Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.036
Filtrar
1.
Intern Med ; 63(3): 413-417, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37344426

RESUMO

A 71-year-old-man was admitted to our hospital with a cerebral embolism and diagnosed with infective endocarditis (IE) caused by Streptococcus sanguinis. Mitral valve replacement was performed. About one month later, he experienced sudden abdominal pain and shock due to a ruptured infected mesenteric artery pseudoaneurysm. Forty-four days after abdominal surgery, he presented with rapidly progressive glomerulonephritis with anti-glomerular basement membrane antibodies. He was treated with plasma exchange and prednisolone, and his renal function gradually improved. Since postoperative complications often occur within a few years after surgery for IE, careful follow-up is important, even after antimicrobial therapy and valve surgery.


Assuntos
Falso Aneurisma , Endocardite Bacteriana , Endocardite , Glomerulonefrite , Nefrite , Acidente Vascular Cerebral , Masculino , Humanos , Idoso , Streptococcus sanguis , Artéria Mesentérica Superior/diagnóstico por imagem , Falso Aneurisma/complicações , Falso Aneurisma/diagnóstico por imagem , Endocardite Bacteriana/complicações , Endocardite Bacteriana/cirurgia , Endocardite/complicações , Glomerulonefrite/complicações , Acidente Vascular Cerebral/complicações
3.
Cell Chem Biol ; 31(2): 298-311.e6, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-37832551

RESUMO

Natural competence is the principal driver of streptococcal evolution. While acquisition of new traits could facilitate rapid fitness improvement for bacteria, entry into the competent state is a highly orchestrated event, involving an interplay between various pathways. We present a new type of competence-predation coordination mechanism in Streptococcus sanguinis. Unlike other streptococci that mediate competence through the ComABCDE regulon, several key components are missing in the S. sanguinis ComCDE circuitry. We assembled two synthetic biology devices linking competence-stimulating peptide (CSP) cleavage and export with a quantifiable readout to unravel the unique features of the S. sanguinis circuitry. Our results revealed the ComC precursor cleavage pattern and the two host ABC transporters implicated in the export of the S. sanguinis CSP. Moreover, we discovered a ComCDE-dependent bacteriocin locus. Overall, this study presents a mechanism for commensal streptococci to maximize transformation outcome in a fluid environment through extensive circuitry rewiring.


Assuntos
Bacteriocinas , Streptococcus sanguis , Streptococcus sanguis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sinais (Psicologia) , Bacteriocinas/metabolismo , Peptídeos
4.
mBio ; 15(1): e0266723, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38095871

RESUMO

IMPORTANCE: Type 4 filaments (T4F) are nanomachines ubiquitous in prokaryotes, centered on filamentous polymers of type 4 pilins. T4F are exceptionally versatile and widespread virulence factors in bacterial pathogens. The mechanisms of filament assembly and the many functions they facilitate remain poorly understood because of the complexity of T4F machineries. This hinders the development of anti-T4F drugs. The significance of our research lies in characterizing the simplest known T4F-the Com pilus that mediates DNA uptake in competent monoderm bacteria-and showing that four protein components universally conserved in T4F are sufficient for filament assembly. The Com pilus becomes a model for elucidating the mechanisms of T4F assembly.


Assuntos
Fímbrias Bacterianas , Streptococcus sanguis , Streptococcus sanguis/genética , Streptococcus sanguis/metabolismo , Fímbrias Bacterianas/genética , Fímbrias Bacterianas/metabolismo , Bactérias/genética , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , DNA/metabolismo
6.
Int J Mol Sci ; 24(21)2023 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-37958670

RESUMO

Bacterial surface proteins assembled into amyloids contribute to biofilm formation and host immune evasion. Streptococcus sanguinis, a pioneer colonizer of teeth commonly involved in cardiovascular infections, expresses about thirty-three proteins anchored to the cell wall by sortase A. Here, we characterized the production of amyloid in S. sanguinis strains differing in biofilm and immune evasion phenotypes and investigated the role of sortase A in amyloidogenesis. Amyloid was identified in biofilms formed by nine strains, using Congo red (CR) staining and cross-polarized light microscopy. Additionally, EGCG, an amyloid inhibitor, impaired biofilm maturation in a strain-specific fashion. The amounts of amyloid-like components quantified in culture fluids of nine strains using thioflavin T and fluorimetry negatively correlated with bacterial binding to complement-activating proteins (SAP, C1q), C3b deposition and rates of opsonophagocytosis in PMNs, implying amyloid production in immune evasion. The deletion of the sortase A gene (srtA) in strain SK36 compromised amyloid production and sucrose-independent biofilm maturation. The srtA mutant further showed increased susceptibility to C3b deposition and altered interactions with PMNs as well as reduced persistence in human blood. These findings highlight the contribution of amyloids to biofilm formation and host immune evasion in S. sanguinis strains, further indicating the participation of sortase A substrates in amyloidogenesis.


Assuntos
Evasão da Resposta Imune , Streptococcus sanguis , Humanos , Streptococcus sanguis/genética , Streptococcus sanguis/metabolismo , Amiloide/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biofilmes
7.
Appl Environ Microbiol ; 89(10): e0108123, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37768099

RESUMO

Biofilms are complex polymicrobial communities which are often associated with human infections such as the oral disease periodontitis. Studying these complex communities under controlled conditions requires in vitro biofilm model systems that mimic the natural environment as close as possible. This study established a multispecies periodontal model in the drip flow biofilm reactor in order to mimic the continuous flow of nutrients at the air-liquid interface in the oral cavity. The design is engineered to enable real-time characterization. A community of five bacteria, Streptococcus gordonii-GFPmut3*, Streptococcus oralis-GFPmut3*, Streptococcus sanguinis-pVMCherry, Fusobacterium nucleatum, and Porphyromonas gingivalis-SNAP26 is visualized using two distinct fluorescent proteins and the SNAP-tag. The biofilm in the reactor develops into a heterogeneous, spatially uniform, dense, and metabolically active biofilm with relative cell abundances similar to those in a healthy individual. Metabolic activity, structural features, and bacterial composition of the biofilm remain stable from 3 to 6 days. As a proof of concept for our periodontal model, the 3 days developed biofilm is exposed to a prebiotic treatment with L-arginine. Multifaceted effects of L-arginine on the oral biofilm were validated by this model setup. L-arginine showed to inhibit growth and incorporation of the pathogenic species and to reduce biofilm thickness and volume. Additionally, L-arginine is metabolized by Streptococcus gordonii-GFPmut3* and Streptococcus sanguinis-pVMCherry, producing high levels of ornithine and ammonium in the biofilm. In conclusion, our drip flow reactor setup is promising in studying spatiotemporal behavior of a multispecies periodontal community.ImportancePeriodontitis is a multifactorial chronic inflammatory disease in the oral cavity associated with the accumulation of microorganisms in a biofilm. Not the presence of the biofilm as such, but changes in the microbiota (i.e., dysbiosis) drive the development of periodontitis, resulting in the destruction of tooth-supporting tissues. In this respect, novel treatment approaches focus on maintaining the health-associated homeostasis of the resident oral microbiota. To get insight in dynamic biofilm responses, our research presents the establishment of a periodontal biofilm model including Streptococcus gordonii, Streptococcus oralis, Streptococcus sanguinis, Fusobacterium nucleatum, and Porphyromonas gingivalis. The added value of the model setup is the combination of simulating continuously changing natural mouth conditions with spatiotemporal biofilm profiling using non-destructive characterization tools. These applications are limited for periodontal biofilm research and would contribute in understanding treatment mechanisms, short- or long-term exposure effects, the adaptation potential of the biofilm and thus treatment strategies.


Assuntos
Bactérias , Periodontite , Humanos , Streptococcus gordonii/fisiologia , Fusobacterium nucleatum , Streptococcus sanguis , Streptococcus oralis , Biofilmes , Arginina/metabolismo , Porphyromonas gingivalis/fisiologia
8.
J Dent Res ; 102(11): 1231-1240, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37698342

RESUMO

Dental caries is a common disease affecting quality of life globally. In the present study, we found that a bacteriophage lysin LysP53 against Acinetobacter baumannii possesses selective activity on Streptococcus mutans, the main etiological agent of dental caries, even in low pH caries microenvironments, whereas only minor LysP53 activity was detected against Streptococcus sanguinis, Streptococcus oralis, and Streptococcus mitis. Testing activity against S. mutans planktonic cells showed that 4 µM LysP53 could kill more than 84% of S. mutans within 1 min in buffer with optimal pHs ranging from 4.0 to 6.5. Daily application of LysP53 on biofilms formed in BHI medium supplemented or not with sucrose could reduce exopolysaccharides, expression of genes related to acid resistance and adhesion, and the number of live bacteria in the biofilms. LysP53 treatment also showed similar effects as 0.12% chlorhexidine in preventing enamel demineralization due to S. mutans biofilms, as well as effective removal of S. mutans colonization of tooth surfaces in mice without observed toxic effects. Because of its selective activity against main cariogenic bacteria and good activity in low pH caries microenvironments, it is advantageous to use LysP53 as an active agent for preventing caries.


Assuntos
Cárie Dentária , Streptococcus mutans , Camundongos , Animais , Cárie Dentária/prevenção & controle , Cárie Dentária/microbiologia , Qualidade de Vida , Streptococcus sanguis/metabolismo , Antibacterianos/farmacologia , Biofilmes
9.
Virulence ; 14(1): 2239519, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37563831

RESUMO

Streptococcus sanguinis is a ubiquitous commensal species of the oral cavity commonly involved as an opportunistic pathogen in cardiovascular infections. In this study, we investigated the functions of endopeptidase O (PepO) and a C3-degrading protease (CppA) in the systemic virulence of S. sanguinis. Isogenic mutants of pepO and cppA obtained in strain SK36 showed increased susceptibility to C3b deposition and to opsonophagocytosis by human polymorphonuclear neutrophils (PMN). These mutants differ, however, in their profiles of binding to serum amyloid P component (SAP) and C1q, whereas both showed reduced interaction with C4b-binding protein (C4BP) and/or factor H (FH) regulators as compared to SK36. The two mutants showed defects in ex vivo persistence in human blood, serum-mediated invasion of HCAEC endothelial cells, and virulence in a Galleria mellonella infection model. The transcriptional activities of pepO and cppA, assessed by RT-qPCR in nine wild-type strains, further indicated strain-specific profiles of pepO/cppA expression. Moreover, non-conserved amino acid substitutions were detected among the strains, mostly in CppA. Phylogenetic comparisons with homologues of streptococcal species of the oral and oropharyngeal sites suggested that S. sanguinis PepO and CppA have independent ancestralities. Thus, this study showed that PepO and CppA are complement evasion proteins expressed by S. sanguinis in a strain-specific manner, which are required for multiple functions associated with cardiovascular virulence.


Assuntos
Células Endoteliais , Streptococcus sanguis , Humanos , Streptococcus sanguis/genética , Streptococcus sanguis/metabolismo , Virulência , Células Endoteliais/metabolismo , Filogenia , Proteínas do Sistema Complemento , Proteínas de Bactérias/metabolismo
10.
Int J Biol Macromol ; 243: 125183, 2023 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-37276901

RESUMO

Dental plaque is a complex microbial biofilm community of many species and a major cause of oral infections and infectious endocarditis. Plaque development begins when primary colonizers attach to oral tissues and undergo coaggregation. Primary colonizers facilitate cellular attachment and inter-bacterial interactions through sortase-dependent pili (or fimbriae) extending out from their cell surface. Consequently, the sortase enzyme is viewed as a potential drug target for controlling biofilm formation and avoiding infection. Streptococcus sanguinis is a primary colonizing bacterium whose pili consist of three different pilin subunits that are assembled together by the pilus-specific (C-type) SsaSrtC sortase. Here, we report on the crystal structure determination of the recombinant wild-type and active-site mutant forms of SsaSrtC. Interestingly, the SsaSrtC structure exhibits an open-lid conformation, although a conserved DPX motif is lacking in the lid. Based on molecular docking and structural analysis, we identified the substrate-binding residues essential for pilin recognition and pilus assembly. We also demonstrated that while recombinant SsaSrtC is enzymatically active toward the five-residue LPNTG sorting motif peptide of the pilins, this activity is significantly reduced by the presence of zinc. We further showed that rutin and α-crocin are potential candidate inhibitors of the SsaSrtC sortase via structure-based virtual screening and inhibition assays. The structural knowledge gained from our study will provide the means to develop new approaches that target pilus-mediated attachment, thereby preventing oral biofilm growth and infection.


Assuntos
Aminoaciltransferases , Proteínas de Fímbrias , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/química , Proteínas de Fímbrias/metabolismo , Proteínas de Bactérias/química , Streptococcus sanguis/metabolismo , Simulação de Acoplamento Molecular , Aminoaciltransferases/química
11.
ISME J ; 17(9): 1430-1444, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37355741

RESUMO

Membrane vesicles are produced by Gram-negative and Gram-positive bacteria. While membrane vesicles are potent elicitors of eukaryotic cells and involved in cell-cell communication, information is scarce about their general biology in the context of community members and the environment. Streptococcus sanguinis, a Gram-positive oral commensal, is prevalent in the oral cavity and well-characterized for its ability to antagonize oral pathobionts. We have found that production and dissemination of membrane vesicles by S. sanguinis is dependent on environmental and community factors. Co-culture with interacting commensal Corynebacterium durum, as well as with the periodontal pathobiont Filifactor alocis had no effect on S. sanguinis vesicle number and size, whereas the periodontal pathobiont Porphyromonas gingivalis abolished S. sanguinis vesicle production. Using both correlation and differential expression analyses to examine the transcriptomic changes underlying vesicle production, we found that differential expression of genes encoding proteins related to the cytoplasmic membrane and peptidoglycan correlate with the abundance of membrane vesicles. Proteomic characterizations of the vesicle cargo identified a variety of proteins, including those predicted to influence host interactions or host immune responses. Cell culture studies of gingival epithelial cells demonstrated that both crude and highly purified membrane vesicles could induce the expression of IL-8, TNF-α, IL-1ß, and Gro-α within 6 hours of inoculation at levels comparable to whole cells. Our findings suggest that production of membrane vesicles by S. sanguinis is heavily influenced by community and environmental factors and plays an important role in communication with host cells.


Assuntos
Proteômica , Streptococcus sanguis , Streptococcus sanguis/genética , Streptococcus sanguis/metabolismo , Boca/microbiologia , Gengiva/microbiologia , Bactérias Gram-Positivas
12.
Microbiol Spectr ; 11(4): e0132223, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37310225

RESUMO

Radiation caries is one of the most common complications of head and neck radiotherapy. A shift in the oral microbiota is the main factor of radiation caries. A new form of biosafe radiation, heavy ion radiation, is increasingly being applied in clinical treatment due to its superior depth-dose distribution and biological effects. However, how heavy ion radiation directly impacts the oral microbiota and the progress of radiation caries are unknown. Here, unstimulated saliva samples from both healthy and caries volunteers and caries-related bacteria were directly exposed to therapeutic doses of heavy ion radiation to determine the effects of radiation on oral microbiota composition and bacterial cariogenicity. Heavy ion radiation significantly decreased the richness and diversity of oral microbiota from both healthy and caries volunteers, and a higher percentage of Streptococcus was detected in radiation groups. In addition, heavy ion radiation significantly enhanced the cariogenicity of saliva-derived biofilms, including the ratios of the genus Streptococcus and biofilm formation. In the Streptococcus mutans-Streptococcus sanguinis dual-species biofilms, heavy ion radiation increased the ratio of S. mutans. Next, S. mutans was directly exposed to heavy ions, and the radiation significantly upregulated the gtfC and gtfD cariogenic virulence genes to enhance the biofilm formation and exopolysaccharides synthesis of S. mutans. Our study demonstrated, for the first time, that direct exposure to heavy ion radiation can disrupt the oral microbial diversity and balance of dual-species biofilms by increasing the virulence of S. mutans, increasing its cariogenicity, indicating a potential correlation between heavy ions and radiation caries. IMPORTANCE The oral microbiome is crucial to understanding the pathogenesis of radiation caries. Although heavy ion radiation has been used to treat head and neck cancers in some proton therapy centers, its correlation with dental caries, especially its direct effects on the oral microbiome and cariogenic pathogens, has not been reported previously. Here, we showed that the heavy ion radiation directly shifted the oral microbiota from a balanced state to a caries-associated state by increasing the cariogenic virulence of S. mutans. Our study highlighted the direct effect of heavy ion radiation on oral microbiota and the cariogenicity of oral microbes for the first time.


Assuntos
Cárie Dentária , Íons Pesados , Microbiota , Humanos , Streptococcus mutans , Streptococcus , Streptococcus sanguis , Biofilmes
13.
ISME J ; 17(7): 1116-1127, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37169870

RESUMO

During oral biofilm development, interspecies interactions drive species distribution and biofilm architecture. To understand what molecular mechanisms determine these interactions, we used information gained from recent biogeographical investigations demonstrating an association of corynebacteria with streptococci. We previously reported that Streptococcus sanguinis and Corynebacterium durum have a close relationship through the production of membrane vesicle and fatty acids leading to S. sanguinis chain elongation and overall increased fitness supporting their commensal state. Here we present the molecular mechanisms of this interspecies interaction. Coculture experiments for transcriptomic analysis identified several differentially expressed genes in S. sanguinis. Due to its connection to fatty acid synthesis, we focused on the glycerol-operon. We further explored the differentially expressed type IV pili genes due to their connection to motility and biofilm adhesion. Gene inactivation of the glycerol kinase glpK had a profound impact on the ability of S. sanguinis to metabolize C. durum secreted glycerol and impaired chain elongation important for their interaction. Investigations on the effect of type IV pili revealed a reduction of S. sanguinis twitching motility in the presence of C. durum, which was caused by a decrease in type IV pili abundance on the surface of S. sanguinis as determined by SEM. In conclusion, we identified that the ability to metabolize C. durum produced glycerol is crucial for the interaction of C. durum and S. sanguinis. Reduced twitching motility could lead to a closer interaction of both species, supporting niche development in the oral cavity and potentially shaping symbiotic health-associated biofilm communities.


Assuntos
Glicerol , Streptococcus , Glicerol/metabolismo , Streptococcus sanguis/genética , Biofilmes , Simbiose , Streptococcus mutans
14.
Nat Microbiol ; 8(6): 1018-1025, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37142775

RESUMO

Training artificial intelligence (AI) systems to perform autonomous experiments would vastly increase the throughput of microbiology; however, few microbes have large enough datasets for training such a system. In the present study, we introduce BacterAI, an automated science platform that maps microbial metabolism but requires no prior knowledge. BacterAI learns by converting scientific questions into simple games that it plays with laboratory robots. The agent then distils its findings into logical rules that can be interpreted by human scientists. We use BacterAI to learn the amino acid requirements for two oral streptococci: Streptococcus gordonii and Streptococcus sanguinis. We then show how transfer learning can accelerate BacterAI when investigating new environments or larger media with up to 39 ingredients. Scientific gameplay and BacterAI enable the unbiased, autonomous study of organisms for which no training data exist.


Assuntos
Inteligência Artificial , Streptococcus sanguis , Humanos , Streptococcus sanguis/metabolismo , Streptococcus gordonii/metabolismo
15.
Int J Mol Sci ; 24(7)2023 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-37047667

RESUMO

This study probed in vitro the mechanisms of competition/coexistence between Streptococcus sanguinis (known for being correlated with health in the oral cavity) and Streptococcus mutans (responsible for aciduric oral environment and formation of caries) by means of quantitative Raman spectroscopy and imaging. In situ Raman assessments of live bacterial culture/coculture focusing on biofilm exopolysaccharides supported the hypothesis that both species engaged in antagonistic interactions. Experiments of simultaneous colonization always resulted in coexistence, but they also revealed fundamental alterations of the biofilm with respect to their water-insoluble glucan structure. Raman spectra (collected at fixed time but different bacterial ratios) showed clear changes in chemical bonds in glucans, which pointed to an action by Streptococcus sanguinis to discontinue the impermeability of the biofilm constructed by Streptococcus mutans. The concurrent effects of glycosidic bond cleavage in water-insoluble α - 1,3-glucan and oxidation at various sites in glucans' molecular chains supported the hypothesis that secretion of oxygen radicals was the main "chemical weapon" used by Streptococcus sanguinis in coculture.


Assuntos
Cárie Dentária , Streptococcus sanguis , Humanos , Streptococcus mutans , Biofilmes , Boca/microbiologia , Glucanos/farmacologia
16.
Front Cell Infect Microbiol ; 13: 1130506, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36949812

RESUMO

Dental biofilms are highly assembled microbial communities surrounded by an extracellular matrix, which protects the resident microbes. The microbes, including commensal bacteria and opportunistic pathogens, coexist with each other to maintain relative balance under healthy conditions. However, under hostile conditions such as sugar intake and poor oral care, biofilms can generate excessive acids. Prolonged low pH in biofilm increases proportions of acidogenic and aciduric microbes, which breaks the ecological equilibrium and finally causes dental caries. Given the complexity of oral microenvironment, controlling the acidic biofilms using antimicrobials that are activated at low pH could be a desirable approach to control dental caries. Therefore, recent researches have focused on designing novel kinds of pH-activated strategies, including pH-responsive antimicrobial agents and pH-sensitive drug delivery systems. These agents exert antibacterial properties only under low pH conditions, so they are able to disrupt acidic biofilms without breaking the neutral microenvironment and biodiversity in the mouth. The mechanisms of low pH activation are mainly based on protonation and deprotonation reactions, acids labile linkages, and H+-triggered reactive oxygen species production. This review summarized pH-activated antibiofilm strategies to control dental caries, concentrating on their effect, mechanisms of action, and biocompatibility, as well as the limitation of current research and the prospects for future study.


Assuntos
Anti-Infecciosos , Cárie Dentária , Humanos , Cárie Dentária/prevenção & controle , Streptococcus sanguis , Streptococcus mutans/fisiologia , Biofilmes , Anti-Infecciosos/farmacologia , Concentração de Íons de Hidrogênio
17.
Dent Mater ; 39(4): 351-361, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36906504

RESUMO

OBJECTIVES: The objective of this study is to develop stoichiometric models of sugar fermentation and cell biosynthesis for model cariogenic Streptococcus mutans and non-cariogenic Streptococcus sanguinis to better understand and predict metabolic product formation. METHODS: Streptococcus mutans (strain UA159) and Streptococcus sanguinis (strain DSS-10) were grown separately in bioreactors fed brain heart infusion broth supplemented with either sucrose or glucose at 37 °C. Cell mass concentration and fermentation products were measured at different hydraulic residence times (HRT) to determine cell growth yield. RESULTS: Sucrose growth yields were 0.080 ± 0.0078 g cell/g and 0.18 ± 0.031 g cell/g for S. sanguinis and S. mutans, respectively. For glucose, this reversed, with S. sanguinis having a yield of 0.10 ± 0.0080 g cell/g and S. mutans 0.053 ± 0.0064 g cell/g. Stoichiometric equations to predict free acid concentrations were developed for each test case. Results demonstrate that S. sanguinis produces more free acid at a given pH than S. mutans due to lesser cell yield and production of more acetic acid. Greater amounts of free acid were produced at the shortest HRT of 2.5 hr compared to longer HRTs for both microorganisms and substrates. SIGNIFICANCE: The finding that the non-cariogenic S. sanguinis produces greater amounts of free acids than S. mutans strongly suggests that bacterial physiology and environmental factors affecting substrate/metabolite mass transfer play a much greater role in tooth or enamel/dentin demineralization than acidogenesis. These findings enhance the understanding of fermentation production by oral streptococci and provide useful data for comparing studies under different environmental conditions.


Assuntos
Cárie Dentária , Desmineralização do Dente , Humanos , Fermentação , Sacarose/metabolismo , Biofilmes , Streptococcus/fisiologia , Streptococcus mutans/metabolismo , Streptococcus sanguis/metabolismo , Esmalte Dentário , Cárie Dentária/microbiologia
18.
Mol Oral Microbiol ; 38(3): 224-236, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36779415

RESUMO

Streptococcus mutans is considered to be a major causative agent of dental caries. VicRK is a two-component signal transduction system (TCSTS) of S. mutans, which can regulate the virulence of S. mutans, such as biofilm formation, exopolysaccharide production, acid production, and acid resistance. Meanwhile, it can also regulate the production of mutacins (nlmC) through the TCSTS ComDE. In this study, we found that the vicR-overexpressing strain was more likely to aggregate to form cell clusters, leading to the formation of abnormal biofilm; the overexpression of vicR increased the length of the chain of S. mutans. Furthermore, the expression of the mutacins in the vicR overexpression strain was increased under aerobic conditions. Compared with the control strain and the parental strain, the vicR overexpression strain was more competitive against Streptococcus gordonii. But there was no significant difference against Streptococcus sanguinis. In clinical strains, the expression level of vicR was positively correlated with their competitive ability against S. gordonii. Transcriptional profiling revealed 24 significantly upregulated genes in the vicR-overexpressing strain, including nlmA, nlmB, nlmC, and nlmD encoding mutacins. Electrophoretic mobility shift assays and DNase I footprinting assays confirmed that VicR can directly bind to the promoter sequence of nlmD. Taken together, our findings further demonstrate that VicRK, an important TCSTS of S. mutans, is involved in S. mutans cell morphology and biofilm formation. VicRK regulates the production of more mutacins in S. mutans in response to oxygen stimulation. VicR can bind to the promoter sequence of nlmD, thereby directly regulating the production of mutacins NlmD.


Assuntos
Proteínas de Bactérias , Cárie Dentária , Humanos , Proteínas de Bactérias/metabolismo , Streptococcus mutans/metabolismo , Biofilmes , Streptococcus sanguis/metabolismo
19.
J Dent Res ; 102(4): 450-458, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36688378

RESUMO

As oral bacteria grow and persist within biofilms attached to the tooth's surface, they interact with other species to form synergistic or antagonistic exchanges that govern homeostasis for the overall population. One example are the interactions between the cariogenic species Streptococcus mutans and oral commensal streptococci. Previously, we showed that the cell-cell signaling pathways of S. mutans were inhibited during coculture with other oral streptococci species, leading us to posit that the S. mutans transcriptome and behaviors are broadly altered during growth with these species. To test this hypothesis, we performed whole transcriptome sequencing (RNA-seq) on cocultures of S. mutans with either Streptococcus gordonii, Streptococcus sanguinis, or Streptococcus oralis and a quadculture containing all 4 species in comparison to S. mutans grown alone. Our results reveal that in addition to species-dependent changes to the S. mutans transcriptome, a conserved response to oral streptococci in general can be observed. We monitored the behavior of S. mutans by both microscopy imaging of biofilms and in a bacteriocin overlay assay and verified that S. mutans acts similarly with each of these species but noted divergences in phenotypes when cocultured with another cariogenic Streptococcus (Streptococcus sobrinus) or with oral nonstreptococci species. RNA-seq with oral nonstreptococci showed lack of a consistent gene expression profile and overlap of differentially expressed genes found with commensal streptococci. Finally, we investigated the role of upregulated S. mutans genes within our data sets to determine if they provided a fitness benefit during interspecies interactions. Eleven total genes were studied, and we found that a majority impacted the fitness of S. mutans in various assays, highlighted by increased biomass of commensal streptococci in mixed-species biofilms. These results confirm a common, species-independent modification of S. mutans behaviors with oral commensal streptococci that emphasizes the need to further evaluate oral bacteria within multispecies settings.


Assuntos
Microbiota , Streptococcus mutans , Streptococcus mutans/genética , Streptococcus sanguis/fisiologia , Streptococcus gordonii/metabolismo , Simbiose , Biofilmes
20.
Proc Natl Acad Sci U S A ; 120(3): e2216237120, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36626560

RESUMO

Type 4 filaments (T4F)-of which type 4 pili (T4P) are the archetype-are a superfamily of nanomachines nearly ubiquitous in prokaryotes. T4F are polymers of one major pilin, which also contain minor pilins whose roles are often poorly understood. Here, we complete the structure/function analysis of the full set of T4P pilins in the opportunistic bacterial pathogen Streptococcus sanguinis. We determined the structure of the minor pilin PilA, which is unexpectedly similar to one of the subunits of a tip-located complex of four minor pilins, widely conserved in T4F. We found that PilA interacts and dramatically stabilizes the minor pilin PilC. We determined the structure of PilC, showing that it is a modular pilin with a lectin module binding a subset of glycans prevalent in the human glycome, the host of S. sanguinis. Altogether, our findings support a model whereby the minor pilins in S. sanguinis T4P form a tip-located complex promoting adhesion to various host receptors. This has general implications for T4F.


Assuntos
Proteínas de Fímbrias , Streptococcus sanguis , Humanos , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/química , Fímbrias Bacterianas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...