1
|
Ellepola K, Shields RC, Kajfasz JK, Zhang H, Lemos JA, Wu H, Wen ZT. MecA in Streptococcus mutans is a multi-functional protein. mSphere 2024:e0030824. [PMID: 39530674 DOI: 10.1128/msphere.00308-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024] Open
Abstract
Our recent studies have shown that deficiency of MecA in Streptococcus mutans significantly affects cell division, growth, and biofilm formation. In this study, an in vitro mixed-species model, proteomics, and affinity pull-down assays were used to further characterize the MecA-mediated regulation in S. mutans. The results showed that compared with the wild type, UA159, the mecA mutant significantly reduced its production of glucans and weakened its ability to facilitate mixed-species biofilm formation. Relative to the wild type, the mecA mutant also displayed unique characteristics, including colony morphology, growth rate, and biofilm formation that did not fully resemble any of the clpP, clpX, clpE, clpCE, and clpC individual or combinational mutants. Deletion of mecA was shown to result in alteration of >337 proteins, including down expression of GtfBC&D and adhesin P1. More than 277 proteins were differentially expressed in response to clpP deletion, including increased expression of GtfB. By cross-referencing the two proteomes, a distinctive set of proteins was found to be altered in the mecA mutant, indicating a ClpP-independent role of MecA in the regulation of S. mutans. When analyzed using affinity pull-down, ClpC, ClpX, ClpE, and CcpA were among the members identified in the MecA-associated complex. Further analysis using a bacterial two-hybrid system confirmed CcpA, ClpX, and ClpE as members of the MecA interactome. These results further suggest that MecA in S. mutans is more than an adapter of the Clp-proteolytic machinery, although the mechanism that underlies the Clp-independent regulation and its impact on S. mutans pathophysiology await further investigation. IMPORTANCE MecA is known as an adaptor protein that works in concerto with ATPase ClpC and protease ClpP in the regulated proteolysis machinery. The results presented here provide further evidence that MecA in S. mutans, a keystone cariogenic bacterium, plays a significant role in its ability to facilitate mixed-species biofilm formation, a trait critical to its cariogenicity. Proteomics analysis, along with affinity pull-down and bacterial two-hybrid system, further confirm that MecA can also regulate S. mutans physiology and biofilm formation through pathways independent of the Clp proteolytic machinery, although how it functions independently of Clp awaits further investigation.
Collapse
Affiliation(s)
- Kassapa Ellepola
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Robert C Shields
- Department of Biological Sciences, Arkansas State University, Jonesboro, Arkansas, USA
| | - Jessica K Kajfasz
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Hua Zhang
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Jose A Lemos
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Hui Wu
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Zezhang T Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
2
|
Treerat P, Rozendal T, de Mattos C, Davis A, Helliwell E, Merritt J, Kreth J. Corynebacterial membrane vesicles disrupt cariogenic interkingdom assemblages. Appl Environ Microbiol 2024:e0088524. [PMID: 39480093 DOI: 10.1128/aem.00885-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 09/26/2024] [Indexed: 11/02/2024] Open
Abstract
Polymicrobial diseases such as periodontal disease and caries pose significant treatment challenges due to their resistance to common approaches like antibiotic therapy. These infections exhibit increased resilience, due to microbial interactions that also disrupt host immune responses. Current research focuses on virulence and disease-promoting interactions, but less is known about interactions that could inhibit or prevent disease development. Normally human-associated microbiomes maintain homeostasis, preventing pathobionts from becoming dominant. In conditions like chronic disseminated candidiasis or severe early childhood caries (s-ECC), an overgrowth of microbes such as Candida albicans disrupts this balance. Typically, C. albicans coexists benignly within the microbial community but can become pathogenic, forming biofilms and interacting with other microbes such as cariogenic Streptococcus mutans. This interaction is particularly significant in s-ECC, where it exacerbates the disease's progression and severity. Here, we present that Corynebacterium durum, itself and through its extracellular membrane vesicles disrupts interkingdom assemblages between C. albicans and S. mutans. Mechanistically the interaction interference occurs at the genetic level with downregulated HWP1 expression, a surface protein specifically induced in the presence of S. mutans promoting the interkingdom interaction. Additionally, we show that C. durum can impede C. albicans systemic virulence in the Galleria mellonella infection model. This suggests that oral corynebacteria may act as a beneficial commensal species, exerting antifungal effects within polymicrobial communities and opening new avenues for managing polymicrobial diseases.IMPORTANCEPolymicrobial diseases such as severe early childhood caries (s-ECC) lack effective treatment options. Prevention, requiring a deeper understanding of ecological processes before the onset of disease symptoms, could be a potential strategy. In this context, we investigated how relatively abundant oral biofilm Corynebacterium species, which are associated with oral health, can interfere with the interkingdom partnership of Streptococcus mutans and Candida albicans. This partnership is a significant driver of tooth decay in s-ECC due to synergistic activities that increase cariogenicity. Our study reveals that oral corynebacteria, through the production of extracellular membrane vesicles, can disrupt the S. mutans and C. albicans partnership by inhibiting fungal hyphae formation. Additionally, the fatty acid cargo within these vesicles exhibits antifungal properties, suggesting that corynebacteria play a role in shaping microbial dynamics within the oral biofilm.
Collapse
Affiliation(s)
- Puthayalai Treerat
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Tanner Rozendal
- Clark Honors College, University of Oregon, Eugene, Oregon, USA
| | - Camilla de Mattos
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Anli Davis
- Lewis & Clark College, Portland, Oregon, USA
| | - Emily Helliwell
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Justin Merritt
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Jens Kreth
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| |
Collapse
|
3
|
Gomberg AF, Grossman AD. It's complicated: relationships between integrative and conjugative elements and their bacterial hosts. Curr Opin Microbiol 2024; 82:102556. [PMID: 39423563 DOI: 10.1016/j.mib.2024.102556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
Integrative and conjugative elements (ICEs) are typically found integrated in a bacterial host chromosome. They can excise, replicate, and transfer from cell to cell. Many contain genes that confer phenotypes to host cells, including antibiotic resistances, specialized metabolisms, phage defense, and symbiosis or pathogenesis determinants. Recent studies revealed that at least three ICEs (ICEclc, Tn916, and TnSmu1) cause growth arrest or death of host cells upon element activation. This review highlights the complex interactions between ICEs and their hosts, including the recent examples of the significant costs to host cells. We contrast two examples of killing, ICEclc and Tn916, in which killing, respectively, benefits or impairs conjugation and emphasize the importance of understanding the impacts of ICE-host relationships on conjugation. ICEs are typically only active in a small fraction of cells in a population, and we discuss how phenotypes normally occurring in a small subset of host cells can be uncovered.
Collapse
Affiliation(s)
- Alexa Fs Gomberg
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Alan D Grossman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.
| |
Collapse
|
4
|
Ma Q, Li J, Yu S, Zhou J, Liu Y, Wang X, Ye D, Wu Y, Gong T, Zhang Q, Wang L, Zou J, Li Y. YkuR functions as a protein deacetylase in Streptococcus mutans. Proc Natl Acad Sci U S A 2024; 121:e2407820121. [PMID: 39356671 PMCID: PMC11474102 DOI: 10.1073/pnas.2407820121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024] Open
Abstract
Protein acetylation is a common and reversible posttranslational modification tightly governed by protein acetyltransferases and deacetylases crucial for various biological processes in both eukaryotes and prokaryotes. Although recent studies have characterized many acetyltransferases in diverse bacterial species, only a few protein deacetylases have been identified in prokaryotes, perhaps in part due to their limited sequence homology. In this study, we identified YkuR, encoded by smu_318, as a unique protein deacetylase in Streptococcus mutans. Through protein acetylome analysis, we demonstrated that the deletion of ykuR significantly upregulated protein acetylation levels, affecting key enzymes in translation processes and metabolic pathways, including starch and sucrose metabolism, glycolysis/gluconeogenesis, and biofilm formation. In particular, YkuR modulated extracellular polysaccharide synthesis and biofilm formation through the direct deacetylation of glucosyltransferases (Gtfs) in the presence of NAD+. Intriguingly, YkuR can be acetylated in a nonenzymatic manner, which then negatively regulated its deacetylase activity, suggesting the presence of a self-regulatory mechanism. Moreover, in vivo studies further demonstrated that the deletion of ykuR attenuated the cariogenicity of S. mutans in the rat caries model, substantiating its involvement in the pathogenesis of dental caries. Therefore, our study revealed a unique regulatory mechanism mediated by YkuR through protein deacetylation that regulates the physiology and pathogenicity of S. mutans.
Collapse
Affiliation(s)
- Qizhao Ma
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Jing Li
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Shuxing Yu
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Jing Zhou
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Yaqi Liu
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Xinyue Wang
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Dingwei Ye
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Yumeng Wu
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Tao Gong
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Qiong Zhang
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Lingyun Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT06510
| | - Jing Zou
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Yuqing Li
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Laboratory of Archaeological Repository, Center for Archaeological Science, Sichuan University, Chengdu610041, China
| |
Collapse
|
5
|
Zhou J, Hu Z, Wang L, Hu Q, Chen Z, Lin T, Zhou R, Cai Y, Wu Z, Zhang Z, Yang Y, Zhang C, Li G, Zeng L, Su K, Li H, Su Q, Zeng G, Cheng B, Wu T. Tumor-colonized Streptococcus mutans metabolically reprograms tumor microenvironment and promotes oral squamous cell carcinoma. MICROBIOME 2024; 12:193. [PMID: 39369210 PMCID: PMC11452938 DOI: 10.1186/s40168-024-01907-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 08/13/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) remains a major death cause in head and neck cancers, but the exact pathogenesis mechanisms of OSCC are largely unclear. RESULTS Saliva derived from OSCC patients but not healthy controls (HCs) significantly promotes OSCC development and progression in rat models, and metabolomic analyses reveal saliva of OSCC patients but not HCs and OSCC tissues but not adjacent non-tumor tissues contain higher levels of kynurenic acid (KYNA). Furthermore, large amounts of Streptococcus mutans (S. mutans) colonize in OSCC tumor tissues, and such intratumoral S. mutans mediates KYNA overproductions via utilizing its protein antigen c (PAc). KYNA shifts the cellular types in the tumor microenvironment (TME) of OSCC and predominantly expedites the expansions of S100a8highS100a9high neutrophils to produce more interleukin 1β (IL-1β), which further expands neutrophils and induces CD8 + T cell exhaustion in TME and therefore promotes OSCC. Also, KYNA compromises the therapeutic effects of programmed cell death ligand 1 (PD-L1) and IL-1β blockades in oral carcinogenesis model. Moreover, KYNA-mediated immunosuppressive program and aryl hydrocarbon receptor (AHR) expression correlate with impaired anti-tumor immunity and poorer survival of OSCC patients. CONCLUSIONS Thus, aberration of oral microbiota and intratumoral colonization of specific oral bacterium such as S. mutans may increase the production of onco-metabolites, exacerbate the oral mucosal carcinogenesis, reprogram a highly immunosuppressive TME, and promote OSCC, highlighting the potential of interfering with oral microbiota and microbial metabolism for OSCC preventions and therapeutics. Video Abstract.
Collapse
Affiliation(s)
- Jiaying Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China
| | - Zixuan Hu
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Province Key Laboratory of Oral Biomedicine, Nanchang, 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, 330006, China
| | - Lei Wang
- BGI Research, Chongqing, 401329, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI Research, Shenzhen, 518083, China
| | - Qinchao Hu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China
| | - Zixu Chen
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tao Lin
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Rui Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China
| | - Yongjie Cai
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhiying Wu
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhiyi Zhang
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yi Yang
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | | | - Guibo Li
- BGI Research, Chongqing, 401329, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI Research, Shenzhen, 518083, China
| | - Lingchan Zeng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
| | - Kai Su
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China
| | - Huan Li
- Department of Intensive Care Unit (ICU), State Key Laboratory of Oncology in South China, CollaborativeInnovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qiao Su
- Animal Experiment Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Gucheng Zeng
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China.
| | - Tong Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China.
| |
Collapse
|
6
|
Chen H, Xu M, Zhang B, Yu S, Weir MD, Melo MAS, Masri RM, Tang Y, Xu HHK, Yang D. Novel strategy of S. mutans gcrR gene over-expression plus antibacterial dimethylaminohexadecyl methacrylate suppresses biofilm acids and reduces dental caries in rats. Dent Mater 2024; 40:e41-e51. [PMID: 38942710 DOI: 10.1016/j.dental.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 06/09/2024] [Indexed: 06/30/2024]
Abstract
OBJECTIVE Streptococcus mutans (S. mutans) is a major contributor to dental caries, with its ability to synthesize extracellular polysaccharides (EPS) and biofilms. The gcrR gene is a regulator of EPS synthesis and biofilm formation. The objectives of this study were to investigate a novel strategy of combining gcrR gene over-expression with dimethylaminohexadecyl methacrylate (DMAHDM), and to determine their in vivo efficacy in reducing caries in rats for the first time. METHODS Two types of S. mutans were tested: Parent S. mutans; and gcrR gene over-expressed S. mutans (gcrR OE S. mutans). Bacterial minimum inhibitory concentration (MIC) and minimal bactericidal concentration (MBC) were measured with DMAHDM and chlorhexidine (CHX). Biofilm biomass, polysaccharide, lactic acid production, live/dead staining, colony-forming units (CFUs), and metabolic activity (MTT) were evaluated. A Sprague-Dawley rat model was used with parent S. mutans and gcrR OE S. mutans colonization to determine caries-inhibition in vivo. RESULTS Drug-susceptibility of gcrR OE S. mutans to DMAHDM or CHX was 2-fold higher than that of parent S. mutans. DMAHDM reduced biofilm CFU by 3-4 logs. Importantly, the combined gcrR OE S. mutans+ DMAHDM dual strategy reduced biofilm CFU by 5 logs. In the rat model, the parent S. mutans group had a higher cariogenicity in dentinal (Dm) and extensive dentinal (Dx) regions. The DMAHDM + gcrR OE group reduced the Dm and Dx caries to only 20 % and 0 %, those of parent S. mutans + PBS control group (p < 0.05). The total caries severity of gcrR OE + DMAHDM group was decreased to 51 % that of parent S. mutans control (p < 0.05). SIGNIFICANCE The strategy of combining S. mutans gcrR over-expression with antibacterial monomer reducing biofilm acids by 97 %, and reduced in vivo total caries in rats by 48 %. The gcrR over-expression + DMAHDM strategy is promising for a wide range of dental applications to inhibit caries and protect tooth structures.
Collapse
Affiliation(s)
- Hong Chen
- Department of Endodontics, the Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 404100, PR China; Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 404100, PR China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 404100, PR China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, 426 Songshi North Road, Yubei Distrinct, Chongqing 401147, PR China
| | - Mengmeng Xu
- Department of Endodontics, the Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 404100, PR China; Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 404100, PR China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 404100, PR China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, 426 Songshi North Road, Yubei Distrinct, Chongqing 401147, PR China
| | - Bin Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Shuang Yu
- Department of Endodontics, the Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 404100, PR China; Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 404100, PR China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 404100, PR China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, 426 Songshi North Road, Yubei Distrinct, Chongqing 401147, PR China
| | - Michael D Weir
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Mary Anne S Melo
- Division of Operative Dentistry, Department of General Dentistry, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Radi M Masri
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Yunhao Tang
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland Dental School, Baltimore, MD 21201, USA
| | - Hockin H K Xu
- Department of Biomaterials and Regenerative Dental Medicine, University of Maryland Dental School, Baltimore, MD 21201, USA.
| | - Deqin Yang
- Department of Endodontics, the Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing 404100, PR China; Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 404100, PR China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 404100, PR China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, 426 Songshi North Road, Yubei Distrinct, Chongqing 401147, PR China.
| |
Collapse
|
7
|
Bao L, Zhu Z, Ismail A, Zhu B, Anandan V, Whiteley M, Kitten T, Xu P. Experimental evolution of gene essentiality in bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.600122. [PMID: 39071448 PMCID: PMC11275930 DOI: 10.1101/2024.07.16.600122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Essential gene products carry out fundamental cellular activities in interaction with other components. However, the lack of essential gene mutants and appropriate methodologies to link essential gene functions with their partners poses significant challenges. Here, we have generated deletion mutants in 32 genes previously identified as essential, with 23 mutants showing extremely slow growth in the SK36 strain of Streptococcus sanguinis. The 23 genes corresponding to these mutants encode components of diverse pathways, are widely conserved among bacteria, and are essential in many other bacterial species. Whole-genome sequencing of 243 independently evolved populations of these mutants has identified >1000 spontaneous suppressor mutations in experimental evolution. Many of these mutations define new gene and pathway relationships, such as F1Fo-ATPase/V1Vo-ATPase/TrkA1-H1 that were demonstrated across multiple Streptococcus species. Patterns of spontaneous mutations occurring in essential gene mutants differed from those found in wildtype. While gene duplications occurred rarely and appeared most often at later stages of evolution, substitutions, deletions, and insertions were prevalent in evolved populations. These essential gene deletion mutants and spontaneous mutations fixed in the mutant populations during evolution establish a foundation for understanding gene essentiality and the interaction of essential genes in networks.
Collapse
Affiliation(s)
- Liang Bao
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Zan Zhu
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Ahmed Ismail
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Bin Zhu
- Massey Cancer Center, Virginia Commonwealth University, Virginia, USA
| | - Vysakh Anandan
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Marvin Whiteley
- School of Biological Sciences, Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Georgia, USA
| | - Todd Kitten
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Ping Xu
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| |
Collapse
|
8
|
Shen L, Sun F, Wang Y, Liu Y, Xin Q, Zhu Z, Zhang H, Xu X, Ding C, Zheng L, Li J. Caries-Prone Primary Teeth: A Hidden Reason and Prophylactic Treatment in the Viewpoint of Materials Science. ACS APPLIED MATERIALS & INTERFACES 2024; 16:41881-41891. [PMID: 39092619 DOI: 10.1021/acsami.4c07388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Dental caries, the most prevalent chronic disease across all age groups, has a high prevalence, particularly among children. However, there is no specific and effective treatment for the prevention of caries in primary teeth (Pr.T.), which stems from a lack of knowledge regarding the basic nature of the tooth surface. Herein, we observed that the adhesion energies of the caries-related bacteria Streptococcus mutans and Streptococcus sanguinis to Pr.T were approximately 10 and 5.5 times higher than those to permanent teeth (Pe.T). A lower degree of mineralization and more hydrophilic characteristics of the Pr.T enamel account for this discrepancy. Accordingly, we proposed that the on-target modification of both hydroxyapatite and organic components on Pr.T by dual modification would render a sufficient hydration layer. This resulted in an approximately 11-time decrease in bacterial adhesion energy after treatment. In contrast, a single hydroxyapatite modification on Pe.T and young permanent teeth (Y.Pe.T) was sufficient to achieve a similar effect. Theoretical simulation further verified the rationality of the approach. Our findings may help understand the reason for Pr.T being caries-prone and provide references for treatment using resin restorations. This strategy offers valuable insights into daily oral hygiene and dental prophylactic treatment in children.
Collapse
Affiliation(s)
- Luxuan Shen
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Fan Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Yu Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yang Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Qiangwei Xin
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Zhongpeng Zhu
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu 215123, China
| | - Hongbo Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Xinyuan Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Chunmei Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Med-X Center for Materials, Sichuan University, Chengdu 610017, China
| |
Collapse
|
9
|
Santiago Narvaez B, Hameer S, Perry JL, Rojas T, Habgood LG. Partial in-vitro dispersal of S. mutans UA159 biofilms by silver-(I)cyanoximate compounds. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001262. [PMID: 39193022 PMCID: PMC11348005 DOI: 10.17912/micropub.biology.001262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024]
Abstract
Silver(I) cyanoximate compounds have antibacterial activity against the oral pathogen Streptococcus mutans, a resident of oral plaque biofilm. As oral biofilm strategies focus on the inhibition of attachment or physical removal of the existing microbes, we were interested in exploring the ability of six different silver(I) cyanoximate compounds to target and disperse a pre-existing biofilm. Here we report that these compounds were only able to partially disperse S. mutans biofilms as the compounds were more effective at inhibiting biofilm formation. None of the six compounds were able to outperform silver nitrate, a commonly used antibacterial in dentistry.
Collapse
Affiliation(s)
| | - Sarah Hameer
- Biology, Rollins College, Winter Park, Florida, United States
| | - Jamie L. Perry
- Biology, Rollins College, Winter Park, Florida, United States
| | - Tiffany Rojas
- Biology, Rollins College, Winter Park, Florida, United States
| | | |
Collapse
|
10
|
Momeni SS, Cao X, Xie B, Rainey K, Childers NK, Wu H. Intraspecies interactions of Streptococcus mutans impact biofilm architecture and virulence determinants in childhood dental caries. mSphere 2024; 9:e0077823. [PMID: 38990043 PMCID: PMC11288028 DOI: 10.1128/msphere.00778-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/11/2024] [Indexed: 07/12/2024] Open
Abstract
Early childhood dental caries (ECC) is the most common chronic disease among children, especially among low socioeconomic populations. Streptococcus mutans is most frequently associated with initiation of ECC. Although many studies report children with multiple S. mutans strains (i.e., genotypes) have greater odds of developing ECC, studies investigating intraspecies interactions in dental caries are lacking. This study investigates the impact of intraspecies interactions on cariogenic and fitness traits of clinical S. mutans isolates using in vitro and in vivo approaches. Association analysis evaluated if presence of multiple S. mutans genotypes within the first year of colonization was associated with caries. Initially, clinical S. mutans isolates from 10 children were evaluated. S. mutans strains (G09 and G18, most prevalent) isolated from one child were used for subsequent analysis. Biofilm analysis was performed for single and mixed cultures to assess cariogenic traits, including biofilm biomass, intra-polysaccharide, pH, and glucan. Confocal laser scanning microscopy (CLSM) and time-lapse imaging were used to evaluate spatial and temporal biofilm dynamics, respectively. A Drosophila model was used to assess colonization in vivo. Results showed the mean biofilm pH was significantly lower in co-cultured biofilms versus monoculture. Doubling of S. mutans biofilms was observed by CLSM and in vivo colonization in Drosophila for co-cultured S. mutans. Individual strains occupied specific domains in co-culture and G09 contributed most to increased co-culture biofilm thickness and colonization in Drosophila. Biofilm formation and acid production displayed distinct signatures in time-lapsed experiments. This study illuminates that intraspecies interactions of S. mutans significantly impacts biofilm acidity, architecture, and colonization.IMPORTANCEThis study sheds light on the complex dynamics of a key contributor to early childhood dental caries (ECC) by exploring intraspecies interactions of different S. mutans strains and their impact on cariogenic traits. Utilizing clinical isolates from children with ECC, the research highlights significant differences in biofilm architecture and acid production in mixed versus single genotype cultures. The findings reveal that co-cultured S. mutans strains exhibit increased cell density and acidity, with individual strains occupying distinct domains. These insights, enhanced by use of time-lapsed confocal laser scanning microscopy and a Drosophila model, offer a deeper understanding of ECC pathogenesis and potential avenues for targeted interventions.
Collapse
Affiliation(s)
- Stephanie S. Momeni
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Xixi Cao
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Baotong Xie
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Katherine Rainey
- Department of Pediatric Dentistry, School of Dentistry, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Noel K. Childers
- Department of Pediatric Dentistry, School of Dentistry, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hui Wu
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
11
|
Treerat P, de Mattos C, Burnside M, Zhang H, Zhu Y, Zou Z, Anderson D, Wu H, Merritt J, Kreth J. Ribosomal-processing cysteine protease homolog modulates Streptococcus mutans glucan production and interkingdom interactions. J Bacteriol 2024; 206:e0010424. [PMID: 38899897 PMCID: PMC11270869 DOI: 10.1128/jb.00104-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Glucan-dependent biofilm formation is a crucial process in the establishment of Streptococcus mutans as a cariogenic oral microbe. The process of glucan formation has been investigated in great detail, with glycosyltransferases GtfB, GtfC, and GtfD shown to be indispensable for the synthesis of glucans from sucrose. Glucan production can be visualized during biofilm formation through fluorescent labeling, and its abundance, as well as the effect of glucans on general biofilm architecture, is a common phenotype to study S. mutans virulence regulation. Here, we describe an entirely new phenotype associated with glucan production, caused by a mutation in the open reading frame SMU_848, which is located in an operon encoding ribosome-associated proteins. This mutation led to the excess production and accumulation of glucan-containing droplets on the surface of biofilms formed on agar plates after prolonged incubation. While not characterized in S. mutans, SMU_848 shows homology to the phage-related ribosomal protease Prp, essential in cleaving off the N-terminal extension of ribosomal protein L27 for functional ribosome assembly in Staphylococcus aureus. We present a further characterization of SMU_848/Prp, demonstrating that the deletion of this gene leads to significant changes in S. mutans gtfBC expression. Surprisingly, it also profoundly impacts the interkingdom interaction between S. mutans and Candida albicans, a relevant dual-species interaction implicated in severe early childhood caries. The presented data support a potential broader role for SMU_848/Prp, possibly extending its functionality beyond the ribosomal network to influence important ecological processes. IMPORTANCE Streptococcus mutans is an important member of the oral biofilm and is implicated in the initiation of caries. One of the main virulence mechanisms is the glucan-dependent formation of biofilms. We identified a new player in the regulation of glucan production, SMU_848, which is part of an operon that also encodes for ribosomal proteins L27 and L21. A mutation in SMU_848, which encodes a phage-related ribosomal protease Prp, leads to a significant accumulation of glucan-containing droplets on S. mutans biofilms, a previously unknown phenotype. Further investigations expanded our knowledge about the role of SMU_848 beyond its role in glucan production, including significant involvement in interkingdom interactions, thus potentially playing a global role in the virulence regulation of S. mutans.
Collapse
Affiliation(s)
- Puthayalai Treerat
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Camilla de Mattos
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Molly Burnside
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Hua Zhang
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Yanting Zhu
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Zhengzhong Zou
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - David Anderson
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Hui Wu
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Justin Merritt
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| | - Jens Kreth
- Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health & Science University (OHSU), Portland, Oregon, USA
| |
Collapse
|
12
|
Lee J, Choi JH, Lee J, Cho E, Lee YJ, Lee HS, Oh KB. Halenaquinol Blocks Staphylococcal Protein A Anchoring on Cell Wall Surface by Inhibiting Sortase A in Staphylococcus aureus. Mar Drugs 2024; 22:266. [PMID: 38921577 PMCID: PMC11204543 DOI: 10.3390/md22060266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Sortase A (SrtA) is a cysteine transpeptidase that binds to the periplasmic membrane and plays a crucial role in attaching surface proteins, including staphylococcal protein A (SpA), to the peptidoglycan cell wall. Six pentacyclic polyketides (1-6) were isolated from the marine sponge Xestospongia sp., and their structures were elucidated using spectroscopic techniques and by comparing them to previously reported data. Among them, halenaquinol (2) was found to be the most potent SrtA inhibitor, with an IC50 of 13.94 μM (4.66 μg/mL). Semi-quantitative reverse transcription PCR data suggest that halenaquinol does not inhibit the transcription of srtA and spA, while Western blot analysis and immunofluorescence microscopy images suggest that it blocks the cell wall surface anchoring of SpA by inhibiting the activity of SrtA. The onset and magnitude of the inhibition of SpA anchoring on the cell wall surface in S. aureus that has been treated with halenaquinol at a value 8× that of the IC50 of SrtA are comparable to those for an srtA-deletion mutant. These findings contribute to the understanding of the mechanism by which marine-derived pentacyclic polyketides inhibit SrtA, highlighting their potential as anti-infective agents targeting S. aureus virulence.
Collapse
Affiliation(s)
- Jaepil Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Jae-Hyeong Choi
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jayho Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Eunji Cho
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Yeon-Ju Lee
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Hyi-Seung Lee
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Ki-Bong Oh
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| |
Collapse
|
13
|
Castellanos JS, Betancourt DE, Díaz-Báez D, Baldión PA. Effect of flavonoids from grape seed and cranberry extracts on the microbiological activity of Streptococcus mutans: a systematic review of in vitro studies. BMC Oral Health 2024; 24:662. [PMID: 38840232 PMCID: PMC11155149 DOI: 10.1186/s12903-024-04263-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 04/15/2024] [Indexed: 06/07/2024] Open
Abstract
OBJECTIVE To provide an overview of the available scientific evidence from in vitro studies regarding the effect induced by the flavonoids contained in grape seed extracts (GSE) and cranberry on the microbiological activity of Streptococcus mutans (S. mutans). METHODS This systematic review was performed following the parameters of the PRISMA statement (Preferred Reporting Items for Systematic Reviews and Meta-Analysis). Electronic and manual searches were conducted using PubMed, ScienceDirect, Web of Science, EBSCO, and Cochrane databases. Reference lists of selected articles were reviewed to identify relevant studies. The search was not limited by year and was conducted solely in English. Eligible studies comprised publications describing in vitro studies that evaluated the effect of flavonoids derived from GSE and cranberry extracts on the microbiological activity of S. mutans. Common variables were identified to consolidate the data. Authors of this review independently screened search results, extracted data, and assessed the risk of bias. RESULTS Of the 420 studies identified from the different databases, 22 publications were finally selected for review. The risk of bias was low in 13 articles and moderate in 9. The studies analyzed in this review revealed that cranberry extract has an inhibitory effect on the bacterial growth of S. mutans in ranges from 0.5 mg/mL to 25 mg/mL, and GSE exerts a similar effect from 0.5 mg/mL to 250 mg/mL. Additionally, the extracts or their fractions showed reduced biofilm formation capacity, decreased polymicrobial biofilm biomass, deregulation of glycosyltransferases (Gtf) B and C expression, and buffering of pH drop. In addition to adequate antioxidant activity related to polyphenol content. CONCLUSIONS The overall results showed that the extracts of cranberry and grape seed were effective in reducing the virulence factors of the oral pathogen. According to the data, proanthocyanidins are the active components in cranberry and grape seed that effectively resist S. mutans. They can inhibit the formation of insoluble polysaccharides in the extracellular matrix and prevent glycan-mediated adhesion, cohesion, and aggregation of the proteins in S. mutans. This suggests that these natural extracts could play an important role in the prevention of cariogenic bacterial colonization, as well as induce a decrease in their microbiological activity.
Collapse
Affiliation(s)
- Jeison Stiven Castellanos
- Departamento de Salud Oral, Facultad de Odontología, Universidad Nacional de Colombia. Av, Cra 30 No. 45-03, Edificio 210, Of. 311, Bogotá, Colombia
| | - Diego Enrique Betancourt
- Departamento de Salud Oral, Facultad de Odontología, Universidad Nacional de Colombia. Av, Cra 30 No. 45-03, Edificio 210, Of. 311, Bogotá, Colombia
| | - David Díaz-Báez
- Unit of Oral Basic Investigation - UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Paula Alejandra Baldión
- Departamento de Salud Oral, Facultad de Odontología, Universidad Nacional de Colombia. Av, Cra 30 No. 45-03, Edificio 210, Of. 311, Bogotá, Colombia.
| |
Collapse
|
14
|
Bernardoni BL, D'Agostino I, La Motta C, Angeli A. An insight into the last 5-year patents on Porphyromonas gingivalis and Streptococcus mutans, the pivotal pathogens in the oral cavity. Expert Opin Ther Pat 2024; 34:433-463. [PMID: 38684444 DOI: 10.1080/13543776.2024.2349739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION The oral cavity harbors an extensive array of over 700 microorganisms, forming the most complex biome of the entire human body, with bacterial species being the most abundant. Oral diseases, e.g. periodontitis and caries, are strictly associated with bacterial dysbiosis. Porphyromonas gingivalis and Streptococcus mutans stand out among bacteria colonizing the oral cavity. AREAS COVERED After a brief overview of the bacterial populations in the oral cavity and their roles in regulating (flora) oral cavity or causing diseases like periodontal and cariogenic pathogens, we focused our attention on P. gingivalis and S. mutans, searching for the last-5-year patents dealing with the proposal of new strategies to fight their infections. Following the PRISMA protocol, we filtered the results and analyzed over 100 applied/granted patents, to provide an in-depth insight into this R&D scenario. EXPERT OPINION Several antibacterial proposals have been patented in this period, from both chemical - peptides and small molecules - and biological - probiotics and antibodies - sources, along with natural extracts, polymers, and drug delivery systems. Most of the inventors are from China and Korea and their studies also investigated anti-inflammatory and antioxidant effects, being beneficial to oral health through a prophylactic, protective, or curative effect.
Collapse
Affiliation(s)
| | | | | | - Andrea Angeli
- Neurofarba Department, University of Florence, Florence, Italy
| |
Collapse
|
15
|
Barbosa CDB, Monici Silva I, Dame-Teixeira N. The action of microbial collagenases in dentinal matrix degradation in root caries and potential strategies for its management: a comprehensive state-of-the-art review. J Appl Oral Sci 2024; 32:e20240013. [PMID: 38775556 PMCID: PMC11182643 DOI: 10.1590/1678-7757-2024-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 05/25/2024] Open
Abstract
Conventional views associate microbial biofilm with demineralization in root caries (RC) onset, while research on their collagenases role in the breakdown of collagen matrix has been sporadically developed, primarily in vitro. Recent discoveries, however, reveal proteolytic bacteria enrichment, specially Porphyromonas and other periodontitis-associated bacteria in subgingivally extended lesions, suggesting a potential role in RC by the catabolism of dentin organic matrix. Moreover, genes encoding proteases and bacterial collagenases, including the U32 family collagenases, were found to be overexpressed in both coronal and root dentinal caries. Despite these advancements, to prove microbial collagenolytic proteases' definitive role in RC remains a significant challenge. A more thorough investigation is warranted to explore the potential of anti-collagenolytic agents in modulating biofilm metabolic processes or inhibiting/reducing the size of RC lesions. Prospective treatments targeting collagenases and promoting biomodification through collagen fibril cross-linking show promise for RC prevention and management. However, these studies are currently in the in vitro phase, necessitating additional research to translate findings into clinical applications. This is a comprehensive state-of-the-art review aimed to explore contributing factors to the formation of RC lesions, particularly focusing on collagen degradation in root tissues by microbial collagenases.
Collapse
Affiliation(s)
- Cecília de Brito Barbosa
- Universidade de Brasília, Faculdade de Ciências da Saúde, Departamento de Odontologia, Brasília, Brasil
| | - Isabela Monici Silva
- Universidade de Brasília, Faculdade de Ciências da Saúde, Departamento de Odontologia, Brasília, Brasil
| | - Naile Dame-Teixeira
- Universidade de Brasília, Faculdade de Ciências da Saúde, Departamento de Odontologia, Brasília, Brasil
| |
Collapse
|
16
|
Wint WY, Miyanohara M, Yamada H, Nakatsuka T, Okamoto M, Ryo K, Tanaka T, Hanada N, Murata T. Rapid multiplex real-time PCR assay using a portable device for the detection of oral pathogens. Diagn Microbiol Infect Dis 2024; 109:116214. [PMID: 38402755 DOI: 10.1016/j.diagmicrobio.2024.116214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
Colonization by several oral pathogens and the onset of oral diseases, such as dental caries and periodontal diseases, are closely related. Therefore, the analysis of pathogens in oral specimens would be helpful for the risk assessment of oral diseases. We developed a rapid multiplex real-time polymerase chain reaction (PCR) method using a portable device and newly designed probe/primer sets to detect the oral pathogens Streptococcus mutans, Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia. The theoretical minimum detectable cell numbers of S. mutans, P. gingivalis, T. denticola, and T. forsythia were 1, 1, 4, and 3, respectively. The multiplex real-time PCR system simultaneously detected the colonization of S. mutans and P. gingivalis in human saliva. These results suggest that the multiplex real-time PCR system may be useful for the risk assessment of oral diseases.
Collapse
Affiliation(s)
- Wit Yee Wint
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Mayu Miyanohara
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Hidenori Yamada
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Takako Nakatsuka
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Masaaki Okamoto
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Koufuchi Ryo
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Tomoko Tanaka
- Department of Oral Health, School of Life Dentistry at Tokyo, Nippon Dental University, Fujimi, Chiyoda-ku, Tokyo, 102-8159 Japan
| | - Nobuhiro Hanada
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan
| | - Takatoshi Murata
- Department of Oral Health Science, Tsurumi University School of Dental Medicine, Tsurumi, 230-8501 Yokohama, Japan.
| |
Collapse
|
17
|
Zhou Y, Liu X, Chen H, Zhao J, Zhang H, Chen W, Yang B. Isolation and Characterisation of Streptococcus spp. with Human Milk Oligosaccharides Utilization Capacity from Human Milk. Foods 2024; 13:1291. [PMID: 38731662 PMCID: PMC11083076 DOI: 10.3390/foods13091291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Human milk oligosaccharides (HMO) that promote the growth of beneficial gut microbes in infants are abundant in human milk. Streptococcus, one of the dominant genera in human milk microbiota, is also highly prevalent in the infant gut microbiota, possibly due to its adeptness at utilizing HMOs. While previous studies have mainly focused on HMO interactions with gut bacteria like Bifidobacterium and Bacteroides spp., the interaction with Streptococcus spp. has not been fully explored. In this study, Streptococcus spp. was isolated from human milk and identified to exhibit extensive capabilities in utilizing HMOs. Their consumption rates of 2'-fucosyllactose (2'-FL), 6'-sialyllactose (6'-SL), and lacto-N-tetraose (LNT) closely matched those of Bifidobacterium longum subsp. infantis ATCC 15697. Furthermore, we assessed the safety-related genes in the genomes of the Streptococcus species capable of utilizing HMOs, revealing potential virulence and resistance genes. In addition, no haemolytic activity was observed. These findings expand the knowledge of metabolic interactions and networks within the microbiota of human milk and the early life human gut.
Collapse
Affiliation(s)
- Ye Zhou
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (Y.Z.); (X.L.); (H.C.); (J.Z.); (H.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
18
|
Mu R, Momeni S, Krieger M, Xie B, Cao X, Merritt J, Wu H. Plasmalogen, a glycerophospholipid crucial for Streptococcus mutans acid tolerance and colonization. Appl Environ Microbiol 2024; 90:e0150023. [PMID: 38456674 PMCID: PMC11022534 DOI: 10.1128/aem.01500-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/23/2024] [Indexed: 03/09/2024] Open
Abstract
Plasmalogen is a specific glycerophospholipid present in both animal and bacterial organisms. It plays a crucial function in eukaryotic cellular processes and is closely related to several human diseases, including neurological disorders and cancers. Nonetheless, the precise biological role of plasmalogen in bacteria is not well understood. In this study, we identified SMU_438c as the enzyme responsible for plasmalogen production in Streptococcus mutans under anaerobic conditions. The heterologous expression of SMU_438c in a plasmalogen-negative strain, Streptococcus sanguinis, resulted in the production of plasmalogen, indicating that this enzyme is sufficient for plasmalogen production. Additionally, the plasmalogen-deficient S. mutans exhibited significantly lower acid tolerance and diminished its colonization in Drosophila flies compared to the wild-type strain and complemented strain. In summary, our data suggest that plasmalogen plays a vital role in bacterial stress tolerance and in vivo colonization. IMPORTANCE This study sheds light on the biological role of plasmalogen, a specific glycerophospholipid, in bacteria, particularly in Streptococcus mutans. Plasmalogens are known for their significant roles in eukaryotic cells and have been linked to human diseases like neurological disorders and cancers. The enzyme SMU_438c, identified as essential for plasmalogen production under anaerobic conditions, was crucial for acid tolerance and in vivo colonization in Drosophila by S. mutans, underscoring its importance in bacterial stress response and colonization. These findings bridge the knowledge gap in bacterial physiology, highlighting plasmalogen's role in microbial survival and offering potential insights into microbial pathogenesis and host-microbe interactions.
Collapse
Affiliation(s)
- Rong Mu
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Stephanie Momeni
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Madeline Krieger
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Baotong Xie
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Xixi Cao
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Justin Merritt
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Hui Wu
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
19
|
Sangha JS, Barrett P, Curtis TP, Métris A, Jakubovics NS, Ofiteru ID. Effects of glucose and lactate on Streptococcus mutans abundance in a novel multispecies oral biofilm model. Microbiol Spectr 2024; 12:e0371323. [PMID: 38376204 PMCID: PMC10986578 DOI: 10.1128/spectrum.03713-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
The oral microbiome plays an important role in protecting oral health. Here, we established a controlled mixed-species in vitro biofilm model and used it to assess the impact of glucose and lactate on the ability of Streptococcus mutans, an acidogenic and aciduric species, to compete with commensal oral bacteria. A chemically defined medium was developed that supported the growth of S. mutans and four common early colonizers of dental plaque: Streptococcus gordonii, Actinomyces oris, Neisseria subflava, and Veillonella parvula. Biofilms containing the early colonizers were developed in a continuous flow bioreactor, exposed to S. mutans, and incubated for up to 7 days. The abundance of bacteria was estimated by quantitative polymerase chain reaction (qPCR). At high glucose and high lactate, the pH in bulk fluid rapidly decreased to approximately 5.2, and S. mutans outgrew other species in biofilms. In low glucose and high lactate, the pH remained above 5.5, and V. parvula was the most abundant species in biofilms. By contrast, in low glucose and low lactate, the pH remained above 6.0 throughout the experiment, and the microbial community in biofilms was relatively balanced. Fluorescence in situ hybridization confirmed that all species were present in the biofilm and the majority of cells were viable using live/dead staining. These data demonstrate that carbon source concentration is critical for microbial homeostasis in model oral biofilms. Furthermore, we established an experimental system that can support the development of computational models to predict transitions to microbial dysbiosis based on metabolic interactions.IMPORTANCEWe developed a controlled (by removing host factor) dynamic system metabolically representative of early colonization of Streptococcus mutans not measurable in vivo. Hypotheses on factors influencing S. mutans colonization, such as community composition and inoculation sequence and the effect of metabolite concentrations, can be tested and used to predict the effect of interventions such as dietary modifications or the use of toothpaste or mouthwash on S. mutans colonization. The defined in vitro model (species and medium) can be simulated in an in silico model to explore more of the parameter space.
Collapse
Affiliation(s)
- Jay S. Sangha
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Paul Barrett
- Safety and Environmental Assurance Centre, Unilever, Colworth Science Park, Sharnbrook, United Kingdom
| | - Thomas P. Curtis
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Aline Métris
- Safety and Environmental Assurance Centre, Unilever, Colworth Science Park, Sharnbrook, United Kingdom
| | - Nicholas S. Jakubovics
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Irina D. Ofiteru
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
20
|
Jackson I, Woodman P, Dowd M, Fibiger L, Cassidy LM. Ancient Genomes From Bronze Age Remains Reveal Deep Diversity and Recent Adaptive Episodes for Human Oral Pathobionts. Mol Biol Evol 2024; 41:msae017. [PMID: 38533900 PMCID: PMC10966897 DOI: 10.1093/molbev/msae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 03/28/2024] Open
Abstract
Ancient microbial genomes can illuminate pathobiont evolution across millenia, with teeth providing a rich substrate. However, the characterization of prehistoric oral pathobiont diversity is limited. In Europe, only preagricultural genomes have been subject to phylogenetic analysis, with none compared to more recent archaeological periods. Here, we report well-preserved microbiomes from two 4,000-year-old teeth from an Irish limestone cave. These contained bacteria implicated in periodontitis, as well as Streptococcus mutans, the major cause of caries and rare in the ancient genomic record. Despite deriving from the same individual, these teeth produced divergent Tannerella forsythia genomes, indicating higher levels of strain diversity in prehistoric populations. We find evidence of microbiome dysbiosis, with a disproportionate quantity of S. mutans sequences relative to other oral streptococci. This high abundance allowed for metagenomic assembly, resulting in its first reported ancient genome. Phylogenetic analysis indicates major postmedieval population expansions for both species, highlighting the inordinate impact of recent dietary changes. In T. forsythia, this expansion is associated with the replacement of older lineages, possibly reflecting a genome-wide selective sweep. Accordingly, we see dramatic changes in T. forsythia's virulence repertoire across this period. S. mutans shows a contrasting pattern, with deeply divergent lineages persisting in modern populations. This may be due to its highly recombining nature, allowing for maintenance of diversity through selective episodes. Nonetheless, an explosion in recent coalescences and significantly shorter branch lengths separating bacteriocin-carrying strains indicate major changes in S. mutans demography and function coinciding with sugar popularization during the industrial period.
Collapse
Affiliation(s)
- Iseult Jackson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
- The SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Peter Woodman
- Department of Archaeology, University College Cork, Cork, Ireland
| | - Marion Dowd
- Faculty of Science, Atlantic Technological University, Sligo, Ireland
| | - Linda Fibiger
- School of History, Classics and Archaeology, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Lara M Cassidy
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
21
|
Hayashida N, Urano-Tashiro Y, Horie T, Saiki K, Yamanaka Y, Takahashi Y. Transcriptome and metabolome analyses of Streptococcus gordonii DL1 under acidic conditions. J Oral Biosci 2024; 66:112-118. [PMID: 38135272 DOI: 10.1016/j.job.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVES Streptococcus gordonii is associated with the formation of biofilms, especially those that comprise dental plaque. Notably, S. gordonii DL1 causes infective endocarditis (IE). Colonization of this bacterium requires a mechanism that can tolerate a drop in environmental pH by producing acid via its own sugar metabolism. The ability to survive acidic environmental conditions might allow the bacterium to establish vegetative colonization even in the endocardium due to inflammation-induced lowering of pH, increasing the risk of IE. At present, the mechanism by which S. gordonii DL1 survives under acidic conditions is not thoroughly elucidated. The present study was thus conducted to elucidate the mechanism(s) by which S. gordonii DL1 survives under acidic conditions. METHODS We analyzed dynamic changes in gene transcription and intracellular metabolites in S. gordonii DL1 exposed to acidic conditions, using transcriptome and metabolome analyses. RESULTS Transcriptome analysis revealed upregulation of genes involved in heat shock response and glycolysis, and down regulation of genes involved in phosphotransferase systems and biosynthesis of amino acids. The most upregulated genes were a beta-strand repeat protein of unknown function (SGO_RS06325), followed by copper-translocating P-type ATPase (SGO_RS09470) and malic enzyme (SGO_RS01850). The latter two of these contribute to cytoplasmic alkalinization. S. gordonii mutant strains lacking each of these genes showed significantly reduced survival under acidic conditions. Metabolome analysis revealed that cytoplasmic levels of several amino acids were reduced. CONCLUSIONS S. gordonii survives the acidic conditions by recovering the acidic cytoplasm using the various activities, which are regulated at the transcriptional level.
Collapse
Affiliation(s)
- Naoto Hayashida
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Yumiko Urano-Tashiro
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Tetsuro Horie
- Research Center for Odontology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Keitarou Saiki
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Yuki Yamanaka
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Yukihiro Takahashi
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| |
Collapse
|
22
|
Yu S, Ma Q, Huang J, Liu Y, Li J, Wang Y, Gong T, Zhang Q, Zou J, Li Y. SMU_1361c regulates the oxidative stress response of Streptococcus mutans. Appl Environ Microbiol 2024; 90:e0187123. [PMID: 38299814 PMCID: PMC10880606 DOI: 10.1128/aem.01871-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/22/2023] [Indexed: 02/02/2024] Open
Abstract
Dental caries is the most common chronic infectious disease around the world and disproportionately affects the marginalized socioeconomic group. Streptococcus mutans, considered a primary etiological agent of caries, depends on the coordinated physiological response to tolerate the oxidative stress generated by commensal species within dental plaque, which is a critical aspect of its pathogenicity. Here, we identified and characterized a novel tetracycline repressor family regulator, SMU_1361c, which appears to be acquired by the bacteria via horizontal gene transfer. Surprisingly, smu_1361c functions as a negative transcriptional regulator to regulate gene expression outside its operon and is involved in the oxidative stress response of S. mutans. The smu_1361c overexpression strain UA159/pDL278-1361c was more susceptible to oxidative stress and less competitive against hydrogen peroxide generated by commensal species Streptococcus gordonii and Streptococcus sanguinis. Transcriptomics analysis revealed that smu_1361c overexpression resulted in the significant downregulation of 22 genes, mainly belonging to three gene clusters responsible for the oxidative stress response. The conversed DNA binding motif of SMU_1361c was determined by electrophoretic mobility shift and DNase I footprinting assay with purified SMU_1361c protein; therefore, smu_1361c is directly involved in gene transcription related to the oxidative stress response. Crucially, our finding provides a new understanding of how S. mutans deals with the oxidative stress that is required for pathogenesis and will facilitate the development of new and improved therapeutic approaches for dental caries.IMPORTANCEStreptococcus mutans is the major organism associated with the development of dental caries, which globally is the most common chronic disease. To persist and survive in biofilms, S. mutans must compete with commensal species that occupy the same ecological niche. Here, we uncover a novel molecular mechanism of how tetracycline repressor family regulator smu_1361c is involved in the oxidative stress response through transcriptomics analysis, electrophoretic mobility shift assay, and DNase I footprinting assay. Furthermore, we demonstrated that smu_1361c mediates S. mutans sensitivity to oxidative stress and competitiveness with commensal streptococci. Therefore, this study has revealed a previously unknown regulation between smu_1361c and genes outside its operon and demonstrated the importance of smu_1361c in the oxidative stress response and the fitness of S. mutans within the plaque biofilms, which can be exploited as a new therapy to modulate ecological homeostasis and prevent dental caries.
Collapse
Affiliation(s)
- Shuxing Yu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qizhao Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaqi Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tao Gong
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiong Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Zou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Qin H, Anderson D, Zou Z, Higashi D, Borland C, Kreth J, Merritt J. Mass spectrometry and split luciferase complementation assays reveal the MecA protein interactome of Streptococcus mutans. Microbiol Spectr 2024; 12:e0369123. [PMID: 38230956 PMCID: PMC10845952 DOI: 10.1128/spectrum.03691-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/11/2023] [Indexed: 01/18/2024] Open
Abstract
MecA is a highly conserved adaptor protein encoded by prokaryotes from the Bacillota phylum. MecA mutants exhibit similar pleiotropic defects in a variety of organisms, although most of these phenotypes currently lack a mechanistic basis. MecA mediates ClpCP-dependent proteolysis of its substrates, but only several such substrates have been reported in the literature and there are suggestions that proteolysis-independent regulatory mechanisms may also exist. Here, we provide the first comprehensive characterization of the MecA interactome and further assess its regulatory role in Clp-dependent proteolysis. Untargeted coimmunoprecipitation assays coupled with mass spectrometry revealed that the MecA ortholog from the oral pathobiont Streptococcus mutans likely serves as a major protein interaction network hub by potentially complexing with >100 distinct protein substrates, most of which function in highly conserved metabolic pathways. The interactome results were independently verified using a newly developed prokaryotic split luciferase complementation assay (SLCA) to detect MecA protein-protein interactions in vivo. In addition, we further develop a new application of SLCA to support in vivo measurements of MecA relative protein binding affinities. SLCA results were independently verified using targeted coimmunoprecipitation assays, suggesting the general utility of this approach for prokaryotic protein-protein interaction studies. Our results indicate that MecA indeed regulates its interactome through both Clp-dependent proteolysis as well as through an as-yet undefined proteolysis-independent mechanism that may affect more than half of its protein interactome. This suggests a significant aspect of the MecA regulatory function still has yet to be discovered.IMPORTANCEDespite multiple decades of study, the regulatory mechanism and function of MecA have remained largely a mystery. The current study provides the first detailed roadmap to investigate these functions in other medically significant bacteria. Furthermore, this study developed new genetic approaches to assay prokaryotic protein-protein interactions via the split luciferase complementation assay (SLCA). SLCA technology is commonly employed in eukaryotic genetic research but has not yet been established for studies of bacterial protein-protein interactions. The SLCA protein binding affinity assay described here is a new technological advance exclusive to the current study and has not been reported elsewhere.
Collapse
Affiliation(s)
- Hua Qin
- Division of Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - David Anderson
- Division of Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - Zhengzhong Zou
- Division of Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - Dustin Higashi
- Division of Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - Christina Borland
- Division of Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - Jens Kreth
- Division of Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Justin Merritt
- Division of Biomaterial and Biomedical Sciences, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
24
|
Mishra S, van Aalst EJ, Wylie BJ, Brady LJ. Cardiolipin occupancy profiles of YidC paralogs reveal the significance of respective TM2 helix residues in determining paralog-specific phenotypes. Front Mol Biosci 2023; 10:1264454. [PMID: 37867558 PMCID: PMC10588454 DOI: 10.3389/fmolb.2023.1264454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
YidC belongs to an evolutionarily conserved family of insertases, YidC/Oxa1/Alb3, in bacteria, mitochondria, and chloroplasts, respectively. Unlike Gram-negative bacteria, Gram-positives including Streptococcus mutans harbor two paralogs of YidC. The mechanism for paralog-specific phenotypes of bacterial YidC1 versus YidC2 has been partially attributed to the differences in their cytoplasmic domains. However, we previously identified a W138R gain-of-function mutation in the YidC1 transmembrane helix 2. YidC1W138R mostly phenocopied YidC2, yet the mechanism remained unknown. Primary sequence comparison of streptococcal YidCs led us to identify and mutate the YidC1W138 analog, YidC2S152 to W/A, which resulted in a loss of YidC2- and acquisition of YidC1-like phenotype. The predicted lipid-facing side chains of YidC1W138/YidC2S152 led us to propose a role for membrane phospholipids in specific-residue dependent phenotypes of S. mutans YidC paralogs. Cardiolipin (CL), a prevalent phospholipid in the S. mutans cytoplasmic membrane during acid stress, is encoded by a single gene, cls. We show a concerted mechanism for cardiolipin and YidC2 under acid stress based on similarly increased promoter activities and similar elimination phenotypes. Using coarse grain molecular dynamics simulations with the Martini2.2 Forcefield, YidC1 and YidC2 wild-type and mutant interactions with CL were assessed in silico. We observed substantially increased CL interaction in dimeric versus monomeric proteins, and variable CL occupancy in YidC1 and YidC2 mutant constructs that mimicked characteristics of the other wild-type paralog. Hence, paralog-specific amino acid- CL interactions contribute to YidC1 and YidC2-associated phenotypes that can be exchanged by point mutation at positions 138 or 152, respectively.
Collapse
Affiliation(s)
- Surabhi Mishra
- Department of Oral Biology, University of Florida, Gainesville, FL, United States
| | - Evan J. van Aalst
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, United States
| | - Benjamin J. Wylie
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, United States
| | - L. Jeannine Brady
- Department of Oral Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
25
|
Jurakova V, Farková V, Kucera J, Dadakova K, Zapletalova M, Paskova K, Reminek R, Glatz Z, Holla LI, Ruzicka F, Lochman J, Linhartova PB. Gene expression and metabolic activity of Streptococcus mutans during exposure to dietary carbohydrates glucose, sucrose, lactose, and xylitol. Mol Oral Microbiol 2023; 38:424-441. [PMID: 37440366 DOI: 10.1111/omi.12428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023]
Abstract
Recent RNA sequencing studies have given us a deeper insight into the cariogenic impact of carbohydrate sources in the bacterium Streptococcus mutans, the principal microbial agent in dental caries etiopathogenesis. The process of dental caries development is facilitated by the ability of this bacterium to ferment some carbohydrates into organic acids contributing to a pH decrease in the oral cavity and the demineralization of the hard tissues of the tooth. Furthermore, in dental caries progression, biofilm formation, which starts and ends with free planktonic cells, plays an important role and has several unique properties called virulence factors. The most cariogenic carbohydrate is sucrose, an easily metabolizable source of energy that induces the acidification and synthesis of glucans, forming typical bacterial cell clumps. We used multifaceted methodological approaches to compare the transcriptomic and metabolomic profiles of S. mutans growing in planktonic culture on preferred and nonpreferred carbohydrates and in fasting conditions. Streptococcus mutans in a planktonic culture with lactose produced the same pH drop as glucose and sucrose. By contrast, xylitol and lactose showed high effectiveness in regulating intracellular polysaccharide metabolism, cell wall structure, and overall virulence involved in the initial phase of biofilm formation and structure but with an opposite pattern compared with sucrose and glucose. Our results confirmed the recent findings that xylitol and lactose play a vital role in biofilm structure. However, they do not reduce its formation, which is related to the creation of a cariogenic environment.
Collapse
Affiliation(s)
- Veronika Jurakova
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Veronika Farková
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jiri Kucera
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Katerina Dadakova
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Martina Zapletalova
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Katerina Paskova
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Roman Reminek
- Institute of Analytical Chemistry of the CAS, Brno, Czech Republic
| | - Zdenek Glatz
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lydie Izakovicova Holla
- Clinic of Stomatology, Institution Shared with St. Anne's University Hospital, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Filip Ruzicka
- Institute for Microbiology, Faculty of Medicine, Masaryk University and St. Anne's University Hospital, Brno, Czech Republic
| | - Jan Lochman
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petra Borilova Linhartova
- Clinic of Stomatology, Institution Shared with St. Anne's University Hospital, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pathophysiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Clinic of Maxillofacial Surgery, Institution Shared with University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
26
|
Dufour D, Li H, Gong SG, Lévesque CM. Transcriptome Analysis of Streptococcus mutans Quorum Sensing-Mediated Persisters Reveals an Enrichment in Genes Related to Stress Defense Mechanisms. Genes (Basel) 2023; 14:1887. [PMID: 37895236 PMCID: PMC10606796 DOI: 10.3390/genes14101887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Persisters are a small fraction of growth-arrested phenotypic variants that can survive lethal concentrations of antibiotics but are able to resume growth once antibiotics are stopped. Their formation can be a stochastic process or one triggered by environmental cues. In the human pathogen Streptococcus mutans, the canonical peptide-based quorum-sensing system is an inducible DNA repair system that is pivotal for bacterial survival. Previous work has shown that the CSP-signaling peptide is a stress-signaling alarmone that promotes the formation of stress-induced persisters. In this study, we exposed S. mutans to the CSP pheromone to mimic DNA damage conditions and isolated the antibiotic persisters by treating the cultures with ofloxacin. A transcriptome analysis was then performed to evaluate the differential gene expression between the normal stationary-phase cells and the persisters. RNA sequencing revealed that triggered persistence was associated with the upregulation of genes related to several stress defense mechanisms, notably, multidrug efflux pumps, the arginine deaminase pathway, and the Opu/Opc system. In addition, we showed that inactivation of the VicK kinase of the YycFG essential two-component regulatory system abolished the formation of triggered persisters via the CSP pheromone. These data contribute to the understanding of the triggered persistence phenotype and may suggest new therapeutic strategies for treating persistent streptococcal infections.
Collapse
Affiliation(s)
| | | | | | - Céline M. Lévesque
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; (D.D.); (H.L.); (S.-G.G.)
| |
Collapse
|
27
|
Drummond IY, DePaolo A, Krieger M, Driscoll H, Eckstrom K, Spatafora GA. Small regulatory RNAs are mediators of the Streptococcus mutans SloR regulon. J Bacteriol 2023; 205:e0017223. [PMID: 37695854 PMCID: PMC10521355 DOI: 10.1128/jb.00172-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/08/2023] [Indexed: 09/13/2023] Open
Abstract
Dental caries is among the most prevalent chronic diseases worldwide. Streptococcus mutans, the chief causative agent of caries, uses a 25-kDa manganese-dependent SloR protein to coordinate the uptake of essential manganese with the transcription of its virulence attributes. Small non-coding RNAs (sRNAs) can either enhance or repress gene expression, and reports in the literature ascribe an emerging role for sRNAs in the environmental stress response. Herein, we focused our attention on 18-50 nt sRNAs as mediators of the S. mutans SloR and manganese regulons. Specifically, the results of RNA sequencing revealed 19 sRNAs in S. mutans, which were differentially transcribed in the SloR-proficient UA159 and SloR-deficient GMS584 strains, and 10 sRNAs that were differentially expressed in UA159 cells grown in the presence of low vs high manganese. We describe SmsR1532 and SmsR1785 as SloR- and manganese-responsive sRNAs that are processed from large transcripts and that bind SloR directly in their promoter regions. The predicted targets of these sRNAs include regulators of metal ion transport, growth management via a toxin-antitoxin operon, and oxidative stress tolerance. These findings support a role for sRNAs in coordinating intracellular metal ion homeostasis with virulence gene control in an important oral cariogen. IMPORTANCE Small regulatory RNAs (sRNAs) are critical mediators of environmental signaling, particularly in bacterial cells under stress, but their role in Streptococcus mutans is poorly understood. S. mutans, the principal causative agent of dental caries, uses a 25-kDa manganese-dependent protein, called SloR, to coordinate the regulated uptake of essential metal ions with the transcription of its virulence genes. In the present study, we identified and characterized sRNAs that are both SloR and manganese responsive. Taken together, this research can elucidate the details of regulatory networks that engage sRNAs in an important oral pathogen and that can enable the development of an effective anti-caries therapeutic.
Collapse
Affiliation(s)
| | | | - Madeline Krieger
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Heather Driscoll
- Department of Biology, Vermont Biomedical Research Network, Norwich University, Northfield, Vermont, USA
| | - Korin Eckstrom
- Department of Microbiology and Molecular Genetics, The Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, USA
| | | |
Collapse
|
28
|
Dornelas-Figueira LM, Ricomini Filho AP, Junges R, Åmdal HA, Cury AADB, Petersen FC. In Vitro Impact of Fluconazole on Oral Microbial Communities, Bacterial Growth, and Biofilm Formation. Antibiotics (Basel) 2023; 12:1433. [PMID: 37760729 PMCID: PMC10525723 DOI: 10.3390/antibiotics12091433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Antifungal agents are widely used to specifically eliminate infections by fungal pathogens. However, the specificity of antifungal agents has been challenged by a few studies demonstrating antibacterial inhibitory effects against Mycobacteria and Streptomyces species. Here, we evaluated for the first time the potential effect of fluconazole, the most clinically used antifungal agent, on a human oral microbiota biofilm model. The results showed that biofilm viability on blood and mitis salivarius agar media was increased over time in the presence of fluconazole at clinically relevant concentrations, despite a reduction in biomass. Targeted PCR revealed a higher abundance of Veillonella atypica, Veillonella dispar, and Lactobacillus spp. in the fluconazole-treated samples compared to the control, while Fusobacterium nucleatum was reduced and Streptococcus spp were not significantly affected. Further, we tested the potential impact of fluconazole using single-species models. Our results, using Streptococcus mutans and Streptococcus mitis luciferase reporters, showed that S. mutans planktonic growth was not significantly affected by fluconazole, whereas for S. mitis, planktonic growth, but not biofilm viability, was inhibited at the highest concentration. Fluconazole's effects on S. mitis biofilm biomass were concentration and time dependent. Exposure for 48 h to the highest concentration of fluconazole was associated with S. mitis biofilms with the most increased biomass. Potential growth inhibitory effects were further tested using four non-streptococcal species. Among these, the planktonic growth of both Escherichia coli and Granulicatella adiacens was inhibited by fluconazole. The data indicate bacterial responses to fluconazole that extend to a broader range of bacterial species than previously anticipated from the literature, with the potential to disturb biofilm communities.
Collapse
Affiliation(s)
- Louise Morais Dornelas-Figueira
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas, Piracicaba 13414-903, SP, Brazil
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0372 Oslo, Norway
| | - Antônio Pedro Ricomini Filho
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Piracicaba 13414-903, SP, Brazil
| | - Roger Junges
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0372 Oslo, Norway
| | - Heidi Aarø Åmdal
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0372 Oslo, Norway
| | - Altair Antoninha Del Bel Cury
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas, Piracicaba 13414-903, SP, Brazil
| | | |
Collapse
|
29
|
Li Y, Li G, Deng X. A tightly controlled gene induction system that contributes to the study of lethal gene function in Streptococcus mutans. J Oral Microbiol 2023; 15:2253675. [PMID: 37691880 PMCID: PMC10486305 DOI: 10.1080/20002297.2023.2253675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/01/2023] [Accepted: 08/24/2023] [Indexed: 09/12/2023] Open
Abstract
Effective control of gene expression is crucial for understanding gene function in both eukaryotic and prokaryotic cells. While several inducible gene expression systems have been reported in Streptococcus mutans, a conditional pathogen that causes dental caries, the significant non-inducible basal expression in these systems seriously limits their utility, especially when studying lethal gene functions and molecular mechanisms. We introduce a tightly controlled xylose-inducible gene expression system, TC-Xyl, for Streptococcus mutans. Western blot results and fluorescence microscopy analysis indicate that TC-Xyl exhibits an extremely low non-inducible basal expression level and a sufficiently high expression level post-induction. Further, by constructing a mutation in which the only source FtsZ is under the control of TC-Xyl, we preliminarily explored the function of the ftsz gene. We found that FtsZ depletion is lethal to Streptococcus mutans, resulting in abnormal round cell shape and mini cell formation, suggesting FtsZ's role in maintaining cell shape stability.
Collapse
Affiliation(s)
- Yongliang Li
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology, Beijing, People’s Republic of China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, People’s Republic of China
| | - Guanwen Li
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, People’s Republic of China
| | - Xuliang Deng
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, People’s Republic of China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, People’s Republic of China
| |
Collapse
|
30
|
Choi A, Dong K, Williams E, Pia L, Batagower J, Bending P, Shin I, Peters DI, Kaspar JR. Human Saliva Modifies Growth, Biofilm Architecture and Competitive Behaviors of Oral Streptococci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.554151. [PMID: 37662325 PMCID: PMC10473590 DOI: 10.1101/2023.08.21.554151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The bacteria within supragingival biofilms participate in complex exchanges with other microbes inhabiting the same niche. One example are the mutans group streptococci (Streptococcus mutans), implicated in the development of tooth decay, and other health-associated commensal streptococci species. Previously, our group transcriptomically characterized intermicrobial interactions between S. mutans and several species of oral bacteria. However, these experiments were carried out in a medium that was absent of human saliva. To better mimic their natural environment, we first evaluated how inclusion of saliva affected growth and biofilm formation of eight streptococci species individually, and found saliva to positively benefit growth rates while negatively influencing biomass accumulation and altering spatial arrangement. These results carried over during evaluation of 29 saliva-derived isolates of various species. Surprisingly, we also found that addition of saliva increased the competitive behaviors of S. mutans in coculture competitions against commensal streptococci that led to increases in biofilm microcolony volumes. Through transcriptomically characterizing mono- and cocultures of S. mutans and Streptococcus oralis with and without saliva, we determined that each species developed a nutritional niche under mixed-species growth, with S. mutans upregulating carbohydrate uptake and utilization pathways while S. oralis upregulated genome features related to peptide uptake and glycan foraging. S. mutans also upregulated genes involved in oxidative stress tolerance, particularly manganese uptake, which we could artificially manipulate by supplementing in manganese to give it an advantage over its opponent. Our report highlights observable changes in microbial behaviors via leveraging environmental- and host-supplied resources over their competitors.
Collapse
Affiliation(s)
- Allen Choi
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| | - Kevin Dong
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| | - Emily Williams
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| | - Lindsey Pia
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| | - Jordan Batagower
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| | - Paige Bending
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| | - Iris Shin
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| | - Daniel I Peters
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| | - Justin R Kaspar
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio
| |
Collapse
|
31
|
Kang DY, Kim A, Kim JN. CcpA and CodY Regulate CRISPR-Cas System of Streptococcus mutans. Microbiol Spectr 2023; 11:e0182623. [PMID: 37367300 PMCID: PMC10434267 DOI: 10.1128/spectrum.01826-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated (Cas) genes are widely recognized as bacterial adaptive immune systems against invading viruses and bacteriophages. The oral pathogen Streptococcus mutans encodes two CRISPR-Cas loci (CRISPR1-Cas and CRISPR2-Cas), and their expression under environmental conditions is still under investigation. In this study, we investigated the transcriptional regulation of cas operons by CcpA and CodY, two global regulators that contribute to carbohydrate and (p)ppGpp metabolism. The possible promoter regions for cas operons and the binding sites for CcpA and CodY in the promoter regions of both CRISPR-Cas loci were predicted using computational algorithms. We found that CcpA could directly bind to the upstream region of both cas operons, and detected an allosteric interaction of CodY within the same region. The binding sequences of the two regulators were identified through footprinting analysis. Our results showed that the promoter activity of CRISPR1-Cas was enhanced under fructose-rich conditions, while deletion of the ccpA gene led to reduced activity of the CRISPR2-Cas promoter under the same conditions. Additionally, deletion of the CRISPR systems resulted in a significant decrease in fructose uptake ability compared to the parental strain. Interestingly, the accumulation of guanosine tetraphosphate (ppGpp) was reduced in the presence of mupirocin, which induces a stringent response, in the CRISPR1-Cas-deleted (ΔCR1cas) and both CRISPR-Cas-deleted (ΔCRDcas) mutant strains. Furthermore, the promoter activity of both CRISPRs was enhanced in response to oxidative or membrane stress, while the CRISPR1 promoter activity was reduced under low-pH conditions. Collectively, our findings demonstrate that the transcription of the CRISPR-Cas system is directly regulated by the binding of CcpA and CodY. These regulatory actions play a crucial role in modulating glycolytic processes and exerting effective CRISPR-mediated immunity in response to nutrient availability and environmental cues. IMPORTANCE An effective immune system has evolved not only in eukaryotic organisms but also in microorganisms, enabling them to rapidly detect and neutralize foreign invaders in the environment. Specifically, the CRISPR-Cas system in bacterial cells is established through a complex and sophisticated regulatory mechanism involving specific factors. In this study, we demonstrate that the expression of two CRISPR systems in S. mutans can be controlled by two global regulators, CcpA and CodY, which play critical roles in carbohydrate metabolism and amino acid biosynthesis. Importantly, our results show that the expression of the CRISPR-Cas system in S. mutans influences (p)ppGpp production during the stringent response, which is a gene expression regulatory response that aids in environmental stress adaptation. This transcriptional regulation by these regulators enables a CRISPR-mediated immune response in a host environment with limited availability of carbon sources or amino acids, while ensuring efficient carbon flux and energy expenditure to support multiple metabolic processes.
Collapse
Affiliation(s)
- Da-Young Kang
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Andy Kim
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas, USA
| | - Jeong Nam Kim
- Department of Integrated Biological Science, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
- Department of Microbiology, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
32
|
Huang X, Laird CG, Riley PP, Wen ZT. Impacts of a DUF2207 Family Protein on Streptococcus mutans Stress Tolerance Responses and Biofilm Formation. Microorganisms 2023; 11:1982. [PMID: 37630542 PMCID: PMC10457818 DOI: 10.3390/microorganisms11081982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Locus SMU.243 in Streptococcus mutans was annotated as a member of the DUF2207 family proteins highly conserved in all bacteria but with unknown function. To investigate its role in S. mutans physiology, a SMU.243-deficient mutant was constructed using allelic exchange mutagenesis, and the impacts of SMU.243 deletion on bacterial growth, stress tolerance response, and biofilm formation were analyzed. Compared to the wild-type UA159, S. mutans lacking SMU.243 displayed a reduced growth rate and a reduced overnight culture density (p < 0.01) when grown at low pH and in the presence of methyl viologen. Relative to the parent strain, the deficient mutant also had a reduced survival rate following incubation in a buffer of pH 2.8 (p < 0.01) and in a buffer containing hydrogen peroxide at 58 mM after 60 min (p < 0.001) and had a reduced capacity in biofilm formation especially in the presence of sucrose (p < 0.01). To study any ensuing functional/phenotypical links between SMU.243 and uppP, which is located immediately downstream of SMU.243 and encodes an undecaprenyl pyrophosphate phosphatase involved in recycling of carrier lipid undecaprenyl phosphate, a uppP deficient mutant was generated using allelic exchange mutagenesis. Unlike the SMU.243 mutant, deletion of uppP affected cell envelope biogenesis and caused major increases in susceptibility to bacitracin. In addition, two variant morphological mutants, one forming rough colonies and the other forming mucoid, smooth colonies, also emerged following the deletion of uppP. The results suggest that the SMU.243-encoded protein of the DUF2207 family in S. mutans plays an important role in stress tolerance response and biofilm formation, but unlike the downstream uppP, does not seem to be involved in cell envelope biogenesis, although the exact roles in S. mutans' physiology awaits further investigation.
Collapse
Affiliation(s)
- Xiaochang Huang
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (X.H.); (P.P.R.)
| | - Camile G. Laird
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (X.H.); (P.P.R.)
| | - Paul P. Riley
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (X.H.); (P.P.R.)
| | - Zezhang Tom Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (X.H.); (P.P.R.)
- Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
33
|
Marimuthu SCV, Murugesan J, Babkiewicz E, Maszczyk P, Sankaranarayanan M, Thangamariappan E, Rosy JC, Ram Kumar Pandian S, Kunjiappan S, Balakrishnan V, Sundar K. Pharmacoinformatics-Based Approach for Uncovering the Quorum-Quenching Activity of Phytocompounds against the Oral Pathogen, Streptococcus mutans. Molecules 2023; 28:5514. [PMID: 37513386 PMCID: PMC10383507 DOI: 10.3390/molecules28145514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Streptococcus mutans, a gram-positive oral pathogen, is the primary causative agent of dental caries. Biofilm formation, a critical characteristic of S. mutans, is regulated by quorum sensing (QS). This study aimed to utilize pharmacoinformatics techniques to screen and identify effective phytochemicals that can target specific proteins involved in the quorum sensing pathway of S. mutans. A computational approach involving homology modeling, model validation, molecular docking, and molecular dynamics (MD) simulation was employed. The 3D structures of the quorum sensing target proteins, namely SecA, SMU1784c, OppC, YidC2, CiaR, SpaR, and LepC, were modeled using SWISS-MODEL and validated using a Ramachandran plot. Metabolites from Azadirachta indica (Neem), Morinda citrifolia (Noni), and Salvadora persica (Miswak) were docked against these proteins using AutoDockTools. MD simulations were conducted to assess stable interactions between the highest-scoring ligands and the target proteins. Additionally, the ADMET properties of the ligands were evaluated using SwissADME and pkCSM tools. The results demonstrated that campesterol, meliantrol, stigmasterol, isofucosterol, and ursolic acid exhibited the strongest binding affinity for CiaR, LepC, OppC, SpaR, and Yidc2, respectively. Furthermore, citrostadienol showed the highest binding affinity for both SMU1784c and SecA. Notably, specific amino acid residues, including ASP86, ARG182, ILE179, GLU143, ASP237, PRO101, and VAL84 from CiaR, LepC, OppC, SecA, SMU1784c, SpaR, and YidC2, respectively, exhibited significant interactions with their respective ligands. While the docking study indicated favorable binding energies, the MD simulations and ADMET studies underscored the substantial binding affinity and stability of the ligands with the target proteins. However, further in vitro studies are necessary to validate the efficacy of these top hits against S. mutans.
Collapse
Affiliation(s)
| | - Jayaprabhakaran Murugesan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India
| | - Ewa Babkiewicz
- Department of Hydrobiology, Faculty of Biology, University of Warsaw, 02-089 Warsaw, Poland
- Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| | - Piotr Maszczyk
- Department of Hydrobiology, Faculty of Biology, University of Warsaw, 02-089 Warsaw, Poland
| | - Murugesan Sankaranarayanan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani 333031, India
| | | | - Joseph Christina Rosy
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India
| | | | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India
| | - Vanavil Balakrishnan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India
| | - Krishnan Sundar
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil 626126, India
| |
Collapse
|
34
|
Liu C, Zhang H, Peng X, Blackledge MS, Furlani RE, Li H, Su Z, Melander RJ, Melander C, Michalek S, Wu H. Small Molecule Attenuates Bacterial Virulence by Targeting Conserved Response Regulator. mBio 2023; 14:e0013723. [PMID: 37074183 PMCID: PMC10294662 DOI: 10.1128/mbio.00137-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 04/20/2023] Open
Abstract
Antibiotic tolerance within a biofilm community presents a serious public health challenge. Here, we report the identification of a 2-aminoimidazole derivative that inhibits biofilm formation by two pathogenic Gram-positive bacteria, Streptococcus mutans and Staphylococcus aureus. In S. mutans, the compound binds to VicR, a key response regulator, at the N-terminal receiver domain, and concurrently inhibits expression of vicR and VicR-regulated genes, including the genes that encode the key biofilm matrix producing enzymes, Gtfs. The compound inhibits S. aureus biofilm formation via binding to a Staphylococcal VicR homolog. In addition, the inhibitor effectively attenuates S. mutans virulence in a rat model of dental caries. As the compound targets bacterial biofilms and virulence through a conserved transcriptional factor, it represents a promising new class of anti-infective agents that can be explored to prevent or treat a host of bacterial infections. IMPORTANCE Antibiotic resistance is a major public health issue due to the growing lack of effective anti-infective therapeutics. New alternatives to treat and prevent biofilm-driven microbial infections, which exhibit high tolerance to clinically available antibiotics, are urgently needed. We report the identification of a small molecule that inhibits biofilm formation by two important pathogenic Gram-positive bacteria, Streptococcus mutans and Staphylococcus aureus. The small molecule selectively targets a transcriptional regulator leading to attenuation of a biofilm regulatory cascade and concurrent reduction of bacterial virulence in vivo. As the regulator is highly conserved, the finding has broad implication for the development of antivirulence therapeutics that selectively target biofilms.
Collapse
Affiliation(s)
- Chang Liu
- Department of Pediatric Dentistry, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, USA
- Department of Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, USA
| | - Hua Zhang
- Department of Pediatric Dentistry, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, USA
- Department of Integrative Biomedical & Diagnostic Sciences, Oregon Health & Science University School of Dentistry, Portland, Oregon, USA
- Department of Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, USA
| | - Xian Peng
- Department of Pediatric Dentistry, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, USA
- Department of Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, USA
| | - Meghan S. Blackledge
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Robert E. Furlani
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Haoting Li
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Zhaoming Su
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Roberta J. Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Suzanne Michalek
- Department of Integrative Biomedical & Diagnostic Sciences, Oregon Health & Science University School of Dentistry, Portland, Oregon, USA
| | - Hui Wu
- Department of Pediatric Dentistry, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, USA
- Department of Integrative Biomedical & Diagnostic Sciences, Oregon Health & Science University School of Dentistry, Portland, Oregon, USA
- Department of Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, USA
| |
Collapse
|
35
|
Drummond IY, DePaolo A, Krieger M, Driscoll H, Eckstrom K, Spatafora GA. Small regulatory RNAs are mediators of the Streptococcus mutans SloR regulon. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543485. [PMID: 37398324 PMCID: PMC10312646 DOI: 10.1101/2023.06.02.543485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Dental caries is among the most prevalent chronic infectious diseases worldwide. Streptococcus mutans , the chief causative agent of caries, uses a 25 kDa manganese dependent SloR protein to coordinate the uptake of essential manganese with the transcription of its virulence attributes. Small non-coding RNAs (sRNAs) can either enhance or repress gene expression and reports in the literature ascribe an emerging role for sRNAs in the environmental stress response. Herein, we identify 18-50 nt sRNAs as mediators of the S. mutans SloR and manganese regulons. Specifically, the results of sRNA-seq revealed 56 sRNAs in S. mutans that were differentially transcribed in the SloR-proficient UA159 and SloR-deficient GMS584 strains, and 109 sRNAs that were differentially expressed in UA159 cells grown in the presence of low versus high manganese. We describe SmsR1532 and SmsR1785 as SloR- and/or manganese-responsive sRNAs that are processed from large transcripts, and that bind SloR directly in their promoter regions. The predicted targets of these sRNAs include regulators of metal ion transport, growth management via a toxin-antitoxin operon, and oxidative stress tolerance. These findings support a role for sRNAs in coordinating intracellular metal ion homeostasis with virulence gene control in an important oral cariogen. IMPORTANCE Small regulatory RNAs (sRNAs) are critical mediators of environmental signaling, particularly in bacterial cells under stress, but their role in Streptococcus mutans is poorly understood. S. mutans, the principal causative agent of dental caries, uses a 25 kDa manganese-dependent protein, called SloR, to coordinate the regulated uptake of essential metal ions with the transcription of its virulence genes. In the present study, we identified and characterize sRNAs that are both SloR- and manganese-responsive. Taken together, this research can elucidate the details of regulatory networks that engage sRNAs in an important oral pathogen, and that can enable the development of an effective anti-caries therapeutic.
Collapse
|
36
|
Martorano-Fernandes L, Goodwine JS, Ricomini-Filho AP, Nobile CJ, Del Bel Cury AA. Candida albicans Adhesins Als1 and Hwp1 Modulate Interactions with Streptococcus mutans. Microorganisms 2023; 11:1391. [PMID: 37374893 DOI: 10.3390/microorganisms11061391] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Candida albicans and Streptococcus mutans are known to synergistically interact with each other in the oral cavity. For example, glucosyltransferase B (GtfB), secreted by S. mutans, can bind to the C. albicans cell surface, promoting dual-species biofilm formation. However, the fungal factors mediating interactions with S. mutans are unknown. The C. albicans adhesins Als1, Als3, and Hwp1 are key players in C. albicans single-species biofilm formation, but their roles, if any, in interacting with S. mutans have not been assessed. Here, we investigated the roles of the C. albicans cell wall adhesins Als1, Als3, and Hwp1 on forming dual-species biofilms with S. mutans. We assessed the abilities of the C. albicans wild-type als1Δ/Δ, als3Δ/Δ, als1Δ/Δ/als3Δ/Δ, and hwp1Δ/Δ strains to form dual-species biofilms with S. mutans by measuring optical density, metabolic activity, cell enumeration, biomass, thickness, and architecture of the biofilms. We observed that the C. albicans wild-type strain formed enhanced dual-species biofilms in the presence of S. mutans in these different biofilm assays, confirming that C. albicans and S. mutans synergistically interact in the context of biofilms. Our results reveal that C. albicans Als1 and Hwp1 are major players in interacting with S. mutans, since dual-species biofilm formation was not enhanced when the als1Δ/Δ or hwp1Δ/Δ strains were cultured with S. mutans in dual-species biofilms. Als3, however, does not seem to play a clear role in interacting with S. mutans in dual-species biofilm formation. Overall, our data suggest that the C. albicans adhesins Als1 and Hwp1 function to modulate interactions with S. mutans and could be potential targets for future therapeutics.
Collapse
Affiliation(s)
- Loyse Martorano-Fernandes
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13414-903, SP, Brazil
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA 95343, USA
| | - James S Goodwine
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA 95343, USA
| | - Antônio Pedro Ricomini-Filho
- Department of Physiological Science, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13414-903, SP, Brazil
| | - Clarissa J Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA 95343, USA
- Health Sciences Research Institute, University of California Merced, Merced, CA 95343, USA
| | - Altair Antoninha Del Bel Cury
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba 13414-903, SP, Brazil
| |
Collapse
|
37
|
Liu J, Carmichael C, Hasturk H, Shi W, Bor B. Rapid specific detection of oral bacteria using Cas13-based SHERLOCK. J Oral Microbiol 2023; 15:2207336. [PMID: 37187674 PMCID: PMC10177689 DOI: 10.1080/20002297.2023.2207336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023] Open
Abstract
Decades of ongoing research has established that oral microbial communities play a role in oral diseases such as periodontitis and caries. Yet the detection of oral bacteria and the profiling of oral polymicrobial communities currently rely on methods that are costly, slow, and technically complex, such as qPCR or next-generation sequencing. For the widescale screening of oral microorganisms suitable for point-of-care settings, there exists the need for a low-cost, rapid detection technique. Here, we tailored the novel CRISPR-Cas-based assay SHERLOCK for the species-specific detection of oral bacteria. We developed a computational pipeline capable of generating constructs suitable for SHERLOCK and experimentally validated the detection of seven oral bacteria. We achieved detection within the single-molecule range that remained specific in the presence of off-target DNA found within saliva. Further, we adapted the assay for detecting target sequences directly from unprocessed saliva samples. The results of our detection, when tested on 30 healthy human saliva samples, fully aligned with 16S rRNA sequencing. Looking forward, this method of detecting oral bacteria is highly scalable and can be easily optimized for implementation at point-of-care settings.
Collapse
Affiliation(s)
- Jett Liu
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Camden Carmichael
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Hatice Hasturk
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA, USA
| | - Wenyuan Shi
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Batbileg Bor
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
38
|
Lei Y, Xu J, Pan M, Chen Y, Li X, Zhu W, Shu C, Fang T, Liao H, Luo Q, Li X. Construction of an antibacterial low-defect hybrid layer by facile PEI electrostatic assembly promotes dentin bonding. J Mater Chem B 2023; 11:335-344. [PMID: 36412982 DOI: 10.1039/d2tb01683d] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Dentin bonding is the most common form of human tissue repair among tissue-biomaterial adhesions, concerning billions of people's oral health worldwide. However, insufficient adhesive infiltration in the demineralized dentin matrix (DDM) always produces numerous defects in the bonding interface termed the hybrid layer, which causes high levels of bacteria-related secondary dental diseases, and less than 50% of the bonding lasts more than 5 years. Therefore, it is urgent and vital to construct an antibacterial low-defect hybrid layer to solve the durability-related problems. A DDM with a hydrogel-like surface formed by the hydration of highly-anionic non-collagenous proteins (NCPs) is firstly used as a template to electrostatically assemble polyethyleneimine (PEI). The formation of a stable antibacterial polyelectrolyte complex of PEI/NCPs rapidly eliminates NCP hydration capacity and significantly improves the infiltration of various adhesives. Simultaneously, both the PEI during the assembly and the PEI-assembled DDM can directly destroy a biofilm of S. Mutans on the DDM. Consequently, a long-term antibacterial and low-defect hybrid layer is successfully created, which greatly improves the bonding effectiveness. This helps to improve the clinical treatment of bacteria-based dental diseases and the tooth-restoration repair effect and prevent secondary dental diseases, having significance in clinical dentistry and providing insights for other tissue-biomaterial adhesions.
Collapse
Affiliation(s)
- Yuqing Lei
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310000, P. R. China. .,Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jiajia Xu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310000, P. R. China. .,Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengqi Pan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310000, P. R. China. .,Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Yadong Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310000, P. R. China. .,Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Xiaojun Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310000, P. R. China. .,Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Weipu Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Chang Shu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310000, P. R. China. .,Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Tianxiang Fang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hongbing Liao
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, College of Stomatology, Guangxi Medical University, Guilin 530021, China
| | - Qiaojie Luo
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310000, P. R. China. .,Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Xiaodong Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310000, P. R. China. .,Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| |
Collapse
|
39
|
Alomeir N, Zeng Y, Fadaak A, Wu TT, Malmstrom H, Xiao J. Effect of Nystatin on Candida albicans - Streptococcus mutans duo-species biofilms. Arch Oral Biol 2023; 145:105582. [PMID: 36395564 PMCID: PMC9729470 DOI: 10.1016/j.archoralbio.2022.105582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/23/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To assess the effect of Nystatin on Candida albicans and Streptococcus mutans duo-species biofilms using an in vitro cariogenic biofilm model. DESIGN Biofilms were formed on saliva-coated hydroxyapatite discs under high sugar challenge (1 % sucrose and 1 % glucose), with inoculation of 105CFU/ml S. mutans and 103CFU/ml C. albicans. Between 20 and 68 h, biofilms were treated with 28,000 IU Nystatin solution, 5 min/application, 4 times/day, to mimic the clinical application. Biofilm's three-dimensional structure was assessed using multi-photon confocal microscopy. The expression of C. albicans and S. mutans virulence genes was assessed via real-time PCR. Duplicate discs were used in 3 independent repeats. t-test and Mann-Whitney U test were used to compare outcomes between treatment and control group. RESULTS Nystatin treatment eliminated C. albicans in biofilms at 44 h. Nystatin-treated group had a significant reduction of biofilm dry-weight and reduced S. mutans abundance by 0.5 log CFU/ml at 44 and 68 h (p < 0.05). Worth noting that biomass distribution across the vertical layout was altered by Nystatin treatment, resulting in less volume on the substrate layers in Nystatin-treated biofilms compared to the control. Reduction of microcolonies size and volume was also observed in Nystatin-treated biofilms (p < 0.05). Nystatin-treated biofilms formed unique halo-shaped microcolonies with reduced core EPS coverage. Furthermore, Nystatin-treated biofilms had significant down-regulations of S. mutans gtfD and atpD genes (p < 0.05). CONCLUSIONS Nystatin application altered the formation and characteristics of C. albicans and S. mutans duo-species biofilms. Therefore, developing clinical regimens for preventing or treating dental caries from an antifungal perspective is warranted.
Collapse
Affiliation(s)
- Nora Alomeir
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, USA
| | - Yan Zeng
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, USA
| | - Ahmed Fadaak
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, USA
| | - Tong Tong Wu
- Department of Biostatistics and computational biology, University of Rochester Medical Center, Rochester, USA
| | - Hans Malmstrom
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, USA
| | - Jin Xiao
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, USA.
| |
Collapse
|
40
|
Sun Y, Chen H, Xu M, He L, Mao H, Yang S, Qiao X, Yang D. Exopolysaccharides metabolism and cariogenesis of Streptococcus mutans biofilm regulated by antisense vicK RNA. J Oral Microbiol 2023; 15:2204250. [PMID: 37138664 PMCID: PMC10150615 DOI: 10.1080/20002297.2023.2204250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Background Streptococcus mutans (S. mutans) is a pivotal cariogenic pathogen contributing to its multiple virulence factors, one of which is synthesizing exopolysaccharides (EPS). VicK, a sensor histidine kinase, plays a major role in regulating genes associated with EPS synthesis and adhesion. Here we first identified an antisense vicK RNA (ASvicK) bound with vicK into double-stranded RNA (dsRNA). Objective This study aims to investigate the effect and mechanism of ASvicK in the EPS metabolism and cariogenesis of S. mutans. Methods The phenotypes of biofilm were detected by scanning electron microscopy (SEM), gas chromatography-mass spectrometery (GC-MS) , gel permeation chromatography (GPC) , transcriptome analysis and Western blot. Co-immunoprecipitation (Co-ip) assay and enzyme activity experiment were adopted to investigate the mechanism of ASvicK regulation. Caries animal models were developed to study the relationship between ASvicK and cariogenicity of S. mutans. Results Overexpression of ASvicK can inhibit the growth of biofilm, reduce the production of EPS and alter genes and protein related to EPS metabolism. ASvicK can adsorb RNase III to regulate vicK and affect the cariogenicity of S. mutans. Conclusions ASvicK regulates vicK at the transcriptional and post-transcriptional levels, effectively inhibits EPS synthesis and biofilm formation and reduces its cariogenicity in vivo.
Collapse
Affiliation(s)
- Yuting Sun
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Hong Chen
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Mengmeng Xu
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Liwen He
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Hongchen Mao
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Shiyao Yang
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xin Qiao
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Deqin Yang
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- CONTACT Deqin Yang Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing4404100, China
| |
Collapse
|
41
|
Cornejo CF, Soken LJ, Salgado PA, Gliosca LA, Squassi AF. Detection of Streptococcus mutans and Streptococcus sobrinus and Their Association with Oral Microbiome Stressors in 6-18-month-old Infants. Int J Clin Pediatr Dent 2023; 16:68-73. [PMID: 37020757 PMCID: PMC10067995 DOI: 10.5005/jp-journals-10005-2489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Abstract
Aim To determine the presence of Streptococcus mutans (S. mutans) and Streptococcus sobrinus (S. sobrinus) and their association with extrinsic and intrinsic variables in 6-18-month-old infants. Methods This was an analytical, cross-sectional study of 65 6-18-month-old infants who visited the Centers for Early Childhood in Buenos Aires City. Three groups were established according to the presence of teeth-group I (GI)-edentulous infants, group II (GII)-infants with 1-8 teeth, and group III (GIII)-infants with 9-16 teeth. Data on the variables, diet, use of artificial teats, and oral hygiene were gathered using a self-administered questionnaire. An oral examination was performed according to the International Caries Detection and Assessment System (ICDAS II) criterion. A saliva sample was taken by aspiration with a sterile plastic syringe. Cariogenic Streptococci (CS) were counted using the adherence test in modified gold broth (AT-MGB). Molecular detection and quantification were performed by quantitative polymerase chain reaction (qPCR) (gtfB, gtfT, and tuf). Results A total of 12% of infants received oral hygiene, 38% used bottles, 30% used pacifiers, and 55% had sugar intake. S. sobrinus and S. mutans were detected in 57.1 and 28.6% of the children with caries, respectively. Groups I, II, and III had CS counts of log 2, 3.4, and 3.7, respectively. S. sobrinus was detected in 26.7% of GI, 52.9% of GII, and 85.7% of GIII, while S. mutans was detected in 13.3%, 35.3%, and 57.7%, respectively. Conclusion The prevalence of S. sobrinus was higher than S. mutans in all groups. The presence of CS was significantly associated with sugar intake. No association was found between S. mutans and S. sobrinus and the presence of caries, hygiene habits, or use of artificial teats. Clinical Significance This study supports the role of diet in developing a cariogenic biofilm in children under 2 years of age. How to cite this article Cornejo CF, Soken LJ, Salgado PA, et al. Detection of Streptococcus mutans and Streptococcus sobrinus and Their Association with Oral Microbiome Stressors in 6-18-month-old Infants. Int J Clin Pediatr Dent 2023;16(1):68-73.
Collapse
Affiliation(s)
- Celina F Cornejo
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Instituto de Investigaciones en Salud Publica, Buenos Aires, Argentina
| | - Luciana J Soken
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Microbiología y Parasitología, Laboratorio de Diagnostico Microbiológico y Molecular, Buenos Aires, Argentina
| | - Pablo A Salgado
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Instituto de Investigaciones en Salud Publica, Buenos Aires, Argentina
| | - Laura A Gliosca
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Microbiología y Parasitología, Laboratorio de Diagnostico Microbiológico y Molecular, Buenos Aires, Argentina
| | - Aldo F Squassi
- Universidad de Buenos Aires, Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Instituto de Investigaciones en Salud Publica, Buenos Aires, Argentina
| |
Collapse
|
42
|
Nagasawa R, Nomura N, Obana N. Identification of a Novel Gene Involved in Cell-to-cell Communication-induced Cell Death and eDNA Production in Streptococcus mutans. Microbes Environ 2023; 38:n/a. [PMID: 37302844 DOI: 10.1264/jsme2.me22085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023] Open
Abstract
Streptococcus mutans is a major caries-causing bacterium that forms firmly attached biofilms on tooth surfaces. Biofilm formation by S. mutans consists of polysaccharide-dependent and polysaccharide-independent processes. Among polysaccharide-independent processes, extracellular DNA (eDNA) mediates the initial attachment of cells to surfaces. We previously reported that the secreted peptide signal, competence-stimulating peptide (CSP) induced cell death in a subpopulation of cells, leading to autolysis-mediated eDNA release. The autolysin gene lytF, the expression of which is stimulated by CSP, has been shown to mediate CSP-dependent cell death, while cell death was not entirely abolished in the lytF deletion mutant, indicating the involvement of other factors. To identify novel genes involved in CSP-dependent cell death, we herein compared transcriptomes between live and dead cells derived from an isogenic population. The results obtained revealed the accumulation of several mRNAs in dead cells. The deletion of SMU_1553c, a putative bacteriocin gene, resulted in significant reductions in CSP-induced cell death and eDNA production levels from those in the parental strain. Moreover, in the double mutant strain of lytF and SMU_1553c, cell death and eDNA production in response to synthetic CSP were completely abolished under both planktonic and biofilm conditions. These results indicate that SMU_1553c is a novel cell death-related factor that contributes to CSP-dependent cell death and eDNA production.
Collapse
Affiliation(s)
- Ryo Nagasawa
- Graduate School of Life and Environmental Sciences, University of Tsukuba
| | - Nobuhiko Nomura
- Faculty of Life and Environmental Sciences, University of Tsukuba
- Microbiology Research Center for Sustainability, University of Tsukuba
| | - Nozomu Obana
- Microbiology Research Center for Sustainability, University of Tsukuba
- Faculty of Medicine, Transborder Medical Research Center, University of Tsukuba
| |
Collapse
|
43
|
Zhang Y, Li Z, Xu X, Peng X. Transposon mutagenesis in oral streptococcus. J Oral Microbiol 2022; 14:2104951. [PMID: 35903085 PMCID: PMC9318214 DOI: 10.1080/20002297.2022.2104951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Oral streptococci are gram-positive facultative anaerobic bacteria that are normal inhabitants of the human oral cavity and play an important role in maintaining oral microecological balance and pathogenesis. Transposon mutagenesis is an effective genetic manipulation strategy for studying the function of genomic features. In order to study cariogenic related genes and crucial biological element genes of oral Streptococcus, transposon mutagenesis was widely used to identify functional genes. With the advent of next-generation sequencing (NGS) technology and the development of transposon random mutation library construction methods, transposon insertion sequencing (TIS) came into being. Benefiting from high-throughput advances in NGS, TIS was able to evaluate the fitness contribution and essentiality of genetic features in the bacterial genome. The application of transposon mutagenesis, including TIS, to oral streptococci provided a massive amount of valuable detailed linkage data between genetic fitness and genetic backgrounds, further clarify the processes of colonization, virulence, and persistence and provides a more reliable basis for investigating relationships with host ecology and disease status. This review focuses on transposon mutagenesis, including TIS, and its applicability in oral streptococci.
Collapse
Affiliation(s)
- Yixin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, Sichuan, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhengyi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, Sichuan, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, Sichuan, China
- Department of Cariology and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xian Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu, Sichuan, China
| |
Collapse
|
44
|
McLellan LK, Anderson ME, Grossman AD. TnSmu1 is a functional integrative and conjugative element in Streptococcus mutans that when expressed causes growth arrest of host bacteria. Mol Microbiol 2022; 118:652-669. [PMID: 36268794 PMCID: PMC10098952 DOI: 10.1111/mmi.14992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 01/18/2023]
Abstract
Integrative and conjugative elements (ICEs) are major drivers of horizontal gene transfer in bacteria. They mediate their own transfer from host cells (donors) to recipients and allow bacteria to acquire new phenotypes, including pathogenic and metabolic capabilities and drug resistances. Streptococcus mutans, a major causative agent of dental caries, contains a putative ICE, TnSmu1, integrated at the 3' end of a leucyl tRNA gene. We found that TnSmu1 is a functional ICE, containing all the genes necessary for ICE function. It excised from the chromosome and excision was stimulated by DNA damage. We identified the DNA junctions generated by excision of TnSmu1, defined the ends of the element, and detected the extrachromosomal circle. We found that TnSmu1 can transfer from S. mutans donors to recipients when co-cultured on solid medium. The presence of TnSmu1 in recipients inhibited successful acquisition of another copy and this inhibition was mediated, at least in part, by the likely transcriptional repressor encoded by the element. Using microscopy to track individual cells, we found that activation of TnSmu1 caused an arrest of cell growth. Our results demonstrate that TnSmu1 is a functional ICE that affects the biology of its host cells.
Collapse
Affiliation(s)
- Lisa K McLellan
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mary E Anderson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Alan D Grossman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
45
|
Tickle ARH, Ledger EVK, Edwards AM. Human serum induces daptomycin tolerance in Enterococcus faecalis and viridans group streptococci. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 36748501 DOI: 10.1099/mic.0.001282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Daptomycin is a membrane-targeting lipopeptide antibiotic used in the treatment of infective endocarditis caused by multidrug-resistant Gram-positive bacteria such as Staphylococcus aureus, enterococci and viridans group streptococci. Despite demonstrating excellent in vitro activity and a low prevalence of resistant isolates, treatment failure is a significant concern, particularly for enterococcal infection. We have shown recently that human serum triggers daptomycin tolerance in S. aureus, but it was not clear if a similar phenotype occurred in other major infective endocarditis pathogens. We found that Enterococcus faecalis, Streptococcus gordonii or Streptococcus mutans grown under standard laboratory conditions were efficiently killed by daptomycin, whereas bacteria pre-incubated in human serum survived exposure to the antibiotic, with >99 % cells remaining viable. Incubation of enterococci or streptococci in serum led to peptidoglycan accumulation, as shown by increased incorporation of the fluorescent d-amino acid analogue HADA. Inhibition of peptidoglycan accumulation using the antibiotic fosfomycin resulted in a >tenfold reduction in serum-induced daptomycin tolerance, demonstrating the important contribution of the cell wall to the phenotype. We also identified a small contribution to daptomycin tolerance in E. faecalis from cardiolipin synthases, although this may reflect the inherent increased susceptibility of cardiolipin-deficient mutants. In summary, serum-induced daptomycin tolerance is a consistent phenomenon between Gram-positive infective endocarditis pathogens, but it may be mitigated using currently available antibiotic combination therapy.
Collapse
Affiliation(s)
- Alicia R H Tickle
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK.,Southmead Hospital, Southmead Road, Westbury-on-Trym, Bristol, Avon, BS10 5NB, UK
| | - Elizabeth V K Ledger
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
| | - Andrew M Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
| |
Collapse
|
46
|
Exploiting Conserved Quorum Sensing Signals in Streptococcus mutans and Streptococcus pneumoniae. Microorganisms 2022; 10:microorganisms10122386. [PMID: 36557639 PMCID: PMC9785397 DOI: 10.3390/microorganisms10122386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Bacterial species of the Streptococcus genera are considered either commensal bacteria or potential pathogens, according to their metabolic evolution and production of quorum sensing (QS)-controlled virulence factors. S. mutans, in particular, has become one of the best-studied examples of bacteria that are able to get along or cheat commensal species, even of the same genera. S. mutans and S. pneumoniae share homolog QS pathways and a competence stimulating peptide (CSP) for regulating bacteriocin production. Intriguingly, the abundance of S. pneumoniae and S. mutans alternates in complex microbial communities, thus opening the role for the fratricide communication of homolog QS systems. Since the inhibition of the QS has been proposed in treating bacterial infections, in this study, we designed and synthesized analogs of S. pneumoniae CSP with precise residual modifications. We reported that S. pneumoniae CSP analogs reduced the expression of genes involved in the QS of S. mutans and biofilm formation without affecting bacterial growth. The CSP analogs inhibited bacteriocin production in S. mutans, as reported by co-cultures with commensal bacteria of the oral cavity. The peptide CSP1AA, bearing substitutions in the residues involved in QS receptor recognition and activation, reported the most significant quorum-quenching activities. Our findings provide new insights into specific chemical drivers in the CSP sequences controlling the interconnection between S. mutans and S. pneumoniae. We think that the results reported in this study open the way for new therapeutic interventions in controlling the virulence factors in complex microbial communities such as the oral microbiota.
Collapse
|
47
|
Li Z, Wu Q, Zhang Y, Zhou X, Peng X. Systematic analysis of lysine malonylation in Streptococcus mutans. Front Cell Infect Microbiol 2022; 12:1078572. [PMID: 36519128 PMCID: PMC9742479 DOI: 10.3389/fcimb.2022.1078572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/11/2022] [Indexed: 11/29/2022] Open
Abstract
Protein lysine malonylation (Kmal) is a novel post-translational modification (PTM) that regulates various biological pathways such as energy metabolism and translation. Malonylation in prokaryotes, however, is still poorly understood. In this study, we performed a global Kmal analysis of the cariogenic organism Streptococcus mutans by combining antibody-based affinity enrichment and high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) analysis. Altogether, 392 malonyllysine sites in 159 proteins were identified. Subsequent bioinformatic analysis revealed that Kmal occurs in proteins involved in various metabolic pathways including translation machinery, energy metabolism, RNA degradation, and biosynthesis of various secondary metabolites. Quantitative analysis demonstrated that Kmal substrates were globally altered in the biofilm growth state compared to the planktonic growth state. Furthermore, a comparative analysis of the lysine malonylome of our study with previously determined lysine acetylome in S. mutans revealed that a small proportion of Kmal sites overlapped with acetylated sites, whereby suggesting that these two acylations have distinct functional implications. These results expand our knowledge of Kmal in prokaryotes, providing a resource for researching metabolic regulation of bacterial virulence and physiological functions by PTM.
Collapse
Affiliation(s)
- Zhengyi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qinrui Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xian Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Proton Pump Inhibitor Pantoprazole Modulates Intestinal Microbiota and Induces TLR4 Signaling and Fibrosis in Mouse Liver. Int J Mol Sci 2022; 23:ijms232213766. [PMID: 36430244 PMCID: PMC9693486 DOI: 10.3390/ijms232213766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/21/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
Proton pump inhibitors (PPIs) are one of the most prescribed drugs around the world. PPIs induce microbiota modulation such as obesity both in humans and in animal models. However, since PPIs can induce microbiota modulation despite the absence of a high-fat diet or weight gain, it is an interesting model to correlate microbiota modulation with the establishment of non-alcoholic fatty liver disease (NAFLD). We investigated the effect of pantoprazole treatment on TLR4 signaling and liver histology in C57BL/6J mice for 60 days, trying to correlate microbiota modulation with some aspects of liver injury. We performed glucose (GTT) and insulin (ITT) tolerance tests, serum lipopolysaccharide (LPS) dosage, liver histology, liver and intestine extraction for Western blot and qPCR. Fecal microbiota were investigated via metagenomics. Chronic treatment with pantoprazole induced microbiota modulation and impaired ileum barrier integrity, without an association with insulin resistance. Furthermore, increased circulating LPS and increased Toll-like receptor 4 (TLR4) and TGFβ downstream signaling may have an important role in the development of the observed liver microvesicular steatosis and fibrosis. Finally, this model of PPI-induced changes in microbiota might be useful to investigate liver microvesicular steatosis and fibrosis.
Collapse
|
49
|
Mansoor A, Khurshid Z, Khan MT, Mansoor E, Butt FA, Jamal A, Palma PJ. Medical and Dental Applications of Titania Nanoparticles: An Overview. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12203670. [PMID: 36296859 PMCID: PMC9611494 DOI: 10.3390/nano12203670] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/08/2022] [Accepted: 10/09/2022] [Indexed: 05/25/2023]
Abstract
Currently, titanium oxide (TiO2) nanoparticles are successfully employed in human food, drugs, cosmetics, advanced medicine, and dentistry because of their non-cytotoxic, non-allergic, and bio-compatible nature when used in direct close contact with the human body. These NPs are the most versatile oxides as a result of their acceptable chemical stability, lower cost, strong oxidation properties, high refractive index, and enhanced aesthetics. These NPs are fabricated by conventional (physical and chemical) methods and the latest biological methods (biological, green, and biological derivatives), with their advantages and disadvantages in this epoch. The significance of TiO2 NPs as a medical material includes drug delivery release, cancer therapy, orthopedic implants, biosensors, instruments, and devices, whereas their significance as a dental biomaterial involves dentifrices, oral antibacterial disinfectants, whitening agents, and adhesives. In addition, TiO2 NPs play an important role in orthodontics (wires and brackets), endodontics (sealers and obturating materials), maxillofacial surgeries (implants and bone plates), prosthodontics (veneers, crowns, bridges, and acrylic resin dentures), and restorative dentistry (GIC and composites).
Collapse
Affiliation(s)
- Afsheen Mansoor
- Department of Dental Material Sciences, School of Dentistry, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad 44080, Pakistan
| | - Zohaib Khurshid
- Department of Prosthodontics and Dental Implantology, College of Dentistry, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | - Muhammad Talal Khan
- Department of Dental Biomaterials, Bakhtawar Amin Medical and Dental College, Multan 60650, Pakistan;
| | - Emaan Mansoor
- Islamic International Dental College, Riphah International University, Islamabad 44000, Pakistan;
| | - Faaz Ahmad Butt
- Department of Materials Engineering, NED University of Engineering & Technology, Karachi 74200, Pakistan;
| | - Asif Jamal
- Department of Microbiology, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Paulo J. Palma
- Center for Innovation and Research in Oral Sciences (CIROS), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Institute of Endodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| |
Collapse
|
50
|
Li X, Zhai Z, Hao Y, Zhang M, Hou C, He J, Shi S, Zhao Z, Sang Y, Ren F, Wang R. The plasmid-encoded lactose operon plays a vital role in the acid production rate of Lacticaseibacillus casei during milk beverage fermentation. Front Microbiol 2022; 13:1016904. [PMID: 36386630 DOI: 10.3389/fmicb.2022.1016904if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/13/2022] [Indexed: 07/26/2024] Open
Abstract
Lacticaseibacillus casei is used extensively in the fermented milk-beverage industry as a starter culture. Acid production capacity during fermentation is the main criterion for evaluating starters although it is strain-dependent. In this study, the acid production rates of 114 L. casei strains were determined and then classified into high acid (HC), medium acid (MC), and low acid (LC) groups. Comparative genomics analysis found that the lac operon genes encoding the phosphoenolpyruvate-lactose phosphotransferase system (PTSLac) were located on plasmids in the HC strains; however, it is notable that the corresponding operons were located on the chromosome in LC strains. Real-time PCR analysis showed that the copy numbers of lac operon genes in HC strains were between 3.1 and 9.3. To investigate the relationship between copy number and acid production rate, the lac operon cluster of the HC group was constitutively expressed in LC strains. The resulting copy numbers of lac operon genes were between 15.8 and 18.1; phospho-β-galactosidase activity increased by 1.68-1.99-fold; and the acid production rates increased by 1.24-1.40-fold, which enhanced the utilization rate of lactose from 17.5 to 42.6% in the recombinant strains. The markedly increased expression of lac operon genes increased lactose catabolism and thereby increased the acid production rate of L. casei.
Collapse
Affiliation(s)
- Xiaoxia Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zhengyuan Zhai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanling Hao
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, China
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Caiyun Hou
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jingjing He
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, China
| | - Shaoqi Shi
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, China
| | - Zhi Zhao
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, China
| | - Yue Sang
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, China
| | - Fazheng Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, China
| | - Ran Wang
- Department of Nutrition and Health, Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, China Agricultural University, Beijing, China
| |
Collapse
|