1
|
Vieira A, Mateus C, Fonseca IM, Domingues F, Oleastro M, Ferreira S. The dual action of probiotic lactobacilli in suppressing virulence and survival of Arcobacter butzleri. Microb Pathog 2025; 204:107589. [PMID: 40239725 DOI: 10.1016/j.micpath.2025.107589] [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: 02/27/2025] [Revised: 03/28/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Arcobacter butzleri is a widely distributed foodborne and waterborne pathogen, increasingly recognized as an emerging enteropathogen. Understanding its survival mechanisms and interactions with probiotics is crucial for developing targeted intervention strategies. A. butzleri must withstand various hostile conditions to successfully colonize the gastrointestinal tract, including inhibition by probiotics, such as Limosilactobacillus reuteri, Lactobacillus acidophilus and Lactiplantibacillus plantarum. Thus, this study aimed to assess the survival of A. butzleri under acidic conditions and determine its minimum inhibitory concentration (MIC) for bile salts. Additionally, the antimicrobial potential of the lactobacilli strains was evaluated by analysing the effects of their culture-free supernatant (CFS) on A. butzleri growth, coculture interactions, and biofilm formation. The influence of lactobacilli on A. butzleri was further investigated through competition, displacement and exclusion assays using Caco-2 cell models. The results indicate that lactobacilli strains exhibit tolerance to acidic environments and physiological bile salt concentrations, whereas A. butzleri was more susceptible to acidic stress. The antagonistic effect of lactobacilli was evidenced by growth inhibition of A. butzleri in the presence of CFS or during coculture. However, CFS from certain lactobacilli strains was found to enhance biofilm formation, highlighting potential consequences. Furthermore, while lactobacilli did not demonstrate significant antagonistic effects in competition assays, they effectively displaced and excluded A. butzleri in the Caco-2 infection model. Overall, these findings suggest that probiotic lactobacilli can inhibit A. butzleri growth, yet their impact on its virulence remains uncertain. This underscores the need for strain-specific probiotic selection to effectively target this pathogen and emphasizes that not every probiotic contribute to the prevention of A. butzleri infections.
Collapse
Affiliation(s)
- Alexandre Vieira
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Cristiana Mateus
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Inês M Fonseca
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Fernanda Domingues
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Mónica Oleastro
- National Reference Laboratory for Gastrointestinal Infections, Department of Infectious Diseases, National Institute of Health Dr. Ricardo Jorge, Lisbon, Portugal
| | - Susana Ferreira
- RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
2
|
Kong L, Hu X, Xia D, Wu J, Zhao Y, Guo H, Zhang S, Qin C, Wang Y, Li L, Su Z, Zhu C, Xu S. Janus PEGylated CuS-engineered Lactobacillus casei combats biofilm infections via metabolic interference and innate immunomodulation. Biomaterials 2025; 317:123060. [PMID: 39736219 DOI: 10.1016/j.biomaterials.2024.123060] [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: 08/02/2024] [Revised: 11/25/2024] [Accepted: 12/25/2024] [Indexed: 01/01/2025]
Abstract
Bacterial implant-associated infections predominantly contribute to the failure of prosthesis implantation. The local biofilm microenvironment (BME), characterized by its hyperacidic condition and high hydrogen peroxide (H2O2) level, inhibits the host's immune response, thereby facilitating recurrent infections. Here, a Janus PEGylated CuS nanoparticle (CuPen) armed engineered Lactobacillus casei (L. casei) denoted as LC@CuPen, is proposed to interfere with bacterial metabolism and arouse macrophage antibiofilm function. Once LC@CuPen reached the BME, NIR irradiation-activated mild heat damages L. casei and biofilm structure. Meanwhile, the BME-responsive LC@CuPen can catalyze local H2O2 to produce toxic •OH, whereas in normal tissues, the effect of •OH production is greatly reduced due to the higher pH and lower H2O2 concentration. The released bacteriocin from damaged L. casei can destroy the bacterial membrane to enhance the penetration of •OH into damaged biofilm. Excessive •OH interferes with normal bacterial metabolism, resulting in reduced resistance of bacteria to heat stress. Finally, under the action of mild heat treatment, the bacterial biofilm lysed and died. Furthermore, the pathogen-associated molecular patterns (PAMPs) in LC@CuPen can induce M1 polarization of macrophages through NF-κB pathway and promote the release of inflammatory factors. Inflammatory factors enhance the migration of macrophages to the site of infection and phagocytose bacteria, thereby inhibiting the recurrence of infection. Generally, this engineered L. casei program presents a novel perspective for the treatment of bacterial implant-associated infections and serves as a valuable reference for future clinical applications of engineered probiotics.
Collapse
Affiliation(s)
- Lingtong Kong
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Xianli Hu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Demeng Xia
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jianghong Wu
- Department of Microbiology, College of Basic Medical Science, Department of Emergency, The Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China.
| | - Yangpeng Zhao
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Hua Guo
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Song Zhang
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Chun Qin
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Yanjun Wang
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Lei Li
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China
| | - Zheng Su
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Chen Zhu
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shuogui Xu
- Department of Orthopedics, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, 200433, China.
| |
Collapse
|
3
|
Pan T, Ren Y, Li J, Liao Y, Xing X. Polymicrobial detection and salivary metabolomics of children with early childhood caries. PeerJ 2025; 13:e19399. [PMID: 40352277 PMCID: PMC12063607 DOI: 10.7717/peerj.19399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Background Early childhood caries (ECC) has been proposed to be associated with various microorganisms and metabolites. This study aims to compare the prevalence of specific microbial species and salivary metabolomics profile in children with and without ECC, and to explore the correlation between salivary metabolites and targeted microbes. Method Five ml of unstimulated saliva was collected from 32 ECC and 22 caries-free children. Clinical indexed were recorded and questionnaires regarding oral health and dietary habits were obtained from the guardians. The presence of eight specific microbial species were examined using species-specific quantitative PCR (qPCR). Untargeted metabolomics was analyzed to identify key differential metabolites and pathways. Correlations among clinical, microbial, and metabolomic data were further explored. Results The prevalence of Scardovia wiggsiae (90.6%, P < 0.001), Streptococcus mutans (43.8%, P = 0.006), Streptococcus sobrinus (62.5%, P < 0.001), Ligilactobacillus salivarius (93.6%, P = 0.01) and Candida albicans (56.3%, P < 0.001) were significantly higher in the ECC group. The prevalence of ECC was higher in children with two targeted species present compared with children with one targeted species. Histidine metabolism and branched-chain amino acids degradation were activated in ECC group, while glyoxylate and dicarboxylate metabolism, purine and pyrimidine metabolism were inhibited. Histidine and glutathione metabolism was activated with enrichment of targeted microbial species, while linoleic acid metabolism and biotin metabolism was inhibited. The duration of each toothbrushing was a significant risk factor for ECC experience. Conclusion The prevalence of Scardovia wiggsiae, Streptococcus mutans, Streptococcus sobrinus and Candida albicans is higher in ECC children compared to caries-free children. Oral habits and salivary metabolites also vary between ECC and caries-free children.
Collapse
Affiliation(s)
- Ting Pan
- Department of Pediatric Dentistry, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, Jiangsu Province, China
| | - YuJia Ren
- Department of Pediatric Dentistry, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, Jiangsu Province, China
| | - JingYi Li
- Department of Pediatric Dentistry, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ying Liao
- Department of Pediatric Dentistry, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, Jiangsu Province, China
| | - XiangHui Xing
- Department of Pediatric Dentistry, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, Jiangsu Province, China
| |
Collapse
|
4
|
Kriswandini IL, Khoswanto C, Luthfi M, Tyas PR, Hermanto AC, Aljunaid MA. Formation of Streptococcus mutans Polymicrobial Biofilms in the Presence of Lactobacillus plantarum and Candida albicans. Eur J Dent 2025. [PMID: 40315863 DOI: 10.1055/s-0045-1806962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025] Open
Abstract
Dental caries is an infectious disease that develops through biofilm. Streptococcus mutans is a cariogenic bacterium that can be found in dental plaque. Streptococcus mutans regulates biofilm formation and communicate with other microbes through a process called quorum sensing. Dental caries prevention can be achieved by inhibiting quorum sensing. This study aimed to investigate the ability of Lactobacillus plantarum and Candida albicans to inhibit the formation of S. mutans polymicrobial biofilms. This study aims to investigate the ability of biofilm formation analyzed through the crystal violet (CV) assay and bacterial metabolic activity analyzed through the methylthiazol tetrazolium (MTT) assay. The bacteria used are S. mutans (serotype C), L. plantarum (FNCC 0020), and C. albicans.CV assay results show that in the presence of L. plantarum, biofilm formation in S. mutans decreases (9.5%). Meanwhile, the formation of S. mutans biofilms increased with the presence of C. albicans (28.8%). MTT assay results showed an increase in the metabolic activity of S. mutans in the presence of L. plantarum (20.2%) and C. albicans (19.4%). Lactobacillus plantarum can inhibit the formation of S. mutans biofilms, while C. albicans can increase S. mutans biofilms.
Collapse
Affiliation(s)
| | - Christian Khoswanto
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Muhammad Luthfi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Pinta Rahayuning Tyas
- Student of Dental Medicine Education Study Program, Faculty of Dental Medicine, Universitas Airlangga Surabaya, Indonesia
| | - Adelheid Chrissanda Hermanto
- Student of Dental Medicine Education Study Program, Faculty of Dental Medicine, Universitas Airlangga Surabaya, Indonesia
| | | |
Collapse
|
5
|
Park S, Shin J, Jun W, Lee D, Kim S, Ha DU, Im J, Han SH. Lipoteichoic acid from a canine probiotic strain Lacticaseibacillus rhamnosus possesses anti-biofilm capacity against clinically isolated canine periodontopathic Porphyromonas species. Microb Pathog 2025; 205:107660. [PMID: 40320055 DOI: 10.1016/j.micpath.2025.107660] [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: 10/15/2024] [Revised: 03/08/2025] [Accepted: 05/02/2025] [Indexed: 05/12/2025]
Abstract
Periodontitis is one of the most prevalent oral infectious diseases in canines which is mainly caused by periodontopathic bacteria such as Porphyromonas spp. Biofilm formation of periodontopathogens is closely related to the development of the disease, as it provides increased resistance against dental medicaments or host immunity. Although we recently demonstrated the anti-biofilm activity of Lactobacillus lipoteichoic acid (LTA), a major cell wall component of Gram-positive bacteria, against various human dental pathogens, the anti-biofilm effects of LTA from canine probiotics on canine periodontopathic Porphyromonas spp. have not been evaluated. Here, we investigated whether LTA purified from healthy canine-derived Lacticaseibacillus rhamnosus (Lr.LTA) affects biofilm formation and pre-formed biofilm of clinically isolated canine periodontopathic Porphyromonas spp., including Porphyromonas gulae, Porphyromonas macacae, and Porphyromonas canoris. We initially purified Lr.LTA through a serial application of butanol extraction, hydrophobic-interaction chromatography, and ion-exchange chromatography and confirmed that the resulting Lr.LTA was of high purity. Lr.LTA effectively suppressed the biofilm formation of P. gulae and P. canoris, but minimally P. macacae, without any effects on the bacterial growth. In addition, the inhibitory effects of Lr.LTA on biofilm formation of P. gulae and P. canoris were more potent than that of P. macacae. Lr.LTA also reduced the pre-formed biofilm of P. gulae and P. canoris, while it rarely affected that of P. macacae. These results suggest that Lr.LTA possesses the anti-biofilm capacity against canine periodontopathic Porphyromonas spp. and can be used as an effective anti-biofilm agent for the prevention and treatment of canine periodontitis caused by infection of Porphyromonas spp.
Collapse
Affiliation(s)
- Somin Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeongmin Shin
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Woohyung Jun
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongwook Lee
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - San Kim
- Bio Interactions Laboratory, BRD Corp., Hwaseong, 18471, Republic of Korea
| | - Dong Uk Ha
- Bio Interactions Laboratory, BRD Corp., Hwaseong, 18471, Republic of Korea
| | - Jintaek Im
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
6
|
Yue B, Gao R, Zhao L, Liu D, Lv C, Wang Z, Ai F, Zhang B, Yu Z, Geng X, Wang H, Wang K, Chen K, Liu C, Wang Z, Dou W. β-Glucuronidase-Expressing Lactobacillus reuteri Triggers Irinotecan Enterotoxicity Through Depleting the Regenerative Epithelial Stem/Progenitor Pool. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2411052. [PMID: 40285661 DOI: 10.1002/advs.202411052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/18/2025] [Indexed: 04/29/2025]
Abstract
Irinotecan (CPT11)-induced diarrhea affects 80-90% of cancer patients due to β-glucuronidase (GUS) converting 7-ethyl-10-hydroxycamptothecin glucuronide (SN38G) to 7-ethyl-10-hydroxycamptothecin (SN38). It remains unclear whether SN38 impacts the homeostasis between gut microbiota and mucosal stem cell niche. This study explores the crosstalk between gut microbiota and intestinal stem cells (ISCs) in intestinal mucositis triggered by CPT11 chemotherapy. CPT11-treated mice exhibited significant colon shortening, inflammatory infiltration, intestinal barrier dysfunction, and ISC impairment, which correlated with gut dysbiosis, enrichment of GUS-expressing bacteria, and intraluminal SN38 accumulation. In contrast, antidiarrheal (Xianglian pill) treatment alleviated SN38-induced enterotoxicity and reduced GUS-expressing bacterial populations. Microbiome profiling of clinical patients and mucositis mice revealed a strong correlation between CPT11/SN38 enterotoxicity and GUS-expressing bacteria, particularly Lactobacillus reuteri. PLS-PM modeling further linked L. reuteri to impaired epithelial regeneration, which is validated using a 3D intestinal organoid model. L. reuteri hindered ISC differentiation into secretory lineages within the organoids. Furthermore, L. reuteri colonization in mice exacerbated mucositis and disrupted epithelial differentiation, while its elimination ameliorated colitis symptoms and preserved crypt cell stemness. These findings suggest that selectively targeting GUS-expressing bacteria, particularly L. reuteri, to protect the regenerative epithelial stem/progenitor pool may serve as an effective strategy for mitigating CPT11-induced enterotoxicity.
Collapse
Affiliation(s)
- Bei Yue
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ruiyang Gao
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ling Zhao
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Donghui Liu
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Cheng Lv
- Centre for Chinese Herbal Medicine Drug Development Limited, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Ziyi Wang
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Fangbin Ai
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Beibei Zhang
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhilun Yu
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaolong Geng
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hao Wang
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Kang Wang
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Kaixian Chen
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chenghai Liu
- Department of Hepatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhengtao Wang
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wei Dou
- The MOE key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
7
|
Merino LE, Llana MN, Garófalo A, Trejo FM, Golowczyc MA. Antibiofilm properties of lactic acid bacteria and their metabolites against Salmonella enterica serotype Enteritidis on eggshell surface. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100384. [PMID: 40255247 PMCID: PMC12008649 DOI: 10.1016/j.crmicr.2025.100384] [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: 04/22/2025] Open
Abstract
Salmonella enteritidis (SE) is a pathogenic bacterium commonly found on the surface of eggshells. In this study, we investigated the biofilm formation of a specific strain of S. enteritidis, CIDCA 115 (SE 115) on eggshells. Additionally, we examined the impact of Lentilactobacillus kefiri strains 83113 and 8321, as well as Lactiplantibacillus plantarum 83114, and their metabolites present in the supernatant on the biofilm formation of SE 115. Scanning electron microscopy revealed that SE 115 formed a mature biofilm structure on the eggshell. During co-incubation and pre-incubation, lactic acid bacteria strains significantly reduced the formation of SE 115 biofilm (p < 0.05 and p < 0.01, respectively) compared to SE 115 grown alone. The cell-free supernatants of lactic acid bacteria also exhibited a reduction in SE 115 biofilm formation and modified its structure. Co-incubation with SE 115 induced alterations in the composition of biofilm matrix components, notably in the levels of fimbria curli and cellulose. The qPCR analysis revealed that, after 48 h of incubation, the expression of the csgD gene, a critical regulator of biofilm formation, remained unchanged compared to planktonic cells. However, genes associated with the production of biofilm matrix components, curli (csgA) and cellulose (bcsA), exhibited heightened expression in the presence of lactic acid bacteria compared to the planktonic state. This study highlights the potential of lactic acid bacteria strains and their metabolites as innovative strategies for managing Salmonella biofilm formation in the context of poultry production.
Collapse
Affiliation(s)
- Lina E. Merino
- Universidad Nacional de Hurlingham, Av. Gdor. Vergara 2222, Hurlingham, Buenos Aires, B1688GEZ, Argentina
- Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CICPBA), Argentina
| | - Mariángeles Noto Llana
- Instituto de investigaciones en Microbiología y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ailín Garófalo
- Instituto de investigaciones en Microbiología y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernando M. Trejo
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA, CONICET-CICPBA-UNLP), Facultad de Ciencias Exactas, UNLP, 47 y 116, La Plata, 1900, Argentina
- Universidad Nacional Arturo Jauretche - UNAJ, Av. Calchaquí 6200 (1888), Florencio Varela, Argentina
| | - Marina A. Golowczyc
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos (CIDCA, CONICET-CICPBA-UNLP), Facultad de Ciencias Exactas, UNLP, 47 y 116, La Plata, 1900, Argentina
| |
Collapse
|
8
|
Singh N, Gulhane RD, Singh A, Goel M, Udelal PP, Sangwan V, Sihag MK, Goel G, Panwar H, Puniya AK. Exploring the antimicrobial potential of lactobacilli against early-stage and mature biofilms of Staphylococcus aureus and Pseudomonas aeruginosa. Front Chem 2025; 13:1425666. [PMID: 40191159 PMCID: PMC11969340 DOI: 10.3389/fchem.2025.1425666] [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: 04/30/2024] [Accepted: 01/31/2025] [Indexed: 04/09/2025] Open
Abstract
Bacterial biofilms are dynamic, complex, and very adaptive, and they can cause health problems in both humans and animals while also posing a serious threat to various industries. This study explores the potential of cell-free preparations of lactobacilli isolated from breast milk (HM; n = 11) and infant fecal (IF; n = 15) samples to impact the growth of Staphylococcus aureus and Pseudomonas aeruginosa biofilms. The anti-biofilm activity of three distinct cell-free preparations, namely, untreated cell-free supernatant (CFS), pH-neutralized CFS (N-CFS), and heat-treated CFS (H-CFS), was examined against both early-stage and mature biofilms. The post-incubation strategy examined the impact on mature biofilms, while the co-incubation treatment assessed the impact of CFS on adhesion and initial colonization. Compared to post-incubation treatment (HM3, 67.12%), the CFSs exhibited greater inhibitory activity during co-incubation (IF9, 85.19%). Based on the findings, untreated CFS exhibited the most promising biofilm inactivation, although its activity was not completely lost upon pH neutralization and heat treatment. Treatment with H-CFSs and N-CFSs moderately reduced the population of S. aureus and P. aeruginosa bacterial cells within the biofilm by 40%-60%. Microscopic observations showed that after CFS treatment, the integrity of the biofilm conformation was disrupted. According to principal component analysis (PCA) (significance level at p < 0.05), the most promising anti-biofilm activity against both test pathogens was found in the CFS of Lacticaseibacillus paracasei HM1.
Collapse
Affiliation(s)
- Niharika Singh
- Department of Dairy Microbiology, College of Dairy and Food Science Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, Punjab, India
- Department of Biotechnology, VSB Engineering College, Karur, Tamil Nadu, India
| | - Rohini Devidas Gulhane
- Department of Dairy Microbiology, College of Dairy and Food Science Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, Punjab, India
| | - Anamika Singh
- Department of Dairy Microbiology, College of Dairy and Food Science Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, Punjab, India
| | - Maitri Goel
- Department of Dairy Microbiology, College of Dairy and Food Science Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, Punjab, India
| | - Pudke Payal Udelal
- Department of Dairy Microbiology, College of Dairy and Food Science Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, Punjab, India
| | - Vikas Sangwan
- Department of Dairy Microbiology, College of Dairy and Food Science Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, Punjab, India
| | - Manvesh Kumar Sihag
- Department of Dairy Chemistry, College of Dairy and Food Science Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, Punjab, India
| | - Gunjan Goel
- Department of Microbiology, School of Interdisciplinary and Applied Science, Central University of Haryana, Mahendergarh, India
| | - Harsh Panwar
- Department of Dairy Microbiology, College of Dairy and Food Science Technology, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Ludhiana, Punjab, India
| | - Anil Kumar Puniya
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| |
Collapse
|
9
|
Wang J, Ma Z, Xu Q, Wei B, Wang M, Liu Y, Tian Y, Zhang H, Xiao L, Zhong Y, Zou Y. Lacticaseibacillus rhamnosus OF44 with Potent Antimicrobial Activity: Evidence from the Complete Genome and Phenotypic Analysis. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10515-4. [PMID: 40106191 DOI: 10.1007/s12602-025-10515-4] [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] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
Lacticaseibacillus rhamnosus is extensively studied, with some strains widely applied for enhancing human health. Complete genome analysis is crucial for the functional exploration of probiotics. This study aimed to investigate the potential of L. rhamnosus OF44 (OF44) to promote human health through complete genome and phenotypic analysis. The complete genome sequence of OF44 was 2,978,769 bp, with 2791 CDSs and an average GC content of 47%. In vitro experiments demonstrated that OF44 had a broad carbon source fermentation capacity. Resistance gene analysis and simulated digestion tests confirmed the favorable tolerance of OF44 under harsh conditions, suggesting its potential as an oral supplement. OF44 possessed various potential genes related to bioactive substances with antimicrobial activity, such as antimicrobial peptides, lactic acid, hydrogen peroxide, and exopolysaccharides, most of which were detected in vitro. Further, OF44 exhibited significant growth inhibition capacities against pathogens from the gut, vagina, skin, and mouth, likely due to high co-aggregation with pathogens, multiple antimicrobial peptide clusters, and adhesin gene clusters. Additionally, oral administration of OF44 was found to reduce the pH and inflammation levels in the vaginal microenvironment of rats with bacterial vaginosis. Therefore, OF44 exhibited probiotic properties in improving reproductive tract bacterial infections by modulating vaginal microbiota balance and inhibiting pathogen growth. In summary, this study provided a new perspective on the application of OF44 as a supplement in the food and pharmaceutical fields.
Collapse
Affiliation(s)
- Jinhong Wang
- BGI Research, Shenzhen, 518083, China
- College of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Zhihui Ma
- BGI Precision Nutrition (Shenzhen) Technology Co., Ltd, Shenzhen, 518083, China
| | - Qianyue Xu
- BGI Research, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Benliang Wei
- BGI Precision Nutrition (Shenzhen) Technology Co., Ltd, Shenzhen, 518083, China
| | - Mengmeng Wang
- BGI Research, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanhong Liu
- BGI Precision Nutrition (Shenzhen) Technology Co., Ltd, Shenzhen, 518083, China
| | - Yu Tian
- BGI Research, Shenzhen, 518083, China
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Haifeng Zhang
- BGI Precision Nutrition (Shenzhen) Technology Co., Ltd, Shenzhen, 518083, China
| | - Liang Xiao
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China
- State Key Laboratory of Genome and Multi-Omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Yiyi Zhong
- BGI Precision Nutrition (Shenzhen) Technology Co., Ltd, Shenzhen, 518083, China.
| | - Yuanqiang Zou
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China.
- State Key Laboratory of Genome and Multi-Omics Technologies, BGI Research, Shenzhen, 518083, China.
| |
Collapse
|
10
|
Ma N, Yang W, Chen B, Bao M, Li Y, Wang M, Yang X, Liu J, Wang C, Qiu L. Exploration of the primary antibiofilm substance and mechanism employed by Lactobacillus salivarius ATCC 11741 to inhibit biofilm of Streptococcus mutans. Front Cell Infect Microbiol 2025; 15:1535539. [PMID: 40134782 PMCID: PMC11933110 DOI: 10.3389/fcimb.2025.1535539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/14/2025] [Indexed: 03/27/2025] Open
Abstract
Introduction Lactobacillus salivarius serves as a probiotic potentially capable of preventing dental caries both in vitro and in vivo. This study focused on understanding the key antibiofilm agents and the mechanisms of action of the Lactobacilli supernatant against Streptococcus mutans. Methods Streptococcus mutans biofilm was constructed and the cell-free supernatant of Lactobacillus salivarius was added. After the biofilm was collected, RNA-seq and qRT-PCR were then performed to get gene information. The influence of temperature, pH and other factors on the supernatant were measured and non-targeted metabolome analysis was performed to analyze the effective components. Results The findings indicated that the supernatant derived from Lactobacillus salivarius could inhibit the biofilm formation of Streptococcus mutans at different times. Through transcriptome analysis, we discovered that the cell-free supernatant reduced biofilm formation, by suppressing phosphoenolpyruvate-dependent phosphotransferase systems along with two ATP-binding cassette transporters, rather than directly affecting the genes that code for glucosyltransferases; additionally, the supernatant was observed to diminish the expression of genes linked to two-component systems, polyketides/non-ribosomal peptides, acid stress response, quorum sensing, and exopolysaccharide formation. Non-targeted LC-MS/MS analysis was employed to discover a variety of potential active compounds present in the cellular filtrate of Lactobacillus salivarius that hinder the growth of S. mutans, including phenyllactic acid, sorbitol, and honokiol. Discussion In summary, our findings support the evaluation of Lactobacillus salivarius as a promising oral probiotic aimed at hindering the formation of biofilms by cariogenic pathogens and the development of dental caries.
Collapse
Affiliation(s)
- Nan Ma
- Department of Periodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
| | - Wei Yang
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Pedodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Bairu Chen
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Meihua Bao
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yimin Li
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Meng Wang
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xiaopeng Yang
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Pedodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Junyi Liu
- Jinzhou Medical University, Jinzhou, China
| | - Chengyue Wang
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Lihong Qiu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
11
|
Ammar HA, Samy R, Reda FM, Hassanein WA. Essential oils and Lactobacillus metabolites as alternative antibiofilm agents against foodborne bacteria and molecular analysis of biofilm regulatory genes. Sci Rep 2025; 15:7576. [PMID: 40038354 PMCID: PMC11880504 DOI: 10.1038/s41598-025-89998-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The formation of biofilm by foodborne pathogens increases the risk of foodborne diseases, resulting in major health risks. Research on strategies for eliminating biofilm formation by foodborne pathogens is urgently needed. Therefore, the objective of this study was to construct a new technique for controlling foodborne bacteria and inhibiting the biosynthesis of biofilm via using natural products. The essential orange oil (EOO) and cell-free filtrate of Lactobacillus pentosus RS2 were used as antibacterial and antibiofilm agents against B. cereus RS1, the strongest biofilm-forming strain. The mixture of cell-free filtrate (CFF) and EOO (CFF/EOO) was the best antibiofilm agent under all tested conditions. The minimal inhibitory concentration (MIC) test revealed that 400 μl ml-1 CFF and 16 μl ml-1 EOO completely inhibited the growth of B. cereus. The treatment of three commercial surfaces with CFF/EOO resulted in a high reduction in biofilm synthesis, with adhesion percentages of 33.3, 36.3, and 40.8% on stainless steel, aluminum foil, and aluminum, respectively. The aluminum surface had the greatest adhesion with B. cereus RS1 among the three tested surfaces. These results were confirmed by expression analysis of three essential coding genes, sinR, calY, and spo0A, participating in biofilm formation in B. cereus. The biofilm-negative regulator gene sinR was overexpressed, whereas the biofilm-positive regulator genes calY and spo0A were down-expressed in B. cereus RS1 after treatment with antibiofilm agents, compared with those in the untreated sample. This study revealed that CFF/EOO was more effective at activating sinR (2.099 ± 0.167-fold increase) and suppressing calY and spo0A (0.314 ± 0.058 and0.238 ± 0.04-fold decrease, respectively) compared to control. This result confirmed the biochemical estimation of biofilm formation in B. cereus after treatment with all the experimental agents. The EOO and CFF of L. pentosus RS2 can be used as strong antibacterial and antibiofilm agents against foodborne bacteria. These products reduced the biofilm formation on trade surfaces affecting the expression of three essential biofilm regulatory genes. This study considered novel research concerning the potential antibiofilm activity of EOO combined with CFF of L. pentosus and the molecular analysis of genes regulating biofilm production under stress of CFF/EOO.
Collapse
Affiliation(s)
- Hala A Ammar
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt.
| | - Ragda Samy
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Fifi M Reda
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Wesam A Hassanein
- Botany and Microbiology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| |
Collapse
|
12
|
Dawan J, Zhang S, Ahn J. Recent Advances in Biofilm Control Technologies for the Food Industry. Antibiotics (Basel) 2025; 14:254. [PMID: 40149064 PMCID: PMC11939704 DOI: 10.3390/antibiotics14030254] [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: 01/28/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Biofilms remain a major challenge in the food industry due to the increased resistance of foodborne pathogens to antimicrobial agents and food processing stresses, leading to food contamination and significant health risks. Their resistance to preservation techniques, antimicrobial treatments, and processing conditions increases concerns regarding food safety. This review discusses recent developments in physical, chemical, and surface modification strategies to control and remove biofilms in food processing environments. Physical methods, such as thermal treatments, electric fields, and ultrasonic systems, have demonstrated their efficacy in disrupting biofilm structure and improving disinfection processes. Chemical treatments, including the use of sanitizers, disinfectants, acidulants, and enzymes, provide targeted approaches to degrade biofilm matrices and inhibit bacterial adhesion. Furthermore, surface modifications of food contact materials provide innovative solutions for preventing biofilm formation and enhancing food safety. These cutting-edge strategies not only improve food safety but also reduce contamination risk in food processing facilities. The review highlights the mechanisms, efficacy, and applicability of these techniques, emphasizing their potential to mitigate biofilm-associated risks and ensure food quality and safety.
Collapse
Affiliation(s)
- Jirapat Dawan
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea; (J.D.); (S.Z.)
| | - Song Zhang
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea; (J.D.); (S.Z.)
| | - Juhee Ahn
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea; (J.D.); (S.Z.)
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| |
Collapse
|
13
|
Liu TH, Hsu YH, Hsiao RY, Cheng MC, Leu YL, Tsai TY. Improved mechanism of the bioactive compounds isolated from Lactobacillus plantarum TWK10-fermented soymilk in periodontal disease. Food Funct 2025; 16:1517-1533. [PMID: 39903172 DOI: 10.1039/d4fo04049j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Lactic acid bacteria (LAB) possess immunomodulatory, antioxidant, and antimicrobial properties, making them promising for oral health applications. This study investigated the antibacterial and anti-biofilm activities of TWK10-fermented soymilk extract (TWK10FSEE) against Porphyromonas gingivalis (Pg) and Aggregatibacter actinomycetemcomitans (Aa). TWK10FSEE disrupted the cell membrane integrity, altered membrane permeability, and inhibited the growth of these periodontal pathogens. In a drip-flow biofilm reactor simulating the oral environment, TWK10FSEE regulated biofilm-related gene expression, reduced fimbriae and extracellular polysaccharide production, and retarded biofilm formation. Structural identification revealed succinic acid (SA) and a mixture of daidzein and genistein (MDG) as the primary antimicrobial components. Both SA and MDG effectively inhibited Pg and Aa growth and biofilm formation. TWK10FSEE shows potential as a functional ingredient for periodontal health.
Collapse
Affiliation(s)
- Te-Hua Liu
- Department of Food Science, Fu Jen Catholic, University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City 242062, Taiwan, Republic of China.
| | - Ya-Hsiang Hsu
- Department of Food Science, Fu Jen Catholic, University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City 242062, Taiwan, Republic of China.
| | - Ru-Yun Hsiao
- Department of Food Science, Fu Jen Catholic, University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City 242062, Taiwan, Republic of China.
| | - Meng-Chun Cheng
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei 10462, Taiwan, Republic of China
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, No.259, Wenhua 1st Rd., Guishan Dist., Taoyuan City 33302, Taiwan, Republic of China
- Biobank, Chang Gung Memorial Hospital, No. 5, Fuxing St., Guishan Dist., Taoyuan City 33305, Taiwan, Republic of China
| | - Tsung-Yu Tsai
- Department of Food Science, Fu Jen Catholic, University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City 242062, Taiwan, Republic of China.
| |
Collapse
|
14
|
Heidari F, Heboyan A, Rokaya D, Fernandes GVO, Heidari M, Banakar M, Zafar MS. Postbiotics and Dental Caries: A Systematic Review. Clin Exp Dent Res 2025; 11:e70114. [PMID: 40066530 PMCID: PMC11894266 DOI: 10.1002/cre2.70114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVE This systematic review aimed to evaluate the current evidence regarding the impact of postbiotics on dental caries, focusing on the effectiveness of postbiotic interventions in caries prevention, mechanisms of action, optimal dosages, and administration protocols. METHODS A literature search was conducted across PubMed/MEDLINE, Scopus, Web of Science, and the Cochrane Library. Eligible studies included randomized controlled trials, quasi-experimental, observational, and in vitro studies. The selection followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. A qualitative synthesis was performed due to heterogeneity in study designs and outcomes. RESULTS Twenty-one studies were included (18 in vitro and three randomized controlled trials). Postbiotics derived from various Lactobacillus species demonstrated inhibitory effects on Streptococcus mutans growth, biofilm formation, and virulence gene expression. Proposed mechanisms include direct antimicrobial activity, inhibition of bacterial adhesion, disruption of biofilm formation, modulation of immune responses, and pH buffering. After postbiotic interventions, human trials showed reduced salivary S. mutans counts and increased salivary pH. CONCLUSIONS Postbiotics offer a promising novel approach to dental caries prevention by targeting cariogenic bacteria and modulating the oral microbiome through multiple mechanisms. Compared to probiotics, postbiotics present additional advantages, including enhanced safety, stability, and ease of incorporation into oral care products.
Collapse
Affiliation(s)
- Faezeh Heidari
- Department of Pediatric Dentistry, Faculty of DentistryShahed UniversityTehranIran
| | - Artak Heboyan
- Department of Research AnalyticsSaveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha UniversityChennaiIndia
- Department of Prosthodontics, Faculty of StomatologyYerevan State Medical University after Mkhitar HeratsiYerevanArmenia
- Department of Prosthodontics, School of DentistryTehran University of Medical SciencesTehranIran
| | - Dinesh Rokaya
- Clinical Sciences Department, College of DentistryAjman UniversityAjmanUnited Arab Emirates
- Center of Medical and Bio‐Allied Health Sciences ResearchAjman UniversityAjmanUnited Arab Emirates
| | | | - Mobina Heidari
- Student Research Committee, School of DentistryShahid Sadoughi University of Medical SciencesYazdIran
| | - Morteza Banakar
- Department of Research AnalyticsSaveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha UniversityChennaiIndia
- Health Policy Research Center, Institute of HealthShiraz University of Medical SciencesShirazIran
- Dental Research Center, Dentistry Research InstituteTehran University of Medical SciencesTehranIran
| | - Muhammad Sohail Zafar
- Clinical Sciences Department, College of DentistryAjman UniversityAjmanUnited Arab Emirates
- Center of Medical and Bio‐Allied Health Sciences ResearchAjman UniversityAjmanUnited Arab Emirates
- School of Dentistry, University of JordanAmmanJordan
| |
Collapse
|
15
|
Fu D, Shu X, Yao L, Zhou G, Ji M, Liao G, Zhu Y, Zou L. Unveiling the dual nature of Lactobacillus: from cariogenic threat to probiotic protector-a critical review with bibliometric analysis. FRONTIERS IN ORAL HEALTH 2025; 6:1535233. [PMID: 39959355 PMCID: PMC11825810 DOI: 10.3389/froh.2025.1535233] [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: 11/27/2024] [Accepted: 01/13/2025] [Indexed: 02/18/2025] Open
Abstract
Introduction Dental caries is a prevalent oral disease with a multifactorial etiology. Lactobacillus has been implicated in caries progression on account of its acidogenic properties; On the other hand, they constitute one of the potential probiotic strategies for preventing dental caries. This complex relationship renders the relationship between Lactobacillus and dental caries remains ambiguous. Methods The Web of Science core collections (WoSCC) were searched to acquire articles relevant to Lactobacillus and dental caries. After retrieval and manual screening, publications were analyzed by VOSviewer. Results Sweden, the US, and China, which have been the center of international cooperation, have produced the most publications in the research area. Caries Research is the main counterpart journal in the field. "Dental caries", "Streptococcus mutans", "Lactobacilli", "Probiotics", and "Children" have been commonly used as keywords. Discussion Based on bibliometric analysis, this study reviews the relationship between lactobacilli and dental caries, emphasizing their dual roles. The detection rate of lactobacilli is closely associated with the incidence and severity of dental caries. However, under specific environmental conditions, these bacteria also exhibit potential probiotic properties that may aid in the prevention of dental caries. Additionally, Lactobacillus is strongly associated with early childhood caries, a specific type of caries.
Collapse
Affiliation(s)
- Di Fu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China
| | - Xingyue Shu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China
| | - Lin Yao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China
| | - Ge Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China
| | - Mengzhen Ji
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, Sichuan, China
| | - Ga Liao
- Department of Information Management, Department of Stomatology Informatics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunwo Zhu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Geriatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
16
|
Sangha JS, Gogulancea V, Curtis TP, Jakubovics NS, Barrett P, Metris A, Ofiţeru ID. Advancing dental biofilm models: the integral role of pH in predicting S. mutans colonization. mSphere 2025; 10:e0074324. [PMID: 39660862 PMCID: PMC11774048 DOI: 10.1128/msphere.00743-24] [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: 09/03/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
Mathematical models can provide insights into complex interactions and dynamics within microbial communities to complement and extend experimental laboratory approaches. For dental biofilms, they can give a basis for evaluating biofilm growth or the transition from health to disease. We have developed mathematical models to simulate the transition toward a cariogenic microbial biofilm, modeled as the overgrowth of Streptococcus mutans within a five-species dental community. This work builds on experimental data from a continuous flow reactor with hydroxyapatite coupons for biofilm growth, in a chemically defined medium with varying concentrations of glucose and lactic acid. The biofilms formed on the coupons were simulated using individual-based models (IbMs), with bacterial growth modeled using experimentally measured kinetic parameters. The IbM assumes that the maximum theoretical growth yield for biomass is dependent on the local concentration of reactants and products, while the growth rates were described using traditional Monod equations. We have simulated all the conditions studied experimentally, considering different initial relative abundance of the five species, and also different initial clustering in the biofilm. The simulation results only reproduced the experimental dominance of S. mutans at high glucose concentration after we considered the species-specific effect of pH on growth rates. This highlights the significance of the aciduric property of S. mutans in the development of caries. Our study demonstrates the potential of combining in vitro and in silico studies to gain a new understanding of the factors that influence dental biofilm dynamics.IMPORTANCEWe have developed in silico models able to reproduce the relative abundance measured in vitro in the synthetic dental biofilm communities growing in a chemically defined medium. The advantage of this combination of in vitro and in silico models is that we can study the influence of one parameter at a time and aim for direct validation. Our work demonstrates the utility of individual-based models for simulating diverse conditions affecting dental biofilm scenarios, such as the frequency of glucose intake, sucrose pulsing, or integration of pathogenic or probiotic species. Although in silico models are reductionist approaches, they have the advantage of not being limited in the scenarios they can test by the ethical consideration of an in vivo system, thus significantly contributing to dental biofilm research.
Collapse
Affiliation(s)
- Jay S. Sangha
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Valentina Gogulancea
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Thomas P. Curtis
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nicholas S. Jakubovics
- Faculty of Medical Sciences, Newcastle University, School of Dental Sciences, Newcastle upon Tyne, United Kingdom
| | - Paul Barrett
- Safety and Environmental Assurance Centre, Unilever, Bedfordshire, United Kingdom
| | - Aline Metris
- Safety and Environmental Assurance Centre, Unilever, Bedfordshire, United Kingdom
| | - Irina D. Ofiţeru
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
17
|
Vernon JJ. Modulation of the Human Microbiome: Probiotics, Prebiotics, and Microbial Transplants. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1472:277-294. [PMID: 40111698 DOI: 10.1007/978-3-031-79146-8_17] [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: 03/22/2025]
Abstract
The balance between health and disease is intrinsically linked to the interactions between microbial communities and the host. This complex environment of antagonism and synergy involves both prokaryotic and eukaryotic cells, whose collaborative metabolic pathways and immunomodulatory elements influence system homeostasis. As with the gut and other niches, the oral microbiome has the capacity to affect distal host sites. The ability to manipulate this environment holds the potential to impact local and systemic disease.With the increasing threat of antimicrobial resistance, novel approaches to reduce the burden of disease are essential. The use of probiotics and prebiotics is one such strategy. Probiotics introduce non-pathogenic bacteria into the environment to compete with pathogens for nutrients and attachment sites, or to produce metabolites that counteract disease aetiologies. Prebiotic compounds enhance the growth of health-associated organisms, offering additional benefits, whilst a conjunctive approach with probiotics potentially holds even greater promise. Though widely studied in the gastrointestinal context, their potential for treating oral diseases, such as dental caries and periodontitis, is less understood. Additionally, the use of microbial transplantations has demonstrated efficacy in other areas, reducing systemic inflammation and recolonising with commensal bacteria. Here we evaluate their use in the oral context and their modulatory impact on overall health.In this chapter, we discuss how pro- and prebiotic strategies seek to modulate both the oral and gut environments to promote oral health and prevent disease. We assess novel approaches for utilising health-associated microorganisms to combat oral disorders, either administered locally in the mouth or imparting influence through immune modulation via the oral-gut axis. By examining available clinical trial data, we aim to further understand the intricacies involved in this discipline. Furthermore, we consider the challenges facing the research community, including optimal candidate organism/compound selection and colonisation retention, as well as considerations for future research.
Collapse
Affiliation(s)
- Jon J Vernon
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK.
| |
Collapse
|
18
|
Yang W, Jiang M, Chen B, Jiang K, Ma N, Li Y, Wang M, Bao M, Wang C, Yang X. Study the effect of Lactobacillus plantarum ATCC 14917 for caries prevention and anti-obesity. Front Nutr 2024; 11:1511660. [PMID: 39777074 PMCID: PMC11703752 DOI: 10.3389/fnut.2024.1511660] [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: 10/20/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction A complicated scenario where "multiple disease threats coexist and multiple health influencing factors are intertwined" is demonstrated by the fact that dental caries, obesity myopia and scoliosis have emerged as global public health issues. The problem of diseases co-existing in living things can be resolved by using probiotics. Lactobacillus plantarum, has gained attention recently due to its probiotic properties, useful traits, and potential medical applications. Objective Examining the anti-obesity and anti-caries effects of L. plantarum ATCC 14917 on dental caries and obese rat models caused by a high-fat and high-sugar diet is the aim of this study. Method In vitro, we assessed the L. plantarum strain's probiotic properties, such as its antibacterial activity and ability to build biofilms, to determine its ability to inhibit Streptococcus mutans. Prior to the in vivo experiment, the subsist test for L. plantarum ATCC 14917 was carried out by mimicking its capacity to lower blood sugar and blood lipid levels as well as its tolerance to gastrointestinal disorders. In order to assess the health promotion effect of L. plantarum in vivo. Three-week-old rats were fed a high-sugar, high-fat diet for 8 weeks. They were split into three groups: the control group (Control), the caries and obesity group (CA _OB) and the caries and obesity treated with L. plantarum ATCC14917 group (LP). L. plantarum ATCC 14917 was applied during the experiment, and the associated indices were then thoroughly assessed. These included the use of Mirco-CT to calculate the enamel volume, the staining of liver and fat cell sections, serological analysis, and 16S rRNA sequencing of feces. Results It was proved that the L. plantarum could inhibit the proliferation of S. mutans and remove dental plaque biofilm in time, which showed the remarkable effects of anti-caries in vitro. The demineralization rate of enamel decreased by 44.10% due to the inhibition of acid production by pathogenic bacteria. Moreover, In intestinal and stomach juice simulations, L. plantarum has a high survival rate. The characteristics of bacterial activity in a wide range of pH could degrade triglycerides and glucose in vitro smoothly. The LP group demonstrated it by reducing animal weight, serum biochemical indices, and HE-stained adipocytes as compared to the CA_OB group. 16S rRNA sequencing data showed that a high-fat and high-sugar diet induced the imbalance of intestinal flora, which showed an increase in microbial abundance, including unclassified_o__Clostridia_UCG-014, unclassified_f__Oscillospiraceae, Turicibacter, unclassified_f__Lachnospiraceae, Clostridium_sensu_stricto_1. After the intervention of L. plantarum, the number of Lactobacillus, Limosilactobacillus, unclassified_f__Muribaculaceae, Blautia, Faecalibaculum increased significantly. Conclusion Therefore, L. plantarum ATCC 14917 performed the potential of reducing tooth decay and controlling weight gain by a single strain. Support the management of dental caries and obesity, and establish a foundation for future functional food research and development.
Collapse
Affiliation(s)
- Wei Yang
- Department of Pedodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
| | - Mingxin Jiang
- Department of Micro-endodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Bairu Chen
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Kongzhao Jiang
- Department of Pedodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Nan Ma
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yimin Li
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Meng Wang
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Meihua Bao
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chengyue Wang
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xiaopeng Yang
- Department of Pedodontics, Affiliated Stomatology Hospital of Jinzhou Medical University, Jinzhou, China
- Collaborative Innovation Center for Health Promotion of Children and Adolescents of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
19
|
Haghighi FH, Farsiani H. Is Lactococcus lactis a Suitable Candidate for Use as a Vaccine Delivery System Against Helicobacter pylori? Curr Microbiol 2024; 82:30. [PMID: 39643816 DOI: 10.1007/s00284-024-03994-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/15/2024] [Indexed: 12/09/2024]
Abstract
Helicobacter pylori was described in 1979. This bacterium, which thrives in the harsh conditions of the stomach, is typically acquired during childhood and can remain colonized for life. Approximately, 90% of the global population is affected, and H. pylori is linked to various conditions, including gastritis, peptic ulcers, lymphoproliferative gastric lymphoma, and even gastric cancer. Currently, antibiotics are the primary treatment method, but the associated challenges of antibiotic use have led to the consideration of oral vaccination as a viable preventive measure against this infection. However, the stomach's harsh environment characterized by its acidic conditions and numerous proteolytic enzymes poses significant obstacles to the development and effectiveness of oral vaccines. To address these challenges, researchers have proposed and evaluated several delivery systems. One of the most promising options is the use of probiotics. Among the various probiotics, Lactococcus lactis stands out as a suitable candidate for oral vaccine delivery against H. pylori due to the advancements in genetic engineering that have been applied to it. This review article discusses the limitations of current treatment strategies and rationalizes the shift toward vaccination, particularly oral vaccination for this infection. It also explores the advantages and challenges of using probiotic bacteria, with a focus on L. lactis as a delivery system. Ultimately, despite the existing challenges, L. lactis continues to be recognized as a promising delivery system. Nonetheless, further research is essential to fully assess its effectiveness and address the challenges associated with this approach.
Collapse
Affiliation(s)
- Faria Hasanzadeh Haghighi
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Farsiani
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Azadi-Square, Medical Campus, Mashhad, 9177948564, Iran.
| |
Collapse
|
20
|
Banakar M, Fernandes GVO, Etemad‐Moghadam S, Frankenberger R, Pourhajibagher M, Mehran M, Yazdi MH, Haghgoo R, Alaeddini M. The strategic role of biotics in dental caries prevention: A scoping review. Food Sci Nutr 2024; 12:8651-8674. [PMID: 39620008 PMCID: PMC11606839 DOI: 10.1002/fsn3.4473] [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: 01/22/2024] [Revised: 07/26/2024] [Accepted: 09/06/2024] [Indexed: 01/31/2025] Open
Abstract
Dental caries is a global oral health issue that is prevalent and preventable. Biotics (probiotics, prebiotics, symbiotics, and postbiotics) are recommended as low-cost methods for preventing dental caries. This scoping review aimed to critically review the scientific evidence concerning the role of biotics in caries prevention and maintaining oral health benefits. A systematic search was conducted in several databases from 2012 onward, using specific keywords. The search resulted in 69 articles. While there is limited research on the mechanism of biotics in preventing caries, numerous studies have investigated the impacts of probiotics on decreasing caries risk factors. Probiotics can reduce cariogenic bacteria, reduce acidogenic bacteria, increase pH, and produce antimicrobial compounds. Probiotics can be used as a therapeutic approach to manage caries by restoring eubiosis at the host-microbial interface, which may not be accomplished with traditional therapies. Its positive effect on reducing dental caries is influenced by the choice of potent probiotic strains, appropriate dosage, treatment period, vehicle, and microbial interaction with the host. Specific oral bacteria have also been shown to utilize prebiotics such as urea and arginine, increasing pH levels. This highlights the potential of combining prebiotic and probiotic bacteria for caries prevention. In addition, this review is focused on bacterial-derived compounds, namely postbiotics, due to their valuable effects in preventing caries. Biotics have demonstrated potential in preventing dental caries and maintaining oral health. Further research is needed to optimize their use and explore the potential of postbiotics for caries prevention.
Collapse
Affiliation(s)
- Morteza Banakar
- Dental Research Center, Dentistry Research InstituteTehran University of Medical SciencesTehranIran
- Department of Pediatric Dentistry, Faculty of DentistryShahed UniversityTehranIran
| | | | - Shahroo Etemad‐Moghadam
- Dental Research Center, Dentistry Research InstituteTehran University of Medical SciencesTehranIran
| | - Roland Frankenberger
- Department of Operative Dentistry and Endodontics, Dental SchoolUniversity of Marburg and University Medical Center Giessen and MarburgMarburgGermany
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research InstituteTehran University of Medical SciencesTehranIran
| | - Majid Mehran
- Department of Pediatric Dentistry, Faculty of DentistryShahed UniversityTehranIran
| | | | - Roza Haghgoo
- Department of Pediatric Dentistry, Faculty of DentistryShahed UniversityTehranIran
| | - Mojgan Alaeddini
- Dental Research Center, Dentistry Research InstituteTehran University of Medical SciencesTehranIran
| |
Collapse
|
21
|
Li IC, Lee YL, Li TJ, Tsai YS, Chen YL, Chen CC. Whole-Genome Sequencing of Three Lactiplantibacillus plantarum Strains Reveals Potential Metabolites for Boosting Host Immunity Safely. J Microbiol Biotechnol 2024; 34:2079-2090. [PMID: 39263794 PMCID: PMC11540610 DOI: 10.4014/jmb.2402.02013] [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: 02/08/2024] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 09/13/2024]
Abstract
In response to the growing demand for immune-related products, this study evaluated the safety and immune-modulating potential of three newly discovered Lactiplantibacillus plantarum strains (GKM3, GKK1, and GKD7) through toxicity tests and whole-genome sequencing. Safety evaluations, including the analysis of antimicrobial resistance genes, virulence factors, plasmids, and prophages, classified these strains as safe for human consumption. Acute oral toxicity tests further supported their safety. To evaluate their immune-modulating potential, dendritic cells were exposed to these strains, and the secretion of key cytokines (IFN-β and IL-12) was measured. Among the strains, GKK1 exhibited the highest enhancement of IFN-β and IL-12 production, suggesting its potential as an immune-stimulating probiotic. Bioinformatics analysis revealed potential metabolic pathways and secondary metabolites, including predicted bacteriocins, associated with immune modulation. The presence of a nitrate reductase region in the GKK1 strain indicated its ability to produce nitric oxide, a critical molecule involved in immune regulation and host defense. The presence of glucorhamnan-related gene clusters in GKK1 also suggested immune-enhancing effects. Nitrate reductase expression was confirmed using qPCR, with the highest levels detected in GKK1. Moreover, this study is the first to show an anti-inflammatory effect of plantaricin A, linked to its presence in strain GKM3 and its potential therapeutic applications due to sequence similarity to known anti-inflammatory peptides. Overall, these three L. plantarum strains demonstrated a safe profile and GKK1 showed potential as an immunity-enhancing probiotic. However, additional investigation is required to confirm the involvement of specific metabolic pathways, secondary metabolites, and bacteriocins in immune responses.
Collapse
Affiliation(s)
- I-Chen Li
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Tsung-Ju Li
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - You-Shan Tsai
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Yen-Lien Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei City 104, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei City 106, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City 320, Taiwan
| |
Collapse
|
22
|
Huang X, Bao J, Yang M, Li Y, Liu Y, Zhai Y. The role of Lactobacillus plantarum in oral health: a review of current studies. J Oral Microbiol 2024; 16:2411815. [PMID: 39444695 PMCID: PMC11497578 DOI: 10.1080/20002297.2024.2411815] [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: 01/25/2024] [Revised: 07/09/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024] Open
Abstract
Background Oral non-communicable diseases, particularly dental caries and periodontal disease, impose a significant global health burden. The underlying microbial dysbiosis is a prominent factor, driving interest in strategies that promote a balanced oral microbiome. Lactobacillus plantarum, a gram-positive lactic acid bacterium known for its adaptability, has gained attention for its potential to enhance oral health. Recent studies have explored the use of probiotic L. plantarum in managing dental caries, periodontal disease, and apical periodontitis. However, a comprehensive review on its effects in this context is still lacking. Aims This narrative review evaluates current literature on L. plantarum's role in promoting oral health and highlights areas for future research. Content In general, the utilization of L. plantarum in managing non-communicable biofilm-dependent oral diseases is promising, but additional investigations are warranted. Key areas for future study include: exploring its mechanisms of action, identifying optimal strains or strain combinations of L. plantarum, determining effective delivery methods and dosages, developing commercial antibacterial agents from L. plantarum, and addressing safety considerations related to its use in oral care.
Collapse
Affiliation(s)
- Xinyan Huang
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | - Jianhang Bao
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| | - Mingzhen Yang
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| | - Yingying Li
- Orthopedic Department, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Youwen Liu
- Orthopedic Department, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Yuankun Zhai
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| |
Collapse
|
23
|
Cheng J, Cho JH, Suh JW. Characterization of Human Breast Milk-Derived Limosilactobacillus reuteri MBHC 10138 with Respect to Purine Degradation, Anti-Biofilm, and Anti-Lipid Accumulation Activities. Antibiotics (Basel) 2024; 13:964. [PMID: 39452230 PMCID: PMC11504937 DOI: 10.3390/antibiotics13100964] [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: 09/05/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Human breast milk is a valuable source of potential probiotic candidates. The bacteria isolated from human breast milk play an important role in the development of the infant gut microbiota, exhibiting diverse biological functions. Methods: In this study, Limosilactobacillus reuteri MBHC 10138 isolated from breast milk was characterized in terms of its probiotic safety characteristics and potential efficacy in hyperuricemia, obesity, lipid liver, and dental caries, conditions which Korean consumers seek to manage using probiotics. Results: Strain MBHC 10138 demonstrated a lack of D-lactate and biogenic amine production as well as a lack of bile salt deconjugation and hemolytic activity. It also exhibited susceptibility to common antibiotics, tolerance to simulated oral-gastric-intestinal conditions, and superior biological activity compared to three L. reuteri reference strains, including KACC 11452 and MJ-1, isolated from feces, and a commercial strain isolated from human breast milk. Notably, L. reuteri MBHC 10138 showed high capabilities in assimilating guanosine (69.48%), inosine (81.92%), and adenosine (95.8%), strongly inhibited 92.74% of biofilm formation by Streptococcus mutans, and reduced lipid accumulation by 32% in HepG2 cells. Conclusions: These findings suggest that strain MBHC 10138, isolated from human breast milk, has potential to be developed as a probiotic for managing hyperuricemia, obesity, and dental caries after appropriate in vivo studies.
Collapse
Affiliation(s)
| | | | - Joo-Won Suh
- Microbio Healthcare, Co., Ltd., Yongin 17058, Republic of Korea; (J.C.); (J.-H.C.)
| |
Collapse
|
24
|
Wang B, Zhang C, Shi C, Zhai T, Zhu J, Wei D, Shen J, Liu Z, Jia K, Zhao L. Mechanisms of oral microflora in Parkinson's disease. Behav Brain Res 2024; 474:115200. [PMID: 39134178 DOI: 10.1016/j.bbr.2024.115200] [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: 03/28/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease with complex pathogenesis and no effective treatment. Recent studies have shown that dysbiosis of the oral microflora is closely related to the development of PD. The abnormally distributed oral microflora of PD patients cause degenerative damage and necrosis of dopamine neurons by releasing their own components and metabolites, intervening in the oral-gut-brain axis, crossing the biofilm, inducing iron dysregulation, activating inter-microflora interactions, and through the mediation of saliva,ultimately influencing the development of the disease. This article reviews the structure of oral microflora in patients with PD, the mechanism of development of PD caused by oral microflora, and the potential value of targeting oral microflora in developing a new strategy for PD prevention, diagnosis and treatment.
Collapse
Affiliation(s)
- Bingbing Wang
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Can Zhang
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Caizhen Shi
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Tianyu Zhai
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Jinghui Zhu
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Dongmin Wei
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Juan Shen
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Zehao Liu
- Medical School of Yan'an University, Yan'an, Shaanxi, China
| | - Kunpeng Jia
- Yan'an University Affiliated Hospital, Yan'an, Shaanxi, China.
| | - Lin Zhao
- Medical School of Yan'an University, Yan'an, Shaanxi, China.
| |
Collapse
|
25
|
Atta L, Mushtaq M, Siddiqui AR, Khalid A, Ul-Haq Z. Targeting glucosyltransferases to combat dental caries: Current perspectives and future prospects. Int J Biol Macromol 2024; 278:134645. [PMID: 39128764 DOI: 10.1016/j.ijbiomac.2024.134645] [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: 05/15/2024] [Revised: 07/23/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The emergence of antimicrobial resistance within bacterial communities poses formidable challenges to existing therapeutic strategies aimed at mitigating biofilm-mediated infections. Recent advancements in this domain have spurred the development of targeted antimicrobial agents, designed to selectively eradicate the primary etiological agents while preserving the beneficial microbial diversity of the oral cavity. Targeting glucosyltransferases (GTFs), which play crucial roles in dental biofilm formation, offers a precise strategy to inhibit extracellular polysaccharide synthesis without compromising oral microbiota. This review article delves into the intricate mechanisms underlying dental caries, with a specific focus on the role of GTFs, enzymes produced by S. mutans. It further provides an overview of current research on GTF inhibitors, exploring their mechanisms of action, efficacy, and potential applications in clinical practice. Furthermore, it discusses the challenges and opportunities in the development of novel GTF inhibitors, emphasizing the need for innovative approaches to combat biofilm-mediated oral diseases effectively.
Collapse
Affiliation(s)
- Lubna Atta
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Mamona Mushtaq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Ali Raza Siddiqui
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Assad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
26
|
Yang X, Che T, Tian S, Zhang Y, Zheng Y, Zhang Y, Zhang X, Wu Z. A Living Microecological Hydrogel with Microbiota Remodeling and Immune Reinstatement for Diabetic Wound Healing. Adv Healthc Mater 2024; 13:e2400856. [PMID: 38744431 DOI: 10.1002/adhm.202400856] [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: 03/06/2024] [Revised: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Dysregulated skin microbiota and compromised immune responses are the major etiological factors for non-healing diabetic wounds. Current antibacterial strategies fail to orchestrate immune responses and indiscriminately eradicate bacteria at the wound site, exacerbating the imbalance of microbiota. Drawing inspiration from the beneficial impacts that probiotics possess on microbiota, a living microecological hydrogel containing Lactobacillus plantarum and fructooligosaccharide (LP/FOS@Gel) is formulated to remodel dysregulated skin microbiota and reinstate compromised immune responses, cultivating a conducive environment for optimal wound healing. LP/FOS@Gel acts as an "evocator," skillfully integrating the skin microecology, promoting the proliferation of Lactobacillus, Ralstonia, Muribaculum, Bacillus, and Allobaculum, while eradicating colonized pathogenic bacteria. Concurrently, LP/FOS@Gel continuously generates lactic acid to elicit a reparative macrophage response and impede the activation of the nuclear factor kappa-B pathway, effectively alleviating inflammation. As an intelligent microecological system, LP/FOS@Gel reinstates the skin's sovereignty during the healing process and effectively orchestrates the harmonious dialogue between the host immune system and microorganisms, thereby fostering the healing of diabetic infectious wounds. These remarkable attributes render LP/FOS@Gel highly advantageous for pragmatic clinical applications.
Collapse
Affiliation(s)
- Xiaopeng Yang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
| | - Tingting Che
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
| | - Shasha Tian
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
| | - Yuanyuan Zhang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
| | - Yin Zheng
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of Functional Polymer Materials, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of Functional Polymer Materials, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhongming Wu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
- Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300134, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Jinan Key Laboratory of Translational Medicine on Metabolic Diseases, Shandong Institute of Endocrine and Metabolic Diseases, Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Jinan, Shandong, 250012, China
| |
Collapse
|
27
|
Algburi AR, Jassim SM, Popov IV, Weeks R, Chikindas ML. Lactobacillus acidophilus VB1 co-aggregates and inhibits biofilm formation of chronic otitis media-associated pathogens. Braz J Microbiol 2024; 55:2581-2592. [PMID: 38789905 PMCID: PMC11405553 DOI: 10.1007/s42770-024-01363-5] [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: 01/23/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
This study aims to evaluate the antibacterial activity of Lactobacillus acidophilus, alone and in combination with ciprofloxacin, against otitis media-associated bacteria. L. acidophilus cells were isolated from Vitalactic B (VB), a commercially available probiotic product containing two lactobacilli species, L. acidophilus and Lactiplantibacillus (formerly Lactobacillus) plantarum. The pathogenic bacterial samples were provided by Al-Shams Medical Laboratory (Baqubah, Iraq). Bacterial identification and antibiotic susceptibility testing for 16 antibiotics were performed using the VITEK2 system. The minimum inhibitory concentration of ciprofloxacin was also determined. The antimicrobial activity of L. acidophilus VB1 cell-free supernatant (La-CFS) was evaluated alone and in combination with ciprofloxacin using a checkerboard assay. Our data showed significant differences in the synergistic activity when La-CFS was combined with ciprofloxacin, in comparison to the use of each compound alone, against Pseudomonas aeruginosa SM17 and Proteus mirabilis SM42. However, an antagonistic effect was observed for the combination against Staphylococcus aureus SM23 and Klebsiella pneumoniae SM9. L. acidophilus VB1 was shown to significantly co-aggregate with the pathogenic bacteria, and the highest co-aggregation percentage was observed after 24 h of incubation. The anti-biofilm activities of CFS and biosurfactant (BS) of L. acidophilus VB1 were evaluated, and we found that the minimum biofilm inhibitory concentration that inhibits 50% of bacterial biofilm (MBIC50) of La-CFS was significantly lower than MBIC50 of La-BS against the tested pathogenic bacterial species. Lactobacillus acidophilus, isolated from Vitane Vitalactic B capsules, demonstrated promising antibacterial and anti-biofilm activities against otitis media pathogens, highlighting its potential as an effective complementary/alternative therapeutic strategy to control bacterial ear infections.
Collapse
Affiliation(s)
- Ammar R Algburi
- Department of Microbiology, Veterinary Medicine College, University of Diyala, Baqubah, Iraq
| | - Shireen M Jassim
- Alkhalis Section for Primary Care/Thoracic and Respiratory Diseases Unit, Alkhalis, Iraq
| | - Igor V Popov
- Centre for Healthy Eating and Food Innovation, Maastricht University-Campus Venlo, Venlo, The Netherlands.
- Agrobiotechnology Center and Faculty "Bioengineering and Veterinary Medicine", Don State Technical University, Rostov-On-Don, Russia.
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Federal Territory Sirius, Sirius University of Science and Technology, Sochi, Russian Federation.
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA
| | - Michael L Chikindas
- Agrobiotechnology Center and Faculty "Bioengineering and Veterinary Medicine", Don State Technical University, Rostov-On-Don, Russia
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
28
|
Saini P, Ayyanna R, Kumar R, Bhowmick SK, Bhaskar V, Dey B. Restriction of growth and biofilm formation of ESKAPE pathogens by caprine gut-derived probiotic bacteria. Front Microbiol 2024; 15:1428808. [PMID: 39135871 PMCID: PMC11317286 DOI: 10.3389/fmicb.2024.1428808] [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: 05/07/2024] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
The accelerated rise in antimicrobial resistance (AMR) poses a significant global health risk, necessitating the exploration of alternative strategies to combat pathogenic infections. Biofilm-related infections that are unresponsive to standard antibiotics often require the use of higher-order antimicrobials with toxic side effects and the potential to disrupt the microbiome. Probiotic therapy, with its diverse benefits and inherent safety, is emerging as a promising approach to prevent and treat various infections, and as an alternative to antibiotic therapy. In this study, we isolated novel probiotic bacteria from the gut of domestic goats (Capra hircus) and evaluated their antimicrobial and anti-biofilm activities against the 'ESKAPE' group of pathogens. We performed comprehensive microbiological, biochemical, and molecular characterizations, including analysis of the 16S-rRNA gene V1-V3 region and the 16S-23S ISR region, on 20 caprine gut-derived lactic acid bacteria (LAB). Among these, six selected Lactobacillus isolates demonstrated substantial biofilm formation under anaerobic conditions and exhibited robust cell surface hydrophobicity and autoaggregation, and epithelial cell adhesion properties highlighting their superior enteric colonization capability. Notably, these Lactobacillus isolates exhibited broad-spectrum growth inhibitory and anti-biofilm properties against 'ESKAPE' pathogens. Additionally, the Lactobacillus isolates were susceptible to antibiotics listed by the European Food Safety Authority (EFSA) within the prescribed Minimum Inhibitory Concentration limits, suggesting their safety as feed additives. The remarkable probiotic characteristics exhibited by the caprine gut-derived Lactobacillus isolates in this study strongly endorse their potential as compelling alternatives to antibiotics and direct-fed microbial (DFM) feed supplements in the livestock industry, addressing the escalating need for antibiotic-free animal products.
Collapse
Affiliation(s)
- Prerna Saini
- National Institute of Animal Biotechnology, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Repally Ayyanna
- National Institute of Animal Biotechnology, Hyderabad, India
| | - Rishi Kumar
- National Institute of Animal Biotechnology, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Sayan Kumar Bhowmick
- National Institute of Animal Biotechnology, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Vinay Bhaskar
- National Institute of Animal Biotechnology, Hyderabad, India
| | - Bappaditya Dey
- National Institute of Animal Biotechnology, Hyderabad, India
- Regional Centre for Biotechnology, Faridabad, India
| |
Collapse
|
29
|
Mukherjee K, Rahman A, Chakraborty J, Pakrashi S. Isolation and characterization of mercury and multidrug-resistant Citrobacter freundii strains from tannery effluents in Kolkata, India. Arch Microbiol 2024; 206:362. [PMID: 39066800 DOI: 10.1007/s00203-024-04090-5] [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: 04/23/2024] [Revised: 07/14/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024]
Abstract
Mercury (Hg) is one of the most potent toxic heavy metals that distresses livestock, humans, and ecological health. Owing to uncontrolled exposure to untreated tannery industrial effluents, metals such as Hg are increasing in nature and are, therefore, becoming a global concern. As a result, understanding the thriving microflora in that severe condition and their characteristics becomes immensely important. During the course of this study, two Hg-resistant bacteria were isolated from tannery wastewater effluents from leather factories in Kolkata, India, which were able to tolerate 2.211 × 10- 3 M (600 µg/ml) Hg. 16 S rDNA analysis revealed strong sequence homology with Citrobacter freundii, were named as BNC22A and BNC22C for this study. In addition they showed high tolerance to nickel (Ni) and Chromium (Cr) at 6.31 × 10- 3 M (1500 µg/ml) and 6.792 × 10- 3 M (2000 µg/ml) respectively. However, both the isolates were sensitive to arsenic (As) and cadmium (Cd). Furthermore, their antibiotic sensitivity profiles reveal a concerning trend towards resistance to multiple drugs. Overuse and misuse of antibiotics in healthcare systems and agriculture has been identified as two of the main reasons for the decline in efficacy of antibiotics. Though their ability to produce lipase makes them industrially potent organisms, their competence to resist several antibiotics and metals that are toxic makes this study immensely relevant. In addition, their ability to negate heavy metal toxicity makes them potential candidates for bioremediation. Finally, the green mung bean seed germination test showed a significant favourable effect of BNC22A and BNC22C against Hg-stimulated toxicity.
Collapse
Affiliation(s)
- Kushal Mukherjee
- Department of Microbiology, Bidhannagar College, Kolkata, West Bengal, 700064, India
| | - Abdul Rahman
- Institute of Genomics and Integrative Biology, New Delhi, India
| | - Joyeeta Chakraborty
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, NH 12, Haringhata, 741249, India
| | - Sourav Pakrashi
- Department of Microbiology, Bidhannagar College, Kolkata, West Bengal, 700064, India.
| |
Collapse
|
30
|
Sandhu S, Sharma V, Kumar S, Rai N, Chand P. Quantifying variations associated with dental caries reveals disparity in effect allele frequencies across diverse populations. BMC Genom Data 2024; 25:50. [PMID: 38831280 PMCID: PMC11149341 DOI: 10.1186/s12863-024-01215-z] [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: 10/11/2023] [Accepted: 03/03/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Dental caries (DC) is a multifaceted oral condition influenced by genetic and environmental factors. Recent advancements in genotyping and sequencing technologies, such as Genome-Wide Association Studies (GWAS) have helped researchers to identify numerous genetic variants associated with DC, but their prevalence and significance across diverse global populations remain poorly understood as most of the studies were conducted in European populations, and very few were conducted in Asians specifically in Indians. AIM This study aimed to evaluate the genetic affinity of effect alleles associated with DC to understand the genetic relationship between global populations with respect to the Indian context. METHODOLOGY This present study used an empirical approach in which variants associated with DC susceptibility were selected. These variants were identified and annotated using the GWAS summary. The genetic affinity was evaluated using Fst. RESULTS The effect of allele frequencies among different populations was examined, revealing variations in allele distribution. African populations exhibited higher frequencies of specific risk alleles, whereas East Asian and European populations displayed distinct profiles. South Asian populations showed a unique genetic cluster. CONCLUSION Our study emphasises the complex genetic landscape of DC and highlights the need for population-specific research as well as validation of GWAS-identified markers in Indians before defining them as established candidate genes.
Collapse
Affiliation(s)
- Sangram Sandhu
- Ancient DNA Lab, Birbal Sahni Institute of Palaeosciences, 226607, Lucknow, Uttar Pradesh, India
- Department of Prosthodontics, Faculty of Dental Sciences, King George's Medical University, 226003, Lucknow, India
| | - Varun Sharma
- NMC Genetics India Pvt Ltd, 122002, Gurugram, Haryana, India
| | - Sachin Kumar
- Ancient DNA Lab, Birbal Sahni Institute of Palaeosciences, 226607, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), 201002, Ghaziabad, India
| | - Niraj Rai
- Ancient DNA Lab, Birbal Sahni Institute of Palaeosciences, 226607, Lucknow, Uttar Pradesh, India.
- Academy of Scientific and Innovative Research (AcSIR), 201002, Ghaziabad, India.
| | - Pooran Chand
- Department of Prosthodontics, Faculty of Dental Sciences, King George's Medical University, 226003, Lucknow, India.
| |
Collapse
|
31
|
Puca V, Marinacci B, Pellegrini B, Campanile F, Santagati M, Grande R. Biofilm and bacterial membrane vesicles: recent advances. Expert Opin Ther Pat 2024; 34:475-491. [PMID: 38578180 DOI: 10.1080/13543776.2024.2338101] [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: 11/14/2023] [Accepted: 03/21/2024] [Indexed: 04/06/2024]
Abstract
INTRODUCTION Bacterial Membrane Vesicles (MVs) play important roles in cell-to-cell communication and transport of several molecules. Such structures are essential components of Extracellular Polymeric Substances (EPS) biofilm matrix of many bacterial species displaying a structural function and a role in virulence and pathogenesis. AREAS COVERED In this review were included original articles from the last ten years by searching the keywords 'biofilm' and 'vesicles' on PUBMED and Scopus databases. The articles available in literature mainly describe a positive correlation between bacterial MVs and biofilms formation. The research on Espacenet and Google Patent databases underlines the available patents related to the application of both biofilm MVs and planktonic MVs in inhibiting biofilm formation. EXPERT OPINION This review covers and analyzes recent advances in the study of the relationship between bacterial vesicles and biofilm. The huge number of papers discussing the role of MVs confirms the interest aimed at developing new applications in the medical field. The study of the MVs composition and biogenesis may contribute to the identification of components which could be (i) the target for the development of new drugs inhibiting the biofilm establishment; (ii) candidates for the development of vaccines; (iii) biomarkers for the diagnosis of bacterial infections.
Collapse
Affiliation(s)
- Valentina Puca
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Beatrice Marinacci
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Benedetta Pellegrini
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Floriana Campanile
- Department of Biomedical and Biotechnological Sciences (BIOMETEC) - Microbiology Section, University of Catania, Catania, Italy
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences (BIOMETEC) - Microbiology Section, University of Catania, Catania, Italy
| | - Rossella Grande
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
32
|
Mato EG, Montaño-Barrientos BJ, Rivas-Mundiña B, Aneiros IV, López LS, Posse JL, Lamas LM. Anti-caries Streptococcus spp.: A potential preventive tool for special needs patients. SPECIAL CARE IN DENTISTRY 2024; 44:813-822. [PMID: 37674277 DOI: 10.1111/scd.12920] [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: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/08/2023]
Abstract
INTRODUCTION Probiotics are living microorganisms that act on the host-microbiome interface to restore the microbiota's physiological homeostasis. Numerous probiotics have been marketed with inhibitory activity against Streptococcus mutans and consequently with a potential anti-caries effect, mainly of the genera Lactobacillus and Bifidobacterium, whose main disadvantage is their limited ability to settle in the oral cavity. METHODS This narrative review describes the main Streptococcus spp. with probiotic anti-Streptococcus mutans activity, whose substantivity is greater than that of Lactobacillus spp. and consequently with anti-caries potentiality. We performed a literature review in the PubMed, Science Direct and Google Scholar databases of articles published in English (without time restriction) related to caries and probiotics. RESULTS The potential identified anti-caries probiotics included Streptococcus spp. A12, Streptococcus oralis (AJ3), Streptococcus oligofermentans, Streptococcus salivarius (K12, M18, JH, LAB813, 24SMB), Streptococcus spp. with arginolytic activity (S. sanguinis, S. gordonii, S. ratti, S. parasanguinis, S. intermedius, S. australis, and S. cristatus), Streptococcus rattus (JH145), Streptococcus dentisani and Streptococcus downii. CONCLUSIONS The possibility of using these Streptococcus spp. as probiotics that inhibit the growth of dental plaque and the development of carious lesions represents a potential tool of particular interest for individuals with physical or intellectual disabilities that impede the routine and effective application of mechanical dental plaque removal techniques.
Collapse
Affiliation(s)
- Eliane García Mato
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Bitalio J Montaño-Barrientos
- Clinical Microbiology, Hospital Álvaro Cunqueiro, University Hospital Complex of Vigo, Microbiology and Infectology Group, Galicia Sur Health Research Institute (IISGS), Vigo, Galicia, Spain
| | - Berta Rivas-Mundiña
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Iván Varela Aneiros
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Lucía Sande López
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Jacobo Limeres Posse
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Lucía Martínez Lamas
- Clinical Microbiology, Hospital Álvaro Cunqueiro, University Hospital Complex of Vigo, Microbiology and Infectology Group, Galicia Sur Health Research Institute (IISGS), Vigo, Galicia, Spain
| |
Collapse
|
33
|
Díaz-Fabregat B, Ramírez-Carmona W, Cannon ML, Monteiro DR, Pessan JP, Antoniali C. Are salivary NO 2- / NO 2- and NO 3- levels biomarkers for dental caries in children? Systematic review and meta-analysis. Nitric Oxide 2024; 144:11-19. [PMID: 38185241 DOI: 10.1016/j.niox.2024.01.001] [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: 07/08/2023] [Revised: 11/15/2023] [Accepted: 01/05/2024] [Indexed: 01/09/2024]
Abstract
The literature is conflicting regarding salivary nitrite (NO2-)/nitrite and nitrate (NO2- and NO3-) levels in children affected by dental caries. For this reason, a systematic review to provide a consensus on the subject was propose, whose objective is to verify whether these molecules could be used as biomarkers in children with caries. A comprehensive search was performed on online database and eleven articles were included in the meta-analysis. The methodological quality of studies was assessed by Newcastle-Ottawa Scale recommended for case-control studies and by AXIS tool for cross-sectional studies. Grading of Recommendations Assessment, Development and Evaluation was used for the assessment of the certainty of the evidence for each outcome. The results showed lower NO2- levels in the group of children affected by dental caries (SMD = -2.18 [-3.24, -1.13], p < 0.01). Age, saliva collection and methods of evaluation can impact the results. When evaluating the severity of the condition, an important variation was detected in relation to the different evaluation methods NO2-/NO2- and NO3-. In conclusion, based on the evidence presented, the results suggest that NO2- levels in saliva are a possible biomarker of dental caries. Results should be evaluated with caution due to the very low evidence from primary studies. Longitudinal studies are necessary to strengthen this hypothesis.
Collapse
Affiliation(s)
- Beatriz Díaz-Fabregat
- São Paulo State University (UNESP), Araçatuba School of Dentistry, Department of Preventive and Restorative Dentistry, Rua José Bonifácio 1193 Araçatuba, SP, Zip code 16015-050, Brazil
| | - Wilmer Ramírez-Carmona
- São Paulo State University (UNESP), Araçatuba School of Dentistry, Department of Preventive and Restorative Dentistry, Rua José Bonifácio 1193 Araçatuba, SP, Zip code 16015-050, Brazil
| | - Mark Lloyd Cannon
- Feinberg School of Medicine, Northwestern University, Ann and Robert Lurie Childrens Hospital, 420 E Superior St, Chicago, IL, 60611, USA
| | - Douglas Roberto Monteiro
- São Paulo State University (UNESP), Araçatuba School of Dentistry, Department of Preventive and Restorative Dentistry, Rua José Bonifácio 1193 Araçatuba, SP, Zip code 16015-050, Brazil; University of Western São Paulo (UNOESTE), Postgraduate Program in Health Sciences, Rua José Bongiovani 700 Presidente Prudente, SP, Zip code 19050-920, Brazil
| | - Juliano Pelim Pessan
- São Paulo State University (UNESP), Araçatuba School of Dentistry, Department of Preventive and Restorative Dentistry, Rua José Bonifácio 1193 Araçatuba, SP, Zip code 16015-050, Brazil
| | - Cristina Antoniali
- São Paulo State University (UNESP), Araçatuba School of Dentistry, Department of Basic Science, Rua José Bonifácio 1193 Araçatuba, SP, Zip code 16015-050, Brazil.
| |
Collapse
|
34
|
Netto de Oliveira da Cunha C, Rodeghiero Collares S, Carvalho Rodrigues D, Walcher DL, Quintana de Moura M, Rodrigues Martins LH, Baracy Klafke G, de Oliveira Arias JL, Carapelli R, do Santos Espinelli Junior JB, Scaini CJ, Farias da Costa de Avila L. The larvicidal effect of the supernatant of Lactobacillus acidophilus ATCC 4356 on Toxocara canis. Exp Parasitol 2024; 258:108720. [PMID: 38367945 DOI: 10.1016/j.exppara.2024.108720] [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: 10/24/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/19/2024]
Abstract
Human toxocariasis is a parasitic anthropozoonosis that is difficult to treat and control. A previous study carried out with Lactobacillus acidophilus ATCC 4356 revealed that the cell free supernatant (CFS) of this probiotic killed 100% of Toxocara canis larvae in vitro. The present study aimed to investigate the characteristics of the CFS of L. acidophilus ATCC 4356, which may be involved in its larvicidal effects on T. canis. L. acidophilus ATCC 4356 was cultured, and lactic and acetic acids present in the CFS were quantified by high performance liquid chromatography (HPLC). The levels of pH and H2O2 were also analyzed. To assess the larvicidal effect of the CFS, this was tested pure and diluted (1:2 to 1:128) on T. canis larvae. High concentrations of lactic and acetic acids were detected in the CFS. The acidity of the pure CFS was observed at pH 3.8, remaining acidic at dilutions of 1:2 to 1:16. Regarding the in vitro larvicidal effect, 100% death of T. canis larvae was observed using the pure CFS and 1:2 dilution. Based on these results, it can be inferred that the presence of higher concentrations of organic acids and low pH of the medium contributed to the larvicidal activity of the CFS of L. acidophilus ATCC 4356. In addition, the maintenance of the larvicidal effect, even after dilution, suggests a greater chance of the larvicidal effect of this CFS against T. canis in vivo.
Collapse
Affiliation(s)
- Carolina Netto de Oliveira da Cunha
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil.
| | | | - Débora Carvalho Rodrigues
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Débora Liliane Walcher
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Micaele Quintana de Moura
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Lourdes Helena Rodrigues Martins
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Gabriel Baracy Klafke
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Jean Lucas de Oliveira Arias
- Integrated Analysis Center, School of Chemistry and Food, Federal University of Rio Grande, Av. Italia, Km 6 - Campus Carreiros, CEP 96203-900, Rio Grande, Rio Grande do Sul, Brazil
| | - Rodolfo Carapelli
- School of Chemistry and Food, Federal University of Rio Grande, Avenida Itália, Km 08 - Campus Carreiros, CEP 96.203-900, Rio Grande, Rio Grande do Sul, Brazil
| | - João Batista do Santos Espinelli Junior
- School of Chemistry and Food, Federal University of Rio Grande, Avenida Itália, Km 08 - Campus Carreiros, CEP 96.203-900, Rio Grande, Rio Grande do Sul, Brazil
| | - Carlos James Scaini
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Luciana Farias da Costa de Avila
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| |
Collapse
|
35
|
Luo SC, Wei SM, Luo XT, Yang QQ, Wong KH, Cheung PCK, Zhang BB. How probiotics, prebiotics, synbiotics, and postbiotics prevent dental caries: an oral microbiota perspective. NPJ Biofilms Microbiomes 2024; 10:14. [PMID: 38402294 PMCID: PMC10894247 DOI: 10.1038/s41522-024-00488-7] [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: 12/07/2023] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
Dental caries, a highly prevalent oral disease, impacts a significant portion of the global population. Conventional approaches that indiscriminately eradicate microbes disrupt the natural equilibrium of the oral microbiota. In contrast, biointervention strategies aim to restore this balance by introducing beneficial microorganisms or inhibiting cariogenic ones. Over the past three decades, microbial preparations have garnered considerable attention in dental research for the prevention and treatment of dental caries. However, unlike related pathologies in the gastrointestinal, vaginal, and respiratory tracts, dental caries occurs on hard tissues such as tooth enamel and is closely associated with localized acid overproduction facilitated by cariogenic biofilms. Therefore, it is insufficient to rely solely on previous mechanisms to delineate the role of microbial preparations in the oral cavity. A more comprehensive perspective should involve considering the concepts of cariogenic biofilms. This review elucidates the latest research progress, mechanisms of action, challenges, and future research directions regarding probiotics, prebiotics, synbiotics, and postbiotics for the prevention and treatment of dental caries, taking into account the unique pathogenic mechanisms of dental caries. With an enhanced understanding of oral microbiota, personalized microbial therapy will emerge as a critical future research trend.
Collapse
Affiliation(s)
- Si-Chen Luo
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Si-Min Wei
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Xin-Tao Luo
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Qiong-Qiong Yang
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Ka-Hing Wong
- Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, PR China
| | - Peter C K Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, PR China
| | - Bo-Bo Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China.
| |
Collapse
|
36
|
Yousif D, Wu Y, Gonzales AA, Mathieu C, Zeng Y, Sample L, Terando S, Li T, Xiao J. Anti-Cariogenic Effects of S. cerevisiae and S. boulardii in S. mutans-C. albicans Cross-Kingdom In Vitro Models. Pharmaceutics 2024; 16:215. [PMID: 38399269 PMCID: PMC10891968 DOI: 10.3390/pharmaceutics16020215] [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: 01/11/2024] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Despite the well-documented health benefits of the probiotic Saccharomyces, its application in oral health has not been comprehensively assessed. Dental caries is a transmissible disease initiated by acid production of cariogenic bacteria and yeast, such as Streptococcus mutans and Candida albicans, on tooth enamel and followed by subsequent enamel demineralization. Here, we investigated the effect of two Saccharomyces strains (Saccharomyces boulardii and Saccharomyces cerevisiae) on S. mutans-C. albicans cross-kingdom interactions using a cariogenic planktonic model. Viable cells, pH changes, and gene expression were measured. S. cerevisiae and S. boulardii inhibited the growth of C. albicans in dual- and multi-species conditions at 4, 6, and 20 h. Saccharomyces also inhibited C. albicans hyphal formation. Furthermore, Saccharomyces reduced the acidity of the culture medium, which usually plummeted below pH 5 when S. mutans and C. albicans were present in the model. The presence of Saccharomyces maintained the culture medium above 6 even after overnight incubation, demonstrating a protective potential against dental enamel demineralization. S. boulardii significantly down-regulated S. mutans atpD and eno gene expression. Overall, our results shed light on a new promising candidate, Saccharomyces, for dental caries prevention due to its potential to create a less cariogenic environment marked by a neutral pH and reduced growth of C. albicans.
Collapse
Affiliation(s)
- Dina Yousif
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.Y.); (Y.W.); (Y.Z.); (L.S.); (T.L.)
| | - Yan Wu
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.Y.); (Y.W.); (Y.Z.); (L.S.); (T.L.)
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430042, China
| | - Alexandria Azul Gonzales
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Christa Mathieu
- VCU College of Health Professions, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Yan Zeng
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.Y.); (Y.W.); (Y.Z.); (L.S.); (T.L.)
| | - Lee Sample
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.Y.); (Y.W.); (Y.Z.); (L.S.); (T.L.)
| | - Sabrina Terando
- School of Arts & Sciences, University of Rochester, Rochester, NY 14627, USA;
| | - Ting Li
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.Y.); (Y.W.); (Y.Z.); (L.S.); (T.L.)
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, Chongqing 400016, China
| | - Jin Xiao
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY 14642, USA; (D.Y.); (Y.W.); (Y.Z.); (L.S.); (T.L.)
| |
Collapse
|
37
|
Rezki S, Sunardi S, Aripin D, Khayan K, Pawarti P, Noeriman AR. Risk factors for dental caries: A case study in peatlands and non-peatlands of West Kalimantan, Indonesia. JOURNAL OF WATER AND HEALTH 2024; 22:321-328. [PMID: 38421626 PMCID: wh_2024_271 DOI: 10.2166/wh.2024.271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The prevalence of dental caries in peatlands and non-peatlands in West Kalimantan require preventive acts based on its natural conditions and the behavior of local communities. The objective was to analyse risk factors for dental caries in communities living in peatlands and non-peatlands in West Kalimantan. The research is a causal comparative study with cross-sectional approach. The samples were chosen by purposive sampling technique among adolescents aged 17-27 years, who were domiciled in Pontianak City (peatland) and Bengkayang (non-peatland) all their lives. The results showed that dental caries is significantly correlated with debris in peatlands (rs = 0.289). On non-peatlands, dental caries is correlated with drinking water phosphate (rs = 0.313) and calculus (rs = 0.034). In West Kalimantan, dental caries is significantly correlated with drinking water minerals (fluoride rs = -0.243; phosphate rs = 0.260), drinking water pH (rs = 0.235), behavior (rs = -0.327), and debris (rs = 0.240). The risk factors for dental caries in peatlands and non-peatlands in West Kalimantan are pH and drinking water minerals (fluoride and calcium), debris, calculus, and behavior.
Collapse
Affiliation(s)
- Sri Rezki
- Doctor Programme of Environmental Science, Padjadjaran University, Bandung, Indonesia; Department of Dental Nursing, Pontianak Health Polytechnic, Pontianak, Indonesia E-mail:
| | - Sunardi Sunardi
- Doctor Programme of Environmental Science, Padjadjaran University, Bandung, Indonesia
| | - Dudi Aripin
- Department of Conservative Dentistry, Faculty of Dentistry, Padjadjaran University, Bandung, Indonesia
| | - Khayan Khayan
- Department of Medical Laboratory, Banten Health Polytechnic, Serang, Indonesia
| | - Pawarti Pawarti
- Department of Dental Nursing, Pontianak Health Polytechnic, Pontianak, Indonesia
| | | |
Collapse
|
38
|
Yu X, Devine D, Vernon J. Manipulating the diseased oral microbiome: the power of probiotics and prebiotics. J Oral Microbiol 2024; 16:2307416. [PMID: 38304119 PMCID: PMC10833113 DOI: 10.1080/20002297.2024.2307416] [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: 10/20/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 02/03/2024] Open
Abstract
Dental caries and periodontal disease are amongst the most prevalent global disorders. Their aetiology is rooted in microbial activity within the oral cavity, through the generation of detrimental metabolites and the instigation of potentially adverse host immune responses. Due to the increasing threat of antimicrobial resistance, alternative approaches to readdress the balance are necessary. Advances in sequencing technologies have established relationships between disease and oral dysbiosis, and commercial enterprises seek to identify probiotic and prebiotic formulations to tackle preventable oral disorders through colonisation with, or promotion of, beneficial microbes. It is the metabolic characteristics and immunomodulatory capabilities of resident species which underlie health status. Research emphasis on the metabolic environment of the oral cavity has elucidated relationships between commensal and pathogenic organisms, for example, the sequential metabolism of fermentable carbohydrates deemed central to acid production in cariogenicity. Therefore, a focus on the preservation of an ecological homeostasis in the oral environment may be the most appropriate approach to health conservation. In this review we discuss an ecological approach to the maintenance of a healthy oral environment and debate the potential use of probiotic and prebiotic supplementation, specifically targeted at sustaining oral niches to preserve the delicately balanced microbiome.
Collapse
Affiliation(s)
- X. Yu
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - D.A. Devine
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - J.J. Vernon
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| |
Collapse
|
39
|
He X, Zhang S, Zhong Y, Huang X, Liu F, He J, Mai S. A low-shrinkage-stress and anti-bacterial adherent dental resin composite: physicochemical properties and biocompatibility. J Mater Chem B 2024; 12:814-827. [PMID: 38189164 DOI: 10.1039/d3tb01556d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Polymerisation shrinkage and biofilm accumulation are the two main problems associated with dental resin composites (DRCs) that induce secondary caries, which can cause restoration failure. Polymerisation shrinkage can lead to microleakage gaps between the tooth and the DRCs, causing the aggregation of bacteria and development of secondary caries. Reducing the shrinkage stress (SS) and improving the resistance to bacterial adhesion have always been the focus of this field in modifying DRCs. A thiol-ene resin system can effectively reduce the polymerisation SS via its step-growth mechanism for delaying the gel point. Fluorinated compounds can reduce the surface free energies, thereby reducing bacterial adhesion. Thus, in this study, a range of mass fractions (0, 10, 20, 30, and 40 wt%) of a fluorinated thiol-ene resin system were added to a fluorinated dimethacrylate resin system/tricyclo decanedimethanol diacrylate to create a fluorinated methacrylate-thiol-ene ternary resin matrix. DRCs were prepared using the obtained ternary resin matrix, and their physical and chemical properties, effect on bacterial adhesion, and biocompatibility were investigated. The results demonstrated that the volumetric shrinkage and SS of the DRCs were reduced with no reduction in conversion degree even after the thiol-ene resin system was added. All DRC-based fluorinated resin systems exhibited an excellent anti-bacterial adhesion effect, as evidenced by the colony-forming unit counts, live/dead bacterial staining, and crystal violet staining tests against Streptococcus mutans (S. mutans). The genetic expressions associated with the bacterial adhesion of S. mutans were substantially affected after being cultured with fluorinated DRCs. All fluorinated DRCs demonstrated good biocompatibility through the in vitro cytotoxicity test and live/dead staining images of the L-929 cells. The above results illustrate that the DRCs based on the fluorinated methacrylate-thiol-ene resin matrix can be potentially applied in clinical practice due to their low SS and anti-bacterial adhesion effect.
Collapse
Affiliation(s)
- Xinlin He
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, People's Republic of China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, 510055, People's Republic of China
- Institute of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, People's Republic of China
| | - Shengcan Zhang
- College of Materials Science and Engineering, South China University of Technology, Guangzhou, Guangdong, 510641, People's Republic of China.
| | - Yewen Zhong
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, People's Republic of China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, 510055, People's Republic of China
- Institute of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, People's Republic of China
| | - Xiangya Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, People's Republic of China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, 510055, People's Republic of China
- Institute of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, People's Republic of China
| | - Fang Liu
- College of Materials Science and Engineering, South China University of Technology, Guangzhou, Guangdong, 510641, People's Republic of China.
| | - Jingwei He
- College of Materials Science and Engineering, South China University of Technology, Guangzhou, Guangdong, 510641, People's Republic of China.
| | - Sui Mai
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, People's Republic of China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, 510055, People's Republic of China
- Institute of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510055, People's Republic of China
| |
Collapse
|
40
|
Jiang H, Wang Z, Jia AQ. Methyl gallate from Camellia nitidissima Chi flowers reduces quorum sensing related virulence and biofilm formation against Aeromonas hydrophila. BIOFOULING 2024; 40:64-75. [PMID: 38373897 DOI: 10.1080/08927014.2024.2316611] [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: 09/07/2023] [Accepted: 02/02/2024] [Indexed: 02/21/2024]
Abstract
Aeromonas hydrophila, a Gram-negative zoonotic bacterium, causes high mortality in fish farming and immunocompromised patients. This study aimed to extract methyl gallate (MG) from the flowers of Camellia nitidissima Chi and evaluate its potential as a quorum sensing inhibitor (QSI) against Aeromonas hydrophila SHAe 115. MG reduced QS-associated virulence factors, including hemolysis, protease, and lipase, while impairing swimming motility and biofilm formation. Additionally, MG down-regulated positive regulatory genes (ahyR, fleQ) and up-regulated negative regulators (litR, fleN). This highlights MG's promise as a potent QSI for A. hydrophila SHAe 115, advancing strategies against infections in aquaculture and human health.
Collapse
Affiliation(s)
- Huan Jiang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhennan Wang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, China
| | - Ai-Qun Jia
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, China
- Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
41
|
Plotniece A, Sobolev A, Supuran CT, Carta F, Björkling F, Franzyk H, Yli-Kauhaluoma J, Augustyns K, Cos P, De Vooght L, Govaerts M, Aizawa J, Tammela P, Žalubovskis R. Selected strategies to fight pathogenic bacteria. J Enzyme Inhib Med Chem 2023; 38:2155816. [PMID: 36629427 PMCID: PMC9848314 DOI: 10.1080/14756366.2022.2155816] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 01/12/2023] Open
Abstract
Natural products and analogues are a source of antibacterial drug discovery. Considering drug resistance levels emerging for antibiotics, identification of bacterial metalloenzymes and the synthesis of selective inhibitors are interesting for antibacterial agent development. Peptide nucleic acids are attractive antisense and antigene agents representing a novel strategy to target pathogens due to their unique mechanism of action. Antisense inhibition and development of antisense peptide nucleic acids is a new approach to antibacterial agents. Due to the increased resistance of biofilms to antibiotics, alternative therapeutic options are necessary. To develop antimicrobial strategies, optimised in vitro and in vivo models are needed. In vivo models to study biofilm-related respiratory infections, device-related infections: ventilator-associated pneumonia, tissue-related infections: chronic infection models based on alginate or agar beads, methods to battle biofilm-related infections are discussed. Drug delivery in case of antibacterials often is a serious issue therefore this review includes overview of drug delivery nanosystems.
Collapse
Affiliation(s)
- Aiva Plotniece
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Riga Stradiņš University, Riga, Latvia
| | | | - Claudiu T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Firenze, Italy
| | - Fabrizio Carta
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Firenze, Italy
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Center for Peptide-Based Antibiotics, University of Copenhagen, Copenhagen East, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Center for Peptide-Based Antibiotics, University of Copenhagen, Copenhagen East, Denmark
| | - Jari Yli-Kauhaluoma
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Koen Augustyns
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Paul Cos
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Linda De Vooght
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Matthias Govaerts
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Juliana Aizawa
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Päivi Tammela
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Raivis Žalubovskis
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Faculty of Materials Science and Applied Chemistry, Institute of Technology of Organic Chemistry, Riga Technical University, Riga, Latvia
| |
Collapse
|
42
|
Yang S, Bai M, Kwok LY, Zhong Z, Sun Z. The intricate symbiotic relationship between lactic acid bacterial starters in the milk fermentation ecosystem. Crit Rev Food Sci Nutr 2023; 65:728-745. [PMID: 37983125 DOI: 10.1080/10408398.2023.2280706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Fermentation is one of the most effective methods of food preservation. Since ancient times, food has been fermented using lactic acid bacteria (LAB). Fermented milk is a very intricate fermentation ecosystem, and the microbial metabolism of fermented milk largely determines its metabolic properties. The two most frequently used dairy starter strains are Streptococcus thermophilus (S. thermophilus) and Lactobacillus delbrueckii subsp. bulgaricus (L. bulgaricus). To enhance both the culture growth rate and the flavor and quality of the fermented milk, it has long been customary to combine S. thermophilus and L. bulgaricus in milk fermentation due to their mutually beneficial and symbiotic relationship. On the one hand, the symbiotic relationship is reflected by the nutrient co-dependence of the two microbes at the metabolic level. On the other hand, more complex interaction mechanisms, such as quorum sensing between cells, are involved. This review summarizes the application of LAB in fermented dairy products and discusses the symbiotic mechanisms and interactions of milk LAB starter strains from the perspective of nutrient supply and intra- and interspecific quorum sensing. This review provides updated information and knowledge on microbial interactions in a fermented milk ecosystem.
Collapse
Affiliation(s)
- Shujuan Yang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Mei Bai
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Zhi Zhong
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, PR China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, PR China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, PR China
| |
Collapse
|
43
|
Adnan M, Siddiqui AJ, Noumi E, Ashraf SA, Awadelkareem AM, Hadi S, Snoussi M, Badraoui R, Bardakci F, Sachidanandan M, Patel M. Biosurfactant derived from probiotic Lactobacillus acidophilus exhibits broad-spectrum antibiofilm activity and inhibits the quorum sensing-regulated virulence. BIOMOLECULES & BIOMEDICINE 2023; 23:1051-1068. [PMID: 37421468 PMCID: PMC10655870 DOI: 10.17305/bb.2023.9324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 07/10/2023]
Abstract
Antimicrobial resistance by pathogenic bacteria has become a global risk to human health in recent years. The most promising approach to combating antimicrobial resistance is to target virulent traits of bacteria. In the present study, a biosurfactant derived from the probiotic strain Lactobacillus acidophilus was tested against three Gram-negative bacteria to evaluate its inhibitory potential on their biofilms, and whether it affected the virulence factors controlled by quorum sensing (QS). A reduction in the virulence factors of Chromobacterium violaceum (violacein production), Serratia marcescens (prodigiosin production) and Pseudomonas aeruginosa (pyocyanin, total protease, LasB elastase and LasA protease production) was observed at different sub-MIC concentrations in a dose-dependent manner. Biofilm development was reduced by 65.76%, 70.64% and 58.12% at the highest sub-MIC levels for C. violaceum, P. aeruginosa and S. marcescens, respectively. Biofilm formation on glass surfaces exhibited significant reduction, with less bacterial aggregation and reduced formation of extracellular polymeric materials. Additionally, swimming motility and exopolysaccharides (EPS) production were shown to be reduced in the presence of the L. acidophilus-derived biosurfactant. Furthermore, molecular docking analysis performed on compounds identified through gas chromatography-mass spectrometry (GC-MS) analysis of QS and biofilm proteins yielded further insights into the mechanism underlying the anti-QS activity. Therefore, the present study has clearly demonstrated that a biosurfactant derived from L. acidophilus can significantly inhibit virulence factors of Gram-negative pathogenic bacteria. This could provide an effective method to inhibit the formation of biofilms and QS in Gram-negative bacteria.
Collapse
Affiliation(s)
- Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Emira Noumi
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Syed Amir Ashraf
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Ha’il, Ha’il, Saudi Arabia
| | - Amir Mahgoub Awadelkareem
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Ha’il, Ha’il, Saudi Arabia
| | - Sibte Hadi
- Department of Forensic Sciences, Naif Arab University for Security Sciences, Riyadh, Saudi Arabia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Fevzi Bardakci
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | | | - Mitesh Patel
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, India
| |
Collapse
|
44
|
Graciette AGC, Hoopes LA, Clauss T, Stewart FJ, Pratte ZA. The microbiome of African penguins (Spheniscus demersus) under managed care resembles that of wild marine mammals and birds. Sci Rep 2023; 13:16679. [PMID: 37794122 PMCID: PMC10551019 DOI: 10.1038/s41598-023-43899-w] [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: 02/07/2023] [Accepted: 09/29/2023] [Indexed: 10/06/2023] Open
Abstract
Animals under managed care in zoos and aquariums are ideal surrogate study subjects for endangered species that are difficult to obtain in the wild. We compared the fecal and oral microbiomes of healthy, managed African penguins (Spheniscus demersus) to those of other domestic and wild vertebrate hosts to determine how host identity, diet, and environment shape the penguin microbiome. The African penguin oral microbiome was more similar to that of piscivorous marine mammals, suggesting that diet and a marine environment together play a strong role in shaping the oral microbiome. Conversely, the penguin cloaca/fecal microbiome was more similar to that of other birds, suggesting that host phylogeny plays a significant role in shaping the gut microbiome. Although the penguins were born under managed care, they had a gut microbiome more similar to that of wild bird species compared to domesticated (factory-farmed) birds, suggesting that the managed care environment and diet resemble those experienced by wild birds. Finally, the microbiome composition at external body sites was broadly similar to that of the habitat, suggesting sharing of microbes between animals and their environment. Future studies should link these results to microbial functional capacity and host health, which will help inform conservation efforts.
Collapse
Affiliation(s)
- Ana G Clavere Graciette
- School of Biological Sciences, Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA, USA
| | | | | | - Frank J Stewart
- School of Biological Sciences, Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT, USA
| | - Zoe A Pratte
- School of Biological Sciences, Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA, USA.
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, MT, USA.
| |
Collapse
|
45
|
Zhang H, Zhang Z, Li J, Qin G. New Strategies for Biocontrol of Bacterial Toxins and Virulence: Focusing on Quorum-Sensing Interference and Biofilm Inhibition. Toxins (Basel) 2023; 15:570. [PMID: 37755996 PMCID: PMC10536320 DOI: 10.3390/toxins15090570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/05/2023] [Accepted: 09/09/2023] [Indexed: 09/28/2023] Open
Abstract
The overuse of antibiotics and the emergence of multiple-antibiotic-resistant pathogens are becoming a serious threat to health security and the economy. Reducing antimicrobial resistance requires replacing antibiotic consumption with more biocontrol strategies to improve the immunity of animals and humans. Probiotics and medicinal plants have been used as alternative treatments or preventative therapies for a variety of diseases caused by bacterial infections. Therefore, we reviewed some of the anti-virulence and bacterial toxin-inhibiting strategies that are currently being developed; this review covers strategies focused on quenching pathogen quorum sensing (QS) systems, the disruption of biofilm formation and bacterial toxin neutralization. It highlights the probable mechanism of action for probiotics and medicinal plants. Although further research is needed before a definitive statement can be made on the efficacy of any of these interventions, the current literature offers new hope and a new tool in the arsenal in the fight against bacterial virulence factors and bacterial toxins.
Collapse
Affiliation(s)
- Hua Zhang
- Henan Key Laboratory of Ion Beam Bio-Engineering, College of Physics, Zhengzhou University, Zhengzhou 450000, China;
- School of Food and Biological Engineering, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China;
| | - Zhen Zhang
- School of Food and Biological Engineering, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China;
| | - Jing Li
- School of Food and Biological Engineering, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China;
| | - Guangyong Qin
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450000, China;
| |
Collapse
|
46
|
Kıray E, Raheel NM. A New Approach to Exopolysaccharides of Post Probiotic Lactobacillus paracasei L1 Strain: Anti-quarum Sensing Activity. Balkan Med J 2023; 40:351-357. [PMID: 37278340 PMCID: PMC10500146 DOI: 10.4274/balkanmedj.galenos.2023.2023-1-96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
Background Multi-antibiotic resistance, which has increased in recent years, poses a serious societal threat as it makes the fight against deadly infection-causing pathogens even more complex and difficult. As such, the search for naturally resistant probiotic microorganisms and metabolic products obtained from these organisms to prevent infections, as an alternative to antibiotics, is crucial. In this context, preventing the quorum sensing (QS) mechanism that provides communication among bacteria is considered a mechanism that can prevent the colonization and progression of deadly infections. Aims To determine the QS mechanism and the immunological effects and various biological and biochemical characterizations of exopolysaccharide (EPS) obtained from the Lactobacillus paracasei L1 strain isolated from the vaginal microflora of healthy women. Study Design Experimental laboratory study. Methods The antibacterial ability, the antibiofilm and QS forming activities, and interferon (IFN)-γ and interleukin (IL)-10 production capacities of EPS were determined. The total antioxidant capacity (TAC), the surface morphology of EPS by scanning electron microscopy (SEM), the presence of functional groups, and the monosaccharide composition were determined by gas chromatographymass spectrometry (GC-MS). Results L. paracasei L1-EPS demonstrated a strong antibiofilm activity on Escherichia coli (65.14%), Staphylococcus aureus (63.27%), and Pseudomonas aeruginosa (54.21%) at a concentration of 5.0 mg/ml. The anti-QS activity of EPS was found to be quite high at 10 mg/ml EPS concentration. In the study performed with human peripheral blood mononuclear cells (hPBMC), the immunostimulatory IFN-γ value was higher (45 ± 0.0.3) than that in the experimental group, while the IL-10 value was lower than that in the control group (36 ± 0.05). The TAC value of L. piracies L1- EPS was found to be 76 μg/ml at 1,000 μg concentration. According to the GC-MS analysis results, glucose constituted 13.80% of the monosaccharide composition of EPS, while alpha-D-galactose constituted 13.89%. Conclusion Interestingly, EPSs of L. paracasei L1 strain, which have not been reported previously, demonstrated high anti-QS and antibiofilm properties, making EPSs a prospective compound for application in the pharmaceutical and food industries owing to their strong antimicrobial and antioxidant capacities.
Collapse
Affiliation(s)
- Esin Kıray
- Department of Medical Services and Techniques, Kırşehir Ahi Evran University Faculty of Medicine, Kırşehir, Turkey
| | - Nadia Masser Raheel
- Graduate School of Natural and Applied Sciences, Kırşehir Ahi Evran University, Kırşehir, Turkey
| |
Collapse
|
47
|
Zaghloul SA, Hashem SN, El-Sayed SR, Badawy MSEM, Bukhari SI, Selim HMRM, Riad OKM. Evaluation of the Cariogenic and Anti-Cariogenic Potential of Human Colostrum and Colostrum-Derived Probiotics: Impact on S. mutans Growth, Biofilm Formation, and L. rhamnosus Growth. Life (Basel) 2023; 13:1869. [PMID: 37763274 PMCID: PMC10532731 DOI: 10.3390/life13091869] [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: 07/23/2023] [Revised: 08/29/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Human colostrum (HC) is essential for oral health as it is rich in probiotics that could affect the growth of the cariogenic S. mutans and its biofilm formation; hindering dental caries in advance. In this study, HC was collected from 36 healthy mothers 1-3 days postpartum. The effect of HC on oral health was carried out by assessing the impact of HC and its derived probiotics' cell-free supernatants (CFS) on the growth of S. mutans (using modified well diffusion) and its biofilm formation (using microtiter plate assay). Moreover, the effect of whole HC on L. rhamnosus, a probiotic oral bacterium, was examined. Probiotics were isolated and identified phenotypically by API 50 CH carbohydrate fermentation and genotypically by 16S rRNA amplification. The in vitro study revealed that HC has cariogenic activity and is associated with biofilm formation. Biofilm strength was inversely proportional to HC dilution (p-value < 0.0001). Nevertheless, HC and colostrum-derived probiotics improve oral health by inhibiting the growth of caries-inducing S. mutans with lower inhibition to L. rhamnosus probiotics. The CFS of isolated probiotics reduced the biofilm formation via the cariogenic S. mutans. These results are not only promising for caries eradication, but they also highlight the importance of breastfeeding infants from their first hours to shape healthy oral microbiota, protecting them from various diseases including dental caries.
Collapse
Affiliation(s)
- Samaa A. Zaghloul
- Department of Operative Dentistry, Faculty of Dental Medicine for Girls, Al-Azhar University, Cairo 11765, Egypt;
| | - Sara N. Hashem
- Department of Pedodontics and Oral Health, Faculty of Dental Medicine for Girls, Al-Azhar University, Cairo 11651, Egypt;
| | - Safaa R. El-Sayed
- Department of Pedodontics and Oral Health, Faculty of Dental Medicine for Girls, Al-Azhar University, Cairo 11651, Egypt;
| | - Mona Shaban E. M. Badawy
- Department of Microbiology and Immunology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo 11884, Egypt; (M.S.E.M.B.); (O.K.M.R.)
| | - Sarah I. Bukhari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Heba Mohammed Refat M. Selim
- Department of Microbiology and Immunology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo 11884, Egypt; (M.S.E.M.B.); (O.K.M.R.)
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Maarefa University, Diriyah, Riyadh 13713, Saudi Arabia
| | - Omnia Karem M. Riad
- Department of Microbiology and Immunology, Faculty of Pharmacy for Girls, Al-Azhar University, Cairo 11884, Egypt; (M.S.E.M.B.); (O.K.M.R.)
| |
Collapse
|
48
|
Lingasamy P, Modhukur V, Mändar R, Salumets A. Exploring Immunome and Microbiome Interplay in Reproductive Health: Current Knowledge, Challenges, and Novel Diagnostic Tools. Semin Reprod Med 2023; 41:172-189. [PMID: 38262441 PMCID: PMC10846929 DOI: 10.1055/s-0043-1778017] [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] [Indexed: 01/25/2024]
Abstract
The dynamic interplay between the immunome and microbiome in reproductive health is a complex and rapidly advancing research field, holding tremendously vast possibilities for the development of reproductive medicine. This immunome-microbiome relationship influences the innate and adaptive immune responses, thereby affecting the onset and progression of reproductive disorders. However, the mechanisms governing these interactions remain elusive and require innovative approaches to gather more understanding. This comprehensive review examines the current knowledge on reproductive microbiomes across various parts of female reproductive tract, with special consideration of bidirectional interactions between microbiomes and the immune system. Additionally, it explores innate and adaptive immunity, focusing on immunoglobulin (Ig) A and IgM antibodies, their regulation, self-antigen tolerance mechanisms, and their roles in immune homeostasis. This review also highlights ongoing technological innovations in microbiota research, emphasizing the need for standardized detection and analysis methods. For instance, we evaluate the clinical utility of innovative technologies such as Phage ImmunoPrecipitation Sequencing (PhIP-Seq) and Microbial Flow Cytometry coupled to Next-Generation Sequencing (mFLOW-Seq). Despite ongoing advancements, we emphasize the need for further exploration in this field, as a deeper understanding of immunome-microbiome interactions holds promise for innovative diagnostic and therapeutic strategies for reproductive health, like infertility treatment and management of pregnancy.
Collapse
Affiliation(s)
| | - Vijayachitra Modhukur
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Reet Mändar
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Microbiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
49
|
Luo Z, Chen A, Xie A, Liu X, Jiang S, Yu R. Limosilactobacillus reuteri in immunomodulation: molecular mechanisms and potential applications. Front Immunol 2023; 14:1228754. [PMID: 37638038 PMCID: PMC10450031 DOI: 10.3389/fimmu.2023.1228754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
Frequent use of hormones and drugs may be associated with side-effects. Recent studies have shown that probiotics have effects on the prevention and treatment of immune-related diseases. Limosilactobacillus reuteri (L. reuteri) had regulatory effects on intestinal microbiota, host epithelial cells, immune cells, cytokines, antibodies (Ab), toll-like receptors (TLRs), tryptophan (Try) metabolism, antioxidant enzymes, and expression of related genes, and exhibits antibacterial and anti-inflammatory effects, leading to alleviation of disease symptoms. Although the specific composition of the cell-free supernatant (CFS) of L. reuteri has not been clarified, its efficacy in animal models has drawn increased attention to its potential use. This review summarizes the effects of L. reuteri on intestinal flora and immune regulation, and discusses the feasibility of its application in atopic dermatitis (AD), asthma, necrotizing enterocolitis (NEC), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS), and provides insights for the prevention and treatment of immune-related diseases.
Collapse
Affiliation(s)
- Zichen Luo
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Ailing Chen
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Anni Xie
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Xueying Liu
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Renqiang Yu
- Department of Neonatology, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
50
|
Guo M, Wu J, Hung W, Sun Z, Zhao W, Lan H, Zhao Z, Wuri G, Fang B, Zhao L, Zhang M. Lactobacillus paracasei ET-22 Suppresses Dental Caries by Regulating Microbiota of Dental Plaques and Inhibiting Biofilm Formation. Nutrients 2023; 15:3316. [PMID: 37571254 PMCID: PMC10421449 DOI: 10.3390/nu15153316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Dental caries is a common and multifactorial biofilm disease that is associated with dietary habits and microbiota. Among the various pathogens inducing caries, S. mutans is the most extensively studied. Promoting oral health with probiotics has gained considerable attention. Lactobacillus paracasei (L. paracasei) strains were reported to modulate the gut microbiota and enhance host resistance to disease. Our previous research has found that L. paracasei ET-22 (ET-22) could inhibit S. mutans biofilms in vitro. However, the preventive effect in vivo and functional mechanism of ET-22 on dental caries were unclear. In this study, the preventive effects of ET-22 on dental caries in mice were checked. Meanwhile, the functional mechanism of ET-22 was further investigated. Results showed that the supplementation of ET-22 in drinking water significantly improved the caries scoring of mice. The microbiota of dental plaques revealed that the live and heat-killed ET-22 similarly regulated the microbial structure in plaque biofilms. Functional prediction of PICRUSt showed that the addition of live and heat-killed ET-22 may inhibit biofilm formation. By the in vitro trials, the live and heat-killed ET-22 indeed inhibited the construction of S. mutans biofilms and EPS productions of biofilms. This evidence suggests that ET-22 can restrain dental caries by regulating the microbiota of dental plaques and inhibiting biofilm formation, which may be partly mediated by the body components of ET-22.
Collapse
Affiliation(s)
- Meng Guo
- School of Food and Health, Beijing Technology and Business University, Beijing 100024, China; (M.G.)
| | - Jianmin Wu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Weilian Hung
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010110, China
| | - Zhe Sun
- School of Food and Health, Beijing Technology and Business University, Beijing 100024, China; (M.G.)
| | - Wen Zhao
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010110, China
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Hanglian Lan
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Zhi Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Guna Wuri
- School of Food and Health, Beijing Technology and Business University, Beijing 100024, China; (M.G.)
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Liang Zhao
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Beijing Laboratory of Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100024, China; (M.G.)
| |
Collapse
|