and activates the Rv3616c-Rv3614c promoter and, unexpectedly, is itself secreted from the bacterial cell by the ESX-1 system that it regulates. Efflux of the DNA-binding regulator results in reduced Rv3616c-Rv3614c transcription, and thus reduced ESX-1 secretion. Our results reveal a direct negative feedback loop that regulates the activity of a secretion system essential for virulence. As the virulence factors secreted by the ESX-1 system are highly antigenic, fine control of secretion may be critical to successful infection.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/pathogenicity , Transcription Factors/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , Operon/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/chemistry , Transcription, Genetic , Transcriptional Activation , Virulence/genetics , Virulence Factors/genetics
16.
Proc Natl Acad Sci U S A ; 108(33): 13450-5, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21795602

ABSTRACT

EspR is a transcriptional regulator that activates the ESX-1 secretion system during Mycobacterium tuberculosis infection and is critical for pathogenesis. It is unique among DNA-binding proteins as it is secreted as part of a feedback regulatory loop that serves to mitigate transcriptional activity. Here we report the crystal structure of a functional EspR dimer at 2.5-Å resolution. The amino-terminal half of EspR is a helix-turn-helix (HTH) DNA-binding domain and the carboxy terminus consists of a dimerization domain with similarity to the SinR:SinI sporulation regulator of Bacillus subtilis. Surprisingly, the HTH domains of EspR are arranged in an unusual conformation in which they are splayed at an oblique angle to each other, suggesting that EspR binds DNA in a profoundly different way than most other known HTH regulators. By mapping the EspR binding sites in the espACD promoter, using both in vivo and in vitro binding assays, we show that the EspR operators are located unusually far from the promoter. The EspR dimer binds to these sites cooperatively, but the two "half-sites" contacted by each DNA recognition motif are separated by 177 base pairs. The distinctive structure of EspR and the exceptional arrangement of its operator contacts suggest that it could promote DNA looping in its target promoter. We hypothesize that direct DNA looping mediated by single-site binding of each EspR monomer may facilitate transcriptional control of this important virulence system.


Subject(s)
Bacterial Proteins/chemistry , DNA-Binding Proteins/chemistry , DNA/chemistry , Helix-Turn-Helix Motifs , Mycobacterium tuberculosis/chemistry , Protein Structure, Quaternary , Transcription Factors/chemistry , Binding Sites , Crystallography, X-Ray , DNA/metabolism , Models, Molecular , Mycobacterium tuberculosis/pathogenicity , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Multimerization , Virulence
17.
bioRxiv ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38260476

ABSTRACT

SARS-CoV-2, the virus responsible for COVID-19, triggers symptoms such as sneezing, aches and pain.1 These symptoms are mediated by a subset of sensory neurons, known as nociceptors, that detect noxious stimuli, densely innervate the airway epithelium, and interact with airway resident epithelial and immune cells.2-6 However, the mechanisms by which viral infection activates these neurons to trigger pain and airway reflexes are unknown. Here, we show that the coronavirus papain-like protease (PLpro) directly activates airway-innervating trigeminal and vagal nociceptors in mice and human iPSC-derived nociceptors. PLpro elicits sneezing and acute pain in mice and triggers the release of neuropeptide calcitonin gene-related peptide (CGRP) from airway afferents. We find that PLpro-induced sneeze and pain requires the host TRPA1 ion channel that has been previously demonstrated to mediate pain, cough, and airway inflammation.7-9 Our findings are the first demonstration of a viral product that directly activates sensory neurons to trigger pain and airway reflexes and highlight a new role for PLpro and nociceptors in COVID-19.

18.
Nat Microbiol ; 8(5): 819-832, 2023 05.
Article in English | MEDLINE | ID: mdl-37037941

ABSTRACT

Whether or not autophagy has a role in defence against Mycobacterium tuberculosis infection remains unresolved. Previously, conditional knockdown of the core autophagy component ATG5 in myeloid cells was reported to confer extreme susceptibility to M. tuberculosis in mice, whereas depletion of other autophagy factors had no effect on infection. We show that doubling cre gene dosage to more robustly deplete ATG16L1 or ATG7 resulted in increased M. tuberculosis growth and host susceptibility in mice, although ATG5-depleted mice are more sensitive than ATG16L1- or ATG7-depleted mice. We imaged individual macrophages infected with M. tuberculosis and identified a shift from apoptosis to rapid necrosis in autophagy-depleted cells. This effect was dependent on phagosome permeabilization by M. tuberculosis. We monitored infected cells by electron microscopy, showing that autophagy protects the host macrophage by partially reducing mycobacterial access to the cytosol. We conclude that autophagy has an important role in defence against M. tuberculosis in mammals.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Mice , Animals , Tuberculosis/microbiology , Autophagy/genetics , Macrophages/microbiology , Autophagy-Related Protein 5/genetics , Mammals
19.
PLoS Genet ; 5(12): e1000767, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20011113

ABSTRACT

Toxin-antitoxin (TA) systems, stress-responsive genetic elements ubiquitous in microbial genomes, are unusually abundant in the major human pathogen Mycobacterium tuberculosis. Why M. tuberculosis has so many TA systems and what role they play in the unique biology of the pathogen is unknown. To address these questions, we have taken a comprehensive approach to identify and functionally characterize all the TA systems encoded in the M. tuberculosis genome. Here we show that 88 putative TA system candidates are present in M. tuberculosis, considerably more than previously thought. Comparative genomic analysis revealed that the vast majority of these systems are conserved in the M. tuberculosis complex (MTBC), but largely absent from other mycobacteria, including close relatives of M. tuberculosis. We found that many of the M. tuberculosis TA systems are located within discernable genomic islands and were thus likely acquired recently via horizontal gene transfer. We discovered a novel TA system located in the core genome that is conserved across the genus, suggesting that it may fulfill a role common to all mycobacteria. By expressing each of the putative TA systems in M. smegmatis, we demonstrate that 30 encode a functional toxin and its cognate antitoxin. We show that the toxins of the largest family of TA systems, VapBC, act by inhibiting translation via mRNA cleavage. Expression profiling demonstrated that four systems are specifically activated during stresses likely encountered in vivo, including hypoxia and phagocytosis by macrophages. The expansion and maintenance of TA genes in the MTBC, coupled with the finding that a subset is transcriptionally activated by stress, suggests that TA systems are important for M. tuberculosis pathogenesis.


Subject(s)
Antitoxins/metabolism , Bacterial Toxins/metabolism , Biological Evolution , Mycobacterium tuberculosis/metabolism , Gene Expression , Genome, Bacterial , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity
20.
mBio ; 13(1): e0272621, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35073755

ABSTRACT

Ceragenins are a family of synthetic amphipathic molecules designed to mimic the properties of naturally occurring cationic antimicrobial peptides (CAMPs). Although ceragenins have potent antimicrobial activity, whether their mode of action is similar to that of CAMPs has remained elusive. Here, we reported the results of a comparative study of the bacterial responses to two well-studied CAMPs, LL37 and colistin, and two ceragenins with related structures, CSA13 and CSA131. Using transcriptomic and proteomic analyses, we found that Escherichia coli responded similarly to both CAMPs and ceragenins by inducing a Cpx envelope stress response. However, whereas E. coli exposed to CAMPs increased expression of genes involved in colanic acid biosynthesis, bacteria exposed to ceragenins specifically modulated functions related to phosphate transport, indicating distinct mechanisms of action between these two classes of molecules. Although traditional genetic approaches failed to identify genes that confer high-level resistance to ceragenins, using a Clustered Regularly Interspaced Short Palindromic Repeats interference (CRISPRi) approach we identified E. coli essential genes that when knocked down modify sensitivity to these molecules. Comparison of the essential gene-antibiotic interactions for each of the CAMPs and ceragenins identified both overlapping and distinct dependencies for their antimicrobial activities. Overall, this study indicated that, while some bacterial responses to ceragenins overlap those induced by naturally occurring CAMPs, these synthetic molecules target the bacterial envelope using a distinctive mode of action. IMPORTANCE The development of novel antibiotics is essential because the current arsenal of antimicrobials will soon be ineffective due to the widespread occurrence of antibiotic resistance. The development of naturally occurring cationic antimicrobial peptides (CAMPs) for therapeutics to combat antibiotic resistance has been hampered by high production costs and protease sensitivity, among other factors. The ceragenins are a family of synthetic CAMP mimics that kill a broad spectrum of bacterial species but are less expensive to produce, resistant to proteolytic degradation, and seemingly resistant to the development of high-level resistance. Determining how ceragenins function may identify new essential biological pathways of bacteria that are less prone to the development of resistance and will further our understanding of the design principles for maximizing the effects of synthetic CAMPs.


Subject(s)
Anti-Infective Agents , Antimicrobial Peptides , Escherichia coli , Proteomics , Bacteria , Anti-Infective Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Microbial Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL