Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 520
Filter
1.
Life Sci ; 277: 119502, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33891941

ABSTRACT

AIM: Chlamydia trachomatis has evolved various strategies to alleviate oxidative stress of host cells to maintain their intracellular survival. However, the exact mechanism of anti-oxidative stress of C. trachomatis is still unclear. The activation of nuclear factor erythroid 2-related factor 2/quinone oxidoreductase (Nrf2/NQO1) signal pathway has been identified as an efficient antioxidant defensive mechanism used by host cells to counteract oxidative stress. Pgp3 is a pivotal virulence factor of C. trachomatis involved in intracellular survival. The aim of this study is to explore the role of Pgp3 on Nrf2/NQO1 signal pathway against oxidative stress. MAIN METHODS: After HeLa cells were stimulated with Pgp3 protein, Nrf2 location and the inclusion bodies of C. trachomatis were detected by indirect immunofluorescence, western blotting and Oxidative stress assay kits were used to separately determine the protein expression and the content of malondialdehyde (MDA), superoxide dismutase (SOD) and total antioxidant capacity (T-AOC) before and after the interference of Nrf-2 and NQO1. KEY FINDINGS: Pgp3 promoted the nuclear translocation of Nrf2 to increase NQO1 expression and reduced oxidative stress induced by LPS to contribute to the survival of C. trachomatis. Inhibition of Nrf2/NQO1 signal pathway with Nrf2 inhibitor and down-regulation of NQO1 with siRNA-NQO1 suppressed oxidative stress resistance induced by Pgp3. SIGNIFICANCE: Here we found that Pgp3 alleviated oxidative stress to promote the infectivity of C. trachomatis through activation of Nrf2/NQO1 signal pathway, which provided a novel understanding of the effects of Pgp3 in the pathogenesis of C. trachomatis.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Chlamydia trachomatis/metabolism , Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Cell Survival/drug effects , HeLa Cells , Heme Oxygenase-1/metabolism , Humans , Malondialdehyde/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Superoxide Dismutase/metabolism
2.
J Biol Chem ; 296: 100460, 2021.
Article in English | MEDLINE | ID: mdl-33639171

ABSTRACT

Bacterial survival during lethal heat stress relies on the cellular ability to reactivate aggregated proteins. This activity is typically executed by the canonical 70-kDa heat shock protein (Hsp70)-ClpB bichaperone disaggregase, which is most widespread in bacteria. The ClpB disaggregase is a member of the ATPase associated with diverse cellular activities protein family and exhibits an ATP-driven threading activity. Substrate binding and stimulation of ATP hydrolysis depends on the Hsp70 partner, which initiates the disaggregation reaction. Recently elevated heat resistance in gamma-proteobacterial species was shown to be mediated by the ATPase associated with diverse cellular activities protein ClpG as an alternative disaggregase. Pseudomonas aeruginosa ClpG functions autonomously and does not cooperate with Hsp70 for substrate binding, enhanced ATPase activity, and disaggregation. With the underlying molecular basis largely unknown, the fundamental differences in ClpG- and ClpB-dependent disaggregation are reflected by the presence of sequence alterations and additional ClpG-specific domains. By analyzing the effects of mutants lacking ClpG-specific domains and harboring mutations in conserved motifs implicated in ATP hydrolysis and substrate threading, we show that the N-terminal, ClpG-specific N1 domain generally mediates protein aggregate binding as the molecular basis of autonomous disaggregation activity. Peptide substrate binding strongly stimulates ClpG ATPase activity by overriding repression by the N-terminal N1 and N2 domains. High ATPase activity requires two functional nucleotide binding domains and drives substrate threading which ultimately extracts polypeptides from the aggregate. ClpG ATPase and disaggregation activity is thereby directly controlled by substrate availability.


Subject(s)
Antigens, Bacterial/metabolism , Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Antigens, Bacterial/physiology , Endopeptidase Clp/physiology , Escherichia coli/metabolism , Escherichia coli Proteins/physiology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/physiology , Protein Aggregates , Protein Binding , Protein Domains/genetics
3.
Mol Immunol ; 130: 77-84, 2021 02.
Article in English | MEDLINE | ID: mdl-33246580

ABSTRACT

Regarding the role of micro RNAs (miRNA) in the proliferation and differentiation of T cells as well as the controversy around the role of bacteria in the pathogenesis of abdominal aortic aneurysm (AAA), the effects of Helicobacter pylori (Hp) and Lactobacillus acidophilus (La) were investigated in the induction of miRNAs and apoptosis in CD4+ memory T (Tem) cells of AAA patients and controls. Signature atherosclerosis miRNAs 21, 92a, 146a, 155, 326 and 663 were measured in the sera and tissues of AAA patients and control. PBMCs separately and in co-culture with HUVEC were treated with Hp-water-extract (HpWE) and La-conditioned-medium (LaCM). Apoptosis and miRNA levels were assessed in the isolated Tem by flowcytometry and real-time-PCR. In single-culture, HpWE increased apoptosis and miR-155 and LaCM decreased apoptosis and increased miR-21. In co-culture, apoptosis decreased in both groups in response to CagA+HpWE. Also, all miRNAs increased in patients Tem but in controls, only miR- 146a and 21 showed changes. Although, apoptosis was similar in Tem of patients and controls, the effects of Hp and La were different on the induction of apoptosis and miRNAs and also these bacteria showed different impacts in single and co-culture conditions. Beyond the direct effects of these bacteria on the pathogenesis of diseases, their effects on miRNAs expression may shed light on their roles in the development and the prevention of AAA.


Subject(s)
Aortic Aneurysm, Abdominal/immunology , Helicobacter pylori/physiology , Lactobacillus acidophilus/physiology , MicroRNAs/genetics , T-Lymphocytes/metabolism , Aged , Antigens, Bacterial/pharmacology , Antigens, Bacterial/physiology , Aortic Aneurysm, Abdominal/pathology , Case-Control Studies , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Immunologic Memory/genetics , Male , MicroRNAs/metabolism , Middle Aged , T-Lymphocytes/drug effects , T-Lymphocytes/pathology
4.
Cells ; 10(1)2020 12 25.
Article in English | MEDLINE | ID: mdl-33375694

ABSTRACT

Gastric cancer constitutes one of the most prevalent malignancies in both sexes; it is currently the fourth major cause of cancer-related deaths worldwide. The pathogenesis of gastric cancer is associated with the interaction between genetic and environmental factors, among which infection by Helicobacter pylori (H. pylori) is of major importance. The invasion, survival, colonization, and stimulation of further inflammation within the gastric mucosa are possible due to several evasive mechanisms induced by the virulence factors that are expressed by the bacterium. The knowledge concerning the mechanisms of H. pylori pathogenicity is crucial to ameliorate eradication strategies preventing the possible induction of carcinogenesis. This review highlights the current state of knowledge and the most recent findings regarding H. pylori virulence factors and their relationship with gastric premalignant lesions and further carcinogenesis.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Virulence Factors/physiology , Animals , Carcinogenesis/pathology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Humans
5.
FASEB J ; 33(11): 12324-12335, 2019 11.
Article in English | MEDLINE | ID: mdl-31442078

ABSTRACT

GNA2091 is one of the components of the 4-component meningococcal serogroup B vaccine (4CMenB) vaccine and is highly conserved in all meningococcal strains. However, its functional role has not been fully characterized. Here we show that nmb2091 is part of an operon and is cotranscribed with the nmb2089, nmb2090, and nmb2092 adjacent genes, and a similar but reduced operon arrangement is conserved in many other gram-negative bacteria. Deletion of the nmb2091 gene causes an aggregative phenotype with a mild defect in cell separation; differences in the outer membrane composition and phospholipid profile, in particular in the phosphoethanolamine levels; an increased level of outer membrane vesicles; and deregulation of the zinc-responsive genes such as znuD. Finally, the ∆2091 strain is attenuated with respect to the wild-type strain in competitive index experiments in the infant rat model of meningococcal infection. Altogether these data suggest that GNA2091 plays important roles in outer membrane architecture, biogenesis, homeostasis, and in meningococcal survival in vivo, and a model for its role is discussed. These findings highlight the importance of GNA2091 as a vaccine component.-Seib, K. L., Haag, A. F., Oriente, F., Fantappiè, L., Borghi, S., Semchenko, E. A., Schulz, B. L., Ferlicca, F., Taddei, A. R., Giuliani, M. M., Pizza, M., Delany, I. The meningococcal vaccine antigen GNA2091 is an analogue of YraP and plays key roles in outer membrane stability and virulence.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Outer Membrane/chemistry , Meningococcal Vaccines , Animals , Antigens, Bacterial/genetics , Bacterial Outer Membrane/physiology , Meningococcal Infections/mortality , Meningococcal Vaccines/genetics , Neisseria meningitidis, Serogroup B/genetics , Neisseria meningitidis, Serogroup B/pathogenicity , Operon , Periplasmic Proteins/physiology , Rats , Rats, Wistar , Regulon , Virulence , Zinc/pharmacology
6.
J Immunol ; 203(5): 1218-1229, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31375544

ABSTRACT

Mycobacterium tuberculosis employs defense mechanisms to protect itself from reactive oxygen species (ROS)-mediated cytotoxicity inside macrophages. In the current study, we found that a secretory protein of M. tuberculosis PPE2 disrupted the assembly of NADPH oxidase complex. PPE2 inhibited NADPH oxidase-mediated ROS generation in RAW 264.7 macrophages and peritoneal macrophages from BALB/c mice. PPE2 interacted with the cytosolic subunit of NADPH oxidase, p67phox, and prevented translocation of p67phox and p47phox to the membrane, resulting in decreased NADPH oxidase activity. Trp236 residue present in the SH3-like domain of PPE2 was found to be critical for its interaction with p67phox Trp236Ala mutant of PPE2 did not interact with p67phox and thereby did not affect ROS generation. M. tuberculosis expressing PPE2 and PPE2-null mutants complemented with PPE2 survived better than PPE2-null mutants in infected RAW 264.7 macrophages. Altogether, this study suggests that PPE2 inhibits NADPH oxidase-mediated ROS production to favor M. tuberculosis survival in macrophages. The findings that M. tuberculosis PPE2 protein is involved in the modulation of oxidative response in macrophages will help us in improving our knowledge of host-pathogen interactions and the application of better therapeutics against tuberculosis.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Phosphoproteins/physiology , Reactive Oxygen Species/metabolism , Animals , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Host-Pathogen Interactions , Macrophages/metabolism , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Tetradecanoylphorbol Acetate/pharmacology , src Homology Domains
7.
FASEB J ; 33(10): 10808-10818, 2019 10.
Article in English | MEDLINE | ID: mdl-31262188

ABSTRACT

Colonization of the oropharynx is the initial step in Group A Streptococcus (GAS) pharyngeal infection. We have previously reported that the highly virulent M1T1 GAS clone attaches to oral epithelial cells via M1 protein interaction with blood group antigen carbohydrate structures. Here, we have identified that colonization of human oral epithelial cells by GAS serotypes M3 and M12 is mediated by human blood group antigens [ABO(H)] and Lewis (Le) antigen expression. Removal of linkage-specific fucose, galactose, N-acetylgalactosamine, and sialic acid modulated GAS colonization, dependent on host ABO(H) blood group and Le expression profile. Furthermore, N-linked glycans from human salivary glycoproteins, when released and purified, were potent inhibitors of M1, M3, and M12 GAS colonization ex vivo. These data highlight the important role played by human protein glycosylation patterns in GAS attachment to oral epithelial cell surfaces.-De Oliveira, D. M. P., Everest-Dass, A., Hartley-Tassell, L., Day, C. J., Indraratna, A., Brouwer, S., Cleary, A., Kautto, L., Gorman, J., Packer, N. H., Jennings, M. P., Walker, M. J., Sanderson-Smith, M. L. Human glycan expression patterns influence Group A streptococcal colonization of epithelial cells.


Subject(s)
Host Microbial Interactions/physiology , Polysaccharides/metabolism , Streptococcus pyogenes/pathogenicity , Antigens, Bacterial/physiology , Bacterial Adhesion/immunology , Bacterial Adhesion/physiology , Bacterial Outer Membrane Proteins/physiology , Blood Group Antigens/chemistry , Carrier Proteins/physiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Glycosylation , Host Microbial Interactions/immunology , Humans , In Vitro Techniques , Polysaccharides/chemistry , Polysaccharides/immunology , Protein Binding , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/immunology , Salivary Proteins and Peptides/metabolism , Streptococcal Infections/etiology , Streptococcal Infections/microbiology , Streptococcus pyogenes/growth & development , Streptococcus pyogenes/physiology , Virulence/physiology
8.
Cell Mol Immunol ; 16(4): 380-391, 2019 04.
Article in English | MEDLINE | ID: mdl-29572547

ABSTRACT

The intracellular pathogen Mycobacterium tuberculosis (Mtb) can survive in the host and cause disease by interfering with a variety of cellular functions. The mammalian cell entry 2 (mce2) operon of Mtb has been shown to contribute to tuberculosis pathogenicity. However, little is known about the regulatory roles of Mtb Mce2 family proteins towards host cellular functions. Here we show that the Mce2 family protein Mce2E suppressed the macrophage innate immune response and promoted epithelial cell proliferation. Mce2E inhibited activation of the extracellular signal-regulated kinase (ERK) and Jun N-terminal kinase (JNK) mitogen-activated protein kinase (MAPK) signaling pathways in a non-canonical D motif (a MAPK-docking motif)-dependent manner, leading to reduced expression of TNF and IL-6 in macrophages. Furthermore, Mce2E promoted proliferation of human lung epithelium-derived lung adenoma A549 cells by inhibiting K48-linked polyubiquitination of eEF1A1 in a ß strand region-dependent manner. In summary, Mce2E is a novel multifunctional Mtb virulence factor that regulates host cellular functions in a niche-dependent manner. Our data suggest a potential novel target for TB therapy.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Epithelial Cells/metabolism , MAP Kinase Signaling System , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Amino Acid Motifs/genetics , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cytokines/metabolism , Female , Humans , Immunity, Innate , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mycobacterium tuberculosis/pathogenicity , Peptide Elongation Factor 1/metabolism , Transplantation, Heterologous , Ubiquitination/genetics , Ubiquitination/immunology , Virulence Factors/metabolism
9.
Biochim Biophys Acta Biomembr ; 1860(11): 2192-2203, 2018 11.
Article in English | MEDLINE | ID: mdl-30409515

ABSTRACT

Anthrax toxin action requires triggering of natural endocytic transport mechanisms whereby the binding component of the toxin forms channels (PA63) within endosomal limiting and intraluminal vesicle membranes to deliver the toxin's enzymatic components into the cytosol. Membrane lipid composition varies at different stages of anthrax toxin internalization, with intraluminal vesicle membranes containing ~70% of anionic bis(monoacylglycero)phosphate lipid. Using model bilayer measurements, we show that membrane lipids can have a strong effect on the anthrax toxin channel properties, including the channel-forming activity, voltage-gating, conductance, selectivity, and enzymatic factor binding. Interestingly, the highest PA63 insertion rate was observed in bis(monoacylglycero)phosphate membranes. The molecular dynamics simulation data show that the conformational properties of the channel are different in bis(monoacylglycero)phosphate compared to PC, PE, and PS lipids. The anthrax toxin protein/lipid bilayer system can be advanced as a novel robust model to directly investigate lipid influence on membrane protein properties and protein/protein interactions.


Subject(s)
Antigens, Bacterial/physiology , Endosomes/metabolism , Lipid Bilayers/metabolism , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Biochemical Phenomena , Biological Transport , Electrophysiological Phenomena , Molecular Dynamics Simulation , Protein Binding
10.
Trends Microbiol ; 26(12): 1008-1021, 2018 12.
Article in English | MEDLINE | ID: mdl-30037568

ABSTRACT

Bacterial pathogens and commensals are surrounded by diverse surface polysaccharides which include capsules and lipopolysaccharides. These carbohydrates play a vital role in bacterial ecology and interactions with the environment. Here, we review recent rapid advancements in this field, which have improved our understanding of the roles, structures, and genetics of bacterial polysaccharide antigens. Genetic loci encoding the biosynthesis of these antigens may have evolved as bacterial diversity-generating machines, driven by selection from a variety of forces, including host immunity, bacteriophages, and cell-cell interactions. We argue that the high adaptive potential of polysaccharide antigens should be taken into account in the design of polysaccharide-targeting medical interventions like conjugate vaccines and phage-based therapies.


Subject(s)
Bacteria/genetics , Bacteria/metabolism , Bacterial Capsules/chemistry , Bacterial Capsules/genetics , Bacterial Capsules/physiology , Biodiversity , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/physiology , Bacteria/pathogenicity , Bacterial Capsules/immunology , Cell Communication , Ecology , Evolution, Molecular , Genetic Loci , Humans , Immunity , Lipopolysaccharides/classification , Lipopolysaccharides/genetics , Lipopolysaccharides/immunology , Lipopolysaccharides/physiology , Phage Therapy , Polysaccharides , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/immunology , Polysaccharides, Bacterial/physiology , Serotyping , Symbiosis , Vaccines, Conjugate
11.
EMBO J ; 37(13)2018 07 02.
Article in English | MEDLINE | ID: mdl-29724755

ABSTRACT

Helicobacter pylori infects half of the world's population, and strains that encode the cag type IV secretion system for injection of the oncoprotein CagA into host gastric epithelial cells are associated with elevated levels of cancer. CagA translocation into host cells is dependent on interactions between the H. pylori adhesin protein HopQ and human CEACAMs. Here, we present high-resolution structures of several HopQ-CEACAM complexes and CEACAMs in their monomeric and dimeric forms establishing that HopQ uses a coupled folding and binding mechanism to engage the canonical CEACAM dimerization interface for CEACAM recognition. By combining mutagenesis with biophysical and functional analyses, we show that the modes of CEACAM recognition by HopQ and CEACAMs themselves are starkly different. Our data describe precise molecular mechanisms by which microbes exploit host CEACAMs for infection and enable future development of novel oncoprotein translocation inhibitors and H. pylori-specific antimicrobial agents.


Subject(s)
Antigens, Bacterial/physiology , Antigens, CD/physiology , Bacterial Proteins/physiology , Cell Adhesion Molecules/physiology , Helicobacter pylori/physiology , Oncogene Proteins/physiology , Antigens, CD/chemistry , Bacterial Proteins/chemistry , Cell Adhesion Molecules/chemistry , HEK293 Cells , Humans , Mutagenesis , Protein Multimerization , Protein Transport
12.
Protein Sci ; 27(8): 1392-1406, 2018 08.
Article in English | MEDLINE | ID: mdl-29672980

ABSTRACT

Bacterial type III secretion systems (T3SS) are used to inject proteins into mammalian cells to subvert cellular functions. The Shigella T3SS apparatus (T3SA) is comprised of a basal body, cytoplasmic sorting platform and exposed needle with needle "tip complex" (TC). TC maturation occurs when the translocator protein IpaB is recruited to the needle tip where both IpaD and IpaB control secretion induction. IpaB insertion into the host membrane is the first step of translocon pore formation and secretion induction. We employed disruptive insertional mutagenesis, using bacteriophage T4 lysozyme (T4L), within predicted IpaB loops to show how topological features affect TC functions (secretion control, translocon formation and effector secretion). Insertions within the N-terminal half of IpaB were most likely to result in a loss of steady-state secretion control, however, all but the two that were not recognized by the T3SA retained nearly wild-type hemolysis (translocon formation) and invasiveness levels (effector secretion). In contrast, all but one insertion in the C-terminal half of IpaB maintained secretion control but were impaired for hemolysis and invasion. These nature of the data suggest the latter mutants are defective in a post-secretion event, most likely due to impaired interactions with the second translocator protein IpaC. Intriguingly, only two insertion mutants displayed readily detectable T4L on the bacterial surface. The data create a picture in which the makeup and structure of a functional T3SA TC is highly amenable to physical perturbation, indicating that the tertiary structure of IpaB within the TC is more plastic than previously realized.


Subject(s)
Bacterial Proteins , Mutagenesis, Insertional/methods , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Antigens, Bacterial/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Cells, Cultured , Erythrocytes , Hemolysis , Sheep , Type III Secretion Systems , X-Ray Diffraction
13.
Microb Pathog ; 113: 176-180, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29038054

ABSTRACT

Erysipelothrix rhusiopathiae is the causative agent of animal erysipelas and human erysipeloid. The major protective antigen SpaA was suggested to play important roles in E. rhusiopathiae adhesion to host cells, but there is no specific study on SpaA pathogenic roles in adhesion. In this study we characterized direct and indirect roles of SpaA in E. rhusiopathiae adhesion to porcine endothelial cells. Recombinant E. rhusiopathiae SpaA (rSpaA) successfully binded to porcine iliac arterial endothelial cells. rSpaA protein pre-incubating endothelial cells or rSpaA antiserum pre-incubating E. rhusiopathiae significantly decreased E. rhusiopathiae adhesion to endothelial cells. rSpaA successfully binded host plasminogen and fibronectin, and rSpaA antiserum significantly decreased plasminogen-recruitment activity but not fibronectin-recruitment activity of E. rhusiopathiae. In conclusion, SpaA acts as adhesin in E. rhusiopathiae adhesion to host cells, and SpaA binding activity to host plasminogen highly likely play roles in this adhesion.


Subject(s)
Adhesins, Bacterial/physiology , Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Endothelial Cells/microbiology , Erysipelothrix/physiology , Adhesins, Bacterial/immunology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Fibronectins/metabolism , Host-Pathogen Interactions/physiology , Humans , Iliac Artery/microbiology , Plasminogen/metabolism , Protein Binding/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Swine , Virulence Factors/physiology
14.
Vet Res ; 48(1): 39, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28705175

ABSTRACT

Streptococcus suis is one of the most important post-weaning porcine bacterial pathogens worldwide. The serotypes 2 and 9 are often considered the most virulent and prevalent serotypes involved in swine infections, especially in Europe. However, knowledge of the bacterial factors involved in the first steps of the pathogenesis of the infection remains scarce. In several pathogenic streptococci, expression of multimodal adhesion proteins known as antigen I/II (AgI/II) have been linked with persistence in the upper respiratory tract and the oral cavity, as well as with bacterial dissemination. Herein, we report expression of these immunostimulatory factors by S. suis serotype 2 and 9 strains and that AgI/II-encoding genes are carried by integrative and conjugative elements. Using mutagenesis and different in vitro assays, we demonstrate that the contribution of AgI/II to the virulence of the serotype 2 strain used herein appears to be modest. In contrast, data demonstrate that the serotype 9 AgI/II participates in self-aggregation, induces salivary glycoprotein 340-related aggregation, contributes to biofilm formation and increased strain resistance to low pH, as well as in bacterial adhesion to extracellular matrix proteins and epithelial cells. Moreover, the use of a porcine infection model revealed that AgI/II contributes to colonization of the upper respiratory tract of pigs. Taken together, these findings suggest that surface exposed AgI/II likely play a key role in the first steps of the pathogenesis of the S. suis serotype 9 infection.


Subject(s)
Streptococcal Infections/veterinary , Streptococcus suis/pathogenicity , Swine Diseases/microbiology , Animals , Antigens, Bacterial/metabolism , Antigens, Bacterial/physiology , Bacterial Adhesion/physiology , Streptococcal Infections/microbiology , Swine
15.
J Immunol ; 198(12): 4772-4780, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28507027

ABSTRACT

Tuberculosis caused by Mycobacterium tuberculosis continues to pose a serious global health threat. The attenuated Mycobacterium bovis bacillus Calmette-Guérin, as the only licensed vaccine, has limited protective efficacy against TB. The development of more effective antituberculosis vaccines is urgent and demands for further identification and understanding of M. tuberculosis Ags. MPT83 (Rv2873), a secreted mycobacterial lipoprotein, has been applied into subunit vaccine development and shown protective effects against M. tuberculosis infection in animals; however, the understanding of the underlying mechanism is limited. In present study, we systematically studied the effect of MPT83 on macrophage apoptosis by constructing Mycobacterium smegmatis strain overexpressing MPT83 (MS_MPT83) and purifying rMPT83 protein. We found that MPT83 induced apoptosis in both human and mouse macrophages. MPT83 induced cyclooxygenase-2 (COX-2) expression at both the transcriptional and protein levels in macrophages, whereas silencing or inhibiting COX-2 blocked rMPT83-induced apoptosis or the enhanced apoptotic response to MS_MPT83 in comparison with M. smegmatis transfected with pMV261 vector (MS_Vec), indicating that COX-2 is required for MPT83-induced apoptosis. Additionally, tlr2 deficiency led to significant reduction of COX-2 expression, accompanied by less apoptosis in macrophages stimulated with rMPT83 or infected with MS_MPT83. Moreover, the activation of p38 accounted for MPT83-induced COX-2 expression. Finally, lower bacteria burdens in the lungs and spleens and enhanced survival were observed in mice i.v. infected with MS_MPT83 compared with MS_Vec. Taken together, our results established a proapoptotic effect of MPT83 and identified the TLR2/p38/COX-2 axis in MPT83-induced macrophage apoptosis.


Subject(s)
Antigens, Bacterial/physiology , Apoptosis , Bacterial Proteins/physiology , Cyclooxygenase 2/metabolism , Macrophages/microbiology , Macrophages/physiology , Membrane Proteins/physiology , Signal Transduction/immunology , Toll-Like Receptor 2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/isolation & purification , Apoptosis/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Cyclooxygenase 2/immunology , Macrophages/drug effects , Macrophages/immunology , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Mice , Mycobacterium smegmatis/immunology , Phosphorylation , Toll-Like Receptor 2/immunology
16.
Microbiol Spectr ; 5(3)2017 05.
Article in English | MEDLINE | ID: mdl-28513416

ABSTRACT

In this article, we have described several cellular pathological effects caused by the Mycobacterium tuberculosis ESX-1. The effects include induction of necrosis, NOD2 signaling, type I interferon production, and autophagy. We then attempted to suggest that these pathological effects are mediated by the cytosolic access of M. tuberculosis-derived materials as a result of the phagosome-disrupting activity of the major ESX-1 substrate ESAT-6. Such activity of ESAT-6 is most likely due to its pore-forming activity at the membrane. The amyloidogenic characteristic of ESAT-6 is reviewed here as a potential mechanism of membrane pore formation. In addition to ESAT-6, the ESX-1 substrate EspB interferes with membrane-mediated innate immune mechanisms such as efferocytosis and autophagy, most likely through its ability to bind phospholipids. Overall, the M. tuberculosis ESX-1 secretion system appears to be a specialized system for the deployment of host membrane-targeting proteins, whose primary function is to interrupt key steps in innate immune mechanisms against pathogens. Inhibitors that block the ESX-1 system or block host factors critical for ESX-1 toxicity have been identified and should represent attractive potential new antituberculosis drugs.


Subject(s)
Antigens, Bacterial/metabolism , Antigens, Bacterial/physiology , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/microbiology , Antigens, Bacterial/immunology , Autophagy , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/immunology , Gene Expression Regulation, Bacterial , Immunity, Innate , Interferon Type I/metabolism , Mycobacterium tuberculosis/immunology , Nod2 Signaling Adaptor Protein , Phagosomes/metabolism , Phospholipids/metabolism , Protein Transport , Tuberculosis/drug therapy , Tuberculosis/immunology , Type VII Secretion Systems
18.
Sci Rep ; 7: 43039, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220899

ABSTRACT

Histones are essential elements of chromatin structure and gene regulation in eukaryotes. An unexpected attribute of these nuclear proteins is their antimicrobial activity. A framework for histone release and function in host defense in vivo was revealed with the discovery of neutrophil extracellular traps, a specialized cell death process in which DNA-based structures containing histones are extruded to ensnare and kill bacteria. Investigating the susceptibility of various Gram-positive pathogens to histones, we found high-level resistance by one leading human pathogen, group A Streptococcus (GAS). A screen of isogenic mutants revealed that the highly surface-expressed M1 protein, a classical GAS virulence factor, was required for high-level histone resistance. Biochemical and microscopic analyses revealed that the N-terminal domain of M1 protein binds and inactivates histones before they reach their cell wall target of action. This finding illustrates a new pathogenic function for this classic GAS virulence factor, and highlights a potential innate immune evasion strategy that may be employed by other bacterial pathogens.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Outer Membrane Proteins/physiology , Carrier Proteins/physiology , Histones/metabolism , Immune Evasion , Neutrophils/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/physiology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , Humans , Neutrophils/metabolism , Streptococcal Infections/metabolism , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/pathogenicity , Virulence Factors/metabolism , Virulence Factors/physiology
19.
Apoptosis ; 22(4): 502-509, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27987050

ABSTRACT

Mycobacterium tuberculosis PE/PPE family proteins, named after the presence of conserved PE (Pro-Glu) and PPE (Pro-Pro-Glu) domains at N-terminal, are prevalent in M. tuberculosis genome. The function of most PE/PPE family proteins remains elusive. To characterize the function of PE_PGRS18, the encoding gene was heterologously expressed in M. smegmatis, a nonpathogenic mycobacterium. The recombinant PE_PGRS18 is cell wall associated. M. smegmatis PE_PGRS18 recombinant showed differential response to stresses and altered the production of host cytokines IL-6, IL-1ß, IL-12p40 and IL-10, as well as enhanced survival within macrophages largely via attenuating the apoptosis of macrophages. In summary, the study firstly unveiled the role of PE_PGRS18 in physiology and pathogenesis of mycobacterium.


Subject(s)
Antigens, Bacterial/physiology , Apoptosis/physiology , Bacterial Proteins/physiology , Cytokines/biosynthesis , Macrophages/microbiology , Membrane Proteins/physiology , Mycobacterium smegmatis/physiology , Mycobacterium tuberculosis/physiology , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Genes, Bacterial , Humans , Macrophages/metabolism , Membrane Proteins/genetics , Mice , Mycobacterium smegmatis/cytology , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/genetics , Nitric Oxide/metabolism , Protein Domains , Recombinant Fusion Proteins/metabolism , THP-1 Cells , Up-Regulation
20.
Oncol Rep ; 36(6): 3087-3094, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27748858

ABSTRACT

Infection with Helicobacter pylori is the strongest risk factor for the development of chronic gastritis, gastric ulcer and gastric carcinoma. The majority of the H. pylori-infected population remains asymptomatic, and only 1% of individuals may progress to gastric cancer. The clinical outcomes caused by H. pylori infection are considered to be associated with bacterial virulence, genetic polymorphism of hosts as well as environmental factors. Most H. pylori strains possess a cytotoxin-associated gene (cag) pathogenicity island (cagPAI), encoding a 120-140 kDa CagA protein, which is the most important bacterial oncoprotein. CagA is translocated into host cells via T4SS system and affects the expression of signaling proteins in a phosphorylation-dependent and independent manner. Thus, this review summarizes the results of relevant studies, discusses the pathogenesis of CagA-mediated gastric cancer.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Helicobacter Infections/complications , Helicobacter pylori/physiology , Stomach Neoplasms/microbiology , Animals , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Helicobacter Infections/microbiology , Host-Pathogen Interactions , Humans , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...