Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.736
Filter
1.
Int J Mol Sci ; 25(15)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39125755

ABSTRACT

The recent increase in Group A Streptococcus (GAS) incidences in several countries across Europe and some areas of the Unites States (U.S.) has raised concerns. To understand GAS diversity and prevalence, we conducted a local genomic surveillance in Eastern North Carolina (ENC) in 2022-2023 with 95 isolates and compared its results to those of the existing national genomic surveillance in the U.S. in 2015-2021 with 13,064 isolates. We observed their epidemiological changes before and during the COVID-19 pandemic and detected a unique sub-lineage in ENC among the most common invasive GAS strain, ST28/emm1. We further discovered a multiple-copy insertion sequence, ISLgar5, in ST399/emm77 and its single-copy variants in some other GAS strains. We discovered ISLgar5 was linked to a Tn5801-like tetM-carrying integrative and conjugative element, and its copy number was associated with an ermT-carrying pRW35-like plasmid. The dynamic insertions of ISLgar5 may play a vital role in genome fitness and adaptation, driving GAS evolution relevant to antimicrobial resistance and potentially GAS virulence.


Subject(s)
Streptococcal Infections , Streptococcus pyogenes , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , North Carolina/epidemiology , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Humans , Genome, Bacterial , COVID-19/epidemiology , COVID-19/virology , Genomics/methods , Phylogeny , DNA Transposable Elements/genetics , SARS-CoV-2/genetics
2.
Front Cell Infect Microbiol ; 14: 1337861, 2024.
Article in English | MEDLINE | ID: mdl-39055978

ABSTRACT

Introduction: It is currently unclear what the role of Group A streptococcus (GAS) virulence factors (VFs) is in contributing to the invasive potential of GAS. This work investigated the evidence for the association of GAS VFs with invasive disease. Methods: We employed a broad search strategy for studies reporting the presence of GAS VFs in invasive and non-invasive GAS disease. Data were independently extracted by two reviewers, quality assessed, and meta-analyzed using Stata®. Results: A total of 32 studies reported on 45 putative virulence factors [invasive (n = 3,236); non-invasive (n = 5,218)], characterized by polymerase chain reaction (PCR) (n = 30) and whole-genome sequencing (WGS) (n = 2). The risk of bias was rated as low and moderate, in 23 and 9 studies, respectively. Meta-,analyses of high-quality studies (n = 23) revealed a significant association of speM [OR, 1.64 (95%CI, 1.06; 2.52)] with invasive infection. Meta-analysis of WGS studies demonstrated a significant association of hasA [OR, 1.91 (95%CI, 1.36; 2.67)] and speG [OR, 2.83 (95%CI, 1.63; 4.92)] with invasive GAS (iGAS). Meta-analysis of PCR studies indicated a significant association of speA [OR, 1.59 (95%CI, 1.10; 2.30)] and speK [OR, 2.95 (95%CI, 1.81; 4.80)] with invasive infection. A significant inverse association was observed between prtf1 [OR, 0.42 (95%CI, 0.20; 0.87)] and invasive infection. Conclusion: This systematic review and genomic meta-analysis provides evidence of a statistically significant association with invasive infection for the hasA gene, while smeZ, ssa, pnga3, sda1, sic, and NaDase show statistically significantly inverse associations with invasive infection. SpeA, speK, and speG are associated with GAS virulence; however, it is unclear if they are markers of invasive infection. This work could possibly aid in developing preventative strategies.


Subject(s)
Streptococcal Infections , Streptococcus pyogenes , Virulence Factors , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Virulence Factors/genetics , Streptococcal Infections/microbiology , Humans , Virulence/genetics , Whole Genome Sequencing , Bacterial Proteins/genetics
4.
mBio ; 15(8): e0084024, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38953375

ABSTRACT

While genome-wide transposon mutagenesis screens have identified numerous essential genes in the significant human pathogen Streptococcus pyogenes (group A Streptococcus or GAS), many of their functions remain elusive. This knowledge gap is attributed in part to the limited molecular toolbox for controlling GAS gene expression and the bacterium's poor genetic transformability. CRISPR interference (CRISPRi), using catalytically inactive GAS Cas9 (dCas9), is a powerful approach to specifically repress gene expression in both bacteria and eukaryotes, but ironically, it has never been harnessed for controlled gene expression in GAS. In this study, we present a highly transformable and fully virulent serotype M1T1 GAS strain and introduce a doxycycline-inducible CRISPRi system for efficient repression of bacterial gene expression. We demonstrate highly efficient, oligo-based single guide RNA cloning directly to GAS, enabling the construction of a gene knockdown strain in just 2 days, in contrast to the several weeks typically required. The system is shown to be titratable and functional both in vitro and in vivo using a murine model of GAS infection. Furthermore, we provide direct in vivo evidence that the expression of the conserved cell division gene ftsZ is essential for GAS virulence, highlighting its promise as a target for emerging FtsZ inhibitors. Finally, we introduce SpyBrowse (https://veeninglab.com/SpyBrowse), a comprehensive and user-friendly online resource for visually inspecting and exploring GAS genetic features. The tools and methodologies described in this work are poised to facilitate fundamental research in GAS, contribute to vaccine development, and aid in the discovery of antibiotic targets. IMPORTANCE: While group A Streptococcus (GAS) remains a predominant cause of bacterial infections worldwide, there are limited genetic tools available to study its basic cell biology. Here, we bridge this gap by creating a highly transformable, fully virulent M1T1 GAS strain. In addition, we established a tight and titratable doxycycline-inducible system and developed CRISPR interference (CRISPRi) for controlled gene expression in GAS. We show that CRISPRi is functional in vivo in a mouse infection model. Additionally, we present SpyBrowse, an intuitive and accessible genome browser (https://veeninglab.com/SpyBrowse). Overall, this work overcomes significant technical challenges of working with GAS and, together with SpyBrowse, represents a valuable resource for researchers in the GAS field.


Subject(s)
CRISPR-Cas Systems , Streptococcal Infections , Streptococcus pyogenes , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Animals , Mice , Streptococcal Infections/microbiology , Virulence/genetics , Gene Expression Regulation, Bacterial , Disease Models, Animal , Female , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
5.
Infect Immun ; 92(7): e0007724, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38869295

ABSTRACT

The interplay between host nutritional immune mechanisms and bacterial nutrient uptake systems has a major impact on the disease outcome. The host immune factor calprotectin (CP) limits the availability of essential transition metals, such as manganese (Mn) and zinc (Zn), to control the growth of invading pathogens. We previously demonstrated that the competition between CP and the human pathogen group A streptococcus (GAS) for Zn impacts GAS pathogenesis. However, the contribution of Mn sequestration by CP in GAS infection control and the role of GAS Mn acquisition systems in overcoming host-imposed Mn limitation remain unknown. Using a combination of in vitro and in vivo studies, we show that GAS-encoded mtsABC is a Mn uptake system that aids bacterial evasion of CP-imposed Mn scarcity and promotes GAS virulence. Mn deficiency caused by either the inactivation of mtsC or CP also impaired the protective function of GAS-encoded Mn-dependent superoxide dismutase. Our ex vivo studies using human saliva show that saliva is a Mn-scant body fluid, and Mn acquisition by MtsABC is critical for GAS survival in human saliva. Finally, animal infection studies using wild-type (WT) and CP-/- mice showed that MtsABC is critical for GAS virulence in WT mice but dispensable in mice lacking CP, indicating the direct interplay between MtsABC and CP in vivo. Together, our studies elucidate the role of the Mn import system in GAS evasion of host-imposed metal sequestration and underscore the translational potential of MtsABC as a therapeutic or prophylactic target.


Subject(s)
Leukocyte L1 Antigen Complex , Manganese , Streptococcal Infections , Streptococcus pyogenes , Manganese/metabolism , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/immunology , Animals , Humans , Mice , Leukocyte L1 Antigen Complex/metabolism , Virulence , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Host-Pathogen Interactions/immunology , Saliva/microbiology , Saliva/immunology , Disease Models, Animal
6.
Infect Immun ; 92(7): e0015224, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38888310

ABSTRACT

The major gram-positive pathogen group A Streptococcus (GAS) is a model organism for studying microbial epidemics as it causes waves of infections. Since 1980, several GAS epidemics have been ascribed to the emergence of clones producing increased amounts of key virulence factors such as streptolysin O (SLO). Herein, we sought to identify mechanisms underlying our recently identified temporal clonal emergence among emm4 GAS, given that emergent strains did not produce augmented levels of virulence factors relative to historic isolates. By creating and analyzing isoallelic strains, we determined that a conserved mutation in a previously undescribed gene encoding a putative carbonic anhydrase was responsible for the defective in vitro growth observed in the emergent strains. We also identified that the emergent strains survived better inside macrophages and killed macrophages at lower rates than the historic strains. Via the creation of isogenic mutant strains, we linked the emergent strain "survival" phenotype to the downregulation of the SLO encoding gene and upregulation of the msrAB operon which encodes proteins involved in defense against extracellular oxidative stress. Our findings are in accord with recent surveillance studies which found a high ratio of mucosal (i.e., pharyngeal) relative to invasive infections among emm4 GAS. Since ever-increasing virulence is unlikely to be evolutionarily advantageous for a microbial pathogen, our data further understanding of the well-described oscillating patterns of virulent GAS infections by demonstrating mechanisms by which emergent strains adapt a "survival" strategy to outcompete previously circulating isolates.


Subject(s)
Bacterial Proteins , Macrophages , Streptococcal Infections , Streptococcus pyogenes , Streptolysins , Virulence Factors , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/mortality , Humans , Macrophages/microbiology , Macrophages/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Streptolysins/genetics , Streptolysins/metabolism , Virulence Factors/genetics , Mutation , Host-Pathogen Interactions/immunology , Virulence/genetics , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Antigens, Bacterial/immunology , Microbial Viability , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Mice , Gene Expression Regulation, Bacterial , Carrier Proteins
7.
Nat Commun ; 15(1): 3916, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729927

ABSTRACT

The UK observed a marked increase in scarlet fever and invasive group A streptococcal infection in 2022 with severe outcomes in children and similar trends worldwide. Here we report lineage M1UK to be the dominant source of invasive infections in this upsurge. Compared with ancestral M1global strains, invasive M1UK strains exhibit reduced genomic diversity and fewer mutations in two-component regulator genes covRS. The emergence of M1UK is dated to 2008. Following a bottleneck coinciding with the COVID-19 pandemic, three emergent M1UK clades underwent rapid nationwide expansion, despite lack of detection in previous years. All M1UK isolates thus-far sequenced globally have a phylogenetic origin in the UK, with dispersal of the new clades in Europe. While waning immunity may promote streptococcal epidemics, the genetic features of M1UK point to a fitness advantage in pathogenicity, and a striking ability to persist through population bottlenecks.


Subject(s)
COVID-19 , Phylogeny , Streptococcal Infections , Streptococcus pyogenes , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/isolation & purification , United Kingdom/epidemiology , Humans , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , COVID-19/epidemiology , Pandemics , Scarlet Fever/epidemiology , Scarlet Fever/microbiology , Mutation , Repressor Proteins/genetics , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Genome, Bacterial , Europe/epidemiology , Bacterial Proteins
8.
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
9.
Infect Immun ; 92(6): e0008324, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38712951

ABSTRACT

Streptococcus pyogenes [group A streptococcus (GAS)] is a human pathogen capable of infecting diverse tissues. To successfully infect these sites, GAS must detect available nutrients and adapt accordingly. The phosphoenolpyruvate transferase system (PTS) mediates carbohydrate uptake and metabolic gene regulation to adapt to the nutritional environment. Regulation by the PTS can occur through phosphorylation of transcriptional regulators at conserved PTS-regulatory domains (PRDs). GAS has several PRD-containing stand-alone regulators with regulons encoding both metabolic genes and virulence factors [PRD-containing virulence regulators (PCVRs)]. One is RofA, which regulates the expression of virulence genes in multiple GAS serotypes. It was hypothesized that RofA is phosphorylated by the PTS in response to carbohydrate levels to coordinate virulence gene expression. In this study, the RofA regulon of M1T1 strain 5448 was determined using RNA sequencing. Two operons were consistently differentially expressed across growth in the absence of RofA; the pilus operon was downregulated, and the capsule operon was upregulated. This correlated with increased capsule production and decreased adherence to keratinocytes. Purified RofA-His was phosphorylated in vitro by PTS proteins EI and HPr, and phosphorylated RofA-FLAG was detected in vivo when GAS was grown in low-glucose C medium. Phosphorylated RofA was not observed when C medium was supplemented 10-fold with glucose. Mutations of select histidine residues within the putative PRDs contributed to the in vivo phosphorylation of RofA, although phosphorylation of RofA was still observed, suggesting other phosphorylation sites exist in the protein. Together, these findings support the hypothesis that RofA is a PCVR that may couple sugar metabolism with virulence regulation.


Subject(s)
Bacterial Proteins , Gene Expression Regulation, Bacterial , Streptococcus pyogenes , Virulence Factors , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/genetics , Streptococcus pyogenes/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Virulence Factors/genetics , Virulence Factors/metabolism , Virulence , Phosphorylation , Humans , Regulon , Operon , Streptococcal Infections/microbiology , Phosphoenolpyruvate Sugar Phosphotransferase System/metabolism , Phosphoenolpyruvate Sugar Phosphotransferase System/genetics , Keratinocytes/microbiology
10.
J Appl Genet ; 65(3): 635-644, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38760644

ABSTRACT

Streptococcus pyogenes (group A Streptococcus, GAS) is a major human pathogen and causes every year over 600 millions upper respiratory tract onfections worldwide. Untreated or repeated infections may lead to post-infectional sequelae such as rheumatic heart disease, a major cause of GAS-mediated mortality. There is no comprehensive, longitudinal analysis of the M type distribution of upper respiratory tract strains isolated in Poland. Single reports describe rather their antibiotic resistance patterns or focus on the invasive isolates. Our goal was to analyse the clonal structure of the upper respiratory tract GAS isolated over multiple years in Poland. Our analysis revealed a clonal structure similar to the ones observed in high-income countries, with M1, M12, M89, M28, and M77 serotypes constituting over 80% of GAS strains. The M77 serotype is a major carrier of erythromycin resistance and is more often correlated with upper respiratory tract infections than other serotypes.


Subject(s)
Respiratory Tract Infections , Streptococcal Infections , Streptococcus pyogenes , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/genetics , Streptococcus pyogenes/isolation & purification , Streptococcus pyogenes/drug effects , Poland/epidemiology , Humans , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/epidemiology , Streptococcal Infections/epidemiology , Streptococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Serogroup , Drug Resistance, Bacterial , Female , Male , Microbial Sensitivity Tests , Erythromycin/therapeutic use , Adult , Child , Child, Preschool
12.
F1000Res ; 12: 209, 2023.
Article in English | MEDLINE | ID: mdl-38778805

ABSTRACT

Background: Invasive group A streptococcus infection (iGAS) is a serious, sometimes life-threatening condition, with high case fatality rates and high morbidity whose incidence is greatly increased in the last years. Despite the increasing importance and frequency of this condition, at the best of our knowledge, no previous reviews have been published focusing on the risk factors for the development of this condition and its early clinical features. This paper reports the study protocol for a scoping review that aims to analyze the early signs and clinical features of invasive group A streptococcus disease in children, to recognize the prodromal stage of the disease. Methods: Comprehensive research combining the terms pediatric and invasive group A streptococcus infection has been performed on PubMed and SCOPUS to identify potential eligible studies. The search strategy for PubMed will be available in this paper. Two reviewers will screen first the abstract and subsequently the full text to identify eligible articles according to the predefined inclusion criteria. Divergences between the reviewers will be resolved by discussion (with a third author if necessary). Two review authors will extract data independently, everyone on a different Excel spreadsheet. Each researcher will be blinded to the decision of the other researcher. When the process will be completed, in case of discordance, any disagreement will be identified and resolved through discussion (with a third author if necessary). Dissemination: The findings of this review will be published in a peer-reviewed journal.


Subject(s)
Streptococcal Infections , Streptococcus pyogenes , Child , Humans , Risk Factors , Streptococcal Infections/microbiology , Streptococcus pyogenes/pathogenicity , Review Literature as Topic
13.
Nature ; 605(7910): 527-531, 2022 05.
Article in English | MEDLINE | ID: mdl-35545676

ABSTRACT

Gasdermins (GSDMs) are a family of pore-forming effectors that permeabilize the cell membrane during the cell death program pyroptosis1. GSDMs are activated by proteolytic removal of autoinhibitory carboxy-terminal domains, typically by caspase regulators1-9. However, no activator is known for one member of this family, GSDMA. Here we show that the major human pathogen group A Streptococcus (GAS) secretes a protease virulence factor, SpeB, that induces GSDMA-dependent pyroptosis. SpeB cleavage of GSDMA releases an active amino-terminal fragment that can insert into membranes to form lytic pores. GSDMA is primarily expressed in the skin10, and keratinocytes infected with SpeB-expressing GAS die of GSDMA-dependent pyroptosis. Mice have three homologues of human GSDMA, and triple-knockout mice are more susceptible to invasive infection by a pandemic hypervirulent M1T1 clone of GAS. These results indicate that GSDMA is critical in the immune defence against invasive skin infections by GAS. Furthermore, they show that GSDMs can act independently of host regulators as direct sensors of exogenous proteases. As SpeB is essential for tissue invasion and survival within skin cells, these results suggest that GSDMA can act akin to a guard protein that directly detects concerning virulence activities of microorganisms that present a severe infectious threat.


Subject(s)
Pyroptosis , Streptococcus pyogenes , Animals , Caspases , Keratinocytes , Mice , Pore Forming Cytotoxic Proteins , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/pathogenicity , Virulence , Virulence Factors
14.
Sci Rep ; 11(1): 19011, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34561464

ABSTRACT

Group A Streptoccocus (GAS) is among the most diverse of all human pathogens, responsible for a range of clinical manifestations, from mild superficial infections such as pharyngitis to serious invasive infections such as necrotising fasciitis and sepsis. The drivers of these different disease phenotypes are not known. The GAS cholesterol-dependent cytolysin, Streptolysin O (SLO), has well established cell and tissue destructive activity. We investigated the role of SLO in determining disease outcome in vivo, by using two different clinical lineages; the recently emerged hypervirulent outbreak emm type 32.2 strains, which result in sepsis, and the emm type 1.0 strains which cause septic arthritis. Using clinically relevant in vivo mouse models of sepsis and a novel septic arthritis model, we found that the amount and activity of SLO was vital in determining the course of infection. The emm type 32.2 strain produced large quantities of highly haemolytic SLO that resulted in rapid development of sepsis. By contrast, the reduced concentration and lower haemolytic activity of emm type 1.0 SLO led to translocation of bacteria from blood to joints. Importantly, sepsis associated strains that were attenuated by deletion or inhibition of SLO, then also translocated to the joint, confirming the key role of SLO in determining infection niche. Our findings demonstrate that SLO is key to in vivo phenotype and disease outcome. Careful consideration should be given to novel therapy or vaccination strategies that target SLO. Whilst neutralising SLO activity may reduce severe invasive disease, it has the potential to promote chronic inflammatory conditions such as septic arthritis.


Subject(s)
Phenotype , Streptococcal Infections/genetics , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Streptolysins/metabolism , Animals , Arthritis, Infectious/microbiology , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Bacterial Translocation , Disease Models, Animal , Fasciitis, Necrotizing/microbiology , Humans , Mice , Molecular Targeted Therapy , Pharyngitis/microbiology , Prognosis , Sepsis/microbiology , Streptococcal Infections/therapy , Streptolysins/physiology
15.
mBio ; 12(4): e0164221, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34253064

ABSTRACT

Adaptation of group A Streptococcus (GAS) to its human host is mediated by two-component systems that transduce external stimuli to regulate bacterial physiology. Among such systems, CsrRS (also known as CovRS) is the most extensively characterized for its role in regulating ∼10% of the GAS genome, including several virulence genes. Here, we show that extracellular magnesium and the human antimicrobial peptide LL-37 have opposing effects on the phosphorylation of the response regulator CsrR by the receptor kinase CsrS. Genetic inactivation of CsrS phosphatase or kinase activity, respectively, had similar but more pronounced effects on CsrR phosphorylation compared to growth in magnesium or LL-37. These changes in CsrR phosphorylation were correlated with the repression or activation of CsrR-regulated genes as assessed by NanoString analysis. Chromatin immunoprecipitation and DNA sequencing (ChIP-seq) revealed CsrR occupancy at CsrRS-regulated promoters and lower-affinity associations at many other locations on the GAS chromosome. Because ChIP-seq did not detect CsrR occupancy at promoters associated with some CsrR-regulated genes, we investigated whether these genes might be controlled indirectly by intermediate regulators whose expression is modulated by CsrR. Transcriptional profiling of mutant strains deficient in the expression of either of two previously uncharacterized transcription regulators in the CsrR regulon indicated that one or both proteins participated in the regulation of 22 of the 42 CsrR-regulated promoters for which no CsrR association was detected by ChIP-seq. Taken together, these results illuminate CsrRS-mediated regulation of GAS gene expression through modulation of CsrR phosphorylation, CsrR association with regulated promoters, and the control of intermediate transcription regulators. IMPORTANCE Group A Streptococcus (GAS) is an important public health threat as a cause of sore throat, skin infections, life-threatening invasive infections, and the postinfectious complications of acute rheumatic fever, a leading cause of acquired heart disease. This work characterizes CsrRS, a GAS system for the detection of environmental signals that enables adaptation of the bacteria for survival in the human throat by regulating the production of products that allow the bacteria to resist clearance by the human immune system. CsrRS consists of two proteins: CsrS, which is on the bacterial surface to detect specific stimuli, and CsrR, which receives signals from CsrS and, in response, represses or activates the expression of genes coding for proteins that enhance bacterial survival. Some of the genes regulated by CsrR encode proteins that are themselves regulators of gene expression, thereby creating a regulatory cascade.


Subject(s)
Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Protein Kinases/genetics , Regulon/genetics , Streptococcus pyogenes/genetics , Adaptation, Physiological/genetics , Antimicrobial Cationic Peptides/pharmacology , Humans , Magnesium/pharmacology , Phosphorylation , Promoter Regions, Genetic , Repressor Proteins/genetics , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Cathelicidins
16.
FEMS Microbiol Lett ; 368(13)2021 07 07.
Article in English | MEDLINE | ID: mdl-34156082

ABSTRACT

Streptococcus pyogenes is a Gram-positive human-specific pathogen that asymptomatically colonizes the human respiratory tract. The factors affecting the colonization to the host is not clearly understood. Adherence of the pathogen to host epithelial cell is the initial step for a successful colonization process. In the host, bacteria live in a polymicrobial community; thus, the signaling mediated between the bacteria plays a significant role in the colonization of the pathogen to the host. Thus, the effect of acyl-homoserine lactone, secreted by Gram-negative bacteria on the adhesion properties of S. pyogenes M3 strain was examined. N-(3-Oxododecanoyl)-L-homoserine lactone (Oxo-C12) increased the cell size as well as hydrophobicity of S. pyogenes. qPCR data revealed that the expression of sagA and hasA was negatively affected by Oxo-C12. Moreover, Oxo-C12 leads to changes in the morphological characteristic of S. pyogenes, further promoting adherence to host epithelia and biofilm formation on abiotic surface. The study demonstrates the role of Oxo-C12 as a factor that can promote virulence in S. pyogenes M3.


Subject(s)
Streptococcal Infections/microbiology , Streptococcus pyogenes/growth & development , Streptococcus pyogenes/pathogenicity , Acyl-Butyrolactones/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Signal Transduction , Species Specificity , Streptococcus pyogenes/classification , Streptococcus pyogenes/metabolism , Virulence
17.
Sci Rep ; 11(1): 12687, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135390

ABSTRACT

Group A Streptococcus (GAS) is a globally significant bacterial pathogen. The GAS genotyping gold standard characterises the nucleotide variation of emm, which encodes a surface-exposed protein that is recombinogenic and under immune-based selection pressure. Within a supervised learning methodology, we tested three random forest (RF) algorithms (Guided, Ordinary, and Regularized) and 53 GAS response regulator (RR) allele types to infer six genomic traits (emm-type, emm-subtype, tissue and country of sample, clinical outcomes, and isolate invasiveness). The Guided, Ordinary, and Regularized RF classifiers inferred the emm-type with accuracies of 96.7%, 95.7%, and 95.2%, using ten, three, and four RR alleles in the feature set, respectively. Notably, we inferred the emm-type with 93.7% accuracy using only mga2 and lrp. We demonstrated a utility for inferring emm-subtype (89.9%), country (88.6%), invasiveness (84.7%), but not clinical (56.9%), or tissue (56.4%), which is consistent with the complexity of GAS pathophysiology. We identified a novel cell wall-spanning domain (SF5), and proposed evolutionary pathways depicting the 'contrariwise' and 'likewise' chimeric deletion-fusion of emm and enn. We identified an intermediate strain, which provides evidence of the time-dependent excision of mga regulon genes. Overall, our workflow advances the understanding of the GAS mga regulon and its plasticity.


Subject(s)
Algorithms , Evolution, Molecular , Genetic Variation , Machine Learning , Regulon , Streptococcus pyogenes/genetics , Alleles , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Carrier Proteins/genetics , Genes, Bacterial , Genome, Bacterial , Humans , Streptococcal Infections/microbiology , Streptococcus pyogenes/classification , Streptococcus pyogenes/pathogenicity
18.
Pediatr Infect Dis J ; 40(7): 612-616, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34097654

ABSTRACT

BACKGROUND: Group A Streptococcus can cause serious and sometimes life-threatening disease in children. The past few years have witnessed a rise in invasive group A Streptococcus infection (iGASi) for unclear reasons. This study attempted to describe the epidemiology, the clinical and demographic characteristics and the outcomes associated with iGASi in hospitalized children in central Israel. METHODS: We retrospectively analyzed the medical records of children <18 years old discharged with a diagnosis of iGASi between January 2012 and December 2019. Clinical, laboratory and microbiologic data, and immunization status were retrieved. The patients were divided into severe and nonsevere groups based on their clinical presentation. The emm type was determined at the national reference center. RESULTS: A total of 167 patients with 206 positive cultures for group A Streptococcus were identified. Hospitalizations for iGASi increased from 701 to 958 per 100,000 admissions between 2012-2015 and 2016-2019, respectively, representing an increase of 37%. The majority of the isolates were from the otolaryngologic system followed by blood, deep soft tissue and respiratory sites. Uncomplicated mastoiditis was the most common diagnosis, followed by bacteremia. Pneumonia was the main diagnosis in the severe group (39.4%). CONCLUSIONS: The admissions because of iGASi in children <18 years old increased during the last 8 years. Surveillance systems and prospective studies should be conducted to expend our understanding of the epidemiology of iGASi in children, better assess the pathogenesis and specific risk factors and monitor changes in emm-type distribution.


Subject(s)
Hospitalization/statistics & numerical data , Streptococcal Infections/blood , Streptococcal Infections/epidemiology , Adolescent , Age Distribution , Anti-Bacterial Agents/therapeutic use , Bacteremia/epidemiology , Child , Child, Preschool , Female , Humans , Incidence , Infant , Israel/epidemiology , Male , Mastoiditis/epidemiology , Mastoiditis/microbiology , Pneumonia, Bacterial/epidemiology , Prospective Studies , Retrospective Studies , Risk Factors , Streptococcal Infections/complications , Streptococcal Infections/drug therapy , Streptococcus pyogenes/pathogenicity , Tertiary Care Centers/statistics & numerical data
20.
Infect Immun ; 89(8): e0018521, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33972369

ABSTRACT

M-type 28 (M28) Streptococcus pyogenes (group A Streptococcus [GAS]) strains are highly associated with life-threatening puerperal infections. Genome sequencing has revealed a large mobile genetic element, RD2, present in most M28 GAS isolates but not found widely in other serotypes. Previous studies have linked RD2 to the ability of M28 GAS to colonize the vaginal tract. A new study by Roshika and colleagues (R. Roshika, I. Jain, J. Medicielo, J. Wächter, J. L. Danger, P. Sumby, Infect Immun 89:e00722-20, 2021, https://doi.org/10.1128/IAI.00722-20) used gain-of-function mutants in three different GAS serotypes to help determine why RD2 appears to have a serotype preference and what that could mean for GAS mucosal colonization and pathogenesis.


Subject(s)
DNA Transposable Elements , Streptococcal Infections/microbiology , Streptococcus pyogenes/classification , Streptococcus pyogenes/genetics , Disease Susceptibility/immunology , Genome, Bacterial , Genomics/methods , Host-Pathogen Interactions/immunology , Humans , Serogroup , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL