Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 908
Filter
1.
Microbiology (Reading) ; 170(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-39177453

ABSTRACT

Escherichia coli (E. coli) is a major cause of urinary tract infections, bacteraemia, and sepsis. CFT073 is a prototypic, urosepsis isolate of sequence type (ST) 73. This laboratory, among others, has shown that strain CFT073 is resistant to serum, with capsule and other extracellular polysaccharides imparting resistance. The interplay of such polysaccharides remains under-explored. This study has shown that CFT073 mutants deficient in lipopolysaccharide (LPS) O-antigen and capsule display exquisite serum sensitivity. Additionally, O-antigen and LPS outer core mutants displayed significantly decreased surface K2 capsule, coupled with increased unbound K2 capsule being detected in the supernatant. The R1 core and O6 antigen are involved in the tethering of K2 capsule to the CFT073 cell surface, highlighting the importance of the R1 core in serum resistance. The dependence of capsule on LPS was shown to be post-transcriptional and related to changes in cell surface hydrophobicity. Furthermore, immunofluorescence microscopy suggested that the surface pattern of capsule is altered in such LPS core mutants, which display a punctate capsule pattern. Finally, targeting LPS biosynthesis using sub-inhibitory concentrations of a WaaG inhibitor resulted in increased serum sensitivity and decreased capsule in CFT073. Interestingly, the dependency of capsule on LPS has been observed previously in other Enterobacteria, indicating that the synergy between these polysaccharides is not just strain, serotype or species-specific but may be conserved across several pathogenic Gram-negative species. Therefore, using WaaG inhibitor derivatives to target LPS is a promising therapeutic strategy to reduce morbidity and mortality by reducing or eliminating surface capsule.


Subject(s)
Bacterial Capsules , Lipopolysaccharides , Lipopolysaccharides/metabolism , Bacterial Capsules/metabolism , Bacterial Capsules/genetics , Humans , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/drug effects , Extraintestinal Pathogenic Escherichia coli/metabolism , O Antigens/genetics , O Antigens/metabolism , Escherichia coli Infections/microbiology , Escherichia coli/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Mutation
2.
Nat Commun ; 15(1): 6946, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138169

ABSTRACT

Hypervirulent Klebsiella pneumoniae (HvKP) is an emerging bacterial pathogen causing invasive infection in immune-competent humans. The hypervirulence is strongly linked to the overproduction of hypermucoviscous capsule, but the underlying regulatory mechanisms of hypermucoviscosity (HMV) have been elusive, especially at the post-transcriptional level mediated by small noncoding RNAs (sRNAs). Using a recently developed RNA interactome profiling approach iRIL-seq, we interrogate the Hfq-associated sRNA regulatory network and establish an intracellular RNA-RNA interactome in HvKP. Our data reveal numerous interactions between sRNAs and HMV-related mRNAs, and identify a plethora of sRNAs that repress or promote HMV. One of the strongest HMV repressors is ArcZ, which is activated by the catabolite regulator CRP and targets many HMV-related genes including mlaA and fbp. We discover that MlaA and its function in phospholipid transport is crucial for capsule retention and HMV, inactivation of which abolishes Klebsiella virulence in mice. ArcZ overexpression drastically reduces bacterial burden in mice and reduces HMV in multiple hypervirulent and carbapenem-resistant clinical isolates, indicating ArcZ is a potent RNA inhibitor of bacterial pneumonia with therapeutic potential. Our work unravels a novel CRP-ArcZ-MlaA regulatory circuit of HMV and provides mechanistic insights into the posttranscriptional virulence control in a superbug of global concern.


Subject(s)
Bacterial Capsules , Bacterial Proteins , Gene Expression Regulation, Bacterial , Klebsiella Infections , Klebsiella pneumoniae , RNA, Bacterial , RNA, Small Untranslated , Klebsiella pneumoniae/pathogenicity , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Animals , Virulence/genetics , Mice , Klebsiella Infections/microbiology , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Capsules/metabolism , Bacterial Capsules/genetics , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Humans , Female , Host Factor 1 Protein/metabolism , Host Factor 1 Protein/genetics
3.
J Appl Microbiol ; 135(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39090973

ABSTRACT

AIMS: Hypervirulent Klebsiella pneumoniae (hvKp) causes invasive community-acquired infections in healthy individuals, and hypermucoviscosity (HMV) is the main phenotype associated with hvKp. This study investigates the impact of microaerobic environment availability on the mucoviscosity of K. pneumoniae. METHODS AND RESULTS: By culturing 25 clinical strains under microaerobic and aerobic environments, we observed a notable reduction in mucoviscosity in microaerobic environments. RNA sequencing and qRT-PCR revealed downregulated expressions of capsule synthesis genes (galf, orf2, wzi, wza, wzb, wzc, wcaj, manC, manB, and ugd) and regulatory genes (rmpA, rmpD, and rmpC) under microaerobic conditions. Transmission electron microscopy and Indian ink staining analysis were performed, revealing that the capsular thickness of K. pneumoniae decreased by half in microaerobic conditions compared to aerobic conditions. Deletion of rmpD and rmpC caused the loss of the HMV phenotype in both aerobic and microaerobic conditions. However, compared to wild-type strain in microaerobic condition, only rmpD overexpression strain, and not rmpC overexpression strain, displayed a significant increase in capsule thickness in microaerobic conditions. CONCLUSIONS: Microaerobic conditions can suppress the mucoviscosity of K. pneumoniae, but this suppression can be overcome by altering the expression of rmpD, indicating a specific function for rmpD in the oxygen environmental adaptation of K. pneumoniae.


Subject(s)
Bacterial Proteins , Klebsiella pneumoniae , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Aerobiosis , Humans , Gene Expression Regulation, Bacterial , Phenotype , Klebsiella Infections/microbiology , Bacterial Capsules/metabolism , Bacterial Capsules/genetics , Virulence/genetics
4.
FASEB J ; 38(13): e23763, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38954404

ABSTRACT

Riemerella anatipestifer is a pathogenic bacterium that causes duck serositis and meningitis, leading to significant harm to the duck industry. To escape from the host immune system, the meningitis-causing bacteria must survive and multiply in the bloodstream, relying on specific virulence factors such as capsules. Therefore, it is essential to study the genes involved in capsule biosynthesis in R. anatipestifer. In this study, we successfully constructed gene deletion mutants Δ3820 and Δ3830, targeting the GE296_RS03820 and GE296_RS03830 genes, respectively, using the RA-LZ01 strain as the parental strain. The growth kinetics analysis revealed that these two genes contribute to bacterial growth. Transmission and scanning electron microscopy (TEM and SEM) and silver staining showed that Δ3820 and Δ3830 produced the altered capsules and compounds of capsular polysaccharides (CPSs). Serum resistance test showed the mutants also exhibited reduced C3b deposition and decreased resistance serum killing. In vivo, Δ3820 and Δ3830 exhibited markedly declining capacity to cross the blood-brain barrier, compared to RA-LZ01. These findings indicate that the GE296_RS03820 and GE296_RS03830 genes are involved in CPSs biosynthesis and play a key role in the pathogenicity of R. anatipestifer. Furthermore, Δ3820 and Δ3830 mutants presented a tendency toward higher survival rates from RA-LZ01 challenge in vivo. Additionally, sera from ducklings immunized with the mutants showed cross-immunoreactivity with different serotypes of R. anatipestifer, including 1, 2, 7 and 10. Western blot and SDS-PAGE assays revealed that the altered CPSs of Δ3820 and Δ3830 resulted in the exposure of some conserved proteins playing the key role in the cross-immunoreactivity. Our study clearly demonstrated that the GE296_RS03820 and GE296_RS03830 genes are involved in CPS biosynthesis in R. anatipestifer and the capsule is a target for attenuation in vaccine development.


Subject(s)
Bacterial Capsules , Ducks , Flavobacteriaceae Infections , Riemerella , Riemerella/genetics , Riemerella/pathogenicity , Riemerella/metabolism , Animals , Ducks/microbiology , Bacterial Capsules/genetics , Bacterial Capsules/metabolism , Flavobacteriaceae Infections/microbiology , Flavobacteriaceae Infections/veterinary , Poultry Diseases/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Polysaccharides, Bacterial/biosynthesis , Virulence Factors/genetics , Gene Deletion
5.
Appl Environ Microbiol ; 90(8): e0035424, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39012166

ABSTRACT

Infections caused by antimicrobial-resistant Escherichia coli are the leading cause of death attributed to antimicrobial resistance (AMR) worldwide, and the known AMR mechanisms involve a range of functional proteins. Here, we employed a pan-genome wide association study (GWAS) approach on over 1,000 E. coli isolates from sick dogs collected across the US and Canada and identified a strong statistical association (empirical P < 0.01) of AMR, involving a range of antibiotics to a group 1 capsular (CPS) gene cluster. This cluster included genes under relaxed selection pressure, had several loci missing, and had pseudogenes for other key loci. Furthermore, this cluster is widespread in E. coli and Klebsiella clinical isolates across multiple host species. Earlier studies demonstrated that the octameric CPS polysaccharide export protein Wza can transmit macrolide antibiotics into the E. coli periplasm. We suggest that the CPS in question, and its highly divergent Wza, functions as an antibiotic trap, preventing antimicrobial penetration. We also highlight the high diversity of lineages circulating in dogs across all regions studied, the overlap with human lineages, and regional prevalence of resistance to multiple antimicrobial classes. IMPORTANCE: Much of the human genomic epidemiology data available for E. coli mechanism discovery studies has been heavily biased toward shiga-toxin producing strains from humans and livestock. E. coli occupies many niches and produces a wide variety of other significant pathotypes, including some implicated in chronic disease. We hypothesized that since dogs tend to share similar strains with their owners and are treated with similar antibiotics, their pathogenic isolates will harbor unexplored AMR mechanisms of importance to humans as well as animals. By comparing over 1,000 genomes with in vitro antimicrobial susceptibility data from sick dogs across the US and Canada, we identified a strong multidrug resistance association with an operon that appears to have once conferred a type 1 capsule production system.


Subject(s)
Anti-Bacterial Agents , Dog Diseases , Drug Resistance, Multiple, Bacterial , Escherichia coli Infections , Escherichia coli , Dogs , Animals , Escherichia coli/genetics , Escherichia coli/drug effects , Dog Diseases/microbiology , Escherichia coli Infections/veterinary , Escherichia coli Infections/microbiology , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Canada , Genome-Wide Association Study , Genome, Bacterial , United States , Bacterial Capsules/genetics , Multigene Family , Evolution, Molecular , Genomics , Escherichia coli Proteins/genetics
6.
Zhonghua Yu Fang Yi Xue Za Zhi ; 58(7): 992-997, 2024 Jul 06.
Article in Chinese | MEDLINE | ID: mdl-39034782

ABSTRACT

Objective: To investigate the effects of wza gene deletion in Klebsiella pneumoniae on capsule formation ability and bacteriophage sensitivity. Methods: The wza deletion mutant strain was constructed through a temperature-sensitive plasmid-mediated homologous recombination. The growth curves of W14 and Δwza were detected by measuring the optical density OD600. In order to analyze the effect of gene wza on bacterial capsule formation, wild-type strain W14 and Δwza mutant strain were detected by transmission electron microscope, and their capsule contents were measured by quantifying the uronic acid contents. The plaque assay was used to detect bacterial sensitivity to bacteriophage in wild-type strain W14 and Δwza mutant strain. The t test was used to compare whether there were differences in the contents of uronic acid in the capsules of wild-type strain W14 and Δwza mutant strain. Results: The PCR results revealed that the Δwza mutant strain was successfully constructed. Compared with wild-type strain W14, the growth curves of Δwza on the solid plates demonstrated a slightly slower growth. However, no difference in growth was observed among wild-type strain W14 and Δwza mutant strains in LB broth. The transmission electron microscope results showed that wza gene deletion resulted in the loss of capsule in bacteria. The uronic acid content assay suggested that the capsule content was significantly decreased in Δwza mutant strain (45.963±2.795) µg/ml compared with wild-type strain W14 (138.800±5.201) µg/ml. There was a statistical difference between the two groups (t=27.233, P<0.001). The plaque assay indicated that bacteria lost its sensitivity to bacteriophage when gene wza was deleted. Conclusion: Deletion of the wza gene impairs bacterial capsule formation ability and can affect bacterial sensitivity to bacteriophage phiW14.


Subject(s)
Bacterial Capsules , Bacteriophages , Gene Deletion , Klebsiella pneumoniae , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/virology , Bacteriophages/genetics , Bacterial Capsules/genetics , Bacterial Proteins/genetics
7.
J Infect Dis ; 230(1): e189-e198, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052729

ABSTRACT

BACKGROUND: Streptococcus pneumoniae serotype 3 remains a problem globally. Malawi introduced 13-valent pneumococcal conjugate vaccine (PCV13) in 2011, but there has been no direct protection against serotype 3 carriage. We explored whether vaccine escape by serotype 3 is due to clonal expansion of a lineage with a competitive advantage. METHODS: The distribution of serotype 3 Global Pneumococcal Sequence Clusters (GPSCs) and sequence types (STs) globally was assessed using sequences from the Global Pneumococcal Sequencing Project. Whole-genome sequences of 135 serotype 3 carriage isolates from Blantyre, Malawi (2015-2019) were analyzed. Comparative analysis of the capsule locus, entire genomes, antimicrobial resistance, and phylogenetic reconstructions were undertaken. Opsonophagocytosis was evaluated using serum samples from vaccinated adults and children. RESULTS: Serotype 3 GPSC10-ST700 isolates were most prominent in Malawi. Compared with the prototypical serotype 3 capsular polysaccharide locus sequence, 6 genes are absent, with retention of capsule polysaccharide biosynthesis. This lineage is characterized by increased antimicrobial resistance and lower susceptibility to opsonophagocytic killing. CONCLUSIONS: A serotype 3 variant in Malawi has genotypic and phenotypic characteristics that could enhance vaccine escape and clonal expansion after post-PCV13 introduction. Genomic surveillance among high-burden populations is essential to improve the effectiveness of next-generation pneumococcal vaccines.


Subject(s)
Bacterial Capsules , Phylogeny , Pneumococcal Infections , Pneumococcal Vaccines , Serogroup , Streptococcus pneumoniae , Humans , Pneumococcal Vaccines/immunology , Pneumococcal Vaccines/administration & dosage , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/classification , Pneumococcal Infections/prevention & control , Pneumococcal Infections/microbiology , Pneumococcal Infections/immunology , Bacterial Capsules/immunology , Bacterial Capsules/genetics , Malawi , Adult , Whole Genome Sequencing , Child, Preschool , Child , Vaccines, Conjugate/immunology , Male , Genome, Bacterial , Female , Young Adult , Infant , Genotype , Carrier State/microbiology
8.
J Infect Dis ; 230(1): 209-220, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052750

ABSTRACT

BACKGROUND: Klebsiella pneumoniae carbapenemase-producing K pneumoniae (KPC-Kp) bloodstream infections are associated with high mortality. We studied clinical bloodstream KPC-Kp isolates to investigate mechanisms of resistance to complement, a key host defense against bloodstream infection. METHODS: We tested growth of KPC-Kp isolates in human serum. In serial isolates from a single patient, we performed whole genome sequencing and tested for complement resistance and binding by mixing study, direct enzyme-linked immunosorbent assay, flow cytometry, and electron microscopy. We utilized an isogenic deletion mutant in phagocytosis assays and an acute lung infection model. RESULTS: We found serum resistance in 16 of 59 (27%) KPC-Kp clinical bloodstream isolates. In 5 genetically related bloodstream isolates from a single patient, we noted a loss-of-function mutation in the capsule biosynthesis gene, wcaJ. Disruption of wcaJ was associated with decreased polysaccharide capsule, resistance to complement-mediated killing, and surprisingly, increased binding of complement proteins. Furthermore, an isogenic wcaJ deletion mutant exhibited increased opsonophagocytosis in vitro and impaired in vivo control in the lung after airspace macrophage depletion in mice. CONCLUSIONS: Loss of function in wcaJ led to increased complement resistance, complement binding, and opsonophagocytosis, which may promote KPC-Kp persistence by enabling coexistence of increased bloodstream fitness and reduced tissue virulence.


Subject(s)
Bacterial Capsules , Complement System Proteins , Klebsiella Infections , Klebsiella pneumoniae , Phagocytosis , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/immunology , Humans , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Animals , Bacterial Capsules/immunology , Bacterial Capsules/genetics , Bacterial Capsules/metabolism , Mice , Complement System Proteins/immunology , Mutation , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Whole Genome Sequencing , Reinfection/microbiology , Reinfection/immunology , Bacteremia/microbiology , Bacteremia/immunology , Female
9.
Microb Genom ; 10(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38896467

ABSTRACT

Since the introduction of the 13-valent pneumococcal conjugate vaccine (PCV13) in Malawi in 2011, there has been persistent carriage of vaccine serotype (VT) Streptococcus pneumoniae, despite high vaccine coverage. To determine if there has been a genetic change within the VT capsule polysaccharide (cps) loci since the vaccine's introduction, we compared 1022 whole-genome-sequenced VT isolates from 1998 to 2019. We identified the clonal expansion of a multidrug-resistant, penicillin non-susceptible serotype 23F GPSC14-ST2059 lineage, a serotype 14 GPSC9-ST782 lineage and a novel serotype 14 sequence type GPSC9-ST18728 lineage. Serotype 23F GPSC14-ST2059 had an I253T mutation within the capsule oligosaccharide repeat unit polymerase Wzy protein, which is predicted in silico to alter the protein pocket cavity. Moreover, serotype 23F GPSC14-ST2059 had SNPs in the DNA binding sites for the cps transcriptional repressors CspR and SpxR. Serotype 14 GPSC9-ST782 harbours a non-truncated version of the large repetitive protein (Lrp), containing a Cna protein B-type domain which is also present in proteins associated with infection and colonisation. These emergent lineages also harboured genes associated with antibiotic resistance, and the promotion of colonisation and infection which were absent in other lineages of the same serotype. Together these data suggest that in addition to serotype replacement, modifications of the capsule locus associated with changes in virulence factor expression and antibiotic resistance may promote vaccine escape. In summary, the study highlights that the persistence of vaccine serotype carriage despite high vaccine coverage in Malawi may be partly caused by expansion of VT lineages post-PCV13 rollout.


Subject(s)
Bacterial Capsules , Pneumococcal Infections , Pneumococcal Vaccines , Serogroup , Streptococcus pneumoniae , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/pathogenicity , Pneumococcal Vaccines/immunology , Humans , Malawi , Bacterial Capsules/genetics , Pneumococcal Infections/microbiology , Pneumococcal Infections/prevention & control , Vaccines, Conjugate , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/immunology , Virulence/genetics , Genotype , Whole Genome Sequencing , Bacterial Proteins/genetics , Virulence Factors/genetics , Child, Preschool , Polymorphism, Single Nucleotide , Infant , Male
10.
Nat Commun ; 15(1): 5258, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898034

ABSTRACT

Many bacterial pathogens, including the human exclusive pathogen Salmonella Typhi, express capsular polysaccharides as a crucial virulence factor. Here, through S. Typhi whole genome sequence analyses and functional studies, we found a list of single point mutations that make S. Typhi hypervirulent. We discovered a single point mutation in the Vi biosynthesis enzymes that control Vi polymerization or acetylation is enough to result in different capsule variants of S. Typhi. All variant strains are pathogenic, but the hyper Vi capsule variants are particularly hypervirulent, as demonstrated by the high morbidity and mortality rates observed in infected mice. The hypo Vi capsule variants have primarily been identified in Africa, whereas the hyper Vi capsule variants are distributed worldwide. Collectively, these studies increase awareness about the existence of different capsule variants of S. Typhi, establish a solid foundation for numerous future studies on S. Typhi capsule variants, and offer valuable insights into strategies to combat capsulated bacteria.


Subject(s)
Bacterial Capsules , Mutation, Missense , Polysaccharides, Bacterial , Salmonella typhi , Typhoid Fever , Salmonella typhi/genetics , Salmonella typhi/pathogenicity , Animals , Mice , Virulence/genetics , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/biosynthesis , Polysaccharides, Bacterial/metabolism , Bacterial Capsules/genetics , Bacterial Capsules/metabolism , Typhoid Fever/microbiology , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Female , Whole Genome Sequencing
11.
mBio ; 15(7): e0077124, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38819157

ABSTRACT

The hyaluronic acid capsule is crucial in protecting group A Streptococcus (GAS) against phagocytic killing. However, there have been reported outbreaks caused by capsule-deficient GAS strains, and the mechanisms underlying their evasion of immune clearance remain unclear. This study demonstrated that the capsule-deficient mutant [Cap(-)] of the emm1 strain increased survival within phagocytic cells compared to the wild-type strain [Cap(+)]. Although both Cap(+) and Cap(-) strains exhibited similar abilities to disrupt the phagosome, only the Cap(+) strain was colocalized with lysosomes and acidified compartments in phagocytic cells, indicating its susceptibility to autophagosome elimination. In contrast, the Cap(-) mutant evaded the recognition of galectin-8 and ubiquitin, impairing selective autophagy-mediated elimination. These findings suggest that a deficiency in the capsule could impair the intracellular elimination of GAS in macrophages, revealing previously unknown aspects of the host's recognition of the GAS capsule in macrophages. IMPORTANCE: Group A Streptococcus (GAS) is a Gram-positive bacterium that causes diseases ranging from mild pharyngitis to severe necrotizing fasciitis. Phagocytic cells serve as the primary defense against bacterial infections, exhibiting remarkable efficiency in eliminating intracellular pathogens. The hyaluronic acid capsule is a critical virulence factor that contributes to the resistance of phagocytosis in GAS. Nevertheless, the outbreaks caused by GAS strains that lack the hyaluronic acid capsule have been reported, and the selective advantage of capsule-deficient strains during infection is not fully understood. This study showed that the autophagic adaptor proteins recognize the capsulated GAS strain but not the capsule-deficient mutant, indicating that the hyaluronic acid capsule could be the autophagic target in macrophages. These findings imply that the hyaluronic acid capsule of GAS actually enhances its elimination within phagocytic cells, subverting the understanding of the capsule in GAS pathogenesis.


Subject(s)
Autophagy , Bacterial Capsules , Macrophages , Streptococcus pyogenes , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/physiology , Macrophages/microbiology , Macrophages/immunology , Bacterial Capsules/metabolism , Bacterial Capsules/genetics , Humans , Immune Evasion , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Phagocytosis , Mice , Hyaluronic Acid/metabolism , Animals
12.
Microbiol Res ; 285: 127783, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38795407

ABSTRACT

The regulator of capsule synthesis (Rcs) system, an atypical two-component system prevalent in numerous gram-negative bacteria, serves as a sophisticated regulatory phosphorylation cascade mechanism. It plays a pivotal role in perceiving environmental stress and regulating the expression of downstream genes to ensure host survival. During the signaling transduction process, various proteins participate in phosphorylation to further modulate signal inputs and outputs. Although the structure of core proteins related to the Rcs system has been partially well-defined, and two models have been proposed to elucidate the intricate molecular mechanisms underlying signal sensing, a systematic characterization of the signal transduction process of the Rcs system remains challenging. Furthermore, exploring its corresponding regulator outputs is also unremitting. This review aimed to shed light on the regulation of bacterial virulence by the Rcs system. Moreover, with the assistance of the Rcs system, biosynthesis technology has developed high-value target production. Additionally, via this review, we propose designing chimeric Rcs biosensor systems to expand their application as synthesis tools. Finally, unsolved challenges are highlighted to provide the basic direction for future development of the Rcs system.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Bacterial , Signal Transduction , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Phosphorylation , Virulence , Bacterial Capsules/metabolism , Bacterial Capsules/genetics , Biosensing Techniques
13.
Microbiol Res ; 285: 127775, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38788350

ABSTRACT

Vibrio alginolyticus is one of the most common opportunistic pathogens in marine animals and humans. In this study, A transposon mutation library of the V. alginolyticus E110 was used to identify motility-related genes, and we found three flagellar and one capsular polysaccharide (CPS) synthesis-related genes were linked to swarming motility. Then, gene deletion and complementation further confirmed that CPS synthesis-related gene ugd is involved in the swarming motility of V. alginolyticus. Phenotype assays showed that the Δugd mutant reduced CPS production, decreased biofilm formation, impaired swimming ability, and increased cytotoxicity compared to the wild-type strain. Transcriptome analysis showed that 655 genes (15%) were upregulated and 914 genes (21%) were downregulated in the Δugd strain. KEGG pathway and heatmap analysis revealed that genes involved in two-component systems (TCSs), chemotaxis, and flagella assembly pathways were downregulated in the Δugd mutant. On the other hand, genes involved in pathways of human diseases, biosynthesis ABC transporters, and metabolism were upregulated in the Δugd mutant. The RT-qPCR further validated that ugd-regulated genes are associated with motility, biofilm formation, virulence, and TCSs. These findings imply that ugd may be an important player in the control of some physiological processes in V. alginolyticus, highlighting its potential as a target for future research and potential therapeutic interventions.


Subject(s)
Bacterial Capsules , Bacterial Proteins , Biofilms , Flagella , Gene Expression Regulation, Bacterial , Vibrio alginolyticus , Vibrio alginolyticus/genetics , Vibrio alginolyticus/physiology , Vibrio alginolyticus/metabolism , Biofilms/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Flagella/genetics , Flagella/metabolism , Flagella/physiology , Bacterial Capsules/metabolism , Bacterial Capsules/genetics , Polysaccharides, Bacterial/biosynthesis , Polysaccharides, Bacterial/metabolism , Polysaccharides, Bacterial/genetics , Virulence , Animals , Gene Expression Profiling , Gene Deletion , Humans , Vibrio Infections/microbiology
14.
Nucleic Acids Res ; 52(15): 9119-9138, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-38804271

ABSTRACT

Hypervirulent Klebsiella pneumoniae (hvKp) can infect healthy individuals, in contrast to classical strains that commonly cause nosocomial infections. The recent convergence of hypervirulence with carbapenem-resistance in K. pneumoniae can potentially create 'superbugs' that are challenging to treat. Understanding virulence regulation of hvKp is thus critical. Accumulating evidence suggest that posttranscriptional regulation by small RNAs (sRNAs) plays a role in bacterial virulence, but it has hardly been studied in K. pneumoniae. We applied RIL-seq to a prototypical clinical isolate of hvKp to unravel the Hfq-dependent RNA-RNA interaction (RRI) network. The RRI network is dominated by sRNAs, including predicted novel sRNAs, three of which we validated experimentally. We constructed a stringent subnetwork composed of RRIs that involve at least one hvKp virulence-associated gene and identified the capsule gene loci as a hub target where multiple sRNAs interact. We found that the sRNA OmrB suppressed both capsule production and hypermucoviscosity when overexpressed. Furthermore, OmrB base-pairs within kvrA coding region and partially suppresses translation of the capsule regulator KvrA. This agrees with current understanding of capsule as a major virulence and fitness factor. It emphasizes the intricate regulatory control of bacterial phenotypes by sRNAs, particularly of genes critical to bacterial physiology and virulence.


Subject(s)
Bacterial Capsules , Gene Expression Regulation, Bacterial , Klebsiella pneumoniae , RNA, Bacterial , RNA, Small Untranslated , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/metabolism , Virulence/genetics , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Bacterial Capsules/genetics , Bacterial Capsules/metabolism , Klebsiella Infections/microbiology , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
15.
Viruses ; 16(5)2024 05 13.
Article in English | MEDLINE | ID: mdl-38793652

ABSTRACT

The genus Acinetobacter comprises both environmental and clinically relevant species associated with hospital-acquired infections. Among them, Acinetobacter baumannii is a critical priority bacterial pathogen, for which the research and development of new strategies for antimicrobial treatment are urgently needed. Acinetobacter spp. produce a variety of structurally diverse capsular polysaccharides (CPSs), which surround the bacterial cells with a thick protective layer. These surface structures are primary receptors for capsule-specific bacteriophages, that is, phages carrying tailspikes with CPS-depolymerizing/modifying activities. Phage tailspike proteins (TSPs) exhibit hydrolase, lyase, or esterase activities toward the corresponding CPSs of a certain structure. In this study, the data on all lytic capsule-specific phages infecting Acinetobacter spp. with genomes deposited in the NCBI GenBank database by January 2024 were summarized. Among the 149 identified TSPs encoded in the genomes of 143 phages, the capsular specificity (K specificity) of 46 proteins has been experimentally determined or predicted previously. The specificity of 63 TSPs toward CPSs, produced by various Acinetobacter K types, was predicted in this study using a bioinformatic analysis. A comprehensive phylogenetic analysis confirmed the prediction and revealed the possibility of the genetic exchange of gene regions corresponding to the CPS-recognizing/degrading parts of different TSPs between morphologically and taxonomically distant groups of capsule-specific Acinetobacter phages.


Subject(s)
Acinetobacter , Bacterial Capsules , Bacteriophages , Genome, Viral , Phylogeny , Bacteriophages/genetics , Bacteriophages/enzymology , Bacteriophages/classification , Acinetobacter/virology , Acinetobacter/genetics , Acinetobacter/enzymology , Bacterial Capsules/metabolism , Bacterial Capsules/genetics , Viral Tail Proteins/genetics , Viral Tail Proteins/metabolism , Polysaccharides/metabolism , Polysaccharides, Bacterial/metabolism , Polysaccharides, Bacterial/genetics , Acinetobacter baumannii/virology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/enzymology , Glycoside Hydrolases
16.
Front Cell Infect Microbiol ; 14: 1373052, 2024.
Article in English | MEDLINE | ID: mdl-38808067

ABSTRACT

Among the Acinetobacter genus, Acinetobacter pittii stands out as an important opportunistic infection causative agent commonly found in hospital settings, which poses a serious threat to human health. Recently, the high prevalence of carbapenem-resistant A. pittii isolates has created significant therapeutic challenges for clinicians. Bacteriophages and their derived enzymes are promising therapeutic alternatives or adjuncts to antibiotics effective against multidrug-resistant bacterial infections. However, studies investigating the depolymerases specific to A. pittii strains are scarce. In this study, we identified and characterized a capsule depolymerase, Dpo27, encoded by the bacteriophage IME-Ap7, which targets A. pittii. A total of 23 clinical isolates of Acinetobacter spp. were identified as A. pittii (21.91%, 23/105), and seven A. pittii strains with various K locus (KL) types (KL14, KL32, KL38, KL111, KL163, KL207, and KL220) were used as host bacteria for phage screening. The lytic phage IME-Ap7 was isolated using A. pittii 7 (KL220) as an indicator bacterium and was observed for depolymerase activity. A putative tail fiber gene encoding a polysaccharide-degrading enzyme (Dpo27) was identified and expressed. The results of the modified single-spot assay showed that both A. pittii 7 and 1492 were sensitive to Dpo27, which was assigned the KL220 type. After incubation with Dpo27, A. pittii strain was susceptible to killing by human serum; moreover, the protein displayed no hemolytic activity against erythrocytes. Furthermore, the protein exhibited sustained activity across a wide pH range (5.0-10.0) and at temperatures between 20 and 50°C. In summary, the identified capsule depolymerase Dpo27 holds promise as an alternative treatment for combating KL220-type A. pittii infections.


Subject(s)
Acinetobacter Infections , Acinetobacter , Bacteriophages , Glycoside Hydrolases , Bacteriophages/genetics , Bacteriophages/enzymology , Bacteriophages/isolation & purification , Humans , Acinetobacter/enzymology , Acinetobacter/genetics , Acinetobacter/virology , Acinetobacter/drug effects , Acinetobacter Infections/microbiology , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Bacterial Capsules/metabolism , Bacterial Capsules/genetics
17.
J Appl Microbiol ; 135(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38688866

ABSTRACT

AIMS: Understanding bacterial phage resistance mechanisms has implications for developing phage-based therapies. This study aimed to explore the development of phage resistance in Escherichia coli K1 isolates' to K1-ULINTec4, a K1-dependent bacteriophage. METHODS AND RESULTS: Resistant colonies were isolated from two different strains (APEC 45 and C5), both previously exposed to K1-ULINTec4. Genome analysis and several parameters were assessed, including growth capacity, phage adsorption, phenotypic impact at capsular level, biofilm production, and virulence in the in vivo Galleria mellonella larvae model. One out of the six resistant isolates exhibited a significantly slower growth rate, suggesting the presence of a resistance mechanism altering its fitness. Comparative genomic analysis revealed insertion sequences in the region 2 of the kps gene cluster involved in the capsule biosynthesis. In addition, an immunoassay targeting the K1 capsule showed a very low positive reaction compared to the control. Nevertheless, microscopic images of resistant strains revealed the presence of capsules with a clustered organization of bacterial cells and biofilm assessment showed an increased biofilm production compared to the sensitive strains. In the G. mellonella model, larvae infected with phage-resistant isolates showed better survival rates than larvae infected with phage-sensitive strains. CONCLUSIONS: A phage resistance mechanism was identified at the genomic level and had a negative impact on the K1 capsule production. The resistant isolates showed an increased biofilm production and a decreased virulence in vivo.


Subject(s)
Bacterial Capsules , Biofilms , Escherichia coli , Animals , Bacterial Capsules/genetics , Bacteriophages/genetics , Bacteriophages/physiology , Biofilms/growth & development , Coliphages/genetics , Coliphages/physiology , Escherichia coli/virology , Escherichia coli/genetics , Escherichia coli Infections/microbiology , Larva/microbiology , Larva/virology , Virulence/genetics , Humans , Moths/microbiology
18.
mBio ; 15(3): e0351923, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38349137

ABSTRACT

K2-capsular Klebsiella pneumoniae is a hypervirulent pathogen that causes fatal infections. Here, we describe a phage tailspike protein, named K2-2, that specifically depolymerizes the K2 capsular polysaccharide (CPS) of K. pneumoniae into tetrasaccharide repeating units. Nearly half of the products contained O-acetylation, which was thought crucial to the immunogenicity of CPS. The product-bound structures of this trimeric enzyme revealed intersubunit carbohydrate-binding grooves, each accommodating three tetrasaccharide units of K2 CPS. The catalytic residues and the key interactions responsible for K2 CPS recognition were identified and verified by site-directed mutagenesis. Further biophysical and functional characterization, along with the structure of a tetrameric form of K2-2, demonstrated that the formation of intersubunit catalytic center does not require trimerization, which could be nearly completely disrupted by a single-residue mutation in the C-terminal domain. Our findings regarding the assembly and catalysis of K2-2 provide cues for the development of glycoconjugate vaccines against K. pneumoniae infection. IMPORTANCE: Generating fragments of capsular polysaccharides from pathogenic bacteria with crucial antigenic determinants for vaccine development continues to pose challenges. The significance of the C-terminal region of phage tailspike protein (TSP) in relation to its folding and trimer formation remains largely unexplored. The polysaccharide depolymerase described here demonstrates the ability to depolymerize the K2 CPS of K. pneumoniae into tetrasaccharide fragments while retaining the vital O-acetylation modification crucial for immunogenicity. By carefully characterizing the enzyme, elucidating its three-dimensional structures, conducting site-directed mutagenesis, and assessing the antimicrobial efficacy of the mutant enzymes against K2 K. pneumoniae, we offer valuable insights into the mechanism by which this enzyme recognizes and depolymerizes the K2 CPS. Our findings, particularly the discovery that trimer formation is not required for depolymerizing activity, challenge the current understanding of trimer-dependent TSP activity and highlight the catalytic mechanism of the TSP with an intersubunit catalytic center.


Subject(s)
Bacteriophages , Klebsiella Infections , Humans , Bacteriophages/genetics , Klebsiella pneumoniae/genetics , Polysaccharides/metabolism , Oligosaccharides/metabolism , Klebsiella Infections/microbiology , Bacterial Capsules/genetics
19.
Front Cell Infect Microbiol ; 13: 1279119, 2023.
Article in English | MEDLINE | ID: mdl-38094742

ABSTRACT

Background: The polysaccharide capsule of Streptococcus pneumoniae plays a major role in virulence, adherence to epithelial cells, and overall survival of the bacterium in the human host. Galactose, mannose, and N-acetylglucosamine (GlcNAc) are likely to be relevant for metabolization in the nasopharynx, while glucose is the primary carbon source in the blood. In this study, we aim to further the understanding of the influence of carbon sources on pneumococcal growth, capsule biosynthesis, and subsequent adherence potential. Methods: We tested the growth behavior of clinical wild-type and capsule knockout S. pneumoniae strains, using galactose, GlcNAc, mannose, and glucose as carbon source for growth. We measured capsule thickness and quantified capsule precursors by fluorescein isothiocyanate (FITC)-dextran exclusion assays and 31P-nuclear magnetic resonance measurements, respectively. We also performed epithelial adherence assays using Detroit 562 cells and performed a transcriptome analysis (RNA sequencing). Results: We observed a reduced growth in galactose, mannose, and GlcNAc compared to growth in glucose and found capsular size reductions in mannose and GlcNAc compared to galactose and glucose. Additionally, capsular precursor measurements of uridine diphosphate-(UDP)-glucose and UDP-galactose showed less accumulation of precursors in GlcNAc or mannose than in glucose and galactose, indicating a possible link with the received capsular thickness measurements. Epithelial adherence assays showed an increase in adherence potential for a pneumococcal strain, when grown in mannose compared to glucose. Finally, transcriptome analysis of four clinical isolates revealed not only strain specific but also common carbon source-specific gene expression. Conclusion: Our findings may indicate a careful adaption of the lifestyle of S. pneumoniae according to the monosaccharides encountered in the respective human niche.


Subject(s)
Galactose , Streptococcus pneumoniae , Humans , Streptococcus pneumoniae/metabolism , Carbon/metabolism , Mannose , Glucose/metabolism , Uridine Diphosphate/metabolism , Bacterial Capsules/genetics
20.
PLoS Pathog ; 19(8): e1011509, 2023 08.
Article in English | MEDLINE | ID: mdl-37540710

ABSTRACT

Among the many oral streptococci, Streptococcus pneumoniae (Spn) stands out for the capacity of encapsulated strains to cause invasive infection. Spread beyond upper airways, however, is a biological dead end for the organism, raising the question of the benefits of expending energy to coat its surface in a thick layer of capsular polysaccharide (CPS). In this study, we compare mutants of two serotypes expressing different amounts of CPS and test these in murine models of colonization, invasion infection and transmission. Our analysis of the effect of CPS amount shows that Spn expresses a capsule of sufficient thickness to shield its surface from the deposition of complement and binding of antibody to underlying epitopes. While effective shielding is permissive for invasive infection, its primary contribution to the organism appears to be in the dynamics of colonization. A thicker capsule increases bacterial retention in the nasopharynx, the first event in colonization, and also impedes IL-17-dependent clearance during late colonization. Enhanced colonization is associated with increased opportunity for host-to-host transmission. Additionally, we document substantial differences in CPS amount among clinical isolates of three common serotypes. Together, our findings show that CPS amount is highly variable among Spn and could be an independent determinant affecting host interactions.


Subject(s)
Pneumococcal Infections , Streptococcus pneumoniae , Animals , Mice , Streptococcus pneumoniae/metabolism , Streptococcus , Polysaccharides/metabolism , Nasopharynx/microbiology , Nose , Pneumococcal Infections/microbiology , Bacterial Capsules/genetics
SELECTION OF CITATIONS
SEARCH DETAIL