1
|
Rebelo MB, Oliveira CS, Tavaria FK. Development of a Postbiotic-Based Orodispersible Film to Prevent Dysbiosis in the Oral Cavity. Front Biosci (Elite Ed) 2025; 17:26987. [PMID: 40150984 DOI: 10.31083/fbe26987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND Oral diseases affect over three billion peopleand are among the most commonly observed infections worldwide. Recent studies have shown that controlling the ecology of the oralome is more effective in reducing the risk of caries than the complete removal of both harmful and beneficial microorganisms. This work aimed to develop a strategy for preventing dysbiosis in the oral cavity by applying a postbiotic-based orodispersible film. METHODS Lactiplantibacillus plantarum 226V and Lacticaseibacillus paracasei L26 were cultured in De Man-Rogosa-Sharpe (MRS) broth for 48 hours, followed by centrifugation and filtration. Then, the resultant postbiotics were then subjected to various dilutions (10% (v/v), 20% (v/v), 40% (v/v), 60% (v/v) and 100% (v/v)) and co-incubated with Streptococcus mutans. Antimicrobial efficacy, minimal inhibitory concentration, the time required to inhibit S. mutans growth, and antibiofilm properties of the postbiotics were assessed. Subsequently, an orodispersible film comprising polymers and plasticizers, namely Xanthan gum, maltodextrin, and glycerol, was developed as a vehicle for postbiotic delivery. Formulation optimization, physical property evaluation, and cytotoxicity against the TR146 human oral cell line (TR146 cell line) were conducted. RESULTS Postbiotics demonstrated antimicrobial and antibiofilm activity against S. mutans following 24-hour co-incubation. The minimal inhibitory concentration for combined postbiotics administration was 20% (v/v). Remarkably, 79.6 ± 8.15% inhibition of biofilm formation was achieved using 100% (v/v) of the postbiotic derived from L. plantarum 226V. Incorporating postbiotics did not compromise the dissolution time of orodispersible films, all exceeding 20 minutes. Furthermore, solubility improved following postbiotic addition, facilitating ease of handling. Importantly, postbiotic-impregnated orodispersible films were non-cytotoxic when exposed to the TR146 cell line. CONCLUSIONS These findings underscore the potential of orodispersible films loaded with postbiotics as a promising potential intervention for oral dysbiosis.
Collapse
Affiliation(s)
- Mariana B Rebelo
- CBQF-Centre for Biotechnology and Fine Chemistry-Associated Laboratory, Portuguese Catholic University, 4169-005 Porto, Portugal
| | - Cláudia S Oliveira
- CBQF-Centre for Biotechnology and Fine Chemistry-Associated Laboratory, Portuguese Catholic University, 4169-005 Porto, Portugal
| | - Freni K Tavaria
- CBQF-Centre for Biotechnology and Fine Chemistry-Associated Laboratory, Portuguese Catholic University, 4169-005 Porto, Portugal
| |
Collapse
|
2
|
Ding J, Tan L, Wu L, Li J, Zhang Y, Shen Z, Zhang C, Zhao C, Gao L. Regulation of tryptophan-indole metabolic pathway in Porphyromonas gingivalis virulence and microbiota dysbiosis in periodontitis. NPJ Biofilms Microbiomes 2025; 11:37. [PMID: 40011497 DOI: 10.1038/s41522-025-00669-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/15/2025] [Indexed: 02/28/2025] Open
Abstract
Pathogenesis of periodontitis is marked by microbiota dysbiosis and disrupted host responses. Porphyromonas gingivalis is a keystone pathogen of periodontitis which expresses various crucial virulence factors. This study aimed to clarify the role and mechanisms of P. gingivalis tryptophan-indole metabolic pathway in the pathogenesis of periodontitis. This study showed that periodontitis patients exhibited elevated tryptophan metabolism and salivary pathogen abundance. Tryptophanase gene-deficiency altered proteome and metabolome of P. gingivalis, inhibited P. gingivalis virulent factors expression, biofilm growth, hemin utilization, cell adhesion/invasion and pro-inflammation ability. Tryptophan-indole pathway of P. gingivalis stimulated periodontitis biofilm formation and induced oral microbiota dysbiosis. In periodontitis mice, this pathway of P. gingivalis aggravated alveolar bone loss and gingival tissue destruction, causing oral and gut microbiota dysbiosis. This study indicates that the tryptophan-indole pathway serves as a significant regulator of P. gingivalis virulence and oral microbiota dysbiosis, which is also associated with gut dysbiosis.
Collapse
Affiliation(s)
- Jing Ding
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lingping Tan
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lingzhi Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jinyu Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yong Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zongshan Shen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chi Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Chuanjiang Zhao
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Li Gao
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Ueda T, Matsuda S, Ninomiya Y, Nakashima F, Yasuda K, Furutama D, Memida T, Yoshimoto T, Kajiya M, Ohta K, Ouhara K, Mizuno N. Nuclear receptor 4A1 (NR4A1) upregulated by n-butylidenephthalide via the mitogen-activated protein kinase (MAPK) pathway ameliorates drug-induced gingival enlargement. Biofactors 2024; 50:1192-1207. [PMID: 38777369 PMCID: PMC11627475 DOI: 10.1002/biof.2077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Drug-induced gingival enlargement (DIGE) is a side effect of ciclosporin, calcium channel blockers, and phenytoin. DIGE is a serious disease that leads to masticatory and esthetic disorders, severe caries, and periodontitis but currently has no standard treatment. We recently reported that nuclear receptor 4A1 (NR4A1) is a potential therapeutic target for DIGE. This study aimed to evaluate the therapeutic effects of n-butylidenephthalide (BP), which increases the expression of NR4A1, on DIGE. In this study, NR4A1 mRNA expression was analyzed in the patients with periodontal disease (PD) and DIGE. We evaluated the effect of BP on NR4A1 expression in gingival fibroblasts and in a DIGE mouse model. RNA sequencing (RNA-seq) was conducted to identify the mechanisms by which BP increases NR4A1 expression. The results showed that NR4A1 mRNA expression in the patients with DIGE was significantly lower than the patients with PD. BP suppressed the upregulation of COL1A1 expression, which was upregulated by TGF-β. BP also ameliorated gingival overgrowth in DIGE mice and reduced Col1a1 and Pai1 expression. BP also decreased Il1β mRNA expression in gingival tissue in DIGE. RNA-seq results showed an increase in the expression of several genes related to mitogen-activated protein kinase including DUSP genes in gingival fibroblasts stimulated by BP. Treatment with ERK and JNK inhibitors suppressed the BP-induced increase in NR4A1 expression. In addition, BP promoted the phosphorylation of ERK in gingival fibroblasts. In conclusion, BP increases NR4A1 expression in gingival fibroblasts through ERK and JNK signaling, demonstrating its potential as a preventive and therapeutic agent against DIGE.
Collapse
Affiliation(s)
- Tomoya Ueda
- Department of Periodontal MedicineGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| | - Shinji Matsuda
- Department of Periodontal MedicineGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| | - Yurika Ninomiya
- Department of Periodontal MedicineGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| | - Fuminori Nakashima
- Department of Periodontal MedicineGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| | - Keisuke Yasuda
- Department of Periodontal MedicineGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| | - Daisuke Furutama
- Department of Biological EndodonticsGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| | - Takumi Memida
- Department of Oral Science and Translation ResearchCollege of Dental Medicine, Nova Southeastern UniversityFort LauderdaleFloridaUSA
| | - Tetsuya Yoshimoto
- Center of Oral Clinical ExaminationHiroshima University HospitalHiroshimaJapan
| | - Mikihito Kajiya
- Center of Oral Clinical ExaminationHiroshima University HospitalHiroshimaJapan
| | - Kouji Ohta
- Department of Public Oral Health, Program of Oral Health SciencesGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| | - Kazuhisa Ouhara
- Department of Periodontal MedicineGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| | - Noriyoshi Mizuno
- Department of Periodontal MedicineGraduate School of Biomedical and Health Sciences, Hiroshima UniversityHiroshimaJapan
| |
Collapse
|
4
|
Nisar S, Shah AH, Nazir R. The clinical praxis of bacteriocins as natural anti-microbial therapeutics. Arch Microbiol 2024; 206:451. [PMID: 39476181 DOI: 10.1007/s00203-024-04152-8] [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: 08/05/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 11/10/2024]
Abstract
In recent decades, the excessive use of antibiotics has resulted in a rise in antimicrobial drug resistance (ADR). Annually, a significant number of human lives are lost due to resistant infectious diseases, leading to around 700,000 deaths, and it is estimated that by 2050, there could be up to 10 million casualties. Apart from their possible application as preservatives in the food sector, bacteriocins are gaining acknowledgment as potential clinical treatments. Not only this, these antimicrobial peptides have revealed in modulating the host immune system producing anti-inflammatory and anti-modulatory responses. At the same time, due to the ever-increasing global threat of antibiotic resistance, bacteriocins have gained attraction among researchers due to their potential clinical applications. Bacteriocins as antimicrobial peptides, represent one of the most important natural defense mechanisms among bacterial species, particularly lactic acid bacteria (LAB), that can fight against infection-causing pathogens. In this review, we are highlighting the potential of bacteriocins as novel therapeutics for inhibiting a wide range of clinically relevant and multi-drug-resistant pathogens (MDR). We also highlight the effectiveness and potential applications of current bacteriocin treatments in combating antimicrobial resistance (AMR), thereby promoting human health.
Collapse
Affiliation(s)
- Safura Nisar
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, India
| | - Abdul Haseeb Shah
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, India.
| | - Ruqeya Nazir
- Centre of Research for Development (CORD), School of Biological Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, India.
| |
Collapse
|
5
|
Dallos Ortega M, Aveyard J, Ciupa A, Poole RJ, Whetnall D, Behnsen JG, D'Sa RA. 3D printable gelatin/nisin biomaterial inks for antimicrobial tissue engineering applications. MATERIALS ADVANCES 2024; 5:7729-7746. [PMID: 39267949 PMCID: PMC11385060 DOI: 10.1039/d4ma00544a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024]
Abstract
Modern regenerative medicine approaches can rely on the fabrication of personalised medical devices and implants; however, many of these can fail due to infections, requiring antibiotics and revision surgeries. Given the rise in multidrug resistant bacteria, developing implants with antimicrobial activity without the use of traditional antibiotics is crucial for successful implant integration and improving patient outcomes. 3D printed gelatin-based implants have a broad range of applications in regenerative medicine due to their biocompatibility, ease of modification and degradability. In this paper, we report on the development of gelatin biomaterial inks loaded with the antimicrobial peptide, nisin, for extrusion-based 3D printing to produce scaffolds with controlled porosity, high shape fidelity, and structural stability. Rheological properties were comprehensively studied to develop inks that had shear thinning behaviour and viscoelastic properties to ensure optimal printability and extrudability, and enable precise deposition and structural integrity during 3D printing. The 3D printed scaffolds fabricated from the gelatin/nisin inks demonstrated excellent antimicrobial efficacy (complete kill) against Gram positive bacteria methicillin-resistant Staphylococcus aureus (MRSA). Overall, this ink's high printability and antimicrobial efficacy with the model antimicrobial peptide, nisin, offers the potential to develop customisable regenerative medicine implants that can effectively combat infection without contributing to the development of multidrug resistant bacteria.
Collapse
Affiliation(s)
- Mateo Dallos Ortega
- School of Engineering, University of Liverpool, Harrison Hughes Building, Brownlow Hill Liverpool L69 3GH UK
| | - Jenny Aveyard
- School of Engineering, University of Liverpool, Harrison Hughes Building, Brownlow Hill Liverpool L69 3GH UK
| | - Alexander Ciupa
- Materials Innovation Factory, University of Liverpool 51 Oxford Street Liverpool L7 3NY UK
| | - Robert J Poole
- School of Engineering, University of Liverpool, Harrison Hughes Building, Brownlow Hill Liverpool L69 3GH UK
| | - David Whetnall
- Materials Innovation Factory, University of Liverpool 51 Oxford Street Liverpool L7 3NY UK
| | - Julia G Behnsen
- School of Engineering, University of Liverpool, Harrison Hughes Building, Brownlow Hill Liverpool L69 3GH UK
| | - Raechelle A D'Sa
- School of Engineering, University of Liverpool, Harrison Hughes Building, Brownlow Hill Liverpool L69 3GH UK
| |
Collapse
|
6
|
Ye C, Zhao C, Kuraji R, Gao L, Rangé H, Kamarajan P, Radaic A, Kapila YL. Nisin, a Probiotic Bacteriocin, Modulates the Inflammatory and Microbiome Changes in Female Reproductive Organs Mediated by Polymicrobial Periodontal Infection. Microorganisms 2024; 12:1647. [PMID: 39203489 PMCID: PMC11357294 DOI: 10.3390/microorganisms12081647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Periodontitis-related oral microbial dysbiosis is thought to contribute to adverse pregnancy outcomes (APOs), infertility, and female reproductive inflammation. Since probiotics can modulate periodontitis and oral microbiome dysbiosis, this study examined the effects of a probiotic bacteriocin, nisin, in modulating the reproductive microbiome and inflammation triggered by periodontitis. A total of 24 eight-week-old BALB/cByJ female mice were randomly divided into four treatment groups (control, infection, nisin, and infection+nisin group), with 6 mice per group. A polymicrobial (Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia, Fusobacterium nucleatum) mouse model of periodontal disease was used to evaluate the effects of this disease on the female reproductive system, with a focus on the microbiome, local inflammation, and nisin's therapeutic potential in this context. Moreover, 16s RNA sequencing was used to evaluate the changes in the microbiome and RT-PCR was used to evaluate the changes in inflammatory cytokines. Periodontal pathogen DNA was detected in the reproductive organs, and in the heart and aorta at the end of the experimental period, and the DNA was especially elevated in the oral cavity in the infection group. Compared to the control groups, only P. gingivalis was significantly higher in the oral cavity and uterus of the infection groups, and T. forsythia and F. nucleatum were significantly higher in the oral cavity of the infection groups. The infection and nisin treatment group had significantly lower levels of P. gingivalis, T. forsythia, and F. nucleatum in the oral cavity compared with the infection group. Since periodontal pathogen DNA was also detected in the heart and aorta, this suggests potential circulatory system transmission. The polymicrobial infection generally decreased the microbiome diversity in the uterus, which was abrogated by nisin treatment. The polymicrobial infection groups, compared to the control groups, generally had lower Firmicutes and higher Bacteroidota in all the reproductive organs, with similar trends revealed in the heart. However, the nisin treatment group and the infection and nisin group, compared to the control or infection groups, generally had higher Proteobacteria and lower Firmicutes and Bacteroidota in the reproductive organs and the heart. Nisin treatment also altered the microbiome community structure in the reproductive tract to a new state that did not mirror the controls. Periodontal disease, compared to the controls, triggered an increase in inflammatory cytokines (IL-6, TNF-α) in the uterus and oral cavity, which was abrogated by nisin treatment. Polymicrobial periodontal disease alters the reproductive tract's microbial profile, microbiome, and inflammatory status. Nisin modulates the microbial profile and microbiome of the reproductive tract and mitigates the elevated uterine inflammatory cytokines triggered by periodontal disease.
Collapse
Affiliation(s)
- Changchang Ye
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, CA 94143, USA; (C.Y.); (C.Z.); (R.K.); (L.G.); (H.R.); (P.K.); (A.R.)
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chuanjiang Zhao
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, CA 94143, USA; (C.Y.); (C.Z.); (R.K.); (L.G.); (H.R.); (P.K.); (A.R.)
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510275, China
| | - Ryutaro Kuraji
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, CA 94143, USA; (C.Y.); (C.Z.); (R.K.); (L.G.); (H.R.); (P.K.); (A.R.)
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo 102-8159, Japan
| | - Li Gao
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, CA 94143, USA; (C.Y.); (C.Z.); (R.K.); (L.G.); (H.R.); (P.K.); (A.R.)
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510275, China
| | - Hélène Rangé
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, CA 94143, USA; (C.Y.); (C.Z.); (R.K.); (L.G.); (H.R.); (P.K.); (A.R.)
- Department of Periodontology, UFR of Odontology, University of Rennes, 35000 Rennes, France
- Service d’Odontologie, CHU de Rennes, 35000 Rennes, France
| | - Pachiyappan Kamarajan
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, CA 94143, USA; (C.Y.); (C.Z.); (R.K.); (L.G.); (H.R.); (P.K.); (A.R.)
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Allan Radaic
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, CA 94143, USA; (C.Y.); (C.Z.); (R.K.); (L.G.); (H.R.); (P.K.); (A.R.)
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Yvonne L. Kapila
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, CA 94143, USA; (C.Y.); (C.Z.); (R.K.); (L.G.); (H.R.); (P.K.); (A.R.)
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Shi Q, Sun L, Gao J, Li F, Chen D, Shi T, Tan Y, Chang H, Liu X, Kang J, Lu F, Huang Z, Zhao H. Effects of sodium lauryl sulfate and postbiotic toothpaste on oral microecology. J Oral Microbiol 2024; 16:2372224. [PMID: 38939048 PMCID: PMC11210412 DOI: 10.1080/20002297.2024.2372224] [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] [Received: 01/29/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024] Open
Abstract
The diversity and delicate balance of the oral microbiome contribute to oral health, with its disruption leading to oral and systemic diseases. Toothpaste includes elements like traditional additives such as sodium lauryl sulfate (SLS) as well as novel postbiotics derived from probiotics, which are commonly employed for maintaining oral hygiene and a healthy oral cavity. However, the response of the oral microbiota to these treatments remains poorly understood. In this study, we systematically investigated the impact of SLS, and toothpaste containing postbiotics (hereafter, postbiotic toothpaste) across three systems: biofilms, animal models, and clinical populations. SLS was found to kill bacteria in both preformed biofilms (mature biofilms) and developing biofilms (immature biofilms), and disturbed the microbial community structure by increasing the number of pathogenic bacteria. SLS also destroyed periodontal tissue, promoted alveolar bone resorption, and enhanced the extent of inflammatory response level. The postbiotic toothpaste favored bacterial homeostasis and the normal development of the two types of biofilms in vitro, and attenuated periodontitis and gingivitis in vivo via modulation of oral microecology. Importantly, the postbiotic toothpaste mitigated the adverse effects of SLS when used in combination, both in vitro and in vivo. Overall, the findings of this study describe the impact of toothpaste components on oral microflora and stress the necessity for obtaining a comprehensive understanding of oral microbial ecology by considering multiple aspects.
Collapse
Affiliation(s)
- Qingying Shi
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, China
| | - Lianlian Sun
- Stomatology Department, Binhai Hospital of Peking University, Tianjin, China
| | - Jing Gao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Fengzhu Li
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Dongxiao Chen
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Tingting Shi
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Youlan Tan
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Huimin Chang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Xiaozhi Liu
- Central Laboratory, Binhai Hospital of Peking University, Tianjin, China
| | - Jian Kang
- Periodontal Disease Department, Tianjin Stomatological Hospital, Tianjin, China
| | - Fuping Lu
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, China
| | - Zhengmei Huang
- Oral and Skin Microecology Institute of Tust & Benzhen, Science and Technology Park of Tianjin University of Science and Technology, Tianjin, China
| | - Huabing Zhao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, China
- Oral and Skin Microecology Institute of Tust & Benzhen, Science and Technology Park of Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
8
|
Wuttke B, Ekat K, Chabanovska O, Jackszis M, Springer A, Vasudevan P, Kreikemeyer B, Lang H. Preparation and In Vitro Characterization of Lactococcus lactis-Loaded Alginate Particles as a Promising Delivery Tool for Periodontal Probiotic Therapy. J Funct Biomater 2024; 15:129. [PMID: 38786639 PMCID: PMC11121860 DOI: 10.3390/jfb15050129] [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: 03/06/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Probiotic microorganisms are used in a variety of food supplements and medical formulations to promote human health. In periodontal therapy, probiotics are mainly used in the form of gels, tablets or rinses that often tend to leak from the periodontal pocket, resulting in a strongly reduced therapeutic effect. In this pilot in vitro study, we present biodegradable alginate-based particles as an alternative, highly efficient system for a periodontal delivery of probiotic bacteria to the inflammation site. For this purpose, Lactococcus (L.) lactis was encapsulated using a standardized pump-controlled extrusion-dripping method. Time-dependent bacterial release in artificial saliva was investigated over 9 days. The effect of freeze drying was explored to ensure long-term storage of L. lactis-loaded particles. Additionally, the particles were bound to dentin surface using approved bioadhesives and subjected to shear stress in a hydrodynamic flow chamber that mimics the oral cavity in vitro. Thus, round particles within the range of 0.80-1.75 mm in radius could be produced, whereby the diameter of the dripping tip had the most significant impact on the size. Although both small and large particles demonstrated a similar release trend of L. lactis, the release rate was significantly higher in the former. Following lyophilization, particles could restore their original shape within 4 h in artificial saliva; thereby, the bacterial viability was not affected. The attachment strength to dentin intensified by an adhesive could resist forces between 10 and 25 N/m2. Full degradation of the particles was observed after 20 days in artificial saliva. Therefore, alginate particles display a valuable probiotic carrier for periodontal applications that have several crucial advantages over existing preparations: a highly stable form, prolonged continuous release of therapeutic bacteria, precise manufacturing according to required dimensions at the application site, strong attachment to the tooth with low risk of dislocation, high biocompatibility and biodegradability.
Collapse
Affiliation(s)
- Bettina Wuttke
- Department of Operative Dentistry and Periodontology, University Medical Center Rostock, 18057 Rostock, Germany
| | - Katharina Ekat
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Rostock, 18057 Rostock, Germany
| | - Oleksandra Chabanovska
- Department of Operative Dentistry and Periodontology, University Medical Center Rostock, 18057 Rostock, Germany
| | - Mario Jackszis
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, University Medical Center Rostock, 18057 Rostock, Germany
| | - Armin Springer
- Medical Biology and Electron Microscopy Centre, University Medical Center Rostock, 18057 Rostock, Germany;
| | - Praveen Vasudevan
- Department of Operative Dentistry and Periodontology, University Medical Center Rostock, 18057 Rostock, Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Rostock, 18057 Rostock, Germany
| | - Hermann Lang
- Department of Operative Dentistry and Periodontology, University Medical Center Rostock, 18057 Rostock, Germany
| |
Collapse
|
9
|
Kuraji R, Ye C, Zhao C, Gao L, Martinez A, Miyashita Y, Radaic A, Kamarajan P, Le C, Zhan L, Range H, Sunohara M, Numabe Y, Kapila YL. Nisin lantibiotic prevents NAFLD liver steatosis and mitochondrial oxidative stress following periodontal disease by abrogating oral, gut and liver dysbiosis. NPJ Biofilms Microbiomes 2024; 10:3. [PMID: 38233485 PMCID: PMC10794237 DOI: 10.1038/s41522-024-00476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Oral microbiome dysbiosis mediates chronic periodontal disease, gut microbial dysbiosis, and mucosal barrier disfunction that leads to steatohepatitis via the enterohepatic circulation. Improving this dysbiosis towards health may improve liver disease. Treatment with antibiotics and probiotics have been used to modulate the microbial, immunological, and clinical landscape of periodontal disease with some success. The aim of the present investigation was to evaluate the potential for nisin, an antimicrobial peptide produced by Lactococcus lactis, to counteract the periodontitis-associated gut dysbiosis and to modulate the glycolipid-metabolism and inflammation in the liver. Periodontal pathogens, namely Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia and Fusobacterium nucleatum, were administrated topically onto the oral cavity to establish polymicrobial periodontal disease in mice. In the context of disease, nisin treatment significantly shifted the microbiome towards a new composition, commensurate with health while preventing the harmful inflammation in the small intestine concomitant with decreased villi structural integrity, and heightened hepatic exposure to bacteria and lipid and malondialdehyde accumulation in the liver. Validation with RNA Seq analyses, confirmed the significant infection-related alteration of several genes involved in mitochondrial dysregulation, oxidative phosphorylation, and metal/iron binding and their restitution following nisin treatment. In support of these in vivo findings indicating that periodontopathogens induce gastrointestinal and liver distant organ lesions, human autopsy specimens demonstrated a correlation between tooth loss and severity of liver disease. Nisin's ability to shift the gut and liver microbiome towards a new state commensurate with health while mitigating enteritis, represents a novel approach to treating NAFLD-steatohepatitis-associated periodontal disease.
Collapse
Affiliation(s)
- Ryutaro Kuraji
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Changchang Ye
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Chuanjiang Zhao
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Li Gao
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - April Martinez
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
| | - Yukihiro Miyashita
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Allan Radaic
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Pachiyappan Kamarajan
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Charles Le
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
| | - Ling Zhan
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
| | - Helene Range
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Periodontology, University of Rennes, UFR of Odontology; Service d'Odontologie, CHU de Rennes, Rennes, France
- INSERM CHU Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer); CIC 1414, Rennes, France
| | - Masataka Sunohara
- Department of Anatomy, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Yukihiro Numabe
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Yvonne L Kapila
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA.
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Zhao C, Kuraji R, Ye C, Gao L, Radaic A, Kamarajan P, Taketani Y, Kapila YL. Nisin a probiotic bacteriocin mitigates brain microbiome dysbiosis and Alzheimer's disease-like neuroinflammation triggered by periodontal disease. J Neuroinflammation 2023; 20:228. [PMID: 37803465 PMCID: PMC10557354 DOI: 10.1186/s12974-023-02915-6] [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: 05/06/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023] Open
Abstract
INTRODUCTION Periodontitis-related oral microbial dysbiosis is thought to contribute to Alzheimer's disease (AD) neuroinflammation and brain amyloid production. Since probiotics can modulate periodontitis/oral dysbiosis, this study examined the effects of a probiotic/lantibiotic, nisin, in modulating brain pathology triggered by periodontitis. METHODS A polymicrobial mouse model of periodontal disease was used to evaluate the effects of this disease on brain microbiome dysbiosis, neuroinflammation, Alzheimer's-related changes, and nisin's therapeutic potential in this context. RESULTS 16S sequencing and real-time PCR data revealed that Nisin treatment mitigated the changes in the brain microbiome composition, diversity, and community structure, and reduced the levels of periodontal pathogen DNA in the brain induced by periodontal disease. Nisin treatment significantly decreased the mRNA expression of pro-inflammatory cytokines (Interleukin-1β/IL-1 β, Interleukin 6/IL-6, and Tumor Necrosis Factor α/TNF-α) in the brain that were elevated by periodontal infection. In addition, the concentrations of amyloid-β 42 (Aβ42), total Tau, and Tau (pS199) (445.69 ± 120.03, 1420.85 ± 331.40, 137.20 ± 36.01) were significantly higher in the infection group compared to the control group (193.01 ± 31.82, 384.27 ± 363.93, 6.09 ± 10.85), respectively. Nisin treatment markedly reduced the Aβ42 (261.80 ± 52.50), total Tau (865.37 ± 304.93), and phosphorylated Tau (82.53 ± 15.77) deposition in the brain of the infection group. DISCUSSION Nisin abrogation of brain microbiome dysbiosis induces beneficial effects on AD-like pathogenic changes and neuroinflammation, and thereby may serve as a potential therapeutic for periodontal-dysbiosis-related AD.
Collapse
Affiliation(s)
- Chuanjiang Zhao
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Periodontology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, 510050, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510050, China
| | - Ryutaro Kuraji
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, 102-8159, Japan
| | - Changchang Ye
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Periodontology, West China School of Stomatology, National Clinical Research Center for Oral Diseases, State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610093, China
| | - Li Gao
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Periodontology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, 510050, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, 510050, China
| | - Allan Radaic
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Pachiyappan Kamarajan
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90024, USA
| | - Yoshimasa Taketani
- Department of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90024, USA
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, 350-0283, Japan
| | - Yvonne L Kapila
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90024, USA.
- Section of Biosystems and Function, Section of Periodontology, UCLA School of Dentistry, 10833 Le Conte Ave, Box 951668, Los Angeles, CA, 90095-1668, USA.
| |
Collapse
|
11
|
Toyoshima K, Ohsugi Y, Lin P, Komatsu K, Shiba T, Takeuchi Y, Hirota T, Shimohira T, Tsuchiya Y, Katagiri S, Iwata T, Aoki A. Blue Light-Emitting Diode Irradiation Without a Photosensitizer Alters Oral Microbiome Composition of Ligature-Induced Periodontitis in Mice. Photobiomodul Photomed Laser Surg 2023; 41:549-559. [PMID: 37788456 DOI: 10.1089/photob.2023.0061] [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: 10/05/2023] Open
Abstract
Objective: This study investigated the suppressive effects of blue light-emitting diode (LED) irradiation on bone resorption and changes in the oral microbiome of mice with ligature-induced periodontitis. Background: Wavelength of blue light has antimicrobial effects; however, whether blue LED irradiation alone inhibits the progression of periodontitis remains unclear. Methods: Nine-week-old male mice ligated ligature around the right maxillary second molar was divided into ligation alone (Li) and ligation with blue LED irradiation (LiBL) groups. The LiBL group underwent blue LED (wavelength, 455 nm) irradiation four times in a week at 150 mW/cm2 without a photosensitizer on the gingival tissue around the ligated tooth at a distance of 5 mm for 5 min. The total energy density per day was 45 J/cm2. Bone resorption was evaluated using micro-computed tomography at 8 days. Differences in the oral microbiome composition of the collected ligatures between the Li and LiBL groups were analyzed using next-generation sequencing based on the 16S rRNA gene from the ligatures. Results: Blue LED irradiation did not suppress bone resorption caused by ligature-induced periodontitis. However, in the LiBL group, the α-diversity, number of observed features, and Chao1 were significantly decreased. The relative abundances in phylum Myxococcota and Bacteroidota were underrepresented, and the genera Staphylococcus, Lactococcus, and Lactobacillus were significantly overrepresented by blue LED exposure. Metagenomic function prediction indicated an increase in the downregulated pathways related to microbial energy metabolism after irradiation. The co-occurrence network was altered to a simpler structure in the LiBL group, and the number of core genera decreased. Conclusions: Blue LED irradiation altered the composition and network of the oral microbiome of ligature-induced periodontitis in mice.
Collapse
Affiliation(s)
- Keita Toyoshima
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yujin Ohsugi
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Peiya Lin
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiji Komatsu
- Department of Lifetime Oral Health Care Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takahiko Shiba
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Yasuo Takeuchi
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Lifetime Oral Health Care Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomomitsu Hirota
- Division of Molecular Genetics, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Shimohira
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yosuke Tsuchiya
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sayaka Katagiri
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Iwata
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akira Aoki
- Department of Periodontology and Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
12
|
Radaic A, Shamir ER, Jones K, Villa A, Garud NR, Tward AD, Kamarajan P, Kapila YL. Specific Oral Microbial Differences in Proteobacteria and Bacteroidetes Are Associated with Distinct Sites When Moving from Healthy Mucosa to Oral Dysplasia-A Microbiome and Gene Profiling Study and Focused Review. Microorganisms 2023; 11:2250. [PMID: 37764094 PMCID: PMC10534919 DOI: 10.3390/microorganisms11092250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/24/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Oral potentially malignant disorders (OPMDs) are a group of conditions that carry a risk of oral squamous cell carcinoma (OSCC) development. Recent studies indicate that periodontal disease-associated pathogenic bacteria may play a role in the transition from healthy mucosa to dysplasia and to OSCC. Yet, the microbial signatures associated with the transition from healthy mucosa to dysplasia have not been established. To characterize oral microbial signatures at these different sites, we performed a 16S sequencing analysis of both oral swab and formalin-fixed, paraffin-embedded tissue (FFPE) samples. We collected oral swabs from healthy mucosa (from healthy patients), histologically normal mucosa adjacent to dysplasia, and low-grade oral dysplasia. Additionally, FFPE samples from histologically normal mucosa adjacent to OSCC, plus low grade and high-grade oral dysplasia samples were also collected. The collected data demonstrate significant differences in the alpha and beta microbial diversities of different sites in oral mucosa, dysplasia, and OSCC, as well as increased dissimilarities within these sites. We found that the Proteobacteria phyla abundance increased, concurrent with a progressive decrease in the Firmicutes phyla abundance, as well as altered levels of Enterococcus cecorum, Fusobacterium periodonticum, Prevotella melaninogenica, and Fusobacterium canifelinum when moving from healthy to diseased sites. Moreover, the swab sample analysis indicates that the oral microbiome may be altered in areas that are histologically normal, including in mucosa adjacent to dysplasia. Furthermore, trends in specific microbiome changes in oral swab samples preceded those in the tissues, signifying early detection opportunities for clinical diagnosis. In addition, we evaluated the gene expression profile of OSCC cells (HSC-3) infected with either P. gingivalis, T. denticola, F. nucelatum, or S. sanguinis and found that the three periodontopathogens enrich genetic processes related to cancer progression, including skin keratinization/cornification, while the commensal enriched processes related to RNA processing and adhesion. Finally, we reviewed the dysplasia microbiome literature and found a significant decrease in commensal bacteria, such as the Streptococci genus, and a simultaneous increase in pathogenic bacteria, mainly Bacteroidetes phyla and Fusobacterium genus. These findings suggest that features of the oral microbiome can serve as novel biomarkers for dysplasia and OSCC disease progression.
Collapse
Affiliation(s)
- Allan Radaic
- School of Dentistry, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; (A.R.); (P.K.)
- School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (K.J.); (A.V.)
| | - Eliah R. Shamir
- School of Medicine, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (E.R.S.); (A.D.T.)
- Genentech, Inc., South San Francisco, CA 94080, USA
| | - Kyle Jones
- School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (K.J.); (A.V.)
- Genentech, Inc., South San Francisco, CA 94080, USA
| | - Alessandro Villa
- School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (K.J.); (A.V.)
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL 33176, USA
| | - Nandita R. Garud
- College of Life Sciences, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA;
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Aaron D. Tward
- School of Medicine, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (E.R.S.); (A.D.T.)
| | - Pachiyappan Kamarajan
- School of Dentistry, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; (A.R.); (P.K.)
- School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (K.J.); (A.V.)
| | - Yvonne L. Kapila
- School of Dentistry, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; (A.R.); (P.K.)
- School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; (K.J.); (A.V.)
| |
Collapse
|
13
|
López-Valverde N, López-Valverde A, Montero J, Rodríguez C, Macedo de Sousa B, Aragoneses JM. Antioxidant, anti-inflammatory and antimicrobial activity of natural products in periodontal disease: a comprehensive review. Front Bioeng Biotechnol 2023; 11:1226907. [PMID: 37600299 PMCID: PMC10435350 DOI: 10.3389/fbioe.2023.1226907] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
Periodontal diseases (PD) are common chronic inflammatory oral pathologies that are strongly linked to others not found in the mouth cavity. The immune system mediates the host response, which includes the upregulation of proinflammatory cytokines, metalloproteinases, and reactive oxygen species (ROS); the latter may play an important role in the establishment and progression of inflammatory diseases, particularly periodontal disease, via the development of oxidative stress (OS). Natural antioxidants have powerful anti-inflammatory properties, and some can reduce serum levels of key PD indicators such tumor necrosis factor (TNF) and interleukin IL-1. This review compiles, through a thorough literature analysis, the antioxidant, anti-inflammatory, and antibacterial effects of a variety of natural products, as well as their therapeutic potential in the treatment of PD.
Collapse
Affiliation(s)
- Nansi López-Valverde
- Department of Medicine and Medical Specialties, Faculty of Health Sciences, Universidad Alcalá de Henares, Madrid, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Madrid, Spain
| | - Antonio López-Valverde
- Department of Surgery, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Javier Montero
- Department of Surgery, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Cinthia Rodríguez
- Department of Dentistry, Universidad Federico Henríquez y Carvajal, Santo Domingo, Dominican Republic
| | - Bruno Macedo de Sousa
- Institute for Occlusion and Orofacial Pain Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | |
Collapse
|
14
|
Guryanova SV. Immunomodulation, Bioavailability and Safety of Bacteriocins. Life (Basel) 2023; 13:1521. [PMID: 37511896 PMCID: PMC10381439 DOI: 10.3390/life13071521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
The rise of antibiotic-resistant bacteria and the emergence of new pathogens have created a need for new strategies to fight against infectious diseases. One promising approach is the use of antimicrobial peptides produced by a certain species of bacteria, known as bacteriocins, which are active against other strains of the same or related species. Bacteriocins can help in the treatment and prevention of infectious diseases. Moreover, bacteriocins can be obtained in prokaryotic organisms, and contribute s to their widespread use. While the use of bacteriocins is currently limited to the food industry (for example, nisin is used as a preservative, E234), a large number of studies on their microbicidal properties suggest that their use in medicine may increase in the foreseeable future. However, for the successful use of bacteriocins in medicine, it is necessary to understand their effect on the immune system, especially in cases where immunity is weakened due to infectious processes, oncological, allergic, or autoimmune diseases. Studies on the immuno-modulatory activity of bacteriocins in animal models and human cells have revealed their ability to induce both pro-inflammatory and anti-inflammatory factors involved in the implementation of innate immunity. The influence of bacteriocins on acquired immunity is revealed by an increase in the number of T-lymphocytes with a simultaneous decrease in B-lymphocyte levels, which makes them attractive substances for reducing inflammation. The widespread use of bacteriocins in the food industry, their low toxicity, and their broad and narrow specificity are reasons for researchers to pay attention to their immunomodulatory properties and explore their medical applications. Inflammation regulation by bacteriocins can be used in the treatment of various pathologies. The aim of the review was to analyze scientific publications on the immunomodulatory activity, bioavailability, and safety of bacteriocins in order to use the data obtained to organize preclinical and clinical studies.
Collapse
Affiliation(s)
- Svetlana V Guryanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- Medical Institute, Peoples' Friendship University of Russia (RUDN University) of the Ministry of Science and Higher Education of the Russian Federation, 117198 Moscow, Russia
| |
Collapse
|
15
|
Chitosan nanoparticles efficiently enhance the dispersibility, stability and selective antibacterial activity of insoluble isoflavonoids. Int J Biol Macromol 2023; 232:123420. [PMID: 36708890 DOI: 10.1016/j.ijbiomac.2023.123420] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
Natural isoflavonoids have attracted much attention in the treatment of oral bacterial infections and other diseases due to their excellent antibacterial activity and safety. However, their poor water solubility, instability and low bioavailability seriously limited the practical application. In this study, licoricidin-loaded chitosan nanoparticles (LC-CSNPs) were synthesized by self-assembly for improving the dispersion of licoricidin (LC) and strengthening antibacterial and anti-biofilm performance. Compared to free LC, the minimum inhibitory concentration of LC-CSNPs against Streptococcus mutans decreased >2-fold to 26 μg/mL, and LC-CSNPs could ablate 70 % biofilms at this concentration. The enhanced antibacterial activity was mainly attributed to the spontaneous surface adsorption of LC-CSNPs on cell membranes through electrostatic interactions. More valuably, LC-CSNPs had no inhibitory effect on the growth of probiotic. Mechanism study indicated that LC-CSNPs altered the transmembrane potential to cause bacterial cells in a hyperpolarized state, generating ROS to cause cells damage and eventually apoptosis. This work demonstrated that the chitosan-based nanoparticles have great potential in enhancing the dispersibility and antibacterial activity of insoluble isoflavonoids, offering a promising therapeutic strategy for oral infections.
Collapse
|
16
|
Jackova Z, Stepan JJ, Coufal S, Kostovcik M, Galanova N, Reiss Z, Pavelka K, Wenchich L, Hruskova H, Kverka M. Interindividual differences contribute to variation in microbiota composition more than hormonal status: A prospective study. Front Endocrinol (Lausanne) 2023; 14:1139056. [PMID: 37033235 PMCID: PMC10081494 DOI: 10.3389/fendo.2023.1139056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/13/2023] [Indexed: 04/11/2023] Open
Abstract
Aims Ovarian hormone deficiency is one of the main risk factors for osteoporosis and bone fractures in women, and these risks can be mitigated by menopausal hormone therapy. Recent evidence suggests that gut microbiota may link changes in estrogen levels and bone metabolism. This study was conducted to investigate the potential relationship between hormonal and bone changes induced by oophorectomy and subsequent hormonal therapy and shifts in gut microbiota composition. Methods We collected 159 stool and blood samples in several intervals from 58 women, who underwent bilateral oophorectomy. Changes in fecal microbiota were assessed in paired samples collected from each woman before and after oophorectomy or the start of hormone therapy. Bacterial composition was determined by sequencing the 16S rRNA gene on Illumina MiSeq. Blood levels of estradiol, FSH, biomarkers of bone metabolism, and indices of low-grade inflammation were measured using laboratory analytical systems and commercial ELISA. Areal bone mineral density (BMD) of the lumbar spine, proximal femur, and femur neck was measured using dual-energy X-ray absorptiometry. Results We found no significant changes in gut microbiota composition 6 months after oophorectomy, despite major changes in hormone levels, BMD, and bone metabolism. A small decrease in bacterial diversity was apparent 18 months after surgery in taxonomy-aware metrics. Hormonal therapy after oophorectomy prevented bone loss but only marginally affected gut microbiota. There were no significant differences in β-diversity related to hormonal status, although several microbes (e.g., Lactococcus lactis) followed estrogen levels. Body mass index (BMI) was the most significantly associated with microbiota variance. Microbiota was not a suitable predictive factor for the state of bone metabolism. Conclusions We conclude that neither the loss of estrogens due to oophorectomy nor their gain due to subsequent hormonal therapy is associated with a specific gut microbiota signature. Sources of variability in microbiota composition are more related to interindividual differences than hormonal status.
Collapse
Affiliation(s)
- Zuzana Jackova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, Prague, Czechia
| | - Jan J. Stepan
- Institute of Rheumatology, Prague, Czechia
- Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Prague, Czechia
| | - Stepan Coufal
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, Prague, Czechia
| | - Martin Kostovcik
- Laboratory of Fungal Genetics and Metabolism, Institute of Microbiology, Czech Academy of Sciences, Prague, Czechia
| | - Natalie Galanova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, Prague, Czechia
| | - Zuzana Reiss
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, Prague, Czechia
| | - Karel Pavelka
- Institute of Rheumatology, Prague, Czechia
- Department of Rheumatology, First Faculty of Medicine, Charles University in Prague, Prague, Czechia
| | | | - Hana Hruskova
- Department of Obstetrics and Gynecology, Charles University in Prague, First Faculty of Medicine, Prague, Czechia
- General University Hospital in Prague, Prague, Czechia
| | - Miloslav Kverka
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology, Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
17
|
Kuraji R, Shiba T, Dong TS, Numabe Y, Kapila YL. Periodontal treatment and microbiome-targeted therapy in management of periodontitis-related nonalcoholic fatty liver disease with oral and gut dysbiosis. World J Gastroenterol 2023; 29:967-996. [PMID: 36844143 PMCID: PMC9950865 DOI: 10.3748/wjg.v29.i6.967] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/14/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
A growing body of evidence from multiple areas proposes that periodontal disease, accompanied by oral inflammation and pathological changes in the microbiome, induces gut dysbiosis and is involved in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). A subgroup of NAFLD patients have a severely progressive form, namely nonalcoholic steatohepatitis (NASH), which is characterized by histological findings that include inflammatory cell infiltration and fibrosis. NASH has a high risk of further progression to cirrhosis and hepatocellular carcinoma. The oral microbiota may serve as an endogenous reservoir for gut microbiota, and transport of oral bacteria through the gastro-intestinal tract can set up a gut microbiome dysbiosis. Gut dysbiosis increases the production of potential hepatotoxins, including lipopolysaccharide, ethanol, and other volatile organic compounds such as acetone, phenol and cyclopentane. Moreover, gut dysbiosis increases intestinal permeability by disrupting tight junctions in the intestinal wall, leading to enhanced translocation of these hepatotoxins and enteric bacteria into the liver through the portal circulation. In particular, many animal studies support that oral administration of Porphyromonas gingivalis, a typical periodontopathic bacterium, induces disturbances in glycolipid metabolism and inflammation in the liver with gut dysbiosis. NAFLD, also known as the hepatic phenotype of metabolic syndrome, is strongly associated with metabolic complications, such as obesity and diabetes. Periodontal disease also has a bidirectional relationship with metabolic syndrome, and both diseases may induce oral and gut microbiome dysbiosis with insulin resistance and systemic chronic inflammation cooperatively. In this review, we will describe the link between periodontal disease and NAFLD with a focus on basic, epidemiological, and clinical studies, and discuss potential mechanisms linking the two diseases and possible therapeutic approaches focused on the microbiome. In conclusion, it is presumed that the pathogenesis of NAFLD involves a complex crosstalk between periodontal disease, gut microbiota, and metabolic syndrome. Thus, the conventional periodontal treatment and novel microbiome-targeted therapies that include probiotics, prebiotics and bacteriocins would hold great promise for preventing the onset and progression of NAFLD and subsequent complications in patients with periodontal disease.
Collapse
Affiliation(s)
- Ryutaro Kuraji
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo 102-0071, Japan
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA 94143, United States
| | - Takahiko Shiba
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, United States
- Department of Periodontology, Tokyo Medical and Dental University, Tokyo 113-8549, Japan
| | - Tien S Dong
- The Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Department of Medicine, University of California David Geffen School of Medicine, Los Angeles, CA 90095, United States
| | - Yukihiro Numabe
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo 102-8159, Japan
| | - Yvonne L Kapila
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA 94143, United States
- Sections of Biosystems and Function and Periodontics, Professor and Associate Dean of Research, Felix and Mildred Yip Endowed Chair in Dentistry, University of California Los Angeles, Los Angeles, CA 90095, United States
| |
Collapse
|
18
|
Radaic A, Brody H, Contreras F, Hajfathalian M, Lucido L, Kamarajan P, Kapila YL. Nisin and Nisin Probiotic Disrupt Oral Pathogenic Biofilms and Restore Their Microbiome Composition towards Healthy Control Levels in a Peri-Implantitis Setting. Microorganisms 2022; 10:1336. [PMID: 35889055 PMCID: PMC9324437 DOI: 10.3390/microorganisms10071336] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023] Open
Abstract
Peri-implantitis is characterized by chronic inflammation of the peri-implant supporting tissues that progressively and irreversibly leads to bone loss and, consequently, implant loss. Similar to periodontal disease, oral dysbiosis is thought to be a driver of peri-implantitis. However, managing peri-implantitis with traditional treatment methods, such as nonsurgical debridement or surgery, is not always successful. Thus, novel strategies have been proposed to address these shortcomings. One strategy is the use of probiotics as antimicrobial agents since they are considered safe for humans and the environment. Specifically, the probiotic Lactococcus lactis produces nisin, which has been used worldwide for food preservation. The objective of this study was to determine whether nisin and the wild-type (WT) nisin-producing L. lactis probiotic can disrupt oral pathogenic biofilms and promote a healthier oral microbiome within these oral biofilms on titanium discs. Using confocal imaging and 16S rRNA sequencing, this study revealed that nisin and WT L. lactis probiotic disrupt oral pathogenic biofilms in a peri-implantitis setting in vitro. More specifically, nisin decreased the viability of the pathogen-spiked biofilms dose-dependently from 62.53 ± 3.69% to 54.26 ± 3.35% and 44.88 ± 2.98%, respectively. Similarly, 105 CFU/mL of WT L. lactis significantly decreased biofilm viability to 52.45 ± 3.41%. Further, both treatments shift the composition, relative abundance, and diversity levels of these biofilms towards healthy control levels. A total of 1 µg/mL of nisin and 103 CFU/mL of WT L. lactis were able to revert the pathogen-mediated changes in the Proteobacteria (from 80.5 ± 2.9% to 75.6 ± 2.0%, 78.0 ± 2.8%, and 75.1 ± 5.3%, respectively) and Firmicutes (from 11.6 ± 1.6% to 15.4 ± 1.3%, 13.8 ± 1.8%, and 13.7 ± 2.6%, respectively) phyla back towards control levels. Thus, nisin and its nisin-producing L. lactis probiotic may be useful in treating peri-implantitis by promoting healthier oral biofilms, which may be useful for improving patient oral health.
Collapse
Affiliation(s)
- Allan Radaic
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Hanna Brody
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Fernando Contreras
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Maryam Hajfathalian
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Luke Lucido
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Pachiyappan Kamarajan
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Yvonne L. Kapila
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
- Division of Oral and Systemic Health Sciences, Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|