Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 810
Filter
1.
Front Cell Infect Microbiol ; 14: 1377993, 2024.
Article in English | MEDLINE | ID: mdl-38711928

ABSTRACT

Introduction: Detailed assessment of the population structure of group B Streptococcus (GBS) among adults is still lacking in Saudi Arabia. Here we characterized a representative collection of isolates from colonized and infected adults. Methods: GBS isolates (n=89) were sequenced by Illumina and screened for virulence and antimicrobial resistance determinants. Genetic diversity was assessed by single nucleotide polymorphisms and core-genome MLST analyses. Results: Genome sequences revealed 28 sequence types (STs) and nine distinct serotypes, including uncommon serotypes VII and VIII. Majority of these STs (n=76) belonged to the human-associated clonal complexes (CCs) CC1 (33.71%), CC19 (25.84%), CC17 (11.24%), CC10/CC12 (7.87%), and CC452 (6.74%). Major CCs exhibited intra-lineage serotype diversity, except for the hypervirulent CC17, which exclusively expressed serotype III. Virulence profiling revealed that nearly all isolates (94.38%) carried at least one of the four alpha family protein genes (i.e., alphaC, alp1, alp2/3, and rib), and 92.13% expressed one of the two serine-rich repeat surface proteins Srr1 or Srr2. In addition, most isolates harbored the pilus island (PI)-2a alone (15.73%) or in combination with PI-1 (62.92%), and those carrying PI-2b alone (10.11%) belonged to CC17. Phylogenetic analysis grouped the sequenced isolates according to CCs and further subdivided them along with their serotypes. Overall, isolates across all CC1 phylogenetic clusters expressed Srr1 and carried the PI-1 and PI-2a loci, but differed in genes encoding the alpha-like proteins. CC19 clusters were dominated by the III/rib/srr1/PI-1+PI-2a (43.48%, 10/23) and V/alp1/srr1/PI-1+PI-2a (34.78%, 8/23) lineages, whereas most CC17 isolates (90%, 9/10) had the same III/rib/srr2/P1-2b genetic background. Interestingly, genes encoding the CC17-specific adhesins HvgA and Srr2 were detected in phylogenetically distant isolates belonging to ST1212, suggesting that other highly virulent strains might be circulating within the species. Resistance to macrolides and/or lincosamides across all major CCs (n=48) was associated with the acquisition of erm(B) (62.5%, 30/48), erm(A) (27.1%, 13/48), lsa(C) (8.3%, 4/48), and mef(A) (2.1%, 1/48) genes, whereas resistance to tetracycline was mainly mediated by presence of tet(M) (64.18%, 43/67) and tet(O) (20.9%, 14/67) alone or in combination (13.43%, 9/67). Discussion: These findings underscore the necessity for more rigorous characterization of GBS isolates causing infections.


Subject(s)
Drug Resistance, Bacterial , Genetic Variation , Genome, Bacterial , Multilocus Sequence Typing , Serogroup , Streptococcal Infections , Streptococcus agalactiae , Virulence Factors , Humans , Saudi Arabia , Streptococcus agalactiae/genetics , Streptococcus agalactiae/drug effects , Streptococcus agalactiae/classification , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/isolation & purification , Streptococcal Infections/microbiology , Virulence/genetics , Drug Resistance, Bacterial/genetics , Virulence Factors/genetics , Polymorphism, Single Nucleotide , Anti-Bacterial Agents/pharmacology , Adult , Phylogeny , Whole Genome Sequencing , Genomics , Genotype , Microbial Sensitivity Tests , Female
2.
Vet Res ; 55(1): 60, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750480

ABSTRACT

Bacterial ClpB is an ATP-dependent disaggregate that belongs to the Hsp100/Clp family and facilitates bacterial survival under hostile environmental conditions. Streptococcus agalactiae, which is regarded as the major bacterial pathogen of farmed Nile tilapia (Oreochromis niloticus), is known to cause high mortality and large economic losses. Here, we report a ClpB homologue of S. agalactiae and explore its functionality. S. agalactiae with a clpB deletion mutant (∆clpB) exhibited defective tolerance against heat and acidic stress, without affecting growth or morphology under optimal conditions. Moreover, the ΔclpB mutant exhibited reduced intracellular survival in RAW264.7 cells, diminished adherence to the brain cells of tilapia, increased sensitivity to leukocytes from the head kidney of tilapia and whole blood killing, and reduced mortality and bacterial loads in a tilapia infection assay. Furthermore, the reduced virulence of the ∆clpB mutant was investigated by transcriptome analysis, which revealed that deletion of clpB altered the expression levels of multiple genes that contribute to the stress response as well as certain metabolic pathways. Collectively, our findings demonstrated that ClpB, a molecular chaperone, plays critical roles in heat and acid stress resistance and virulence in S. agalactiae. This finding provides an enhanced understanding of the functionality of this ClpB homologue in gram-positive bacteria and the survival strategy of S. agalactiae against immune clearance during infection.


Subject(s)
Bacterial Proteins , Fish Diseases , Streptococcal Infections , Streptococcus agalactiae , Stress, Physiological , Streptococcus agalactiae/physiology , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/genetics , Virulence , Animals , Streptococcal Infections/veterinary , Streptococcal Infections/microbiology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Fish Diseases/microbiology , Cichlids , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mice , RAW 264.7 Cells
3.
Microb Pathog ; 191: 106675, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705216

ABSTRACT

Bovine mastitis, caused by Streptococcus agalactiae (Group B Streptococcus; GBS), poses significant economic challenges to the global dairy industry. Mouse models serves as valuable tools for assessing GBS-induced infections as an alternative to large animals. This study aimed to investigate the LD50 dose, organ bacterial load, and quantification of peritoneal leukocyte populations for GBS serotypes Ia and II isolates from China and Pakistan. Additionally, we measured indicators such as lactoferrin, albumin, and myeloperoxidase (MPO) activity. Pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6, and IL-2) and anti-inflammatory cytokines (IL-10 and TGF-ß) in serum and tissue samples were evaluated using ELISA and qPCR, respectively. BALB/c mice (4 mice per group) received individual intraperitoneal injections of 100 µl containing specific bacterial inoculum concentrations (ranging from 105 to 109 CFU per mouse) of Chinese and Pakistani GBS isolates (serotypes Ia and II). Control groups received 100 µL of sterile PBS. Results revealed that the LD50 bacterial dose causing 50 % mortality in mice was 107 CFU. The highest bacterial load in all experimental groups was quantified in the peritoneum, followed by blood, mammary gland, liver, spleen, lungs, and brain. The most significant bacterial dissemination was observed in mice inoculated with Pakistani serotype Ia at 24 h, with a subsequent notable decline in bacterial counts at day 3. Notably, infection with Pakistani serotype Ia showed a trend of increased total leukocyte counts, significantly higher than Pakistani serotype II, Chinese Serotype Ia, and Chinese serotype II. A substantial influx of neutrophils and lymphocytes was observed in response to all tested serotypes, with Pakistani serotype Ia inducing a significantly higher influx compared to other groups (Pakistani serotype II, Chinese serotype Ia, and Chinese serotype II). Furthermore, TNF-α, IL-1ß, IL-2, and IL-6 expressions were significantly increased in mice one day after infection with the Pakistani serotype Ia. Compared to mice infected with the Pakistani serotype II, Chinese Serotype Ia, and Chinese serotype II, those infected with the Pakistani serotype Ia isolate exhibited the highest production of IL-10 and TGF-ß, along with significantly increased concentrations of lactoferrin, albumin, and MPO. These findings suggest that the persistence and severity of infection caused by the Pakistani serotype Ia may be linked to its ability to spread to deeper tissues. This study enhances our understanding of the clinical characteristics of bovine mastitis caused by S. agalactiae in China and Pakistan.


Subject(s)
Cytokines , Disease Models, Animal , Mice, Inbred BALB C , Serogroup , Streptococcal Infections , Streptococcus agalactiae , Animals , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/classification , Streptococcus agalactiae/immunology , Streptococcus agalactiae/genetics , Mice , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , China , Cytokines/metabolism , Cytokines/blood , Female , Pakistan , Bacterial Load , Cattle , Lethal Dose 50 , Mastitis, Bovine/microbiology
4.
Vaccine ; 42(16): 3564-3571, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38692955

ABSTRACT

BACKGROUND: Group B Streptococcus (GBS) is a leading cause of morbidity and mortality in young infants worldwide. This study aimed to investigate candidate GBS vaccine targets, virulence factors, and antimicrobial resistance determinants. METHODS: We used whole-genome sequencing to characterize invasive GBS isolates from infants < 3 months of age obtained from a multicenter population-based study conducted from 2015 to 2021 in China. RESULTS: Overall, seven serotypes were detected from 278 GBS isolates, four (Ia, Ib, III, V) of which accounted for 97.8 %. We detected 30 sequence types (including 10 novel types) that were grouped into six clonal complexes (CCs: CC1, CC10, CC17, CC19, CC23 and CC651); three novel ST groups in CC17 were detected, and the rate of CC17, considered a hyperinvasive neonatal clone complex, was attached to 40.6 % (113/278). A total of 98.9 % (275/278) of isolates harbored at least one alpha-like protein gene. All GBS isolates contained at least one of three pilus backbone determinants and the pilus types PI-2b and PI-1 + PI-2a accounted for 79.8 % of the isolates. The 112 serotype III/CC17 GBS isolates were all positive for hvgA. Most of the isolates (75.2 %) were positive for serine-rich repeat glycoprotein determinants (srr1or srr2). Almost all isolates possessed cfb (99.6 %), c1IE (100 %), lmb (95.3 %) or pavA (100 %) gene. Seventy-seven percent of isolates harboured more than three antimicrobial resistance genes with 28.4 % (79/278) gyrA quinoloneresistancedeterminants mutation, 83.8 % (233/278) carrying tet cluster genes and 77.3 % (215/278) carrying erm genes which mediated fluoroquinolone, tetracycline and clindamycin resistance, respectively." CONCLUSIONS: The findings from this large whole-genome sequence of GBS isolates establish important baseline data required for further surveillance and evaluating the impact of future vaccine candidates.


Subject(s)
Streptococcal Infections , Streptococcal Vaccines , Streptococcus agalactiae , Virulence Factors , Whole Genome Sequencing , Humans , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/drug effects , Streptococcus agalactiae/immunology , Streptococcus agalactiae/isolation & purification , Streptococcus agalactiae/classification , Whole Genome Sequencing/methods , Virulence Factors/genetics , Infant , Streptococcal Infections/microbiology , Streptococcal Infections/epidemiology , Streptococcal Infections/prevention & control , Streptococcal Vaccines/immunology , Infant, Newborn , China/epidemiology , Female , Serogroup , Male , Drug Resistance, Bacterial/genetics , Genome, Bacterial , Anti-Bacterial Agents/pharmacology
5.
Microbiology (Reading) ; 170(4)2024 Apr.
Article in English | MEDLINE | ID: mdl-38656296

ABSTRACT

Group B streptococcus (GBS) is a chain-forming commensal bacterium and opportunistic pathogen that resides in the gastrointestinal and genitourinary tract of healthy adults. GBS can cause various infections and related complications in pregnant and nonpregnant women, adults, and newborns. Investigations of the mechanisms by which GBS causes disease pathogenesis often utilize colony count assays to estimate bacterial population size in experimental models. In other streptococci, such as group A streptococcus and pneumococcus, variation in the chain length of the bacteria that can occur naturally or due to mutation can affect facets of pathogenesis, such as adherence to or colonization of a host. No studies have reported a relationship between GBS chain length and pathogenicity. Here, we used GBS strain 874391 and several derivative strains displaying longer chain-forming phenotypes (874391pgapC, 874391ΔcovR, 874391Δstp1) to assess the impact of chain length on bacterial population estimates based on the colony-forming unit (c.f.u.) assay. Disruption of GBS chains via bead beating or sonication in conjunction with fluorescence microscopy was used to compare chaining phenotypes pre- and post-disruption to detect long- and short-chain forms, respectively. We used a murine model of GBS colonization of the female reproductive tract to assess whether chaining may affect bacterial colonization dynamics in the host during chronic infection in vivo. Overall, we found that GBS exhibiting long-chain form can significantly affect population size estimates based on the colony count assay. Additionally, we found that the length of chaining of GBS can affect virulence in the reproductive tract colonization model. Collectively, these findings have implications for studies of GBS that utilize colony count assays to measure GBS populations and establish that chain length can affect infection dynamics and disease pathogenesis for this important opportunistic pathogen.


Subject(s)
Streptococcal Infections , Streptococcus agalactiae , Virulence Factors , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Female , Streptococcal Infections/microbiology , Mice , Animals , Virulence Factors/genetics , Virulence Factors/metabolism , Humans , Colony Count, Microbial , Virulence , Disease Models, Animal , Pregnancy
6.
FEMS Microbiol Rev ; 48(3)2024 May 08.
Article in English | MEDLINE | ID: mdl-38678005

ABSTRACT

Group B Streptococcus (GBS; also known as Streptococcus agalactiae) is an opportunistic bacterial pathogen that causes sepsis, meningitis, pneumonia, and skin and soft tissue infections in neonates and healthy or immunocompromised adults. GBS is well-adapted to survive in humans due to a plethora of virulence mechanisms that afford responses to support bacterial survival in dynamic host environments. These mechanisms and responses include counteraction of cell death from exposure to excess metal ions that can cause mismetallation and cytotoxicity, and strategies to combat molecules such as reactive oxygen and nitrogen species that are generated as part of innate host defence. Cytotoxicity from reactive molecules can stem from damage to proteins, DNA, and membrane lipids, potentially leading to bacterial cell death inside phagocytic cells or within extracellular spaces within the host. Deciphering the ways in which GBS responds to the stress of cytotoxic reactive molecules within the host will benefit the development of novel therapeutic and preventative strategies to manage the burden of GBS disease. This review summarizes knowledge of GBS carriage in humans and the mechanisms used by the bacteria to circumvent killing by these important elements of host immune defence: oxidative stress, nitrosative stress, and stress from metal ion intoxication/mismetallation.


Subject(s)
Metals , Streptococcal Infections , Streptococcus agalactiae , Streptococcus agalactiae/physiology , Streptococcus agalactiae/pathogenicity , Humans , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Metals/metabolism , Metals/toxicity , Oxidative Stress , Reactive Oxygen Species/metabolism , Stress, Physiological , Virulence , Opportunistic Infections/microbiology
7.
Dis Aquat Organ ; 158: 27-36, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38661135

ABSTRACT

Streptococcus agalactiae infection is one of the major factors limiting the expansion of tilapia farming globally. In this study, we investigated the serotype distribution, virulence and antimicrobial resistance of S. agalactiae isolates from tilapia farmed in Lake Volta, Ghana. Isolates from 300 moribund fish were characterised by Gram staining, MALDI-TOF/MS and 16S rRNA sequencing. Serotype identification was based on multiplex polymerase chain reaction (PCR) amplification of the capsular polysaccharide genes. Detection of virulence genes (cfb, fbsA and cspA) and histopathology were used to infer the pathogenicity of the isolates. The susceptibility of isolates to antibiotics was tested using the Kirby-Bauer disk diffusion assay. All 32 isolates identified as S. agalactiae were of serotype Ia. This was notably different from isolates previously collected from the farms in 2017, which belonged to serotype Ib, suggesting a possible serotype replacement. The prevalence of the pathogen was related to the scale of farm operation, with large-scale farms showing higher S. agalactiae positivity. Data from histopathological analysis and PCR amplification of targeted virulence genes confirmed the virulence potential and ability of the isolates to cause systemic infection in tilapia. Except for gentamicin, the majority of the isolates were less resistant to the tested antibiotics. All isolates were fully sensitive to oxytetracycline, erythromycin, florfenicol, enrofloxacin, ampicillin and amoxicillin. This study has improved our understanding of the specific S. agalactiae serotypes circulating in Lake Volta and demonstrates the need for continuous monitoring to guide the use of antimicrobials and vaccines against streptococcal infections in Ghanaian aquaculture systems.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Fish Diseases , Serogroup , Streptococcal Infections , Streptococcus agalactiae , Animals , Streptococcus agalactiae/drug effects , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Ghana/epidemiology , Fish Diseases/microbiology , Streptococcal Infections/veterinary , Streptococcal Infections/microbiology , Streptococcal Infections/epidemiology , Virulence , Anti-Bacterial Agents/pharmacology , Lakes/microbiology , Cichlids , Aquaculture
8.
Nature ; 615(7952): 472-481, 2023 03.
Article in English | MEDLINE | ID: mdl-36859544

ABSTRACT

The meninges are densely innervated by nociceptive sensory neurons that mediate pain and headache1,2. Bacterial meningitis causes life-threatening infections of the meninges and central nervous system, affecting more than 2.5 million people a year3-5. How pain and neuroimmune interactions impact meningeal antibacterial host defences are unclear. Here we show that Nav1.8+ nociceptors signal to immune cells in the meninges through the neuropeptide calcitonin gene-related peptide (CGRP) during infection. This neuroimmune axis inhibits host defences and exacerbates bacterial meningitis. Nociceptor neuron ablation reduced meningeal and brain invasion by two bacterial pathogens: Streptococcus pneumoniae and Streptococcus agalactiae. S. pneumoniae activated nociceptors through its pore-forming toxin pneumolysin to release CGRP from nerve terminals. CGRP acted through receptor activity modifying protein 1 (RAMP1) on meningeal macrophages to polarize their transcriptional responses, suppressing macrophage chemokine expression, neutrophil recruitment and dural antimicrobial defences. Macrophage-specific RAMP1 deficiency or pharmacological blockade of RAMP1 enhanced immune responses and bacterial clearance in the meninges and brain. Therefore, bacteria hijack CGRP-RAMP1 signalling in meningeal macrophages to facilitate brain invasion. Targeting this neuroimmune axis in the meninges can enhance host defences and potentially produce treatments for bacterial meningitis.


Subject(s)
Brain , Meninges , Meningitis, Bacterial , Neuroimmunomodulation , Humans , Brain/immunology , Brain/microbiology , Calcitonin Gene-Related Peptide/metabolism , Meninges/immunology , Meninges/microbiology , Meninges/physiopathology , Pain/etiology , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Meningitis, Bacterial/complications , Meningitis, Bacterial/immunology , Meningitis, Bacterial/microbiology , Meningitis, Bacterial/pathology , Streptococcus agalactiae/immunology , Streptococcus agalactiae/pathogenicity , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity , Nociceptors/metabolism , Receptor Activity-Modifying Protein 1/metabolism , Macrophages/immunology , Macrophages/metabolism
9.
Front Immunol ; 12: 786602, 2021.
Article in English | MEDLINE | ID: mdl-34899755

ABSTRACT

Streptococcus agalactiae, also known as group B streptococcus (GBS), can cause pneumonia, meningitis, and bacteremia, making it a pathogen that can increase the risk of death in newborns and immunodeficient individuals. Neutrophils are the first barrier to a host's innate immune defense against these infections. Fpr2(Formyl peptide receptor 2) is an important chemotactic receptor of neutrophils, though its activation would cause pro- and anti-inflammatory effects. In this study, we found that mice without Fpr2 receptor were highly susceptible to GBS infections. These mice demonstrated decreased chemotaxis to neutrophils, decreased bactericidal ability of neutrophils, and high mortality. RNA-seq and Luminex assay indicated that Fpr2 activates key signal molecules downstream and produces chemokines CXCL1/2 to chemotaxis neutrophils. Like Fpr2-/-, CXCL1/2 or neutrophil depletion impairs host's ability to defend against GBS infection. Altogether, these data indicate that Fpr2 contributes to a host's ability to control GBS infection and that a lack of Fpr2 was associated with selective impairment during the production of chemokines CXCL1 and CXCL2 as well as neutrophil recruitment. Here, We clarified that Fpr2, as a chemotactic receptor, could not only directly chemotactic neutrophils, but also regulate the production of chemokines to control infection by chemotactic neutrophils.


Subject(s)
Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Chemotaxis, Leukocyte , Neutrophil Infiltration , Neutrophils/immunology , Receptors, Formyl Peptide/metabolism , Streptococcal Infections/prevention & control , Streptococcus agalactiae/immunology , Animals , Disease Models, Animal , Host-Pathogen Interactions , Immunity, Innate , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Neutrophils/microbiology , Receptors, Formyl Peptide/genetics , Signal Transduction , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Streptococcal Infections/microbiology , Streptococcus agalactiae/pathogenicity , Time Factors
10.
Microbiologyopen ; 10(6): e1256, 2021 11.
Article in English | MEDLINE | ID: mdl-34964296

ABSTRACT

Although Streptococcus agalactiae periprosthetic joint infection (PJI) is not as prevalent as staphylococcal PJI, invasive S. agalactiae infection is not uncommon. Here, RNA-seq was used to perform transcriptomic analysis of S. agalactiae PJI using fluid derived from sonication of explanted arthroplasties of subjects with S. agalactiae PJI, with results compared to those of S. agalactiae strain NEM316 grown in vitro. A total of 227 genes with outlier expression were found (164 upregulated and 63 downregulated) between PJI sonicate fluid and in vitro conditions. Functional enrichment analysis showed genes involved in mobilome and inorganic ion transport and metabolism to be most enriched. Genes involved in nickel, copper, and zinc transport, were upregulated. Among known virulence factors, cyl operon genes, encoding ß-hemolysin/cytolysin, were consistently highly expressed in PJI versus in vitro. The data presented provide insight into S. agalactiae PJI pathogenesis and may be a resource for identification of novel PJI therapeutics or vaccines against invasive S. agalactiae infections.


Subject(s)
Joint Prosthesis/adverse effects , Prosthesis-Related Infections/microbiology , Streptococcal Infections/microbiology , Streptococcus agalactiae/genetics , Transcriptome , Adult , Aged , Bacterial Adhesion/genetics , Biofilms/growth & development , Female , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genome, Bacterial , Humans , Male , Middle Aged , RNA-Seq , Streptococcus agalactiae/pathogenicity , Streptococcus agalactiae/physiology , Virulence Factors/genetics , Whole Genome Sequencing
11.
PLoS Pathog ; 17(12): e1010121, 2021 12.
Article in English | MEDLINE | ID: mdl-34871327

ABSTRACT

Type VII secretion systems (T7SS) have been identified in Actinobacteria and Firmicutes and have been shown to secrete effector proteins with functions in virulence, host toxicity, and/or interbacterial killing in a few genera. Bioinformatic analysis indicates that isolates of Group B Streptococcus (GBS) encode at least four distinct subtypes of T7SS machinery, three of which encode adjacent putative T7SS effectors with WXG and LXG motifs. However, the function of T7SS in GBS pathogenesis is unknown. Here we assessed the role of the most abundant GBS T7SS subtype during GBS pathogenesis. In a murine model of hematogenous meningitis, mice infected with GBS lacking a functional T7SS or lacking the secreted WXG100 effector EsxA exhibited less mortality, lower bacterial burdens in tissues, and decreased inflammation in the brain compared to mice infected with the parental GBS strain. We further showed that this T7SS induces cytotoxicity in brain endothelium and that EsxA contributes to these cytotoxicity phenotypes in a WXG motif-dependent manner. Finally, we determined that EsxA is a pore-forming protein, thus demonstrating the first role for a non-mycobacterial EsxA homolog in pore formation. This work reveals the importance of a T7SS in host-GBS interactions and has implications for T7SS effector function in other Gram-positive bacteria.


Subject(s)
Streptococcal Infections/metabolism , Streptococcus agalactiae/pathogenicity , Type VII Secretion Systems/metabolism , Virulence/physiology , Animals , Bacterial Proteins/metabolism , Cells, Cultured , Humans , Mice , Streptococcus agalactiae/metabolism
12.
Emerg Microbes Infect ; 10(1): 2113-2124, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34727007

ABSTRACT

The clustered regularly interspaced palindromic repeats (CRISPR)-Cas (CRISPR-associated) system is a prokaryotic defence against invading mobile genetic elements, such as bacteriophages or exogenous plasmids. Beyond this, this system has been shown to play an important role in controlling the virulence of some bacterial pathogens. Streptococcus agalactiae strain GD201008-001, a causative agent of septicemia and meningitis in tilapia, contains a single type II CRISPR-Cas system with Cas9 as a signature protein. In this study, we found that the deletion of CRISPR significantly reduced adhesion, invasion, cytotoxicity and haemolysis, and caused severely attenuated virulence in the piscine S. agalactiae strain. RNA-Seq identified 236 endogenous genes regulated by CRISPR, with 159 genes upregulated and 77 genes downregulated. The resulting change in gene transcription by CRISPR was much more pronounced than that by cas9 in this bacterium, indicating CRISPR-mediated endogenous gene regulation was mostly independently of cas9. Subsequent studies showed that CovR/S two-component system was transcriptionally upregulated due to CRISPR deletion, which repressed the expression of the cylE gene coding for a cytolytic toxin, and thus decreased the activity of ß-haemolysin/cytolysin. However, upregulation of CovR/S was not the contributor to the attenuation phenotype of ΔCRISPR. Further, we demonstrated that CRISPR is capable of repressing the expression of Toll-like receptor 2 (TLR2)-activating lipoprotein Sag0671 and thus dampens the innate immune response. This study revealed that the CRISPR system of S. agalactiae exhibited extraordinary potential capability in the regulation of endogenous transcripts, which contributes to bacterial innate immune evasion and virulence.


Subject(s)
Bacterial Proteins/genetics , Fish Diseases/microbiology , Gene Expression Regulation, Bacterial , Streptococcal Infections/veterinary , Streptococcus agalactiae/pathogenicity , Animals , Bacterial Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Female , Fishes , Mice , Mice, Inbred C57BL , Perforin/genetics , Perforin/metabolism , Streptococcal Infections/microbiology , Streptococcus agalactiae/genetics , Streptococcus agalactiae/physiology , Virulence
13.
Biomolecules ; 11(10)2021 10 13.
Article in English | MEDLINE | ID: mdl-34680142

ABSTRACT

DnaJ proteins or heat shock protein 40s (HSP40s) form one of the largest heat shock protein families. In this study, 2 cDNAs encoding Nile tilapia (Oreochromis niloticus) DnaJ proteins (On-DnaJ B9b and On-DnaJ C3a) were successfully cloned and characterized. The structures and organizations of these two genes are first reported in the present study. On-DnaJ B9b is approximately 2.1 kb long and contains 2 exons and 1 intron, while On-DnaJ C3a is approximately 12 kb long and contains 12 exons and 11 introns. Under normal conditions, On-DnaJ B9b mRNA is highly expressed in gonad and trunk kidney tissues, while On-DnaJ C3a transcripts are abundantly expressed in gills, intestine, liver, and trunk kidney tissues. Following pathogenic infections, the expression of both genes is induced in the liver, spleen and head kidney tissues of Nile tilapia that were infected with two virulent pathogenic bacteria, Streptococcus agalactiae and Flavobacterium columnare. Silencing of these two genes was first carried out, and the results clearly indicated their crucial roles under both heat and bacterial stress conditions. The fundamental knowledge obtained from this study indicates the characteristic basic biofunctions of heat shock proteins in the regulation of intracellular proteins during infection, which involve preventing protein aggregation, promoting protein refolding, and activating unfolded protein degradation.


Subject(s)
Cichlids/genetics , HSP40 Heat-Shock Proteins/genetics , Heat-Shock Proteins/genetics , Immunity, Innate/genetics , Animals , Cichlids/immunology , Cichlids/microbiology , Cichlids/physiology , Flavobacterium/pathogenicity , Gene Expression Regulation/immunology , Hot Temperature/adverse effects , Kidney/metabolism , Kidney/microbiology , Liver/metabolism , Liver/microbiology , Spleen/metabolism , Spleen/microbiology , Streptococcus agalactiae/pathogenicity
14.
PLoS Genet ; 17(9): e1009761, 2021 09.
Article in English | MEDLINE | ID: mdl-34491998

ABSTRACT

Virulence of the neonatal pathogen Group B Streptococcus is under the control of the master regulator CovR. Inactivation of CovR is associated with large-scale transcriptome remodeling and impairs almost every step of the interaction between the pathogen and the host. However, transcriptome analyses suggested a plasticity of the CovR signaling pathway in clinical isolates leading to phenotypic heterogeneity in the bacterial population. In this study, we characterized the CovR regulatory network in a strain representative of the CC-17 hypervirulent lineage responsible of the majority of neonatal meningitis. Transcriptome and genome-wide binding analysis reveal the architecture of the CovR network characterized by the direct repression of a large array of virulence-associated genes and the extent of co-regulation at specific loci. Comparative functional analysis of the signaling network links strain-specificities to the regulation of the pan-genome, including the two specific hypervirulent adhesins and horizontally acquired genes, to mutations in CovR-regulated promoters, and to variability in CovR activation by phosphorylation. This regulatory adaptation occurs at the level of genes, promoters, and of CovR itself, and allows to globally reshape the expression of virulence genes. Overall, our results reveal the direct, coordinated, and strain-specific regulation of virulence genes by the master regulator CovR and suggest that the intra-species evolution of the signaling network is as important as the expression of specific virulence factors in the emergence of clone associated with specific diseases.


Subject(s)
Bacterial Proteins/physiology , Gene Regulatory Networks , Streptococcus agalactiae/pathogenicity , Virulence Factors/physiology , Virulence/genetics , Bacterial Proteins/genetics , Chromosomes, Bacterial , Genes, Bacterial , Host-Pathogen Interactions , Humans , Promoter Regions, Genetic , Prophages/genetics , Streptococcus agalactiae/genetics , Transcription, Genetic/physiology , Virulence Factors/genetics
15.
PLoS Pathog ; 17(8): e1009791, 2021 08.
Article in English | MEDLINE | ID: mdl-34370789

ABSTRACT

In many Gram-positive bacteria, the redox-sensing transcriptional repressor Rex controls central carbon and energy metabolism by sensing the intra cellular balance between the reduced and oxidized forms of nicotinamide adenine dinucleotide; the NADH/NAD+ ratio. Here, we report high-resolution crystal structures and characterization of a Rex ortholog (Gbs1167) in the opportunistic pathogen, Streptococcus agalactiae, also known as group B streptococcus (GBS). We present structures of Rex bound to NAD+ and to a DNA operator which are the first structures of a Rex-family member from a pathogenic bacterium. The structures reveal the molecular basis of DNA binding and the conformation alterations between the free NAD+ complex and DNA-bound form of Rex. Transcriptomic analysis revealed that GBS Rex controls not only central metabolism, but also expression of the monocistronic rex gene as well as virulence gene expression. Rex enhances GBS virulence after disseminated infection in mice. Mechanistically, NAD+ stabilizes Rex as a repressor in the absence of NADH. However, GBS Rex is unique compared to Rex regulators previously characterized because of its sensing mechanism: we show that it primarily responds to NAD+ levels (or growth rate) rather than to the NADH/NAD+ ratio. These results indicate that Rex plays a key role in GBS pathogenicity by modulating virulence factor gene expression and carbon metabolism to harvest nutrients from the host.


Subject(s)
Bacterial Proteins/genetics , Gene Products, rex/genetics , NAD/deficiency , Regulon , Streptococcal Infections/microbiology , Streptococcus agalactiae/pathogenicity , Virulence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Female , Gene Expression Profiling , Gene Products, rex/chemistry , Gene Products, rex/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Binding , Protein Conformation , Streptococcal Infections/metabolism
16.
Acta Obstet Gynecol Scand ; 100(10): 1814-1821, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34255864

ABSTRACT

INTRODUCTION: Vaginal colonization with Streptococcus agalactiae (group B streptococci) is hypothesized to constitute a risk factor for preterm prelabor rupture of membranes. In vitro studies have shown that S. agalactiae strains isolated from infants with neonatal sepsis adhere to chorion cells of the human chorioamniotic membrane. However, it is still unknown whether S. agalactiae strains penetrate the chorioamniotic membranes and whether S. agalactiae colonization affects the biomechanical properties of the membranes and thus contributes to increased risk of preterm prelabor rupture. The aim of this in vitro study was to explore if different strains of S. agalactiae penetrate and affect the biomechanical properties of human chorioamniotic membranes. MATERIAL AND METHODS: Three different strains of S. agalactiae were obtained, one from an early-onset neonatal infection, one from a case of preterm prelabor rupture of membranes and one from a healthy pregnant carrier. Chorioamniotic membranes from elective cesarean deliveries were either incubated with S. agalactiae or mounted in a two-chamber incubation cell generating a "maternal" and a "fetal" chamber and incubated with S. agalactiae in the maternal chamber. Subsequently the membranes were examined to evaluate S. agalactiae attachment, penetration and the effect on the biomechanical properties. RESULTS: At 5 h after incubation, S. agalactiae adhered to the chorioamniotic membranes with increased number at 20 h. Streptococcus agalactiae did not penetrate the membranes even after 20 h of incubation. Streptococcus agalactiae increased the ultimate tensile stress needed to rupture the membranes and increased the work needed to rupture the membranes as well as the elastic modulus. CONCLUSIONS: Human chorioamniotic membranes constitute a physical barrier against S. agalactiae infections. Moreover, S. agalactiae infection leads to increased strength of the membranes.


Subject(s)
Chorion/microbiology , Fetal Membranes, Premature Rupture/microbiology , Pregnancy Complications, Infectious , Streptococcal Infections , Streptococcus agalactiae/pathogenicity , Female , Humans , In Vitro Techniques , Pregnancy
17.
Front Immunol ; 12: 667781, 2021.
Article in English | MEDLINE | ID: mdl-34093564

ABSTRACT

Leukocyte cell-derived chemotaxin 2 (LECT2) is a multifunctional cytokine that especially plays an important role in innate immune. However, the roles of LECT2 in the immune response of the economically important fish Nile tilapia (Oreochromis niloticus) against bacterial infection remains unclear. In this study, a lect2 gene from Nile tilapia (On-lect2) was identified, and its roles in the fish's immune response against bacterial infection were determined and characterised. On-lect2 contains an open reading frame of 456 bp that encodes a peptide of 151 amino acids, as well as the conservative peptidase M23 domain. On-LECT2 is 62%-84% identical to other fish species and about 50% identical to mammals. The highest transcriptional level of On-lect2 was detected in the liver, whereas the lowest levels were detected in the other tissues. Moreover, the On-LECT2 protein is located mainly in the brain and head kidney. The transcriptional levels of On-lect2 substantially increased in the head kidney, brain, liver and spleen after Streptococcus agalactiae infection. Knockdown On-lect2 led to higher mortality due to liver necrosis or haemorrhage and splenomegaly. In vitro analysis indicated that the recombinant protein of On-LECT2 improved phagocytic activity of head kidney-derived macrophages. In vivo challenge experiments revealed several functions of On-LECT2 in the immune response of Nile tilapia against bacterial infection, including promotion of inflammation, reduction of tissue damages and improvement of survival rate.


Subject(s)
Cichlids/microbiology , Fish Diseases/prevention & control , Fish Proteins/metabolism , Immunity, Innate , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/microbiology , Streptococcal Infections/veterinary , Streptococcus agalactiae/pathogenicity , Animals , Cichlids/genetics , Cichlids/immunology , Cichlids/metabolism , Fish Diseases/immunology , Fish Diseases/metabolism , Fish Diseases/microbiology , Fish Proteins/genetics , Host-Pathogen Interactions , Intercellular Signaling Peptides and Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Streptococcal Infections/metabolism , Streptococcal Infections/microbiology , Streptococcal Infections/prevention & control , Streptococcus agalactiae/immunology , Transcription, Genetic
18.
PLoS One ; 16(6): e0253242, 2021.
Article in English | MEDLINE | ID: mdl-34129624

ABSTRACT

Streptococcus agalactiae (group B streptococcus or GBS) is a commensal bacterium that can frequently behave as a pathogen, particularly in the neonatal period and in the elderly. The gut is a primary site of GBS colonization and a potential port of entry during neonatal infections caused by hypervirulent clonal complex 17 (CC17) strains. Here we studied the interactions between the prototypical CC17 BM110 strain and polarized enterocytes using the Caco-2 cell line. GBS could adhere to and invade these cells through their apical or basolateral surfaces. Basolateral invasion was considerably more efficient than apical invasion and predominated under conditions resulting in weakening of cell-to-cell junctions. Bacterial internalization occurred by a mechanism involving caveolae- and lipid raft-dependent endocytosis and actin re-organization, but not clathrin-dependent endocytosis. In the first steps of Caco-2 invasion, GBS colocalized with the early endocytic marker EEA-1, to later reside in acidic vacuoles. Taken together, these data suggest that CC17 GBS selectively adheres to the lateral surface of enterocytes from which it enters through caveolar lipid rafts using a classical, actin-dependent endocytic pathway. These data may be useful to develop alternative preventive strategies aimed at blocking GBS invasion of the intestinal barrier.


Subject(s)
Enterocytes/microbiology , Streptococcal Infections/microbiology , Streptococcus agalactiae/pathogenicity , Bacterial Adhesion , Caco-2 Cells/microbiology , Endocytosis , Humans , Intercellular Junctions/microbiology , Microscopy, Fluorescence , Streptococcus agalactiae/physiology , Virulence
19.
Res Vet Sci ; 138: 109-115, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34126449

ABSTRACT

Streptococcus agalactiae (GBS) is an important pathogen that has increasingly received attention for its role in invasive infections and its broad host range. Research on the regulation of gene expression could illuminate GBS pathogenesis. We previously identified a novel transcriptional regulator XtgS, which is a negative regulator of GBS pathogenicity. Here, we demonstrate that XtgS overexpression significantly attenuated GBS virulence in zebrafish infection tests, and XtgS indirectly downregulated the transcription of two iron transport systems based on the results of transcriptomic analysis, electrophoretic mobility shift assays (EMSAs) and lacZ fusion assays. Subsequent studies verified that the inactivation of iron transport system 1 resulted in GBS excessive iron accumulation and attenuated virulence. Thus, we infer that the downregulation of iron transport system 1 caused by XtgS overexpression probably attenuates bacterial virulence, which partially clarifies the mechanism by which XtgS alleviates the pathogenesis. These findings provide new insights into the relationship between exogenous transcriptional regulation and bacterial virulence.


Subject(s)
Bacterial Proteins/genetics , Fish Diseases/microbiology , Iron/metabolism , Streptococcal Infections/veterinary , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Transcription Factors/genetics , Zebrafish , Animals , Bacterial Proteins/metabolism , Streptococcal Infections/microbiology , Transcription Factors/metabolism , Virulence/genetics
20.
Pediatr Infect Dis J ; 40(7): 663-668, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34097659

ABSTRACT

BACKGROUND: Prevention strategies can reduce the incidence of early-onset group B Streptococcus (GBS) neonatal sepsis (EOGBS). Rates of GBS colonization and infection vary among regions within China. China has not adopted a unified prevention strategy. METHODS: To assess strategies to reduce EOGBS in China, models were developed to quantify residual EOGBS rates with intrapartum antibiotic prophylaxis in infants ≥ 35 weeks' gestation in risk factor-based and antepartum screening-based strategies. Maternal GBS colonization rates and EOGBS incidence in 3 regions of China (A: Xiamen of Fujian province, B: Shanghai and C: Liuzhou of Guangxi province) were estimated from published data. RESULTS: Estimates for GBS colonization and attack rates were 21.6%, 11.7% and 6.1% and 1.79, 1.79 and 0.58 per 1000 live births for regions A, B and C, respectively. Modeling predicted that strategies including screening cultures beginning at 36 weeks' gestation and intrapartum antibiotic prophylaxis in 90% of eligible parturients could reduce EOGBS incidence to 0.44, 0.50 and 0.16 per 1000 live births in these regions. In region C, the expected EOGBS rate could be reduced to 0.28 per 1000 using a risk factor-based strategy. CONCLUSIONS: Different strategies for preventing EOGBS may be needed in different regions of mainland China. Screening strategies may be most appropriate in regions with higher attack rates, even with moderate levels of maternal GBS colonization. In areas with low attack rates, risk factor strategies that reduce morbidity by at least one-third may suffice.


Subject(s)
Infectious Disease Transmission, Vertical/prevention & control , Neonatal Sepsis/prevention & control , Streptococcal Infections/prevention & control , Streptococcus agalactiae/pathogenicity , Antibiotic Prophylaxis , China/epidemiology , Female , Geography , Gestational Age , Humans , Incidence , Infant, Newborn , Neonatal Sepsis/epidemiology , Pregnancy , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/prevention & control , Risk Factors , Streptococcal Infections/blood , Streptococcal Infections/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...