1
|
Wang H, Fan Q, Wang Y, Yi L, Wang Y. Rethinking the control of Streptococcus suis infection: Biofilm formation. Vet Microbiol 2024; 290:110005. [PMID: 38280304 DOI: 10.1016/j.vetmic.2024.110005] [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: 05/30/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
Streptococcus suis is an emerging zoonotic pathogen that is widespread in swine populations. The control of S. suis infection and its associated diseases is a daunting challenge worldwide. Biofilm formation appears to be the main reason for the persistence of S. suis. In this review we gather existing knowledge on S. suis biofilm, describing the role of biofilm formation in S. suis virulence and drug resistance, the regulatory factors of S. suis biofilm formation, and the research progress of inhibiting S. suis biofilm formation, with the aim of providing guidance for future studies related to the field of S. suis biofilms.
Collapse
Affiliation(s)
- Haikun Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Qingying Fan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Li Yi
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China; College of Life Science, Luoyang Normal University, Luoyang, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China.
| |
Collapse
|
2
|
Mirhashemi AH, Pourhajibagher M, Zebardast B, Bahrami R, Kharazi Fard MJ. In vitro effects of antimicrobial properties and shear bond strength of different concentrations of Emodin nanoparticles incorporated orthodontic composites. Int Orthod 2024; 22:100836. [PMID: 38134823 DOI: 10.1016/j.ortho.2023.100836] [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: 10/02/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVE Fixed appliances used in orthodontic treatment are accompanied by some drawbacks, including the development of white spots or enamel demineralization in the vicinity of the brackets and bonding failures. This study aims to evaluate the effect of combining different wt.% of Emodin nanoparticles (ENPs) with orthodontic adhesives to attain adhesives with improved antimicrobial and mechanical properties. METHODS ENPs were synthesized and added to orthodontic composite at different concentrations (0.5%, 1%, and 2%). The distribution of ENPs within the composite was evaluated using a field emission scanning electron microscope (FESEM). A total of 216 disks were prepared, with 144 subjected to an eluted components test, 36 used for disk agar diffusion (DAD) test, and 36 for biofilm inhibition test. These tests aimed to assess the antimicrobial activity of the composites against Streptococcus mutans, Lactobacillus acidophilus, and Candida albicans. Additionally, the bond strength between stainless-steel brackets and teeth was evaluated using the shear bond strength (SBS) test, and the adhesive remnant index (ARI) score was determined. One-way analysis of variance and Kruskal-Wallis test were used to analyse the SBS and ARI, respectively. For pairwise group comparison concerning the biofilm inhibition, DAD, and eluted components tests, the Tamhane and Games-Howell tests for data with unequal variances and the post-hoc Tukey's HSD and Scheffe tests for data with equal variances were used. RESULTS The FESEM results confirmed the synthesis and even distribution of ENPs in the composite. Only the 2% group showed significant biofilm inhibition against all microorganisms studied (P<0.05). The DAD test revealed that a 1% concentration of ENPs is sufficient to inhibit growth for all microorganisms. The eluted components test demonstrated that the 2% concentration of ENPs performed significantly better against S. mutans compared to the control group (P<0.05). The highest mean SBS was observed with the 0.5% ENP concentration, while no significant differences in SBS and ARI were found among the groups (P>0.05). CONCLUSIONS This in vitro study showed that the 2% concentration of ENP produced significantly improved antimicrobial activity without adversely affecting SBS and ARI score. This would support the addition of 2% ENP to orthodontic adhesives.
Collapse
Affiliation(s)
- Amir Hossein Mirhashemi
- Department of Orthodontics, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Bardia Zebardast
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran.
| | - Rashin Bahrami
- Dental Sciences Research Center, Department of Orthodontics, School of Dentistry, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Javad Kharazi Fard
- Department of Epidemiology and Biostatistics, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Bai J, Xie Y, Li M, Huang X, Guo Y, Sun J, Tang Y, Liu X, Wei C, Li J, Yang Y. Ultrasound-assisted extraction of emodin from Rheum officinale Baill and its antibacterial mechanism against Streptococcus suis based on CcpA. ULTRASONICS SONOCHEMISTRY 2024; 102:106733. [PMID: 38150957 PMCID: PMC10765492 DOI: 10.1016/j.ultsonch.2023.106733] [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: 03/01/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 12/29/2023]
Abstract
Emodin was extracted from Rheum officinale Baill by ultrasound-assisted extraction (UAE), and ethanol was chosen as the suitable solvent through SEM and molecular dynamic simulation. Under the optimum conditions (power 541 W, time 23 min, liquid to material ratio 13:1 mL/g, ethanol concentration 83 %) predicted by RSM, the yield of emodin was 2.18 ± 0.11 mg/g. Moreover, ultrasound power and time displayed the significant effects on the extraction process. Extracting dynamics analysis indicated that the extraction process of emodin by UAE conformed to Fick's second diffusion law. The results of antibacterial experiments suggested that emodin can damage cell membrane and inhibit the expression of cps2A, sao, mrp, epf, neu and the hemolytic activity of S. suis. Biolayer interferometry and FT-IR multi-peak fitting assays demonstrated that emodin induced a secondary conformational shift in CcpA. Molecular docking and molecular dynamics confirmed that emodin bound to CcpA through hydrogen bonding (ALA248, GLU249, GLY129 and ASN196) and π-π T-shaped interaction (TYR225 and TYR130), and the mutation of amino acid residues affected the affinity of CcpA to emodin. Therefore, emodin inhibited the sugar utilization of S. suis through binding to CcpA, and CcpA may be a potential target to inhibit the growth of S. suis.
Collapse
Affiliation(s)
- Jingwen Bai
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Yu Xie
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Miao Li
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Xianjun Huang
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Yujia Guo
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Jingwen Sun
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Yang Tang
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Xuantong Liu
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Chi Wei
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Jianqiang Li
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Yu Yang
- College of Art and Science, Northeast Agricultural University, Harbin 150030, People's Republic of China.
| |
Collapse
|
4
|
An R, Guo Y, Gao M, Wang J. Subcutaneous Streptococcus dysgalactiae GAPDH vaccine in mice induces a proficient innate immune response. J Vet Sci 2023; 24:e72. [PMID: 38031651 PMCID: PMC10556295 DOI: 10.4142/jvs.23103] [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: 04/11/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) on the surface of Streptococcus dysgalactiae, coded with gapC, is a glycolytic enzyme that was reported to be a moonlighting protein and virulence factor. OBJECTIVE This study assessed GAPDH as a potential immunization candidate protein to prevent streptococcus infections. METHODS Mice were vaccinated subcutaneously with recombinant GAPDH and challenged with S. dysgalactiae in vivo. They were then evaluated using histological methods. rGAPDH of mouse bone marrow-derived dendritic cells (BMDCs) was evaluated using immunoblotting, reverse transcription quantitative polymerase chain reaction, and enzyme-linked immunosorbent assay methods. RESULTS Vaccination with rGAPDH improved the survival rates and decreased the bacterial burdens in the mammary glands compared to the control group. The mechanism by which rGAPDH vaccination protects against S. dysgalactiae was investigated. In vitro experiments showed that rGAPDH boosted the generation of interleukin-10 and tumor necrosis factor-α. Treatment of BMDCs with TAK-242, a toll-like receptor 4 inhibitor, or C29, a toll-like receptor 2 inhibitor, reduced cytokines substantially, suggesting that rGAPDH may be a potential ligand for both TLR2 and TLR4. Subsequent investigations showed that rGAPDH may activate the phosphorylation of MAPKs and nuclear factor-κB. CONCLUSIONS GAPDH is a promising immunization candidate protein for targeting virulence and enhancing immune-mediated protection. Further investigations are warranted to understand the mechanisms underlying the activation of BMDCs by rGAPDH in a TLR2- and TLR4-dependent manner and the regulation of inflammatory cytokines contributing to mastitis pathogenesis.
Collapse
Affiliation(s)
- Ran An
- Heilongjiang Provincial Key Laboratory of Zoonosis, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China
| | - Yongli Guo
- Department of Immunology, Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin Medical University, Harbin 150000, China
| | - Mingchun Gao
- Heilongjiang Provincial Key Laboratory of Zoonosis, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China.
| | - Junwei Wang
- Heilongjiang Provincial Key Laboratory of Zoonosis, Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, China.
| |
Collapse
|
5
|
Qun T, Zhou T, Hao J, Wang C, Zhang K, Xu J, Wang X, Zhou W. Antibacterial activities of anthraquinones: structure-activity relationships and action mechanisms. RSC Med Chem 2023; 14:1446-1471. [PMID: 37593578 PMCID: PMC10429894 DOI: 10.1039/d3md00116d] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/24/2023] [Indexed: 08/19/2023] Open
Abstract
With the increasing prevalence of untreatable infections caused by antibiotic-resistant bacteria, the discovery of new drugs from natural products has become a hot research topic. The antibacterial activity of anthraquinones widely distributed in traditional Chinese medicine has attracted much attention. Herein, the structure and activity relationships (SARs) of anthraquinones as bacteriostatic agents are reviewed and elucidated. The substituents of anthraquinone and its derivatives are closely related to their antibacterial activities. The stronger the polarity of anthraquinone substituents is, the more potent the antibacterial effects appear. The presence of hydroxyl groups is not necessary for the antibacterial activity of hydroxyanthraquinone derivatives. Substitution of di-isopentenyl groups can improve the antibacterial activity of anthraquinone derivatives. The rigid plane structure of anthraquinone lowers its water solubility and results in the reduced activity. Meanwhile, the antibacterial mechanisms of anthraquinone and its analogs are explored, mainly including biofilm formation inhibition, destruction of the cell wall, endotoxin inhibition, inhibition of nucleic acid and protein synthesis, and blockage of energy metabolism and other substances.
Collapse
Affiliation(s)
- Tang Qun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| | - Tiantian Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University 440113 Guangzhou China
| | - Jiongkai Hao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
| | - Chunmei Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Keyu Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Jing Xu
- Huanghua Agricultural and Rural Development Bureau Bohai New Area 061100 Hebei China
| | - Xiaoyang Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences 200241 Shanghai China
- Key laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Research Institute, Chinese Academy of Agricultural Sciences Shanghai 200241 China
| |
Collapse
|
6
|
Raghuveer D, Pai VV, Murali TS, Nayak R. Exploring Anthraquinones as Antibacterial and Antifungal agents. ChemistrySelect 2023. [DOI: 10.1002/slct.202204537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Affiliation(s)
- Dhanush Raghuveer
- Department of Biotechnology Manipal School of Life Sciences Manipal Academy of Higher Education Manipal 576104 India
| | - V. Varsha Pai
- Department of Biotechnology Manipal School of Life Sciences Manipal Academy of Higher Education Manipal 576104 India
| | - Thokur Sreepathy Murali
- Department of Biotechnology Manipal School of Life Sciences Manipal Academy of Higher Education Manipal 576104 India
| | - Roopa Nayak
- Department of Biotechnology Manipal School of Life Sciences Manipal Academy of Higher Education Manipal 576104 India
| |
Collapse
|
7
|
Xue B, Shen Y, Zuo J, Song D, Fan Q, Zhang X, Yi L, Wang Y. Bringing Antimicrobial Strategies to a New Level: The Quorum Sensing System as a Target to Control Streptococcus suis. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122006. [PMID: 36556371 PMCID: PMC9782415 DOI: 10.3390/life12122006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
Streptococcus suis (S. suis) is an important zoonotic pathogen. It mainly uses quorum sensing (QS) to adapt to complex and changeable environments. QS is a universal cell-to-cell communication system that has been widely studied for its physiological functions, including the regulation of bacterial adhesion, virulence, and biofilm formation. Quorum sensing inhibitors (QSIs) are highly effective at interfering with the QS system and bacteria have trouble developing resistance to them. We review the current research status of the S. suis LuxS/AI-2 QS system and QSIs. Studies showed that by inhibiting the formation of AI-2, targeting the LuxS protein, inhibiting the expression of luxs gene can control the LuxS/AI-2 QS system of S. suis. Other potential QSIs targets are summarized, which may be preventing and treating S. suis infections, including AI-2 production, transmission, LuxS protein, blockage of AI-2 binding to receptors, AI-2-mediated QS. Since antibiotics are becoming increasingly ineffective due to the emergence of resistant bacteria, including S. suis, it is thus critical to find new antibacterial drugs with different mechanisms of action. QSIs provide hope for the development of such drugs.
Collapse
Affiliation(s)
- Bingqian Xue
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang 471000, China
| | - Yamin Shen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang 471000, China
| | - Jing Zuo
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang 471000, China
| | - Dong Song
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang 471000, China
| | - Qingying Fan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang 471000, China
| | - Xiaoling Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang 471000, China
| | - Li Yi
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang 471000, China
- College of Life Science, Luoyang Normal University, Luoyang 471000, China
- Correspondence: (L.Y.); (Y.W.)
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang 471000, China
- Correspondence: (L.Y.); (Y.W.)
| |
Collapse
|
8
|
Quinones as an Efficient Molecular Scaffold in the Antibacterial/Antifungal or Antitumoral Arsenal. Int J Mol Sci 2022; 23:ijms232214108. [PMID: 36430585 PMCID: PMC9697455 DOI: 10.3390/ijms232214108] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
Quinone-based compounds constitute several general classes of antibiotics that have long shown unwavering efficiency against both Gram-positive and Gram-negative microbial infections. These quinone-based antibiotics are increasingly popular due to their natural origins and are used in natural beverages from herbs or plants in African, Chinese and Indian traditional medicines to treat and prevent various diseases. Quinone-based antibiotics display different bioactive profiles depending on their structures and exert specific biocidal and anti-biofilm properties, and based on recent literature, will be discussed herein.
Collapse
|
9
|
Zhou Y, Yu F, Chen M, Zhang Y, Qu Q, Wei Y, Xie C, Wu T, Liu Y, Zhang Z, Chen X, Dong C, Che R, Li Y. Tylosin Inhibits Streptococcus suis Biofilm Formation by Interacting With the O-acetylserine (thiol)-lyase B CysM. Front Vet Sci 2022; 8:829899. [PMID: 35155655 PMCID: PMC8832016 DOI: 10.3389/fvets.2021.829899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/23/2022] Open
Abstract
Streptococcus suis (S. suis) can decrease its virulence or modify local conditions through biofilm formation, which promotes infection persistence in vivo. Biofilm formation is an important cause of chronic drug-resistant S. suis infection. The aim of this study was to evaluate whether tylosin effectively inhibits S. suis biofilm formation by interacting with O-acetylserine (thiol)-lyase B (CysM), a key enzymatic regulator of cysteine synthesis. Biofilm formation of the mutant (ΔcysM) strain was significantly lower compared to the wild-type ATCC 700794 strain. Tylosin inhibited cysM gene expression, decreased extracellular matrix contents, and reduced cysteine, homocysteine, and S-adenosylmethionine levels, indicating its potential value as an effective inhibitor of S. suis biofilm formation. Furthermore, using biolayer interferometry technology and fourier-transform infrared spectroscopy, we found that tylosin and CysM could be combined directly. Overall, these results provide evidence that tylosin inhibits S. suis biofilm formation by interacting with CysM.
Collapse
Affiliation(s)
- Yonghui Zhou
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Fei Yu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mo Chen
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuefeng Zhang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qianwei Qu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanru Wei
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chunmei Xie
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Tong Wu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanyan Liu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhiyun Zhang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xueying Chen
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chunliu Dong
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ruixiang Che
- College of Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- *Correspondence: Ruixiang Che
| | - Yanhua Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Yanhua Li
| |
Collapse
|
10
|
Li J, Fan Q, Jin M, Mao C, Zhang H, Zhang X, Sun L, Grenier D, Yi L, Hou X, Wang Y. Paeoniflorin reduce luxS/AI-2 system-controlled biofilm formation and virulence in Streptococcus suis. Virulence 2021; 12:3062-3073. [PMID: 34923916 PMCID: PMC8923065 DOI: 10.1080/21505594.2021.2010398] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Streptococcus suis (S. suis), more specifically serotype 2, is a bacterial pathogen that threatens the lives of pigs and humans. Like many other pathogens, S. suis exhibits quorum sensing (QS) system-controlled virulence factors, such as biofilm formation that complicates treatment. Therefore, impairing the QS involving LuxS/AI-2 cycle in S. suis, may be a promising alternative strategy for overcoming S. suis infections. In this study, we investigated paeoniflorin (PF), a monoterpenoid glycoside compound extracted from peony, as an inhibitor of S. suis LuxS/AI-2 system. At a sub-minimal inhibitory concentration (MIC) (1/16 MIC; 25 μg/ml), PF significantly reduced biofilm formation by S. suis through inhibition of extracellular polysaccharide (EPS) production, without affecting bacterial growth. Moreover, evidence was brought that PF reduces AI-2 activity in S. suis biofilm. Molecular docking indicated that LuxS may be the target of PF. Monitoring LuxS enzymatic activity confirmed that PF had a partial inhibitory effect. Finally, we showed that the use of PF in a mouse model can relieve S. suis infections. This study highlighted the anti-biofilm potential of PF against S. suis, and brought evidence that it may as an inhibitor of the LuxS/AI-2 system to prevent S. suis biofilm-related infections. PF can thus be used as a new type of natural biofilm inhibitor for clinical application.
Collapse
Affiliation(s)
- Jinpeng Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Qingying Fan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Manyu Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Chenlong Mao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Hui Zhang
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Xiaoling Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Liyun Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| | - Daniel Grenier
- Groupe de Recherche En Écologie Buccale (Greb), Faculté de Médecine Dentaire, Université Laval, Quebec City, Canada
| | - Li Yi
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China.,College of Life Science, Luoyang Normal University, Luoyang, China
| | - Xiaogai Hou
- College of Agriculture/College of Tree Peony, Henan University of Science and Technology Luoyang China
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, China
| |
Collapse
|
11
|
Đukanović S, Ganić T, Lončarević B, Cvetković S, Nikolić B, Tenji D, Randjelović D, Mitić-Ćulafić D. Elucidating the antibiofilm activity of Frangula emodin against Staphylococcus aureus biofilms. J Appl Microbiol 2021; 132:1840-1855. [PMID: 34779074 DOI: 10.1111/jam.15360] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/11/2021] [Accepted: 11/04/2021] [Indexed: 11/30/2022]
Abstract
AIMS Because the Staphylococcus aureus is one of the most well-known pathogens associated with medical devices and nosocomial infections, the aim of the study was to examine antibiofilm potential of emodin against it. METHODS AND RESULTS Antibacterial activity was examined through microdilution assay. Antibiofilm testing included crystal violet staining of biofilm biomass and morphology analysis by Atomic force microscopy (AFM). Furthermore, aerobic respiration was monitored using the Micro-Oxymax respirometer. For investigation of gene expression qRT-PCR was performed. Emodin demonstrated strong antibacterial activity and ability to inhibit biofilm formation of all tested strains. The effect on preformed biofilms was spotted in few strains. AFM revealed that emodin affects biofilm structure and roughness. Monitoring of respiration under emodin treatment in planktonic and biofilm form revealed that emodin influenced aerobic respiration. Moreover, qRT-PCR showed that emodin modulates expression of icaA, icaD, srrA and srrB genes, as well as RNAIII, and that this activity was strain-specific. CONCLUSION The results obtained in this study indicate the novel antibiofilm activity of emodin and its multiple pathways of action. SIGNIFICANCE AND IMPACT OF STUDY This is the first study that examined pathways through which emodin expressed its antibiofilm activity.
Collapse
Affiliation(s)
| | - Tea Ganić
- Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Branka Lončarević
- Institute for Chemistry, Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | | | - Biljana Nikolić
- Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Dina Tenji
- Faculty of Science, University of Novi Sad, Novi Sad, Serbia
| | - Danijela Randjelović
- Institute for Chemistry, Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
12
|
Zheng Q, Li S, Li X, Liu R. Advances in the study of emodin: an update on pharmacological properties and mechanistic basis. Chin Med 2021; 16:102. [PMID: 34629100 PMCID: PMC8504117 DOI: 10.1186/s13020-021-00509-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/17/2021] [Indexed: 12/21/2022] Open
Abstract
Rhei Radix et Rhizoma, also known as rhubarb or Da Huang, has been widely used as a spice and as traditional herbal medicine for centuries, and is currently marketed in China as the principal herbs in various prescriptions, such as Da-Huang-Zhe-Chong pills and Da-Huang-Qing-Wei pills. Emodin, a major bioactive anthraquinone derivative extracted from rhubarb, represents multiple health benefits in the treatment of a host of diseases, such as immune-inflammatory abnormality, tumor progression, bacterial or viral infections, and metabolic syndrome. Emerging evidence has made great strides in clarifying the multi-targeting therapeutic mechanisms underlying the efficacious therapeutic potential of emodin, including anti-inflammatory, immunomodulatory, anti-fibrosis, anti-tumor, anti-viral, anti-bacterial, and anti-diabetic properties. This comprehensive review aims to provide an updated summary of recent developments on these pharmacological efficacies and molecular mechanisms of emodin, with a focus on the underlying molecular targets and signaling networks. We also reviewed recent attempts to improve the pharmacokinetic properties and biological activities of emodin by structural modification and novel material-based targeted delivery. In conclusion, emodin still has great potential to become promising therapeutic options to immune and inflammation abnormality, organ fibrosis, common malignancy, pathogenic bacteria or virus infections, and endocrine disease or disorder. Scientifically addressing concerns regarding the poor bioavailability and vague molecular targets would significantly contribute to the widespread acceptance of rhubarb not only as a dietary supplement in food flavorings and colorings but also as a health-promoting TCM in the coming years.
Collapse
Affiliation(s)
- Qi Zheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Shuo Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China.
| |
Collapse
|
13
|
Pourhajibagher M, Rahimi-Esboei B, Ahmadi H, Bahador A. The anti-biofilm capability of nano-emodin-mediated sonodynamic therapy on multi-species biofilms produced by burn wound bacterial strains. Photodiagnosis Photodyn Ther 2021; 34:102288. [PMID: 33836275 DOI: 10.1016/j.pdpdt.2021.102288] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/10/2021] [Accepted: 04/02/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Management of burn wound infections (BWIs) is difficult due to the emergence of multidrug-resistant microorganisms. This study aimed to explore the anti-biofilm efficacy of sonodynamic therapy (SDT) using nano-emodin (N-EMO) against multi-species bacterial biofilms containing Staphylococcus aureus, Pseudomonas aeruginosa, and Acinetobacter baumannii. METHODS Following synthesis and confirmation of N-EMO as a sonosensitizer, the anti-biofilm efficacy of SDT against multi-species bacterial biofilms was determined using minimum inhibitory concentrations (MIC), minimum biofilm inhibitory concentration (MBIC), and minimal biofilm eradication concentration (MBEC) of N-EMO. The reduction of multi-species bacterial biofilms was then evaluated following the treatments using Log reduction and crystal violet (CV) assays. In addition, the expression profiling of abaI, agrA, and lasI genes using SDT with sub-MIC, sub-MBIC, and sub-MBEC of N-EMO was assessed. RESULTS Successful synthesis of N-EMO was confirmed through several characterization tests. As the results demonstrated, the MIC value of N-EMO for the multi-species bacterial suspension was 0.15 × 10-4 g/L, as well as, the MBEC value of N-EMO was 2.5 × 10-4 g/L, approximately 4-fold higher than that of MBIC (0.62 × 10-4 g/L). According to the CV assay, there were 57.8 %, 71.0 %, and 81.5 % reduction in the biofilm of multi-species bacterial growth following SDT using 1/128 MBEC, 1/16 MBIC, and 1/2 MIC of N-EMO, respectively. Log reductions analysis demonstrated that 1/2 MIC of N-EMO was more potent in inhibiting the biofilm growth of multi-species test bacteria by 5.725 ± 0.12 (99.9993 %). In this study, N-EMO-mediated SDT could obviously downregulate the gene expression of virulence factors (P < 0.05). The gene expression of lasI, agrA, and abaI were downregulated about 2.5-, 3.6-, and 5.5-fold; and 3.0-, 5.2-, and 7.4-fold following SDT with sub-MBIC and sub-MBEC of N-EMO, respectively. CONCLUSION These results highlight the potential of N-EMO-mediated SDT in inhibition of biofilm formation, degradation of formed biofilms, and reduction of virulence factor associated with biofilms of multi-species bacterial biofilms in BWIs.
Collapse
Affiliation(s)
- Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Rahimi-Esboei
- Department of Parasitology and Mycology, School of Medicine, Tonekabone Branch, Islamic Azad University, Tonekabone, Iran; Toxoplasmosis Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hanie Ahmadi
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology, Tehran, Iran
| | - Abbas Bahador
- Department of Medical Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Fellowship in Clinical Laboratory Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Tang Y, Bai J, Yang Y, Bai X, Bello-Onaghise G, Xu Y, Li Y. Effect of Syringopicroside Extracted from Syringa oblata Lindl on the Biofilm Formation of Streptococcus suis. Molecules 2021; 26:1295. [PMID: 33673668 PMCID: PMC7957517 DOI: 10.3390/molecules26051295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/21/2021] [Accepted: 02/24/2021] [Indexed: 11/29/2022] Open
Abstract
Syringopicroside is a natural drug with antibacterial activity, which is the main ingredient of Syringa oblata Lindl (S. oblata). In order to further develop the application of S. oblata and evaluate the ability of syringopicroside against Streptococcus suis (S. suis), this investigation first applied an ultrasonic-assisted method to extract syringopicroside, and then response surface methodology (RSM) was performed to get the optimum condition. Based on RSM analysis, a second-order polynomial equation about the syringopicroside yield and four variables, including ultrasonic power, time, temperature, and liquid-to-solid ratio, was purposed. Through RSM prediction and model verification experiments, the optimum conditions were determined, as follows: ultrasonic time was 63 min, temperature was 60 °C, a liquid-to-solid ratio was set to 63 mL/g, and ultrasonic power was 835 W. Under this condition, a high syringopicroside yield was obtained (3.07 ± 0.13 mg/g), which was not significantly different with a predicated value. After separation and purification by HPD 500 microporous resin, then mass spectrum was applied to identify the main ingredient in aqueous extract. A minimal inhibitory concentration (MIC) assay revealed the value against S. suis of syringopicroside was 2.56 µg/µL and syringopicroside with sub-inhibitory concentrations that could effectively inhibit biofilm formation of S. suis. Besides, scanning electron microscopy analysis indicated syringopicroside could destroy the multi-layered aggregation structure of S. suis. Finally, molecular docking analysis confirmed that syringopicroside was combined with Orfy protein of S. suis through hydrogen bonds, hydrophobic interaction, and π-π stacking.
Collapse
Affiliation(s)
- Yang Tang
- Department of Applied Chemistry, College of Art and Science, Northeast Agricultural University, Harbin 150030, China; (Y.T.); (J.B.); (Y.Y.); (X.B.)
| | - Jingwen Bai
- Department of Applied Chemistry, College of Art and Science, Northeast Agricultural University, Harbin 150030, China; (Y.T.); (J.B.); (Y.Y.); (X.B.)
| | - Yu Yang
- Department of Applied Chemistry, College of Art and Science, Northeast Agricultural University, Harbin 150030, China; (Y.T.); (J.B.); (Y.Y.); (X.B.)
| | - Xuedong Bai
- Department of Applied Chemistry, College of Art and Science, Northeast Agricultural University, Harbin 150030, China; (Y.T.); (J.B.); (Y.Y.); (X.B.)
| | - God’spower Bello-Onaghise
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China;
| | - Yaqin Xu
- Department of Applied Chemistry, College of Art and Science, Northeast Agricultural University, Harbin 150030, China; (Y.T.); (J.B.); (Y.Y.); (X.B.)
| | - Yanhua Li
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China;
| |
Collapse
|
15
|
Mishra R, Panda AK, De Mandal S, Shakeel M, Bisht SS, Khan J. Natural Anti-biofilm Agents: Strategies to Control Biofilm-Forming Pathogens. Front Microbiol 2020; 11:566325. [PMID: 33193155 PMCID: PMC7658412 DOI: 10.3389/fmicb.2020.566325] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Pathogenic microorganisms and their chronic pathogenicity are significant concerns in biomedical research. Biofilm-linked persistent infections are not easy to treat due to resident multidrug-resistant microbes. Low efficiency of various treatments and in vivo toxicity of available antibiotics drive the researchers toward the discovery of many effective natural anti-biofilm agents. Natural extracts and natural product-based anti-biofilm agents are more efficient than the chemically synthesized counterparts with lesser side effects. The present review primarily focuses on various natural anti-biofilm agents, i.e., phytochemicals, biosurfactants, antimicrobial peptides, and microbial enzymes along with their sources, mechanism of action via interfering in the quorum-sensing pathways, disruption of extracellular polymeric substance, adhesion mechanism, and their inhibitory concentrations existing in literature so far. This study provides a better understanding that a particular natural anti-biofilm molecule exhibits a different mode of actions and biofilm inhibitory activity against more than one pathogenic species. This information can be exploited further to improve the therapeutic strategy by a combination of more than one natural anti-biofilm compounds from diverse sources.
Collapse
Affiliation(s)
- Rojita Mishra
- Department of Botany, Polasara Science College, Polasara, India
| | | | - Surajit De Mandal
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, College of Agriculture, South China Agricultural University, Guangzhou, China
| | - Muhammad Shakeel
- Key Laboratory of Bio-Pesticide Innovation and Application of Guangdong Province, College of Agriculture, South China Agricultural University, Guangzhou, China
| | | | - Junaid Khan
- Department of Pharmacy, Sant Gahira Guru University, Ambikapur, India
| |
Collapse
|
16
|
Qu Q, Wang J, Cui W, Zhou Y, Xing X, Che R, Liu X, Chen X, Bello-Onaghise G, Dong C, Li Z, Li X, Li Y. In vitro activity and In vivo efficacy of Isoliquiritigenin against Staphylococcus xylosus ATCC 700404 by IGPD target. PLoS One 2019; 14:e0226260. [PMID: 31860659 PMCID: PMC6924684 DOI: 10.1371/journal.pone.0226260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/23/2019] [Indexed: 12/16/2022] Open
Abstract
Staphylococcus xylosus (S. xylosus) is a type of coagulase-negative Staphylococcus, which was previously considered as non-pathogenic. However, recent studies have linked it with cases of mastitis in cows. Isoliquiritigenin (ISL) is a bioactive compound with pharmacological functions including antibacterial activity. In this study, we evaluated the effect of ISL on S. xylosus in vitro and in vivo. The MIC of ISL against S. xylosus was 80 μg/mL. It was observed that sub-MICs of ISL (1/2MIC, 1/4MIC, 1/8MIC) significantly inhibited the formation of S. xylosus biofilm in vitro. Previous studies have observed that inhibiting imidazole glycerol phosphate dehydratase (IGPD) concomitantly inhibited biofilm formation in S. xylosus. So, we designed experiments to target the formation of IGPD or inhibits its activities in S. xylosus ATCC 700404. The results indicated that the activity of IGPD and its histidine content decreased significantly under 1/2 MIC (40 μg/mL) ISL, and the expression of IGPD gene (hisB) and IGPD protein was significantly down-regulated. Furthermore, Bio-layer interferometry experiments showed that ISL directly interacted with IGPD protein (with strong affinity; KD = 234 μM). In addition, molecular docking was used to predict the binding mode of ISL and IGPD. In vivo tests revealed that, ISL significantly reduced TNF-α and IL-6 levels, mitigated the destruction of the mammary glands and reversed the production of inflammatory cells in mice. The results of the study suggest that, ISL may inhibit S. xylosus growth by acting on IGPD, which can be used as a target protein to treat infections caused by S. xylosus.
Collapse
Affiliation(s)
- Qianwei Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Jinpeng Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Wenqiang Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Yonghui Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Xiaoxu Xing
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Ruixiang Che
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Xin Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Xueying Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - God’spower Bello-Onaghise
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Chunliu Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
| | - Zhengze Li
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, P. R. China
| | - Xiubo Li
- Feed Research Institute Chinese Academy of Agricultural Science, Harbin, Heilongjiang, P. R. China
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, P. R. China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, Heilongjiang, P. R. China
- * E-mail:
| |
Collapse
|
17
|
Liu YY, Chen XR, Wang JP, Cui WQ, Xing XX, Chen XY, Ding WY, God'spower BO, Eliphaz N, Sun MQ, Li YH. Transcriptomic analysis reveals flavonoid biosynthesis of Syringa oblata Lindl. in response to different light intensity. BMC PLANT BIOLOGY 2019; 19:487. [PMID: 31711412 PMCID: PMC6849326 DOI: 10.1186/s12870-019-2100-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/25/2019] [Indexed: 05/04/2023]
Abstract
BACKGROUND Hazy weather significantly increase air pollution and affect light intensity which may also affect medicinal plants growth. Syringa oblata Lindl. (S. oblata), an effective anti-biofilm medicinal plants, is also vulnerable to changes in plant photoperiods and other abiotic stress responses. Rutin, one of the flavonoids, is the main bioactive ingredient in S. oblata that inhibits Streptococcus suis biofilm formation. Thus, the present study aims to explore the biosynthesis and molecular basis of flavonoids in S. oblata in response to different light intensity. RESULTS In this study, it was shown that compared with natural (Z0) and 25% ~ 35% (Z2) light intensities, the rutin content of S. oblata under 50% ~ 60% (Z1) light intensity increased significantly. In addition, an integrated analysis of metabolome and transcriptome was performed using light intensity stress conditions from two kinds of light intensities which S. oblata was subjected to: Z0 and Z1. The results revealed that differential metabolites and genes were mainly related to the flavonoid biosynthetic pathway. We found out that 13 putative structural genes and a transcription factor bHLH were significantly up-regulated in Z1. Among them, integration analysis showed that 3 putative structural genes including 4CL1, CYP73A and CYP75B1 significantly up-regulated the rutin biosynthesis, suggesting that these putative genes may be involved in regulating the flavonoid biosynthetic pathway, thereby making them key target genes in the whole metabolic process. CONCLUSIONS The present study provided helpful information to search for the novel putative genes that are potential targets for S. oblata in response to light intensity.
Collapse
Affiliation(s)
- Yan-Yan Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xing-Ru Chen
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Jin-Peng Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Wen-Qiang Cui
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xiao-Xu Xing
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xue-Ying Chen
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Wen-Ya Ding
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Bello-Onaghise God'spower
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Nsabimana Eliphaz
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Meng-Qing Sun
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang, Harbin, Heilongjiang, 150030, People's Republic of China.
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China.
| |
Collapse
|
18
|
Papadopoulou A, Dalsgaard I, Wiklund T. Inhibition Activity of Compounds and Bacteriophages against Flavobacterium psychrophilum Biofilms In Vitro. JOURNAL OF AQUATIC ANIMAL HEALTH 2019; 31:225-238. [PMID: 31216387 DOI: 10.1002/aah.10069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 03/24/2019] [Indexed: 06/09/2023]
Abstract
Flavobacterium psychrophilum produces biofilms under laboratory conditions, and it has been inconclusively suggested that F. psychrophilum biofilms can be a potential reservoir for transmission of the pathogen to a fish population under fish farming conditions. Therefore, there is a need for anti-biofilm compounds. The main aim of this study was to determine the anti-biofilm properties of certain compounds and bacteriophages on F. psychrophilum biofilms under static conditions using a standard 96-well microtiter plate biofilm assay in vitro. Eight compounds (A-type proanthocyanidins, D-leucine, EDTA, emodin, fucoidan, L-alliin, parthenolide, and 2-aminoimidazole) at three sub-minimum inhibitory concentrations (sub-MICs), four bacteriophages (Fpv-3, Fpv-9, Fpv-10, and Fpv-21), and a phage combination (Fpv-9 + Fpv-10) were tested for inhibition of biofilm formation and reduction of the biomass of mature biofilms formed by two smooth isolates (P7-9/10 and P1-10B/10) and two rough isolates (P7-9/2R/10 and P1-10B/2R/10) of F. psychrophilum. The crystal violet staining method was used to stain the biofilms. Most of the compounds at sub-MICs inhibited the biofilm formation of mainly smooth isolates, attaining up to 80% inhibition. Additionally, the same reduction trend was also observed for 2-aminoimidazole, emodin, parthenolide, and D-leucine on the biomass of mature biofilms in a concentration-dependent manner. The anti-biofilm properties of the compounds are believed to lie in their ability to disturb the cellular interactions during biofilm formation and probably to cause cell dispersal in already formed biofilms. Lytic bacteriophages efficiently inhibited biofilm formation of F. psychrophilum, while they partially reduced the biomass of mature biofilms. However, the phage combination (Fpv-9 + Fpv-10) showed a successful reduction in the biomass of F. psychrophilum mature biofilms. We conclude that inhibiting compounds together with bacteriophages may supplement the use of disinfectants against bacterial biofilms (e.g., F. psychrophilum biofilms), leading to a reduced occurrence of bacterial coldwater disease outbreaks at fish farms.
Collapse
Affiliation(s)
- Anna Papadopoulou
- Laboratory of Aquatic Pathobiology, Environmental and Marine Biology, Åbo Akademi University, Tykistokatu 6, FI-20520, Turku, Finland
| | - Inger Dalsgaard
- National Institute of Aquatic Resources, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Tom Wiklund
- Laboratory of Aquatic Pathobiology, Environmental and Marine Biology, Åbo Akademi University, Tykistokatu 6, FI-20520, Turku, Finland
| |
Collapse
|
19
|
Ding WY, Zheng SD, Qin Y, Yu F, Bai JW, Cui WQ, Yu T, Chen XR, Bello-Onaghise G, Li YH. Chitosan Grafted With β-Cyclodextrin: Synthesis, Characterization, Antimicrobial Activity, and Role as Absorbefacient and Solubilizer. Front Chem 2019; 6:657. [PMID: 30687698 PMCID: PMC6335354 DOI: 10.3389/fchem.2018.00657] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/18/2018] [Indexed: 11/13/2022] Open
Abstract
We synthesized chitosan grafted with β-cyclodextrin (CD-g-CS) from mono-6-deoxy-6-(p-toluenesulfonyl)-β-cyclodextrin and chitosan. Two different amounts of immobilized β-cyclodextrin (β-CD) on CD-g-CS (QCD: 0.643 × 103 and 0.6 × 102 μmol/g) were investigated. The results showed that the amino contents of CD-g-CS with QCD = 0.643 × 103 and 0.6 × 102 μmol/g were 6.34 ± 0.072 and 9.41 ± 0.055%, respectively. Agar diffusion bioassay revealed that CD-g-CS (QCD = 0.6 × 102 μmol/g) was more active against Staphylococcus xylosus and Escherichia coli than CD-g-CS (QCD = 0.643 × 103 μmol/g). Cell membrane integrity tests and scanning electron microscopy observation revealed that the antimicrobial activity of CD-g-CS was attributed to membrane disruption and cell lysis. Uptake tests showed that CD-g-CS promoted the uptake of doxorubicin hydrochloride by S. xylosus, particularly for CD-g-CS with QCD = 0.6 × 102 μmol/g, and the effect was concentration dependent. CD-g-CS (QCD = 0.6 × 102 and 0.643 × 103 μmol/g) also improved the aqueous solubilities of sulfadiazine, sulfamonomethoxine, and sulfamethoxazole. These findings provide a clear understanding of CD-g-CS and are of great importance for reducing the dosage of antibiotics and antibiotic residues in animal-derived foods. The results also provide a reliable, direct, and scientific theoretical basis for its wide application in the livestock industry.
Collapse
Affiliation(s)
- Wen-Ya Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China.,Northeastern Science Inspection Station, China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology, Northeast Agricultural University, Harbin, China
| | - Si-Di Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yue Qin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Fei Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Jing-Wen Bai
- College of Science, Northeast Agricultural University, Harbin, China
| | - Wen-Qiang Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Tao Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xing-Ru Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - God'spower Bello-Onaghise
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
20
|
Shi C, Li M, Muhammad I, Ma X, Chang Y, Li R, Li C, He J, Liu F. Combination of berberine and ciprofloxacin reduces multi-resistant Salmonella strain biofilm formation by depressing mRNA expressions of luxS, rpoE, and ompR. J Vet Sci 2018; 19:808-816. [PMID: 30304890 PMCID: PMC6265579 DOI: 10.4142/jvs.2018.19.6.808] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/13/2018] [Accepted: 10/01/2018] [Indexed: 12/30/2022] Open
Abstract
Bacterial biofilms have been demonstrated to be closely related to clinical infections and contribute to drug resistance. Berberine, which is the main component of Coptis chinensis, has been reported to have efficient antibacterial activity. This study aimed to investigate the potential effect of a combination of berberine with ciprofloxacin (CIP) to inhibit Salmonella biofilm formation and its effect on expressions of related genes (rpoE, luxS, and ompR). The fractional inhibitory concentration (FIC) index of the combination of berberine with CIP is 0.75 showing a synergistic antibacterial effect. The biofilm's adhesion rate and growth curve showed that the multi-resistant Salmonella strain had the potential to form a biofilm relative to that of strain CVCC528, and the antibiofilm effects were in a dose-dependent manner. Biofilm microstructures were rarely observed at 1/2 × MIC/FIC concentrations (MIC, minimal inhibition concentration), and the combination had a stronger antibiofilm effect than each of the antimicrobial agents used alone at 1/4 × FIC concentration. LuxS, rpoE, and ompR mRNA expressions were significantly repressed (p < 0.01) at 1/2 × MIC/FIC concentrations, and the berberine and CIP combination repressed mRNA expressions more strongly at the 1/4 × FIC concentration. The results indicate that the combination of berberine and CIP has a synergistic effect and is effective in inhibiting Salmonella biofilm formation via repression of luxS, rpoE, and ompR mRNA expressions.
Collapse
Affiliation(s)
- Chenxi Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Minmin Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Ishfaq Muhammad
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Xin Ma
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yicong Chang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Rui Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Changwen Li
- Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Jingshan He
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| | - Fangping Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin 150030, China
| |
Collapse
|
21
|
Liu YY, Chen XR, Gao LF, Chen M, Cui WQ, Ding WY, Chen XY, God'spower BO, Li YH. Spectrum-Effect Relationships Between the Bioactive Ingredient of Syringa oblata Lindl. Leaves and Its Role in Inhibiting the Biofilm Formation of Streptococcus suis. Front Pharmacol 2018; 9:570. [PMID: 29922159 PMCID: PMC5996274 DOI: 10.3389/fphar.2018.00570] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022] Open
Abstract
Syringa oblata Lindl. (S. oblata) has been used in herbal medicines for treating bacterial diseases. It is also thought to inhibit Streptococcus suis (S. suis) biofilm formation. However, due to the inherent nature of the complexity in its chemical properties, it is difficult to understand the possible bioactive ingredients of S. oblata. The spectrum-effect relationships method was applied to screen the main active ingredients in S. oblata obtained from Heilongjiang Province based on gray relational analysis. The results revealed that Sub-MICs obtained from 10 batches of S. oblata could inhibit biofilm formation by S. suis. Gray relational analysis revealed variations in the contents of 15 main peaks and rutin was discovered to be the main active ingredient. Then, the function of rutin was further verified by inhibiting S. suis biofilm formation using crystal violet staining. Computational studies revealed that rutin may target the chloramphenicol acetyltransferase protein in the biofilm formation of S. suis. In conclusion, this study revealed that the spectrum-effect relationships and computational studies are useful tools to associate the active ingredient with the potential anti-biofilm effects of S. oblata. Here, our findings would provide foundation for the further understanding of the mechanism of S. oblata intervention in biofilm formation.
Collapse
Affiliation(s)
- Yan-Yan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xing-Ru Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Ling-Fei Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Mo Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Wen-Qiang Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Wen-Ya Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xue-Ying Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Bello-Onaghise God'spower
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
22
|
Li YH, Zhou YH, Ren YZ, Xu CG, Liu X, Liu B, Chen JQ, Ding WY, Zhao YL, Yang YB, Wang S, Liu D. Inhibition of Streptococcus suis Adhesion and Biofilm Formation in Vitro by Water Extracts of Rhizoma Coptidis. Front Pharmacol 2018; 9:371. [PMID: 29713285 PMCID: PMC5911698 DOI: 10.3389/fphar.2018.00371] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 03/29/2018] [Indexed: 12/12/2022] Open
Abstract
Streptococcus suis is difficult to treat and responsible for various infections in humans and pigs. It can also form biofilms and induce persistent infections. Rhizoma Coptidis is a medicinal plant widely used in Traditional Chinese Medicine. Although the inhibitory effects of Rhizoma Coptidis on biofilm formation have been investigated in several studies, the ability of Rhizoma Coptidis to inhibit S. suis biofilm formation and the underlying mechanisms have not yet been reported. In this study, we showed that sub-minimal inhibitory concentrations (25 and 50 μg mL-1) of water extracts of Rhizoma Coptidis (Coptis deltoidea C.Y.Cheng & P.K.Hsiao, obtained from Sichuan Province) were sufficient to inhibit biofilm formation, as shown in the tissue culture plate (TCP) method and scanning electron microscopy. Real-time PCR and iTRAQ were used to measure gene and protein expression in S. suis. Sub-minimum inhibitory concentrations (25 and 50 μg mL-1) of Rhizoma Coptidis water extracts inhibited S. suis adhesion significantly in an anti-adherence assay. Some genes, such as gapdh, sly, and mrp, and proteins, such as antigen-like protein, CPS16V, and methyltransferase H, involved in adhesion were significantly modulated in cells treated with 50 μg mL-1 of Rhizoma Coptidis water extracts compared to untreated cells. The results from this study suggest that compounds in Rhizoma Coptidis water extracts play an important role in inhibiting adhesion of S. suis cells and, therefore, biofilm formation.
Collapse
Affiliation(s)
- Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yong-Hui Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yong-Zhi Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Chang-Geng Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xin Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Bing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Jian-Qing Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Wen-Ya Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yu-Lin Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yan-Bei Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Shuai Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Di Liu
- Heilongjiang Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
23
|
Ding W, Sun J, Lian H, Xu C, Liu X, Zheng S, Zhang D, Han X, Liu Y, Chen X, God′spower BO, Li Y. The Influence of Shuttle-Shape Emodin Nanoparticles on the Streptococcus suis Biofilm. Front Pharmacol 2018; 9:227. [PMID: 29593544 PMCID: PMC5859365 DOI: 10.3389/fphar.2018.00227] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/28/2018] [Indexed: 12/12/2022] Open
Abstract
Biofilm is one of the most important physiological protective barriers of the Streptococcus suis (S. suis), and it is also one of the primary causes of hindrance to drug infiltration, reduction of bactericidal effects, and the development of antibiotic resistance. In order to intervene or eliminate S. suis biofilm, shuttle-shape emodin-loaded nanoparticles were developed in our study. The emodin nanoparticles were prepared by emodin and gelatin-cyclodextrin which was synthesized as drug carrier, and the nanoparticles were 174 nm in size, -4.64 mv in zeta potential, and exhibited a sustained emodin release. Moreover, the delivery kinetics of nanoparticles were also explored in our study. The confocal laser scanning microscopy and colony forming unit enumeration experiment indicated that nanoparticles could increase drug infiltration and uptake by biofilm. The flow cytometry system analysis showed that nanoparticles could be up taken by 99% of the bacteria cells. TCP assay and scanning electron microscopy showed that the nanoparticles had better effect on biofilm inhibition and elimination when compared with emodin solution. These results revealed that the emodin nanoparticles had a better therapeutic effect on the S. suis biofilm in vitro.
Collapse
Affiliation(s)
- Wenya Ding
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Jin Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - He Lian
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Changgeng Xu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xin Liu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Sidi Zheng
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Dong Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaopeng Han
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanyan Liu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xueying Chen
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Bello O. God′spower
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yanhua Li
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
24
|
Wang Y, Wang X, Jiang W, Wang K, Luo J, Li W, Zhou X, Zhang L. Antimicrobial peptide GH12 suppresses cariogenic virulence factors of Streptococcus mutans. J Oral Microbiol 2018; 10:1442089. [PMID: 29503706 PMCID: PMC5827641 DOI: 10.1080/20002297.2018.1442089] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/14/2018] [Indexed: 02/05/2023] Open
Abstract
Cariogenic virulence factors of Streptococcus mutans include acidogenicity, aciduricity, and extracellular polysaccharides (EPS) synthesis. The de novo designed antimicrobial peptide GH12 has shown bactericidal effects on S. mutans, but its interaction with virulence and regulatory systems of S. mutans remains to be elucidated. The objectives were to investigate the effects of GH12 on virulence factors of S. mutans, and further explore the function mechanisms at enzymatic and transcriptional levels. To avoid decrease in bacterial viability, we limited GH12 to subinhibitory levels. We evaluated effects of GH12 on acidogenicity of S. mutans by pH drop, lactic acid measurement and lactate dehydrogenase (LDH) assay, on aciduricity through survival rate at pH 5.0 and F1F0-ATPase assay, and on EPS synthesis using quantitative measurement, morphology observation, vertical distribution analyses and biomass calculation. Afterwards, we conducted quantitative real-time PCR to acquire the expression profile of related genes. GH12 at 1/2 MIC (4 mg/L) inhibited acid production, survival rate, EPS synthesis, and biofilm formation. The enzymatic activity of LDH and F1F0-ATPase was inhibited, and ldh, gtfBCD, vicR, liaR, and comDE genes were significantly downregulated. In conclusion, GH12 inhibited virulence factors of S. mutans, through reducing the activity of related enzymes, downregulating virulence genes, and inactivating specific regulatory systems.
Collapse
Affiliation(s)
- Yufei Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiuqing Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wentao Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kun Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junyuan Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Chen XR, Wang XT, Hao MQ, Zhou YH, Cui WQ, Xing XX, Xu CG, Bai JW, Li YH. Homology Modeling and Virtual Screening to Discover Potent Inhibitors Targeting the Imidazole Glycerophosphate Dehydratase Protein in Staphylococcus xylosus. Front Chem 2017; 5:98. [PMID: 29177138 PMCID: PMC5686052 DOI: 10.3389/fchem.2017.00098] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/30/2017] [Indexed: 01/11/2023] Open
Abstract
The imidazole glycerophosphate dehydratase (IGPD) protein is a therapeutic target for herbicide discovery. It is also regarded as a possible target in Staphylococcus xylosus (S. xylosus) for solving mastitis in the dairy cow. The 3D structure of IGPD protein is essential for discovering novel inhibitors during high-throughput virtual screening. However, to date, the 3D structure of IGPD protein of S. xylosus has not been solved. In this study, a series of computational techniques including homology modeling, Ramachandran Plots, and Verify 3D were performed in order to construct an appropriate 3D model of IGPD protein of S. xylosus. Nine hits were identified from 2,500 compounds by docking studies. Then, these nine compounds were first tested in vitro in S. xylosus biofilm formation using crystal violet staining. One of the potential compounds, baicalin was shown to significantly inhibit S. xylosus biofilm formation. Finally, the baicalin was further evaluated, which showed better inhibition of biofilm formation capability in S. xylosus by scanning electron microscopy. Hence, we have predicted the structure of IGPD protein of S. xylosus using computational techniques. We further discovered the IGPD protein was targeted by baicalin compound which inhibited the biofilm formation in S. xylosus. Our findings here would provide implications for the further development of novel IGPD inhibitors for the treatment of dairy mastitis.
Collapse
Affiliation(s)
- Xing-Ru Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xiao-Ting Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Mei-Qi Hao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Yong-Hui Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Wen-Qiang Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Xiao-Xu Xing
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Chang-Geng Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Jing-Wen Bai
- College of Science, Northeast Agricultural University, Harbin, China
| | - Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
26
|
Li L, Tian Y, Yu J, Song X, Jia R, Cui Q, Tong W, Zou Y, Li L, Yin L, Liang X, He C, Yue G, Ye G, Zhao L, Shi F, Lv C, Cao S, Yin Z. iTRAQ-based quantitative proteomic analysis reveals multiple effects of Emodin to Haemophilus parasuis. J Proteomics 2017; 166:39-47. [PMID: 28679110 DOI: 10.1016/j.jprot.2017.06.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 06/09/2017] [Accepted: 06/21/2017] [Indexed: 12/20/2022]
Abstract
UNLABELLED Haemophilus parasuis, a symbiotic bacteria of upper respiratory tract of swine, is the etiological agent of Glässer's disease, which is characterized by fibrinous polyserositis. Emodin, exhibits antibacterial activity against H. parasuis, yet the action mode has not been fully understood. In present study, isobaric tag for relative and absolute quantification (iTRAQ) method was applied to analyze the global protein alteration of H. parasuis in response to 16μg/mL Emodin. In total, 338 proteins exhibiting significant differential expressions were identified. It was speculated that, through application of bioinformatics analysis to theses differentially expressed proteins, Emodin mainly inhibited some key proteins expression of ABC transport system, carbohydrate metabolism pathway and bacterial cell division by inhibiting the ribosome synthesis, resulting in the growth inhibition of H. parasuis. Remarkably, nine virulence-associated proteins were detected differently expressed, further experiments revealed that after treatment with Emodin, H. parasuis could be inhibited to adhere to and invade into porcine kidney epithelial cells (PK-15 line) and exhibited increased sensitivity to serum complement in a concentration-dependent manner. Phagocytosis assay showed Emodin also could enhance phagocytic activity of porcine alveolar macrophages PAM to H. parasuis. These results indicated that Emodin also can attenuate virulence of H. parasuis and reduce infection. BIOLOGICAL SIGNIFICANCE The Glässer's disease caused by H. parasuis has become a typical bacterial disease and cause serious economic loss to the swine industry around the world. Antibiotics are extensively used to control the infection, but increasing antibiotic resistance has been a severe problem. Hence, novel treatment agents are needed. So far, few antibacterial agents were reported that could control H. parasuis infection. In the present study, the state-of-the-art quantitative proteomic technology was applied to uncover underlying action mechanism of Emodin. This study extends understanding of antibacterial effect of Emodin to H. parasuis at molecular level and provides useful information for further investigations. Moreover, our results provide theoretical foundation for the practical application of Emodin.
Collapse
Affiliation(s)
- Li Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ye Tian
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiankang Yu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Renyong Jia
- Key laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Qiankun Cui
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wenzhi Tong
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Changliang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Guizhou Yue
- College of Science, Sichuan Agricultural University, Ya'an 625014, China
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ling Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Fei Shi
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Cheng Lv
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Sanjie Cao
- Key laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; Key laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
27
|
Xu CG, Yang YB, Zhou YH, Hao MQ, Ren YZ, Wang XT, Chen JQ, Muhammad I, Wang S, Liu D, Li XB, Li YH. Comparative Proteomic Analysis Provides insight into the Key Proteins as Possible Targets Involved in Aspirin Inhibiting Biofilm Formation of Staphylococcus xylosus. Front Pharmacol 2017; 8:543. [PMID: 28871227 PMCID: PMC5566577 DOI: 10.3389/fphar.2017.00543] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 08/04/2017] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus xylosus is an opportunistic pathogen that causes infection in humans and cow mastitis. And S. xylosus possesses a strong ability to form biofilms in vitro. As biofilm formation facilitates resistance to antimicrobial agents, the discovery of new medicinal properties for classic drugs is highly desired. Aspirin, which is the most common active component of non-steroidal anti-inflammatory compounds, affects the biofilm-forming capacity of various bacterial species. We have found that aspirin effectively inhibits biofilm formation of S. xylosus by Crystal violet (CV) staining and scanning electron microscopy analyses. The present study sought to elucidate possible targets of aspirin in suppressing S. xylosus biofilm formation. Based on an isobaric tag for relative and absolute quantitation (iTRAQ) fold-change of >1.2 or <0.8 (P-value < 0.05), 178 differentially expressed proteins, 111 down-regulated and 67 up-regulated, were identified after application of aspirin to cells at a 1/2 minimal inhibitory concentration. Gene ontology analysis indicated enrichment in metabolic processes for the majority of the differentially expressed proteins. We then used the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database to analyze a large number of differentially expressed proteins and identified genes involved in biosynthesis of amino acids pathway, carbon metabolism (pentose phosphate and glycolytic pathways, tricarboxylic acid cycle) and nitrogen metabolism (histidine metabolism). These novel proteins represent candidate targets in aspirin-mediated inhibition of S. xylosus biofilm formation at sub-MIC levels. The findings lay the foundation for further studies to identify potential aspirin targets.
Collapse
Affiliation(s)
- Chang-Geng Xu
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Yan-Bei Yang
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Yong-Hui Zhou
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Mei-Qi Hao
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Yong-Zhi Ren
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Xiao-Ting Wang
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Jian-Qing Chen
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Ishfaq Muhammad
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Shuai Wang
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Di Liu
- Heilongjiang Academy of Agricultural SciencesHarbin, China
| | - Xiu-Bo Li
- Feed Research Institute, Chinese Academy of Agricultural SciencesBeijing, China
| | - Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| |
Collapse
|
28
|
Wang S, Wang C, Gao L, Cai H, Zhou Y, Yang Y, Xu C, Ding W, Chen J, Muhammad I, Chen X, He X, Liu D, Li Y. Rutin Inhibits Streptococcus suis Biofilm Formation by Affecting CPS Biosynthesis. Front Pharmacol 2017; 8:379. [PMID: 28670278 PMCID: PMC5472726 DOI: 10.3389/fphar.2017.00379] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/31/2017] [Indexed: 11/28/2022] Open
Abstract
Streptococcus suis (S. suis) form biofilms and causes severe diseases in humans and pigs. Biofilms are communities of microbes embedded in a matrix of extracellular polymeric substances. Eradicating biofilms with the use of antibiotics or biocides is often ineffective and needs replacement with other potential agents. Compared to conventional agents, promising and potential alternatives are biofilm-inhibiting compounds without impairing growth. Here, we screened a S. suis adhesion inhibitor, rutin, derived from Syringa. Rutin, a kind of flavonoids, shows efficient biofilm inhibition of S. suis without impairing its growth. Capsular polysaccharides(CPS) are reported to be involved in its adherence to influence bacterial biofilm formation. We investigated the effect of rutin on S. suis CPS content and structure. The results showed that rutin was beneficial to improve the CPS content of S. suis without changing its structure. We further provided evidence that rutin specifically affected S. suis biofilm susceptibility by affecting CPS biosynthesis in vitro. The study explores the antibiofilm potential of rutin against S. suis which can be used as an adhesion inhibitor for the prevention of S. suis biofilm-related infections. Nevertheless, rutin could be used as a novel natural inhibitor of biolfilm and its molecular mechanism provide basis for its pharmacological and clinical applications.
Collapse
Affiliation(s)
- Shuai Wang
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Chang Wang
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Lingfei Gao
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Hua Cai
- Harbin Pharmaceutical Group Bio-Vaccine Co. Ltd. (Hayao Vaccine),Harbin, China
| | - Yonghui Zhou
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Yanbei Yang
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Changgeng Xu
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Wenya Ding
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Jianqing Chen
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Ishfaq Muhammad
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China
| | - Xueying Chen
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| | - Xinmiao He
- Harbin Pharmaceutical Group Bio-Vaccine Co. Ltd. (Hayao Vaccine),Harbin, China
| | - Di Liu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural UniversityHarbin, China
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural UniversityHarbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical DevelopmentHarbin, China
| |
Collapse
|
29
|
Yan X, Gu S, Shi Y, Cui X, Wen S, Ge J. The effect of emodin on Staphylococcus aureus strains in planktonic form and biofilm formation in vitro. Arch Microbiol 2017; 199:1267-1275. [PMID: 28616631 DOI: 10.1007/s00203-017-1396-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 05/06/2017] [Accepted: 06/05/2017] [Indexed: 10/19/2022]
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive pathogen and forms biofilm easily. Bacteria inside biofilms display an increased resistance to antibiotics and disinfectants. The objective of the current study was to assess the antimicrobial activities of emodin, 1,2,8-trihydroxy-6-methylanthraquinone, an anthraquinone derivative isolated from Polygonum cuspidatum and Rheum palmatum, against S. aureus CMCC26003 grown in planktonic and biofilm cultures in vitro. In addition, a possible synergistic effect between emodin and berberine chloride was evaluated. As quantified by crystal violet method, emodin significantly decreased S. aureus biofilm growth in a dose-dependent manner. The above findings were further supported by scanning electron microscopy. Moreover, the present study demonstrated that sub-MICs emodin obviously intervened the release of extracellular DNA and inhibited expression of the biofilm-related genes (cidA, icaA, dltB, agrA, sortaseA and sarA) by real-time RT-PCR. These results revealed a promising application for emodin as a therapeutic agent and an effective strategy to prevent S. aureus biofilm-related infections.
Collapse
Affiliation(s)
- Xin Yan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shanshan Gu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Yunjia Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xingyang Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shanshan Wen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Junwei Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
30
|
Bai J, Yang Y, Wang S, Gao L, Chen J, Ren Y, Ding W, Muhammad I, Li Y. Syringa oblata Lindl. Aqueous Extract Is a Potential Biofilm Inhibitor in S. suis. Front Pharmacol 2017; 8:26. [PMID: 28194111 PMCID: PMC5278344 DOI: 10.3389/fphar.2017.00026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/16/2017] [Indexed: 11/13/2022] Open
Abstract
Streptococcus suis (S. suis) is a zoonotic pathogen that causes severe disease symptoms in pigs and humans. Syringa oblata Lindl. distributed in the middle latitudes of Eurasia and North America were proved as the most development potential of Chinese Medicine. In this study, biofilm formation by S. suis decreased after growth with 1/2 MIC, 1/4 MIC, or 1/8 MIC of Syringa oblata Lindl. aqueous extract and rutin. Scanning electron microscopy analysis revealed the potential effect of Syringa oblata Lindl. aqueous extract and rutin against biofilm formation by S. suis. Using iTRAQ technology, comparative proteomic analyses was performed at two conditions: 1/2 MIC of Syringa oblata Lindl. aqueous extract treated and non-treated cells. The results revealed the existence of 28 proteins of varying amounts. We found that the majority of the proteins were related to cell growth and metabolism. We also found that Syringa oblata Lindl. Aqueous extract affected the synthesis enzymes. In summary, Syringa oblata Lindl. aqueous extract might be used to inhibit the biofilm formation effectively by S. suis, and the active ingredients of the Syringa oblate Lindl. aqueous extract is rutin. The content of rutin is 9.9 ± 0.089 mg/g dry weight.
Collapse
Affiliation(s)
- Jingwen Bai
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yanbei Yang
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Shuai Wang
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Lingfei Gao
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Jianqing Chen
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yongzhi Ren
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Wenya Ding
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Ishfaq Muhammad
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| |
Collapse
|
31
|
Yang YB, Chen JQ, Zhao YL, Bai JW, Ding WY, Zhou YH, Chen XY, Liu D, Li YH. Sub-MICs of Azithromycin Decrease Biofilm Formation of Streptococcus suis and Increase Capsular Polysaccharide Content of S. suis. Front Microbiol 2016; 7:1659. [PMID: 27812354 PMCID: PMC5072222 DOI: 10.3389/fmicb.2016.01659] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/05/2016] [Indexed: 01/13/2023] Open
Abstract
Streptococcus suis (S. suis) caused serious disease symptoms in humans and pigs. S. suis is able to form thick biofilms and this increases the difficulty of treatment. After growth with 1/2 minimal inhibitory concentration (MIC) of azithromycin, 1/4 MIC of azithromycin, or 1/8 MIC of azithromycin, biofilm formation of S. suis dose-dependently decreased in the present study. Furthermore, scanning electron microscopy analysis revealed the obvious effect of azithromycin against biofilm formation of S. suis. Especially, at two different conditions (1/2 MIC of azithromycin non-treated cells and treated cells), we carried out comparative proteomic analyses of cells by using iTRAQ technology. Finally, the results revealed the existence of 19 proteins of varying amounts. Interestingly, several cell surface proteins (such as ATP-binding cassette superfamily ATP-binding cassette transporter (G7SD52), CpsR (K0FG35), Cps1/2H (G8DTL7), CPS16F (E9NQ13), putative uncharacterized protein (G7SER0), NADP-dependent glyceraldehyde-3-phosphate dehydrogenase (G5L259), putative uncharacterized protein (G7S2D6), amino acid permease (B0M0G6), and NsuB (G5L351)) were found to be implicated in biofilm formation. More importantly, we also found that azithromycin affected expression of the genes cps1/2H, cpsR and cps16F. Especially, after growth with 1/2 MIC of azithromycin and 1/4 MIC of azithromycin, the capsular polysaccharide content of S. suis was significantly higher.
Collapse
Affiliation(s)
- Yan-Bei Yang
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Jian-Qing Chen
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yu-Lin Zhao
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Jing-Wen Bai
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Wen-Ya Ding
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yong-Hui Zhou
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Xue-Ying Chen
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Di Liu
- Heilongjiang Academy of Agricultural Sciences Harbin, China
| | - Yan-Hua Li
- College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| |
Collapse
|
32
|
Wang S, Yang Y, Zhao Y, Zhao H, Bai J, Chen J, Zhou Y, Wang C, Li Y. Sub-MIC Tylosin Inhibits Streptococcus suis Biofilm Formation and Results in Differential Protein Expression. Front Microbiol 2016; 7:384. [PMID: 27065957 PMCID: PMC4811924 DOI: 10.3389/fmicb.2016.00384] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 03/11/2016] [Indexed: 12/25/2022] Open
Abstract
Streptococcus suis (S.suis) is an important zoonotic pathogen that causes severe diseases in humans and pigs. Biofilms of S. suis can induce persistent infections that are difficult to treat. In this study, the effect of tylosin on biofilm formation of S. suis was investigated. 1/2 minimal inhibitory concentration (MIC) and 1/4 MIC of tylosin were shown to inhibit S. suis biofilm formation in vitro. By using the iTRAQ strategy, we compared the protein expression profiles of S. suis grown with sub-MIC tylosin treatment and with no treatment. A total of 1501 proteins were identified by iTRAQ. Ninety-six differentially expressed proteins were identified (Ratio > ±1.5, p < 0.05). Several metabolism proteins (such as phosphoglycerate kinase) and surface proteins (such as ABC transporter proteins) were found to be involved in biofilm formation. Our results indicated that S. suis metabolic regulation, cell surface proteins, and virulence proteins appear to be of importance in biofilm growth with sub-MIC tylosin treatment. Thus, our data revealed the rough regulation of biofilm formation that may provide a foundation for future research into mechanisms and targets.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yanbei Yang
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yulin Zhao
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Honghai Zhao
- Department of Biotechnology, Heilongjiang Vocational College for Nationalities Harbin, China
| | - Jingwen Bai
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Jianqing Chen
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yonghui Zhou
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Chang Wang
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| | - Yanhua Li
- Department of Veterinary Pharmacy, College of Veterinary Medicine, Northeast Agricultural University Harbin, China
| |
Collapse
|