Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Microbiol Immunol ; 65(3): 115-124, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33368645

ABSTRACT

We previously isolated a symbiotic environmental amoeba, harboring an environmental chlamydia, Neochlamydia S13. Interestingly, this bacterium failed to survive outside of host cells and was immediately digested inside other amoebae, indicating bacterial distribution via cytokinesis. This may provide a model for understanding organelle development and chlamydial pathogenesis and evolution; therefore, we assessed our hypothesis of Neochlamydia S13 distribution via cytokinesis by comparative analysis with other environmental Chlamydiae (Protochlamydia R18 and Parachlamydia Bn9 ). Dual staining with 4',6-diamidino-2-phenylindole and phalloidin revealed that the progeny of Neochlamydia S13 and Protochlamydia R18 existed in both daughter cells with a contractile ring on the verge of separation. However, in contrast to other environmental Chlamydiae, little Neochlamydia S13 16S ribosomal DNA was amplified from the culture supernatant. Interestingly, Neochlamydia S13 failed to infect aposymbiotic amoebae, indicating an intimate interaction with the host cells. Furthermore, its infectious rates in cultures expanded from a single amoeba were always maintained at 100%, indicating distribution via cytokinesis. We concluded that unlike other environmental Chlamydiae, Neochlamydia S13 has a unique ability to divide its progeny only via host amoebal cytokinesis. This may be a suitable model to elucidate the mechanism of cell organelle distribution and of chlamydial pathogenesis and evolution.


Subject(s)
Amoeba , Chlamydiales , Cytokinesis , Amoeba/microbiology , RNA, Ribosomal, 16S/genetics , Symbiosis
2.
Can J Microbiol ; 67(9): 651-656, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33756093

ABSTRACT

Bacteria acquire genetic variations that help them to adapt to stressful environmental conditions, and these changes may be associated with the development of antimicrobial resistance. In this study, we investigated the mutation frequencies of 270 isolates of Escherichia coli from river water, which represents a relatively unstressful environment. As we predicted, mutation frequencies of the E. coli isolates ranged from <1 × 10-11 to 6.3 × 10-8 (median, 1.7 × 10-9), and a strong mutator (≥ 4 × 10-7) was not detected. To better understand the role of mutation frequency in the development of antimicrobial resistance, we assessed antimicrobial sensitivity after exposure of the E. coli isolates to subinhibitory concentrations of ciprofloxacin, as a surrogate for stress. We found that antimicrobial resistance increased in bacteria with a low mutation frequency after exposure, and the relative increase in antimicrobial resistance generally increased, depending on the mutation frequency. Thus, mutation frequency may contribute to the development of antimicrobial resistance of bacteria in natural environments.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Microbial Sensitivity Tests , Mutation Rate , Rivers , Water
3.
J Hum Genet ; 65(1): 41-48, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31723216

ABSTRACT

Neochlamydia strain S13 is an amoebal symbiont of an Acanthamoeba sp. The symbiont confers resistance to Legionella pneumophila on its host; however, the molecular mechanism underlying this resistance is not completely understood. Genome analyses have been crucial for understanding the complicated host-symbiont relationship but segregating the host's genome DNA from the symbiont's DNA is often challenging. In this study, we successfully identified a bimodal genomic structure in Neochlamydia strain S13 using PacBio RS II supported by ultra-long reads derived from MinION. One mode consisted of circular sequences of 2,586,667 and 231,307 bp; the other was an integrated sequence of the two via long homologous regions. They encoded 2175 protein-coding regions, some of which were implied to be acquired via horizontal gene transfer. They were specifically conserved in the genus Neochlamydia and formed a cluster in the genome, presumably by multiplication through genome replication. Moreover, it was notable that the sequenced DNA was obtained without segregating the symbiont DNA from the host. This is an easy and versatile technique that facilitates the characterization of diverse hosts and symbionts in nature.


Subject(s)
Genome, Bacterial , Gram-Negative Bacteria/genetics , Sequence Analysis, DNA/instrumentation , Acanthamoeba/microbiology , Genomics/methods , Gram-Negative Bacteria/growth & development , Gram-Negative Bacteria/isolation & purification , Phylogeny , Sequence Analysis, DNA/methods
4.
Can J Microbiol ; 65(2): 135-143, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30336068

ABSTRACT

The obligate intracellular bacterium Chlamydia trachomatis activates the host cell apoptosis pathway at a late stage of its developmental cycle. However, whether caspase-3, which is a key enzyme of apoptosis, is activated in Chlamydia-infected cells remains unknown. Here, we established HEp-2 cells stably expressing cFluc-DEVD, which is a caspase-3 substrate sequence inserted into cyclic firefly luciferase, and then monitored the dynamics of caspase-3 activity in cells infected with Chlamydia. Transfected cells without infection showed a significant increase in luciferase activity due to stimulation with staurosporine, an inducer of apoptosis. Activation was significantly blocked by addition of caspase inhibitor z-VAD-fmk. Furthermore, as expected, Chlamydia infection caused a significant increase in luciferase activation at 36-48 h postinfection with a contrastive decrease at 24 h postinfection, which is already well known. Such activation caused by the infection was much stronger when the amount of bacteria was increased. Thus, caspase-3 activation was accurately monitored by the luciferase activity in HEp-2 cells constitutively expressing the cFluc-DEVD probe. Furthermore, our data showed that C. trachomatis activates caspase-3 in host cells at a late stage of infection.


Subject(s)
Apoptosis , Caspase 3/metabolism , Chlamydia trachomatis/physiology , Cells, Cultured , Enzyme Activation , Humans
5.
BMC Med Educ ; 19(1): 53, 2019 Feb 11.
Article in English | MEDLINE | ID: mdl-30744704

ABSTRACT

BACKGROUND: Nurses are responsible for implementing appropriate measures to reduce hospital infections, especially with multidrug resistant bacteria, so nursing students should learn about microbiology. This helps them to understand bacterial dissemination and infectious disease control. Because of tight schedules, however, its teaching is limited in undergraduate nursing classes in Japan. We therefore tested whether a simple short practical session in a microbiology class could help to improve undergraduate nursing students' awareness of bacterial traits and how to prevent infections. METHODS: This study involved second-grade nursing students (n = 76). Two short practical sessions (a total of 3 h, across 2 days) were used to assess the effectiveness of washing or disinfection on hand bacteria in a 16-class microbiology course (total class time was 24 h, plus an exam). Hand bacteria were sampled on LB agar plates with orientation during the first half-day, and the plates examined for colonies with distinct color or morphological traits, and discussed, in the second session, a week later. Questionnaires before and after the exercise were used to assess changes in awareness of unseen bacteria inhabiting around us connecting bacterial traits and how to prevent infections. RESULTS: The results showed that the practical increased the nursing students' awareness of fomites (utensils) (p = 0.0115), fomites (contact-based) (p = 0.0016), habitats (body surface) (p = 0.0127), action facilitating hospital infection (p = 0.0166), and changes in physical condition caused by bacterial infections (p = 0.0136). There were no changes in word associations (p = 0.627) or habitats (inside body) (p = 0.308). Difficulty score, which is an element in questionnaire psychometric properties, tended to be close to the expected score through the practical, but not statistical significant. In addition, regardless of before or after practical, Cronbach α score, which is an indicator of the reliability among items of multi-choice questions, showed > 0.8, indicating validity of evaluation items. Thus, the student's awareness of unseen bacteria inhabiting around us was significantly increased as compared to those before practical in microbiology class. CONCLUSIONS: The simple short practical effectively improved nursing students' awareness of unseen bacteria inhabiting around us in microbiology course, useful for even tight teaching schedules.


Subject(s)
Cross Infection/nursing , Cross Infection/prevention & control , Education, Medical, Undergraduate/standards , Education, Nursing/standards , Microbiology/education , Students, Nursing , Health Knowledge, Attitudes, Practice , Humans , Reproducibility of Results
6.
Microbiol Immunol ; 62(11): 720-728, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30357893

ABSTRACT

Tetrahymena can facilitate plasmid transfer among Escherichia coli or from E. coli to Salmonella Enteritidis via vesicle accumulation. In this study, whether ciliates promote the interactive transfer of plasmids encoding blaIMP-1 between fecal E. coli and environmental Aeromonas caviae was investigated. Both bacteria were mixed with or without ciliates and incubated overnight at 30°C. The frequency of plasmid-acquired bacteria was estimated by colony counts using an agar plate containing ceftazidim (CAZ) followed by determination of the minimum inhibitory concentration (MIC). Cultures containing ciliates interactively transferred the plasmid between E. coli and Aeromonas with a frequency of 10-4 to 10-5 . All plasmid-acquired bacteria showed a MIC against CAZ of >128 µg/mL and the plasmid transfer was confirmed by PCR amplification of the blaIMP-1 gene. Fluorescent observation showed that both bacteria accumulated in the same vesicle and that transwell sequestering significantly decreased the transfer frequency. Although ciliates preferentially ingested E. coli rather than A. caviae, both bacteria were co-localized into the same vesicles of ciliates, indicating that their meeting is associated with the gene transfer. Thus, ciliates interactively promote plasmid transfer between E. coli and A. caviae. The results of this study will facilitate control of the spread of multiple-antibiotic resistant bacteria.


Subject(s)
Aeromonas caviae/genetics , Bacterial Proteins/genetics , Feces/microbiology , Gene Transfer, Horizontal , Plasmids/genetics , Tetrahymena/microbiology , beta-Lactamases/genetics , Aeromonas caviae/drug effects , Anti-Bacterial Agents/pharmacology , DNA, Bacterial/genetics , Drug Resistance, Bacterial/genetics , Environmental Microbiology , Escherichia coli/genetics , Genes, Bacterial/genetics , Microbial Sensitivity Tests , Tetrahymena/physiology
7.
J Infect Chemother ; 24(2): 130-137, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29132924

ABSTRACT

Chlamydia trachomatis is the leading cause of sexually transmitted infections worldwide. Capsaicin, a component of chili pepper, which can stimulate actin remodeling via capsaicin receptor TRPV1 (transient receptor potential vanilloid 1) and anti-inflammatory effects via PPARγ (peroxisome proliferator-activated receptor-γ) and LXRα (liver X receptor α), is a potential candidate to control chlamydial growth in host cells. We examined whether capsaicin could inhibit C. trachomatis growth in immortal human epithelial HeLa cells. Inclusion forming unit and quantitative PCR assays showed that capsaicin significantly inhibited bacterial growth in cells in a dose-dependent manner, even in the presence of cycloheximide, a eukaryotic protein synthesis inhibitor. Confocal microscopic and transmission electron microscopic observations revealed an obvious decrease in bacterial numbers to inclusions bodies formed in the cells. Although capsaicin can stimulate the apoptosis of cells, no increase in cleaved PARP (poly (ADP-ribose) polymerase), an apoptotic indicator, was observed at a working concentration. All of the drugs tested (capsazepine, a TRPV1 antagonist; 5CPPSS-50, an LXRα inhibitor; and T0070907, a PPARγ inhibitor) had no effect on chlamydial inhibition in the presence of capsaicin. In addition, we also confirmed that capsaicin inhibited Chlamydia pneumoniae growth, indicating a phenomena not specific to C. trachomatis. Thus, we conclude that capsaicin can block chlamydial growth without the requirement of host cell protein synthesis, but by another, yet to be defined, mechanism.


Subject(s)
Capsaicin/pharmacology , Chlamydia trachomatis/drug effects , Chlamydophila pneumoniae/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Chlamydia trachomatis/growth & development , Chlamydophila pneumoniae/growth & development , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , HeLa Cells , Humans , Inclusion Bodies/drug effects , Liver X Receptors/antagonists & inhibitors , PPAR gamma/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology , TRPV Cation Channels/antagonists & inhibitors
8.
J Infect Chemother ; 24(10): 815-821, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30082190

ABSTRACT

We explored the bacteria present in the vaginal microbiota facilitating the prevalence of Chlamydia trachomatis in women visiting a community hospital in Sapporo, Japan, by amplicon sequencing. A total of 273 cervical swab samples were collected, and bacterial vaginosis was evaluated in all specimens by assessment of the Nugent score. In 16 of the samples, bacterial 16S rDNA could not be detected and they were therefore omitted from subsequent experiments (n = 257). A significant negative correlation was observed between the Nugent scores and the amount of Lactobacillus 16S rDNA. Among the 257 samples, chlamydial plasmid was detected in 20 samples and was used for amplicon sequencing. No significant association between the Nugent score and the prevalence of C. trachomatis was detected. Based on the results of chlamydial plasmid detection and the Nugent score, chlamydia-negative samples (n = 27) were randomly selected. Finally, the number of operational taxonomic units (OTUs) obtained from amplicon sequencing was compared between chlamydia-positive (n = 20) and -negative samples (n = 27), revealing that a significant difference was only detected for the OTU numbers of Enterobacteriaceae between the C. trachomatis-positive and -negative groups. However, almost all of the samples utilized for amplicon sequencing failed to grow on MacConkey agar plates and produce indole. Taken together, we concluded that traces of bacteria, not live bacteria, belonging to the Enterobacteriaceae indicated the flow of bacteria through the anogenital route along with gut indole, and the resulting impact on the prevalence of C. trachomatis in the cervicogenital tract of women in Japan.


Subject(s)
Chlamydia Infections/epidemiology , Chlamydia trachomatis , Enterobacteriaceae/isolation & purification , Vaginosis, Bacterial/epidemiology , Vaginosis, Bacterial/microbiology , Adult , Correlation of Data , Enterobacteriaceae/classification , Enterobacteriaceae/genetics , Female , Hospitals, Community , Humans , Japan/epidemiology , Lactobacillus/genetics , Lactobacillus/isolation & purification , Microbiota/genetics , Phylogeny , Prevalence , RNA, Ribosomal, 16S/genetics , Vagina/microbiology
9.
Parasitol Res ; 117(3): 937-941, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29380051

ABSTRACT

A free-living amoeba, Naegleria is ubiquitously distributed in various natural environments. Since some Naegleria spp. are exclusively distributed in the Arctic and sub-Antarctic regions, we hypothesized that the amoeba may be useful to determine long-term survival of Naegleria in laboratory conditions at 4 °C. The main objective of the study is to determine that a species of an environmental amoebal isolated can live at low temperatures after a long time. Here, we therefore show long-term survival of an amoeba, Naegleria polaris isolated from a sediment sample, which was collected from Antarctica 10 years ago, and since stored at 4 °C. The sample was put on non-nutrient agar plates with heat-killed Escherichia coli, and then the plate was incubated at 4, 15, or 30 °C. Motile amoebae were seen only when the plate was incubated at 15 °C. The sequencing of ribosomal DNA including internal transcribed spacers (ITS) 1, 5.8S rDNA, and ITS2 region revealed the amoebae to be N. polaris, which is exclusively distributed in the Arctic and sub-Antarctic regions. Scanning electron microscopic observation showed that no typical sucker-like structure was seen on the surface of N. polaris, but the cysts were similar to those of Naegleria fowleri. Thus, our result shows, for the first time, that N. polaris can survive after 10 years of storage at 4 °C. This finding may help us understand the still undescribed effects of environmental samples on viability of amoebae.


Subject(s)
Longevity , Naegleria/physiology , Animals , Antarctic Regions , Arctic Regions , Cold Temperature , DNA, Ribosomal , Naegleria/classification
10.
Cytometry A ; 91(8): 822-829, 2017 08.
Article in English | MEDLINE | ID: mdl-28715618

ABSTRACT

Neutrophil extracellular traps (NETs) are extracellular chromatin fibers adorned with antimicrobial proteins, such as myeloperoxidase (MPO), which are extruded from activated neutrophils. NETosis is the metamorphosis of neutrophils with NET formation that follows decondensation of DNA and rupture of the plasma membrane. Although NETs play important roles in innate immunity, excessive formation of NETs can be harmful to the hosts. Until now, various methods for evaluation of NETs have been reported. Although each has a virtue, the gold standard has not been established. Here we demonstrate a simple, objective, and quantitative method to detect NETs using flow cytometry. This method uses a plasma membrane-impermeable DNA-binding dye, SYTOX Green. SYTOX Green-positive cells were detected in human peripheral polymorphonuclear cells exposed to a NET inducer, phorbol 12-myristate 13-acetate (PMA). The number of SYTOX Green-positive cells was increased depending on the exposure duration and concentrations of PMA. Furthermore, co-localization of MPO and plasma membrane-appendant DNA of SYTOX Green-positive cells was demonstrated. Moreover, a NET inhibitor, diphenylene iodonium, could significantly reduce the number of SYTOX Green-positive cells induced by PMA. The collective evidence suggests that SYTOX Green-positive cells include neutrophils that formed NETs. The established method could detect neutrophils that underwent NETosis but not early apoptosis with equivalence in quantification to another well-used image analysis, which is based on fluorescent staining. Additionally, NETs that were formed in vivo were also detectable by this method. It is conceivable that the established method will bring us better understanding of the relation between NETosis and human diseases. © 2017 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of ISAC.


Subject(s)
Extracellular Traps/physiology , Apoptosis/physiology , DNA/metabolism , Extracellular Traps/metabolism , Flow Cytometry/methods , Humans , Neutrophils/metabolism , Neutrophils/physiology , Peroxidase/metabolism , Tetradecanoylphorbol Acetate/metabolism
11.
J Infect Chemother ; 23(7): 439-445, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28431935

ABSTRACT

Previous works have demonstrated considerable variability in hospital cleanliness in Japan, suggesting that contamination is driven by factors that are currently poorly controlled. We undertook 16S rRNA sequence analysis to study population structures of hospital environmental microbiomes to see which factor(s) impacted contamination. One hundred forty-four samples were collected from surfaces of three hospitals with distinct sizes ("A": >500 beds, "B": 100-500 beds, "C": <100 beds). Sample locations of two ward types (Surgical and Internal) included patient room bed table (multiple) (4BT), patient overbed table (multiple) (4OT), patient room sink (multiple) (4S), patient room bed table (single) (SBT), patient overbed table (single) (SOT), patient room sink (single) (SS), nurse desk (ND), and nurse wagon (NW). Total DNA was extracted from each sample, and the 50 samples that yielded sufficient DNA were used for further 16S rRNA sequencing of hospital microbiome populations with cluster analysis. The number of assigned bacterial OTU populations was significantly decreased in hospital "C" compared to the other hospitals. Cluster analysis of sampling locations revealed that the population structure in almost all locations of hospital "C" and some locations in the other hospitals was very similar and unusually skewed with a family, Enterobacteriaceae. Interestingly, locations included patient area (4OT, 4BT, SBT) and nurse area (ND), with a device (NW) bridging the two and a place (4S and SS) shared between patients or visitors. We demonstrated diversity changes of hospital environmental microbiomes with a skewed population, presumably by medical staff pushing NWs or sinks shared by patients or visitors.


Subject(s)
Bacteria/classification , Bacteria/genetics , Equipment and Supplies, Hospital/microbiology , Hospitals/statistics & numerical data , Microbial Consortia/genetics , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Environmental Microbiology , Genetic Variation , Humans , Japan/epidemiology , Patients' Rooms , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
12.
BMC Microbiol ; 16(1): 292, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27978822

ABSTRACT

BACKGROUND: Environmental chlamydiae belonging to the Parachlamydiaceae are obligate intracellular bacteria that infect Acanthamoeba, a free-living amoeba, and are a risk for hospital-acquired pneumonia. However, whether amoebae harboring environmental chlamydiae actually survive in hospital environments is unknown. We therefore isolated living amoebae with symbiotic chlamydiae from hospital environments. RESULTS: One hundred smear samples were collected from Hokkaido University Hospital, Sapporo, Japan; 50 in winter (February to March, 2012) and 50 in summer (August, 2012), and used for the study. Acanthamoebae were isolated from the smear samples, and endosymbiotic chlamydial traits were assessed by infectivity, cytokine induction, and draft genomic analysis. From these, 23 amoebae were enriched on agar plates spread with heat-killed Escherichia coli. Amoeba prevalence was greater in the summer-collected samples (15/30, 50%) than those of the winter season (8/30, 26.7%), possibly indicating a seasonal variation (p = 0.096). Morphological assessment of cysts revealed 21 amoebae (21/23, 91%) to be Acanthamoeba, and cultures in PYG medium were established for 11 of these amoebae. Three amoebae contained environmental chlamydiae; however, only one amoeba (Acanthamoeba T4) with an environmental chlamydia (Protochlamydia W-9) was shown the infectious ability to Acanthamoeba C3 (reference amoebae). While Protochlamydia W-9 could infect C3 amoeba, it failed to replicate in immortal human epithelial, although exposure of HEp-2 cells to living bacteria induced the proinflammatory cytokine, IL-8. Comparative genome analysis with KEGG revealed similar genomic features compared with other Protochlamydia genomes (UWE25 and R18), except for a lack of genes encoding the type IV secretion system. Interestingly, resistance genes associated with several antibiotics and toxic compounds were identified. CONCLUSION: These findings are the first demonstration of the distribution in a hospital of a living Acanthamoeba carrying an endosymbiotic chlamydial pathogen.


Subject(s)
Acanthamoeba/isolation & purification , Acanthamoeba/microbiology , Chlamydia/isolation & purification , Environmental Microbiology , Hospitals , Anti-Bacterial Agents/pharmacology , Base Sequence , Chlamydia/genetics , Cytokines/metabolism , DNA, Bacterial/genetics , Genes, Bacterial , Humans , Phylogeny , RNA, Ribosomal, 16S/genetics , Seasons , Symbiosis
13.
Infect Immun ; 83(7): 2917-25, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25939513

ABSTRACT

The obligate intracellular bacterium Chlamydia pneumoniae is not only a causative agent of community-acquired pneumonia but is also associated with a more serious chronic disease, asthma, which might be exacerbated by air pollution containing carbon nanoparticles. Although a detailed mechanism of exacerbation remains unknown, the proinflammatory cytokine interleukin-1ß (IL-1ß) is a critical player in the pathogenesis of asthma. C. pneumoniae induces IL-1ß in macrophages via NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome activation and Toll-like receptor 2/4 (TLR2/4) stimulation. Carbon nanoparticles, such as carbon nanotubes (CNTs), can also evoke the NLRP3 inflammasome to trigger IL-1ß secretion from lipopolysaccharide-primed macrophages. This study assessed whether costimulation of C. pneumoniae with CNTs synergistically enhanced IL-1ß secretion from macrophages, and determined the molecular mechanism involved. Enhanced IL-1ß secretion from C. pneumoniae-infected macrophages by CNTs was dose and time dependent. Transmission electron microscopy revealed that C. pneumoniae and CNTs were engulfed concurrently by macrophages. Inhibitors of actin polymerization or caspase-1, a component of the inflammasome, significantly blocked IL-1ß secretion. Gene silencing using small interfering RNA (siRNA) targeting the NLRP3 gene also abolished IL-1ß secretion. Other inhibitors (K(+) efflux inhibitor, cathepsin B inhibitor, and reactive oxygen species-generating inhibitor) also blocked IL-1ß secretion. Taken together, these findings demonstrated that CNTs synergistically enhanced IL-1ß secretion from C. pneumoniae-infected macrophages via the NLRP3 inflammasome and caspase-1 activation, providing novel insight into our understanding of how C. pneumoniae infection can exacerbate asthma.


Subject(s)
Carbon/immunology , Carrier Proteins/metabolism , Chlamydophila pneumoniae/immunology , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Macrophages/immunology , Nanoparticles/metabolism , Caspase 1/metabolism , Cell Line , Endocytosis , Gene Silencing , Humans , Macrophages/microbiology , Microscopy, Electron, Transmission , NLR Family, Pyrin Domain-Containing 3 Protein
14.
J Infect Chemother ; 21(12): 842-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26429774

ABSTRACT

Although sexually transmitted disease due to Chlamydia trachomatis occurs similarly in both men and women, the female urogenital tract differs from that of males anatomically and physiologically, possibly leading to specific polymorphisms of the bacterial surface molecules. In the present study, we therefore characterized polymorphic features in a high-definition phylogenetic marker, polymorphic outer membrane protein (Pmp) F of C. trachomatis strains isolated from male urogenital tracts in Japan (Category: Japan-males, n = 12), when compared with those isolated from female cervical ducts in Japan (Category: Japan-females, n = 11), female cervical ducts in the other country (Category: Ref-females, n = 12) or homosexual male rectums in the other country (Category: Ref-males, n = 7), by general bioinformatics analysis tool with MAFFT software. As a result, phylogenetic reconstruction of the PmpF amino acid sequences showing three distinct clusters revealed that the Japan-males were limited into cluster 1 and 2, although there were only four clusters even though including an outgroup. Meanwhile, the phylogenetic distance values of PmpF passenger domain without hinge region, but not its full-length sequence, showed that the Japan-males were more stable and displayed less diversity when compared with the other categories, supported by the sequence conservation features. Thus, PmpF passenger domain is a useful phylogenetic maker, and the phylogenic features indicate that C. trachomatis strains isolated from male urogenital tracts in Japan may be unique, suggesting an adaptation depending on selective pressure, such as the presence or absence of microbial flora, furthermore possibly connecting to sexual differentiation.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Chlamydia trachomatis/isolation & purification , Chlamydia trachomatis/metabolism , Urinary Tract/microbiology , Chlamydia Infections/microbiology , Female , Humans , Japan , Male , Sexually Transmitted Diseases/microbiology
15.
Environ Microbiol ; 16(2): 486-97, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24460765

ABSTRACT

To elucidate how ancient pathogenic chlamydiae could overcome temperature barriers to adapt to human cells, we characterized a primitive chlamydia found in HS-T3 amoebae (Acanthamoeba) isolated from a hot spring. Phylogenetic analysis revealed the primitive species to be Protochlamydia. In situ hybridization staining showed broad distribution into the amoebal cytoplasm, which was supported by transmission electron microscopic analysis showing typical chlamydial features, with inclusion bodies including both elementary and reticular bodies. Interestingly, although most amoebae isolated from natural environments show reduced growth at 37°C, the HS-T3 amoebae harbouring the Protochlamydia grew well at body temperature. Although infection with Protochlamydia did not confer temperature tolerance to the C3 amoebae, the number of infectious progenies rapidly increased at 37°C with amoebal lysis. In immortalized human epithelial HEp-2 cells, fluorescence microscopic study revealed atypical inclusion of the Protochlamydia, and quantitative real-time polymerase chain reaction analyses also showed an increase in 16S ribosomal RNA DNA amounts. Together, these results showed that the Protochlamydia found in HS-T3 amoebae isolated from a hot spring successfully adapted to immortalized human HEp-2 cells at 37°C, providing further information on the evolution of ancient Protochlamydia to the present pathogenic chlamydiae.


Subject(s)
Acanthamoeba/microbiology , Adaptation, Physiological , Chlamydiales/growth & development , Hot Springs/microbiology , Phylogeny , Cell Line , Chlamydiales/genetics , Chlamydiales/ultrastructure , Hot Temperature , Humans , RNA, Ribosomal, 16S/genetics , Symbiosis
16.
BMC Microbiol ; 14: 330, 2014 Dec 21.
Article in English | MEDLINE | ID: mdl-25528659

ABSTRACT

BACKGROUND: Pathogenic chlamydiae are obligate intracellular pathogens and have adapted successfully to human cells, causing sexually transmitted diseases or pneumonia. Chlamydial outer protein N (CopN) is likely a critical effector protein secreted by the type III secretion system in chlamydiae, which manipulates host cells. However, the mechanisms of its action remain to be clarified. In this work, we aimed to identify previously unidentified CopN effector target in host cells. RESULTS: We first performed a pull-down assay with recombinant glutathione S-transferase (GST) fusion CopN proteins (GST-CpCopN: Chlamydia pneumoniae TW183, GST-CtCopN: Chlamydia trachomatis D/UW-3/CX) as "bait" and soluble lysates obtained from human immortal epithelial HEp-2 cells as "prey", followed by SDS-PAGE with mass spectroscopy (MS). We found that a host cell protein specifically bound to GST-CpCopN, but not GST-CtCopN. MS revealed the host protein to be fructose bisphosphate aldolase A (aldolase A), which plays a key role in glycolytic metabolism. We also confirmed the role of aldolase A in chlamydia-infected HEp-2 cells by using two distinct experiments for gene knockdown with an siRNA specific to aldolase A transcripts, and for assessment of glycolytic enzyme gene expression levels. As a result, both the numbers of chlamydial inclusion-forming units and RpoD transcripts were increased in the chlamydia-infected aldolase A knockdown cells, as compared with the wild-type HEp-2 cells. Meanwhile, chlamydial infection tended to enhance expression of aldolase A. CONCLUSIONS: We discovered that one of the C. pneumoniae CopN targets is the glycolytic enzyme aldolase A. Sequestering aldolase A may be beneficial to bacterial growth in infected host cells.


Subject(s)
Chlamydophila pneumoniae/metabolism , Fructose-Bisphosphate Aldolase/metabolism , Bacterial Outer Membrane Proteins/metabolism , Cell Line , Chlamydia Infections/metabolism , Chlamydia trachomatis/metabolism , Glycolysis/physiology , Humans
17.
J Infect Chemother ; 20(8): 460-4, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24855914

ABSTRACT

Chlamydia trachomatis is an obligate intracellular bacterium that causes a sexually transmitted disease. Ureaplasma parvum is commensal in the human genital tract, with a minimal contribution to urogenital infection. We have recently found that U. parvum has a significant effect on the presence of C. trachomatis in the genital tract of healthy women. We therefore assessed the effect of U. parvum co-incubation on C. trachomatis maturation from reticulate bodies (RBs) to elementary bodies (EBs) in HeLa cells in the absence or presence of interferon (IFN)-γ, which is a critical host defense factor. IFN-γ stimulation of viable U. parvum significantly prompted chlamydial growth with an increase in infectious particles, EBs, in HeLa cells. IFN-γ treatment of killed U. parvum had a similar effect on C. trachomatis maturation in HeLa cells. There was no change in expression of indoleamine 2,3-dioxygenase (IDO) in cultures of viable or killed U. parvum. We concluded that U. parvum co-incubation by IFN-γ helped C. trachomatis to mature from RBs to EBs in HeLa cells, independent of IDO expression. This suggests a novel survival strategy of C. trachomatis against IFN-γ exposure, prompting secondary infection of the genital mucosa, with possible clinical implications.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia trachomatis , Coinfection/microbiology , Interferon-gamma/pharmacology , Ureaplasma Infections/microbiology , Ureaplasma/drug effects , Chlamydia trachomatis/drug effects , Chlamydia trachomatis/growth & development , Coculture Techniques , Female , HeLa Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Models, Biological
18.
Acta Parasitol ; 69(2): 1324-1328, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38775916

ABSTRACT

PURPOSE: Trichomonas vaginalis is a causative agent of common non-viral sexually transmitted infections worldwide. However, the biological features, such as genotypes and endosymbionts, of T. vaginalis isolated in Japan remain unclear. The aim of this study was to characterize the actin-based genotypes and the endosymbionts of T. vaginalis isolated in Sapporo, Japan. METHODS: Three T. vaginalis clinical strains were isolated in Sapporo, Japan between 2019 and 2022. Actin-based genotyping was conducted by sequencing and phylogenetic analyses. The endosymbionts, such as Mycoplasma sp. and Trichomonasvirus, were detected using PCR and RT-PCR, respectively. Furthermore, the detected Mycoplasma spp. were identified using 16S rRNA gene sequencing. RESULTS: Of the three T. vaginalis strains, two belonged to genotype E, whereas one was genotype G as determined by actin-based genotyping. Two of the T. vaginalis strains harbored Mycoplasma spp. Using nearly full-length 16S rRNA gene sequencing, both were identified as Candidatus Mycoplasma girerdii. In contrast, the Trichomonasvirus was not found in the T. vaginalis strains. CONCLUSION: To our knowledge, this is the first report on the characterization of actin-based genotypes and the presence of endosymbiotic Ca. M. girerdii in T. vaginalis strains in Japan. Thus, this study will provide an important impetus for future research.


Subject(s)
Actins , Genotype , Mycoplasma , Phylogeny , RNA, Ribosomal, 16S , Symbiosis , Trichomonas vaginalis , Trichomonas vaginalis/genetics , Trichomonas vaginalis/isolation & purification , Japan , Mycoplasma/genetics , Mycoplasma/isolation & purification , Mycoplasma/classification , Actins/genetics , Humans , RNA, Ribosomal, 16S/genetics , Female , Trichomonas Vaginitis/parasitology
19.
J Infect Dis ; 206(1): 110-6, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22539814

ABSTRACT

BACKGROUND: Human monocytic ehrlichiosis is one of the most prevalent tick-borne zoonoses caused by infection with Ehrlichia chaffeensis. Although E. chaffeensis lacks entire lipopolysaccharide and most peptidoglycan biosynthesis genes, it induces inflammatory cytokines and chemokines. Ehrlichia chaffeensis components that induce inflammation and the responsive host cell pathway are not known. METHODS: Expression of penicillin-binding protein (PBP) in E. chaffeensis was analyzed by reverse-transcription polymerase chain reaction and Bocillin FL binding assay. Next, recombinant PBP, which was high-pressure liquid chromatography purified, and native PBP of E. chaffeensis were investigated for their ability to induce proinflammatory cytokines in the human monocytic leukemia cell line THP-1 and bone marrow-derived macrophages (BMDMs) from wild-type and MyD88 knockout mice. RESULTS: Expression of PBP by E. chaffeensis was upregulated during its intracellular life cycle. PBP induced interleukin 8 or CXCL2, tumor necrosis factor α, interleukin 1ß, and interleukin 10 in THP-1 cells and BMDMs. Cytokine induction by PBP was MyD88-dependent. Removal of PBP from E. chaffeensis lysate using penicillin affinity column and a complementation assay confirmed cytokine-inducing activity of native PBP. CONCLUSIONS: The cytokine-inducing activity by E. chaffeensis PBP provides novel insights into pathogen-associated molecular patterns and pathogenesis of E. chaffeensis infection.


Subject(s)
Cytokines/biosynthesis , Ehrlichia chaffeensis/metabolism , Myeloid Differentiation Factor 88/metabolism , Penicillin-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL2/biosynthesis , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Cytokines/genetics , Cytokines/immunology , Ehrlichia chaffeensis/genetics , Ehrlichia chaffeensis/immunology , Ehrlichiosis/genetics , Ehrlichiosis/metabolism , Humans , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-8/biosynthesis , Interleukin-8/genetics , Interleukin-8/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
20.
Microbiology (Reading) ; 158(Pt 6): 1607-1614, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22403190

ABSTRACT

Parachlamydia acanthamoebae is an obligate intracellular bacterium that infects free-living amoebae (Acanthamoeba), and is a potential human pathogen associated with hospital-acquired pneumonia. The attachment mechanism of this bacteria to host cells is crucial in bacterial pathogenesis, yet remains undetermined. Hence, we obtained monoclonal antibodies (mAbs) specific to either P. acanthamoebae or amoebae in an attempt to elucidate the attachment mechanism involved. Hybridomas of 954 clones were assessed, and we found that four mAbs (mAb38, mAb300, mAb311, mAb562) that were reactive to the amoebae significantly inhibited bacterial attachment. All mAbs recognized amoebal released molecules, and mAb311 also recognized the amoebal surface. mAbs reacted with the bacteria not only within amoebae, but also when they were released from amoebae (except mAb311). Furthermore, a serine protease inhibitor had an inhibitory effect on the bacterial attachment to amoebae, although none of the mAbs had any synergistic effect on the inhibition of attachment by the protease inhibitor. Taken together, we conclude that concurrent P. acanthoamebae attachment to amoebae is required for several amoebal released molecules and serine protease activity, implying the existence of a complicated host-parasite relationship.


Subject(s)
Acanthamoeba/microbiology , Bacterial Adhesion , Chlamydiales/physiology , Protozoan Proteins/metabolism , Serine Proteases/metabolism , Acanthamoeba/enzymology , Acanthamoeba/genetics , Acanthamoeba/metabolism , Chlamydiales/genetics , Host-Parasite Interactions , Protozoan Proteins/genetics , Serine Proteases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL