Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
PLoS Pathog ; 14(4): e1007011, 2018 04.
Article in English | MEDLINE | ID: mdl-29709019

ABSTRACT

To subvert host defenses, Mycobacterium tuberculosis (Mtb) avoids being delivered to degradative phagolysosomes in macrophages by arresting the normal host process of phagosome maturation. Phagosome maturation arrest by Mtb involves multiple effectors and much remains unknown about this important aspect of Mtb pathogenesis. The SecA2 dependent protein export system is required for phagosome maturation arrest and consequently growth of Mtb in macrophages. To better understand the role of the SecA2 pathway in phagosome maturation arrest, we identified two effectors exported by SecA2 that contribute to this process: the phosphatase SapM and the kinase PknG. Then, utilizing the secA2 mutant of Mtb as a platform to study effector functions, we identified specific steps in phagosome maturation inhibited by SapM and/or PknG. By identifying a histidine residue that is essential for SapM phosphatase activity, we confirmed for the first time that the phosphatase activity of SapM is required for its effects on phagosome maturation in macrophages. We further demonstrated that SecA2 export of SapM and PknG contributes to the ability of Mtb to replicate in macrophages. Finally, we extended our understanding of the SecA2 pathway, SapM, and PknG by revealing that their contribution goes beyond preventing Mtb delivery to mature phagolysosomes and includes inhibiting Mtb delivery to autophagolysosomes. Together, our results revealed SapM and PknG to be two effectors exported by the SecA2 pathway of Mtb with distinct as well as cumulative effects on phagosome and autophagosome maturation. Our results further reveal that Mtb must have additional mechanisms of limiting acidification of the phagosome, beyond inhibiting recruitment of the V-ATPase proton pump to the phagosome, and they indicate differences between effects of Mtb on phagosome and autophagosome maturation.


Subject(s)
Adenosine Triphosphatases/metabolism , Autophagosomes/microbiology , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Macrophages/microbiology , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/pathogenicity , Phagosomes/microbiology , Tuberculosis/microbiology , Adenosine Triphosphatases/genetics , Animals , Autophagosomes/immunology , Autophagosomes/metabolism , Autophagy , Bacterial Proteins/genetics , Female , Lysosomes/immunology , Lysosomes/metabolism , Lysosomes/microbiology , Macrophages/immunology , Macrophages/metabolism , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Phagosomes/immunology , Phagosomes/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proton Pumps , Tuberculosis/immunology , Tuberculosis/metabolism
2.
Mol Microbiol ; 100(1): 90-107, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26712165

ABSTRACT

Mycobacterium tuberculosis proteins that are exported out of the bacterial cytoplasm are ideally positioned to be virulence factors; however, the functions of individual exported proteins remain largely unknown. Previous studies identified Rv0199 as an exported membrane protein of unknown function. Here, we characterized the role of Rv0199 in M. tuberculosis virulence using an aerosol model of murine infection. Rv0199 appears to be a member of a Mce-associated membrane (Mam) protein family leading us to rename it OmamA, for orphaned Mam protein A. Consistent with a role in Mce transport, we showed OmamA is required for cholesterol import, which is a Mce4-dependent process. We further demonstrated a function for OmamA in stabilizing protein components of the Mce1 transporter complex. These results indicate a function of OmamA in multiple Mce transporters and one that may be analogous to the role of VirB8 in stabilizing Type IV secretion systems, as structural similarities between Mam proteins and VirB8 proteins are predicted by the Phyre 2 program. In this study, we provide functional information about OmamA and shed light on the function of Mam family proteins in Mce transporters.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Animals , Bacterial Proteins/genetics , Cholesterol/metabolism , Disease Models, Animal , Gene Deletion , Gene Order , Membrane Proteins/genetics , Mice , Mutation , Mycobacterium tuberculosis/genetics , Phenotype , Protein Binding , Protein Transport , Tuberculosis/microbiology , Tuberculosis/mortality , Tuberculosis/pathology , Virulence Factors
3.
Infect Immun ; 80(3): 996-1006, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22215736

ABSTRACT

The ability of Mycobacterium tuberculosis to grow in macrophages is critical to the virulence of this important pathogen. One way M. tuberculosis is thought to maintain a hospitable niche in macrophages is by arresting the normal process of phagosomes maturing into acidified phagolysosomes. The process of phagosome maturation arrest by M. tuberculosis is not fully understood, and there has remained a need to firmly establish a requirement for phagosome maturation arrest for M. tuberculosis growth in macrophages. Other intracellular pathogens that control the phagosomal environment use specialized protein export systems to deliver effectors of phagosome trafficking to the host cell. In M. tuberculosis, the accessory SecA2 system is a specialized protein export system that is required for intracellular growth in macrophages. In studying the importance of the SecA2 system in macrophages, we discovered that SecA2 is required for phagosome maturation arrest. Shortly after infection, phagosomes containing a ΔsecA2 mutant of M. tuberculosis were more acidified and showed greater association with markers of late endosomes than phagosomes containing wild-type M. tuberculosis. We further showed that inhibitors of phagosome acidification rescued the intracellular growth defect of the ΔsecA2 mutant, which demonstrated that the phagosome maturation arrest defect of the ΔsecA2 mutant is responsible for the intracellular growth defect. This study demonstrates the importance of phagosome maturation arrest for M. tuberculosis growth in macrophages, and it suggests there are effectors of phagosome maturation that are exported into the host environment by the accessory SecA2 system.


Subject(s)
Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Immune Evasion , Macrophages/microbiology , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/pathogenicity , Phagosomes/microbiology , Virulence Factors/metabolism , Adenosine Triphosphatases/genetics , Animals , Bacterial Proteins/genetics , Gene Deletion , Macrophages/immunology , Membrane Transport Proteins/genetics , Mice , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Phagosomes/immunology , Virulence Factors/genetics
4.
J Bacteriol ; 193(4): 854-61, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21148733

ABSTRACT

The exported proteins of Mycobacterium tuberculosis that are localized at the bacterial cell surface or secreted into the environment are ideally situated to interact with host factors and to function in virulence. In this study, we constructed a novel ß-lactamase reporter transposon and used it directly in M. tuberculosis for genome-wide identification of exported proteins. From 177 ß-lactam-resistant transposon mutants, we identified 111 different exported proteins. The majority of these proteins have no known function, and for nearly half of the proteins, our demonstration that they are exported when fused to a ß-lactamase reporter is the first experimental proof of their extracytoplasmic localization. The transposon mutants in our banked library were of further value as a collection of mutants lacking individual exported proteins. By individually testing each of 111 mutants for growth in macrophages, six attenuated mutants with insertions in mce1A, mce1B, mce2F, rv0199, ctaC, and lppX were identified. Given that much of the M. tuberculosis genome encodes proteins of unknown function, our library of mapped transposon mutants is a valuable resource for efforts in functional genomics. This work also demonstrates the power of a ß-lactamase reporter transposon that could be applied similarly to other bacterial pathogens.


Subject(s)
Bacterial Proteins/metabolism , Extracellular Space/metabolism , Genome, Bacterial , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Tuberculosis/microbiology , Animals , Bacterial Proteins/genetics , Cells, Cultured , Extracellular Space/genetics , Female , Gene Expression Regulation, Bacterial , Genes, Reporter , Humans , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mycobacterium tuberculosis/genetics , Protein Transport
5.
PLoS One ; 15(7): e0234792, 2020.
Article in English | MEDLINE | ID: mdl-32614850

ABSTRACT

The Myo/Nog cell lineage was discovered in the chick embryo and is also present in adult mammalian tissues. The cells are named for their expression of mRNA for the skeletal muscle specific transcription factor MyoD and bone morphogenetic protein inhibitor Noggin. A third marker for Myo/Nog cells is the cell surface molecule recognized by the G8 monoclonal antibody (mAb). G8 has been used to detect, track, isolate and kill Myo/Nog cells. In this study, we screened a membrane proteome array for the target of the G8 mAb. The array consisted of >5,000 molecules, each synthesized in their native confirmation with appropriate post-translational modifications in a single clone of HEK-293T cells. G8 mAb binding to the clone expressing brain-specific angiogenesis inhibitor 1 (BAI1) was detected by flow cytometry, re-verified by sequencing and validated by transfection with the plasmid construct for BAI1. Further validation of the G8 target was provided by enzyme-linked immunosorbent assay. The G8 epitope was identified by screening a high-throughput, site directed mutagenesis library designed to cover 95-100% of the 954 amino acids of the extracellular domain of the BAI1 protein. The G8 mAb binds within the third thrombospondin repeat of the extracellular domain of human BAI1. Immunofluorescence localization experiments revealed that G8 and a commercially available BAI1 mAb co-localize to the subpopulation of Myo/Nog cells in the skin, eyes and brain. Expression of the multi-functional BAI1 protein in Myo/Nog cells introduces new possibilities for the roles of Myo/Nog cells in normal and diseased tissues.


Subject(s)
Angiogenic Proteins/biosynthesis , Myofibroblasts/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Amino Acid Substitution , Angiogenic Proteins/chemistry , Angiogenic Proteins/genetics , Angiogenic Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Antigen-Antibody Reactions , Brain/cytology , Carrier Proteins/analysis , Cell Lineage , Epitopes/immunology , Eye Proteins/biosynthesis , Eye Proteins/chemistry , Eye Proteins/genetics , Eye Proteins/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Muscle Development , MyoD Protein/analysis , Organ Specificity , Protein Conformation , Protein Domains , Rabbits , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Repetitive Sequences, Amino Acid , Skin/cytology , Species Specificity , Tattooing , Young Adult
6.
Future Microbiol ; 5(10): 1581-97, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21073315

ABSTRACT

Protein export is essential in all bacteria and many bacterial pathogens depend on specialized protein export systems for virulence. In Mycobacterium tuberculosis, the etiological agent of the disease tuberculosis, the conserved general secretion (Sec) and twin-arginine translocation (Tat) pathways perform the bulk of protein export and are both essential. M. tuberculosis also has specialized export pathways that transport specific subsets of proteins. One such pathway is the accessory SecA2 system, which is important for M. tuberculosis virulence. There are also specialized ESX export systems that function in virulence (ESX-1) or essential physiologic processes (ESX-3). The increasing prevalence of drug-resistant M. tuberculosis strains makes the development of novel drugs for tuberculosis an urgent priority. In this article, we discuss our current understanding of the protein export systems of M. tuberculosis and consider the potential of these pathways to be novel targets for tuberculosis drugs.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Bacterial Proteins/antagonists & inhibitors , Humans , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity
7.
J Bacteriol ; 188(11): 3785-95, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16707671

ABSTRACT

We determined that LVS and Schu S4 strains of the human pathogen Francisella tularensis express a siderophore when grown under iron-limiting conditions. We purified this siderophore by conventional column chromatography and high-pressure liquid chromatography and used mass spectrometric analysis to demonstrate that it is structurally similar to the polycarboxylate siderophore rhizoferrin. The siderophore promoted the growth of LVS and Schu S4 strains in iron-limiting media. We identified a potential siderophore biosynthetic gene cluster encoded by fslABCD in the F. tularensis genome. The first gene in the cluster, fslA, encodes a member of the superfamily of nonribosomal peptide synthetase-independent siderophore synthetases (NIS synthetases) characterized by the aerobactin synthetases IucA and IucC. We determined that fslA is transcribed as part of an operon with downstream gene fslB and that the expression of the locus is induced by iron starvation. A targeted in-frame nonpolar deletion of fslA in LVS resulted in the loss of siderophore expression and in a reduced ability of F. tularensis to grow under conditions of iron limitation. Siderophore activity and the ability to grow under iron limitation could be regained by introducing the fslA(+) gene on a complementing plasmid. Our results suggest that the fslA-dependent siderophore is important for survival of F. tularensis in an iron-deficient environment.


Subject(s)
Francisella tularensis/metabolism , Siderophores/metabolism , Base Sequence , Chromatography, High Pressure Liquid , DNA Primers , Ferric Compounds/metabolism , Francisella tularensis/growth & development , Iron/metabolism , Kinetics , Mass Spectrometry , Molecular Sequence Data , Polymerase Chain Reaction , Siderophores/chemistry , Siderophores/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL