Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Gut Pathog ; 15(1): 32, 2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37415157

ABSTRACT

Listeria monocytogenes (Lm) is a facultative, intracellular Gram-positive pathogenic bacterium that causes sepsis, a condition characterized by persistent excessive inflammation and organ dysfunction. However, the pathogenesis of Lm-induced sepsis is unknown. In this research, we discovered that TRIM32 is required for innate immune regulation during Lm infection. Trim32 deficiency remarkably reduced bacteremia and proinflammatory cytokine secretion in mice with severe Lm infection, preventing sepsis. Trim32-/- mice had a lower bacterial burden after Lm infection and survived significantly longer than wild-type (WT) mice, as well as lower serum levels of inflammatory cytokines TNF-α, IL-6, IL-18, IL-12p70, IFN-ß, and IFN-γ at 1 day post infection (dpi) compared to WT mice. On the other hand, the chemokines CXCL1, CCL2, CCL7, and CCL5 were enhanced at 3 dpi in Trim32-/- mice than WT mice, reflecting increased recruitment of neutrophils and macrophages. Furthermore, Trim32-/- mice had higher levels of macrophage-associated iNOS to kill Lm. Collectively, our findings suggest that TRIM32 reduces innate immune cells recruitment and Lm killing capabilities via iNOS production.

2.
Front Immunol ; 13: 874922, 2022.
Article in English | MEDLINE | ID: mdl-35911731

ABSTRACT

Anti-PD-1 immunotherapy has saved numerous lives of cancer patients; however, it only exerts efficacy in 10-15% of patients with colorectal cancer. Fecal microbiota transplantation (FMT) is a potential approach to improving the efficacy of anti-PD-1 therapy, whereas the detailed mechanisms and the applicability of this combination therapy remain unclear. In this study, we evaluated the synergistic effect of FMT with anti-PD-1 in curing colorectal tumor-bearing mice using a multi-omics approach. Mice treated with the combination therapy showed superior survival rate and tumor control, compared to the mice received anti-PD-1 therapy or FMT alone. Metagenomic analysis showed that composition of gut microbiota in tumor-bearing mice treated with anti-PD-1 therapy was remarkably altered through receiving FMT. Particularly, Bacteroides genus, including FMT-increased B. thetaiotaomicron, B. fragilis, and FMT-decreased B. ovatus might contribute to the enhanced efficacy of anti-PD-1 therapy. Furthermore, metabolomic analysis upon mouse plasma revealed several potential metabolites that upregulated after FMT, including punicic acid and aspirin, might promote the response to anti-PD-1 therapy via their immunomodulatory functions. This work broadens our understanding of the mechanism by which FMT improves the efficacy of anti-PD-1 therapy, which may contribute to the development of novel microbiota-based anti-cancer therapies.


Subject(s)
Colorectal Neoplasms , Gastrointestinal Microbiome , Animals , Bacteroides , Colorectal Neoplasms/etiology , Colorectal Neoplasms/therapy , Fecal Microbiota Transplantation/adverse effects , Metagenome , Mice
3.
Front Immunol ; 13: 785422, 2022.
Article in English | MEDLINE | ID: mdl-35145519

ABSTRACT

Human tumors harbor a plethora of microbiota. It has been shown that the composition and diversity of intratumor microbiome are significantly associated with the survival of patients with pancreatic ductal adenocarcinoma (PDAC). However, the association in Chinese patients as well as the effect of different microorganisms on inhibiting tumor growth are unclear. In this study, we collected tumor samples resected from long-term and short-term PDAC survivors and performed 16S rRNA amplicon sequencing. We found that the microbiome in samples with different survival time were significantly different, and the differential bacterial composition was associated with the metabolic pathways in the tumor microenvironment. Furthermore, administration of Megasphaera, one of the differential bacteria, induced a better tumor growth inhibition effect when combined with the immune checkpoint inhibitor anti-programmed cell death-1 (anti-PD-1) treatment in mice bearing 4T1 tumor. These results indicate that specific intratumor microbiome can enhance the anti-tumor effect in the host, laying a foundation for further clarifying the underlying detailed mechanism.


Subject(s)
Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Megasphaera , Microbiota , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Aged , Aged, 80 and over , Animals , Biomarkers , Carcinoma, Pancreatic Ductal/therapy , China , Cytokines/metabolism , Disease Models, Animal , Dysbiosis , Female , Humans , Immunohistochemistry , Male , Megasphaera/classification , Megasphaera/genetics , Metabolic Networks and Pathways , Metagenome , Metagenomics/methods , Mice , Middle Aged , Molecular Targeted Therapy , Neoplasm Staging , Pancreatic Neoplasms/therapy , Prognosis , Treatment Outcome , Pancreatic Neoplasms
4.
Virulence ; 11(1): 769-780, 2020 12.
Article in English | MEDLINE | ID: mdl-32507026

ABSTRACT

BACILLUS CEREUS: is an opportunistic pathogen that can cause emetic or diarrheal foodborne illness. Previous studies have identified multiple pathogenic B. cereus strains and characterized a variety of virulence factors. Here, we demonstrate that the virulence and lethality of B. cereus for mammalian cells and host animals involve the interaction of B. cereus flagellin proteins and the host-cell-surface-localized glycosphingolipid Gb3 (CD77, Galα1-4Galß1-4Glcß1-Cer). We initially found that B. cereus infection was less lethal for Gb3-deficiencient A4galt-/- mice than for wild-type mice. Subsequent experiments established that some factor other than secreted toxins must account of the observed differential lethality: Gb3-deficiencient A4galt-/- mice were equally susceptible to secreted-virulence-factor-mediated death as WT mice, and we observed no differences in the bacterial loads of spleens or livers of mice treated with B. cereus strain vs. mice infected with a mutant variant of incapable of producing many secreted toxins. A screen for host-interacting B. cereus cell wall components identified the well-known flagellin protein, and both flagellin knockout strain assays and Gb3 inhibitor studies confirmed that flagellin does interact with Gb3 in a manner that affects B. cereus infection of host cells. Finally, we show that treatment with polyclonal antibody against flagellin can protect mice against B. cereus infection. Thus, beyond demonstrating a previously unappreciated interaction between a bacterial motor protein and a mammalian cell wall glycosphingolipid, our study will provide useful information for the development of therapies to treat infection of B. cereus.


Subject(s)
Bacillus cereus/metabolism , Bacillus cereus/pathogenicity , Bacterial Adhesion , Flagellin/metabolism , Host-Pathogen Interactions , Trihexosylceramides/metabolism , Animals , Bacterial Load , Cell Line , Gram-Positive Bacterial Infections/microbiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Trihexosylceramides/genetics , Virulence , Virulence Factors/metabolism
5.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-31988176

ABSTRACT

Streptococcus suis is an emerging zoonotic agent that causes streptococcal toxic shock-like syndrome (STSLS) and meningitis in humans, with high mortality and morbidity. The pathogenesis of both STSLS and central nervous system (CNS) infections caused by S. suis is not well understood. TRIM32, a member of the tripartite motif (TRIM) protein family, has been reported to regulate host inflammatory responses. In this study, we showed that TRIM32 deficiency significantly reduced the level of bacteremia and the production of proinflammatory cytokines following severe S. suis infection, protecting infected mice from STSLS. The influence of TRIM32 gene deletion on a range of processes known to be involved in S. suis meningitis was also examined. Both levels of bacterial loads and indications of brain hemorrhage were reduced in infected Trim32-/- mice compared with infected wild-type (WT) controls. We also found that TRIM32 deficiency increased the permeability of the blood-brain barrier (BBB) and the recruitment of inflammatory monocytes during the early course of S. suis infection, potentially limiting the development of S. suis meningitis. Our results suggest that TRIM32 sensitizes S. suis-induced infection via innate immune response regulation.


Subject(s)
Host-Pathogen Interactions , Meningitis, Bacterial/physiopathology , Shock, Septic/physiopathology , Streptococcus suis/growth & development , Ubiquitin-Protein Ligases/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Immunity, Innate , Meningitis, Bacterial/immunology , Mice , Mice, Knockout , Shock, Septic/immunology , Ubiquitin-Protein Ligases/deficiency
6.
Infect Genet Evol ; 68: 249-252, 2019 03.
Article in English | MEDLINE | ID: mdl-30597207

ABSTRACT

In June 2016, a Streptococcus suis outbreak occurred in Guangxi, China. We determined the genetic characteristics of six clinically isolated strains by serotyping, PCR, and whole-genome sequencing, performing genome epidemiology analysis on these and 961 public available S. suis genomes. We also classified the first sequence type ST665 human case. Sporadic and outbreak cases were distinguished by whole-genome sequencing and phylogenomics. This approach could help to prevent and control S. suis epidemics in Guangxi and the wider region.


Subject(s)
Disease Outbreaks , Genome, Bacterial , Genomics , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Streptococcus suis/classification , Streptococcus suis/genetics , China/epidemiology , Female , Genomics/methods , Geography, Medical , History, 21st Century , Humans , Male , Molecular Epidemiology , Phylogeny , Public Health Surveillance , Streptococcal Infections/history , Streptococcus suis/isolation & purification
7.
Sci Rep ; 7(1): 12099, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28935867

ABSTRACT

S100A4 has been implicated in cancer and several inflammatory diseases, but its role in inflammatory bowel disease has not been well investigated. Here, upon infection with Citrobacter rodentium, a model for enteropathogenic Escherichia coli infection in humans, induced the infiltration of a large number of S100A4+ cells into the colon in wild type (WT) mice. Deficiency of S100A4 reduced weight loss, bacterial colonization and colonic pathology. Furthermore, the expression of inflammatory cytokines and the recruitment of macrophages and neutrophils also decreased significantly in S100A4 knock out (S100A4 -/-) mice. In vitro, soluble S100A4 directly up-regulated expression of integrin ß-1 in intestinal epithelial cells and significantly increased the adherence of C. rodentium to intestinal epithelial cells. Additionally, the effects of S100A4 on the adherence of C. rodentium to epithelial cells could be abolished by a receptor for advanced glycation end products (RAGE)-specific inhibitor (FPS-ZM1). Therefore, these data indicate a novel mechanism for S100A4 that promotes colitis development by enhancing host adhesion and colonization of Citrobacter rodentium through the S100A4-mediated host inflammatory responses.


Subject(s)
Bacterial Adhesion/physiology , Colitis/metabolism , Enterobacteriaceae Infections/metabolism , Epithelial Cells/metabolism , S100 Calcium-Binding Protein A4/metabolism , Animals , Cell Line, Tumor , Citrobacter rodentium/physiology , Colitis/complications , Colitis/genetics , Cytokines/metabolism , Enterobacteriaceae Infections/complications , Enterobacteriaceae Infections/microbiology , Epithelial Cells/microbiology , Host-Pathogen Interactions , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Macrophages/microbiology , Macrophages/physiology , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/physiology , S100 Calcium-Binding Protein A4/genetics
8.
Virulence ; 8(7): 1290-1302, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28402705

ABSTRACT

Streptococcus suis is an important emerging zoonotic agent that causes acute bacterial meningitis in humans with high mortality and morbidity. Our previous work showed that factor H-binding protein (Fhb) contributed to virulence of S. suis, but the role of Fhb in the development of S. suis meningitis remained unclear. In this study, we demonstrated for the first time that Fhb contributed to the traversal of S. suis across the human blood-brain barrier by allelic-exchange mutagenesis, complementation and specific antibody blocking studies. We also showed that globotriaosylceramide (Gb3), the receptor of Fhb, was involved in this process and affected S. suis infection-induced activation of myosin light chain 2 through Rho/ROCK signaling in hCMEC/D3 cells. Using a murine model of S. suis meningitis, we further demonstrated that Gb3-deficiency prevented the mice from developing severe brain inflammation or injury. Our results demonstrate that the Fhb-Gb3 interaction plays an important role in the development of S. suis meningitis and might be a potential therapeutic target against S. suis infection.


Subject(s)
Bacterial Proteins/metabolism , Meningitis, Bacterial/metabolism , Streptococcal Infections/metabolism , Streptococcus suis/metabolism , Trihexosylceramides/metabolism , Animals , Bacterial Proteins/genetics , Carrier Proteins , Female , Host-Pathogen Interactions , Humans , Male , Meningitis, Bacterial/microbiology , Mice , Mice, Inbred C57BL , Streptococcal Infections/microbiology , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Virulence
9.
Sci Rep ; 6: 29373, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27383625

ABSTRACT

Vascular leakage frequently occurs in patients with severe Staphylococcus aureus infection. However, the mechanism underlying S. aureus infection-induced vascular leakage remains unclear. Here, we identified the S. aureus virulence factor phenol-soluble modulin (PSM)α4 from the culture supernatant of strain USA300 as a stimulator of heparin-binding protein (HBP) release from polymorphonuclear neutrophils (PMNs) and demonstrated that PSMα4-induced HBP release from PMNs leads to vascular leakage. PSMα4 appeared less cytolytic than PSMα1-3 and was insensitive to lipoproteins; it significantly increased myeloperoxidase and elastase release from PMNs and cell surface CD63 expression in PMNs. PSMα4-induced HBP release required formyl peptide receptor 2 (FPR2) and phosphoinositide 3-kinase (PI3K) and depended on Ca(2+) influx and cytoskeleton rearrangement. Thus, PSMα4 may stimulate HBP release by activating FPR2 and PI3K to initiate PMN degranulation. PSMα4-induced HBP release from PMNs increased endothelial cell monolayer permeability in vitro and induced vascular leakage in mice. This novel function of PSMα4 may contribute to the pathogenesis of S. aureus and may be a potential therapeutic target.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Bacterial Toxins/metabolism , Blood Proteins/metabolism , Capillary Permeability/physiology , Carrier Proteins/metabolism , Neutrophils/drug effects , Staphylococcus aureus/physiology , Virulence Factors/physiology , Animals , Bacterial Toxins/chemistry , Case-Control Studies , Culture Media , Exocytosis/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neutrophils/metabolism , Spectrometry, Mass, Electrospray Ionization , Staphylococcal Infections/metabolism , Staphylococcus aureus/pathogenicity
10.
FEBS Lett ; 590(9): 1384-92, 2016 05.
Article in English | MEDLINE | ID: mdl-27086582

ABSTRACT

The recently identified Streptococcus suis adhesin factor H-binding protein (Fhb) targets the host cellular receptor glycolipid GbO3 through its N terminus. However, it is unclear how Fhb interacts with its receptor. Here, we determined the complex structure of factor H-binding protein receptor-binding domain (Fhb RBD) with Gb2, an analog of its receptor, revealing that Gb2 binds in a pocket of the ß sandwich core domain. We identified the key residues for Fhb RBD receptor binding using mutagenesis and isothermal titration calorimetry. Mutagenesis analyses indicated that Fhb binds to Gb2 mainly through hydrogen and hydrophobic interactions. Our findings provided structural insights into the Fhb-mediated host-pathogen interactions of S. suis.


Subject(s)
Adhesins, Bacterial/chemistry , Streptococcus suis/chemistry , Trihexosylceramides/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Binding Sites , Humans , Point Mutation , Protein Binding , Streptococcus suis/pathogenicity , Trihexosylceramides/chemistry
11.
J Microbiol Methods ; 124: 35-40, 2016 May.
Article in English | MEDLINE | ID: mdl-26979644

ABSTRACT

Early diagnosis of Mycoplasma pneumoniae (MP) infection is crucial for prompt treatment and good patient outcome. However, serological tests to detect MP rapidly and conveniently are still lacking. This study aimed to use the fluorescent dye Alexa Fluor® 647 as the detection marker to develop a lateral flow immunochromatographic assay (LFIA) for detection of MP-specific IgM in serum specimen. Monoclonal mouse antibody against human IgM (µ-chain specific) and goat anti-rabbit IgG were labeled with Alexa Fluor® 647 (anti-IgM-AF647 and anti-IgG-AF647). A mixture of natural MP antigen and recombinant P1 antigen was coated as the test line (T line) and rabbit IgG was coated as the control line (C line) on a nitrocellulose (NC) membrane. The MP antigens captured IgM-anti-IgM-AF647 complex on the T line. Rabbit IgG captured anti-IgG-AF647 on the C line. The fluorescence intensity on the T line and C line was measured. Sartorius CN140 NC membrane showed higher sensitivity than CN95. The optimal reaction time for the LFIA was 10min. The area under the receiver operating characteristic curve based on 34 MP positive and 166 MP negative serum samples was 0.986 (p<0.001). The cutoff value of T/C area ratio was 0.3830. The LFIA strips did not react with serum from patients infected with non-MP pathogens including influenza viruses and bacteria causing respiratory tract infection. The intra-assay and inter-assay coefficients of variation were between 3.28% and 10.14%. The shelf life was calculated to be 2years at room temperature. The LFIA strips and the commercial EUROIMMUN kit showed consistent results on 372 serum specimens. The overall consistency rate was 96.37% with a Kappa value of 0.842 (p<0.001). The LFIA in the current study may be a sensitive and specific approach to detect early MP infection rapidly and conveniently.


Subject(s)
Antibodies, Bacterial/blood , Chromatography, Affinity/methods , Immunoglobulin M/blood , Mycoplasma pneumoniae/immunology , Pneumonia, Mycoplasma/blood , Animals , Humans , Mice , Mycoplasma pneumoniae/isolation & purification , Pneumonia, Mycoplasma/microbiology , Rabbits , Sensitivity and Specificity
12.
Toxicon ; 113: 55-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26792715

ABSTRACT

Staphylococcus enterotoxin B (SEB) is a superantigen that can induce massive activation of T cells with specific Vß and inflammatory cytokine cascades, which mediate shock. To date, no SEB vaccine has been developed for preventing toxic shock syndrome (TSS). Here, we evaluated the therapeutic effect of a fusion protein human serum albumin-Vß (HSA-Vß) on TSS induced by SEB. Compared with Vß, the preparation of HSA-Vß was much easier to handle owing to its solubility. Affinity testing showed that HSA-Vß had high affinity for SEB. In vitro results showed that HSA-Vß could effectively inhibit interferon (IFN)-γ and tumor necrosis factor (TNF)-α secretion by human peripheral blood mononuclear cells. Moreover, in vivo, HSA-Vß reduced IFN-γ and TNF-α levels in the serum and protected mice from SEB lethal challenge when administered simultaneously with SEB or 30 min after SEB. In summary, we simplified the preparation of Vß by fusion with HSA, creating the HSA-Vß protein, which effectively inhibited cytokine production and protected mice from lethal challenge with SEB. These data indicated that HSA-Vß may represent a novel therapeutic strategy for the treatment of SEB-induced TSS.


Subject(s)
Enterotoxins/toxicity , Receptors, Antigen, T-Cell/immunology , Serum Albumin/chemistry , Serum Albumin/immunology , Shock, Septic/chemically induced , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/immunology , Humans , Interferon-gamma , Leukocytes, Mononuclear , Mice , Protein Subunits , Tumor Necrosis Factor-alpha
13.
Front Microbiol ; 6: 1001, 2015.
Article in English | MEDLINE | ID: mdl-26441928

ABSTRACT

Muramidase-released protein (MRP) is as an important virulence marker of Streptococcus suis (S. suis) serotype 2. Our previous works have shown that MRP can bind human fibrinogen (hFg); however, the function of this interaction in S. suis meningitis is not known. In this study, we found that the deletion of mrp significantly impairs the hFg-mediated adherence and traversal ability of S. suis across human cerebral microvascular endothelial cells (hCMEC/D3). Measurement of the permeability to Lucifer yellow in vitro and Evans blue extravasation in vivo show that the MRP-hFg interaction significantly increases the permeability of the blood-brain barrier (BBB). In the mouse meningitis model, wild type S. suis caused higher bacterial loads in the brain and more severe histopathological signs of meningitis than the mrp mutant at day 3 post-infection. Western blot analysis and immunofluorescence observations reveal that the MRP-hFg interaction can destroy the cell adherens junction protein p120-catenin of hCMEC/D3. These results indicate that the MRP-hFg interaction is important in the development of S. suis meningitis.

14.
Wei Sheng Wu Xue Bao ; 55(5): 643-9, 2015 May 04.
Article in Chinese | MEDLINE | ID: mdl-26259489

ABSTRACT

OBJECTIVE: In order to study the role of SS2 Type IV Secretion System VirD4 in evasion of the host innate immune killing, we constructed a knockout mutant AVirD4. Then we studied its biological activity and virulence. METHODS: The two VirD4 flanking DNA sequences were amplified using genome of 05ZYH33 as template. We also amplified the Cm sequence of shuttle vector pSET1, and through overlap extension PCR we connected the three fragments together. Using suicide vector pSET4s, we constructed the recombinant gene knockout vector pSET4s::VirD4. The mutant AVirD4 was successfully constructed by allelic replacement. Virulence of mutant strain was compared with wild type strain 05ZYH33 through in vitro bactericidal assays, competitive infection and challenge experiment of CD1 mice. RESULTS: Mutant strain AVirD4 was constructed successfully, its virulence attenuated compared to the wild type strain. CONCLUSION: These findings indicated that Type IV Secretion System component VirD4 contributed to the virulence of S. suis with important functions in evading innate immunocyte killing.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems , Streptococcal Infections/microbiology , Streptococcus suis/metabolism , Streptococcus suis/pathogenicity , Animals , Bacterial Proteins/genetics , Female , Humans , Mice , Streptococcus suis/genetics , Virulence
15.
Front Microbiol ; 6: 644, 2015.
Article in English | MEDLINE | ID: mdl-26167160

ABSTRACT

Streptococcus suis serotype 2 (SS2) is an emerging human pathogen worldwide. A large outbreak occurred in the summer of 2005 in China. Serum samples from this outbreak revealed that levels of the main proinflammatory cytokines were significantly higher in patients with streptococcal toxic-shock-like syndrome (STSLS) than in patients with meningitis only. However, the mechanism underlying the cytokine storm in STSLS caused by SS2 remained unclear. In this study, we found that suilysin (SLY) is the main protein inflammatory stimulus of SS2 and that native SLY (nSLY) stimulated cytokines independently of its haemolytic ability. Interestingly, a small amount of SLY (Å Mol/L) induced strong, long-term TNF-α release from human PBMCs. We also found that nSLY stimulated TNF-α in wild-type macrophages but not in macrophages from mice that carried a spontaneous mutation in TLR4 (P712H). We demonstrated for the first time that SLY stimulates immune cells through TLR4. In addition, the Myd88 adaptor-p38-MAPK pathway was involved in this process. The present study suggested that the TLR4-dependent inflammatory responses induced by SLY in host might contribute to the STSLS caused by SS2 and that p38-MAPK could be used as a target to control the release of excess TNF-α induced by SS2.

16.
J Biol Chem ; 290(31): 19081-92, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26085092

ABSTRACT

Streptococcus suis serotype 2 (Ss2) is an important swine and human zoonotic pathogen. In the present study, we identified a novel secreted immunogenic protein, SsTGase, containing a highly conserved eukaryotic-like transglutaminase (TGase) domain at the N terminus. We found that inactivation of SsTGase significantly reduced the virulence of Ss2 in a pig infection model and impaired its antiphagocytosis in human blood. We further solved the crystal structure of the N-terminal portion of the protein in homodimer form at 2.1 Å. Structure-based mutagenesis and biochemical studies suggested that disruption of the homodimer directly resulted in the loss of its TGase activity and antiphagocytic ability. Characterization of SsTGase as a novel virulence factor of Ss2 by acting as a TGase would be beneficial for developing new therapeutic agents against Ss2 infections.


Subject(s)
Bacterial Proteins/chemistry , Streptococcus suis/enzymology , Transglutaminases/chemistry , Amino Acid Sequence , Animals , Bacterial Proteins/physiology , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Phagocytosis , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Sus scrofa , Transglutaminases/physiology , Virulence Factors
17.
Biomed Res Int ; 2015: 580240, 2015.
Article in English | MEDLINE | ID: mdl-25654114

ABSTRACT

An S. maltophilia strain named WJ66 was isolated from a patient; WJ66 showed resistance to more antibiotics than the other S. maltophilia strains. This bacteraemia is resistant to sulphonamides, or fluoroquinolones, while the representative strain of S. maltophilia, K279a, is sensitive to both. To explore drug resistance determinants of this strain, the draft genome sequence of WJ66 was determined and compared to other S. maltophilia sequences. Genome sequencing and genome-wide evolutionary analysis revealed that WJ66 was highly homologous with the strain K279a, but strain WJ66 contained additional antibiotic resistance genes. Further analysis confirmed that strain WJ66 contained an amino acid substitution (Q83L) in fluoroquinolone target GyrA and carried a class 1 integron, with an aadA2 gene in the resistance gene cassette. Homology analysis from the pathogen-host interaction database showed that strain WJ66 lacks raxST and raxA, which is consistent with K279a. Comparative genomic analyses revealed that subtle nucleotide differences contribute to various significant phenotypes in close genetic relationship strains.


Subject(s)
Drug Resistance, Multiple, Bacterial , Stenotrophomonas maltophilia/genetics , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , China , Databases, Genetic , Drug Resistance, Microbial/drug effects , Drug Resistance, Microbial/genetics , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Genes, Bacterial , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Integrons/genetics , Molecular Sequence Data , Phylogeny , Sequence Alignment , Sequence Analysis, DNA , Stenotrophomonas maltophilia/drug effects
18.
Int J Oncol ; 46(1): 281-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25353649

ABSTRACT

Circulating tumor cells (CTCs) were recognized as novel tumor biomarker for prognostic and predictive purposes in various cancers. Various detection technologies and devices have been developed to enumerate and characterize CTCs. Most of those approaches are based on the positive enrichment strategy and immunocytological techniques. However, the sensitivity of these approaches proved to be limited in metastatic tumors and the detection of early tumor cell dissemination was problematic. In the present study, we developed a novel CTC detection method by real-time RT-PCR technique in combination of negative enrichment strategy. The developed enrichment approach could recover more than 75% of spiked breast cancer cells from peripheral blood. The detection limit of duplex real-time RT-PCR assay using KRT19 and ERBB2 as targeted genes was consistently one breast tumor cell. Moreover, CTC detection by duplex real-time RT-PCR assay had higher detection sensitivity than that by immunostaining, especially in early breast cancer. In summary, the results of the present study indicated the potential clinical utilities of CTCs identification on breast cancer by duplex real-time RT-PCR in combination with negative enrichment.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , DNA, Neoplasm/analysis , Neoplastic Cells, Circulating/pathology , Real-Time Polymerase Chain Reaction/methods , Biomarkers, Tumor/blood , Breast Neoplasms/genetics , Cell Separation/methods , Female , HL-60 Cells , Humans , Jurkat Cells , MCF-7 Cells , Neoplastic Cells, Circulating/metabolism , Tumor Cells, Cultured
20.
PLoS One ; 8(7): e67728, 2013.
Article in English | MEDLINE | ID: mdl-23935843

ABSTRACT

Streptococcus suis (S.suis) is an important emerging worldwide pig pathogen and zoonotic agent with rapid evolution of virulence and drug resistance. In this study, we wanted to investigate the effect of licochalcone A on growth and properties of Streptococcus suis. The antimicrobial activity of licochalcone A was tested by growth inhibition assay and the minimal inhibitory concentrations (MICs) also were determined. The effect of licochalcone A on S.suis biofilm formation was characterized by crystal violet staining. The effect of licochalcone A on suilysin secretion was evaluated by titration of hemolytic activity. To understand the antimicrobial effect, gene expression profile of S.suis treated by licochalcone A was analyzed by DNA microarray. Our results demonstrated that licochalcone A showed antimicrobial activity on S.suis with MICs of 4 µg/ml for S.suis serotype 2 strains and 8 µg/ml for S.suis serotype 7 strains. Biofilm formation was inhibited by 30-40% in the presence of licochalcone A (3 µg/ml) and suilysin secretion was also significantly inhibited in the presence of licochalcone A (1.5 µg/ml). The gene expression profile of S.suis in the presence of licochalcone A showed that 132 genes were differentially regulated, and we analyzed the regulated genes in the aspect of the bacterial cell cycle control. Among the deregulated genes, the genes responsible for the mass doubling was increased expression, but the genes responsible for DNA replication and cell division were inhibited the expression. So, we think the regulation of the cell cycle genes might provide a mechanistic understanding of licochalcone A mediated antimicrobial effect against S.suis.


Subject(s)
Chalcones/pharmacology , Streptococcus suis/drug effects , Streptococcus suis/growth & development , Biofilms/drug effects , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Genes, Bacterial/genetics , Hemolysin Proteins/metabolism , Hemolysis/drug effects , Humans , Microbial Sensitivity Tests , Oligonucleotide Array Sequence Analysis , Streptococcus suis/genetics , Streptococcus suis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...