1
|
Scharnow A, Solinski AE, Rowe S, Drechsel I, Zhang H, Shaw E, Page JE, Wu H, Sieber SA, Wuest WM. In Situ Biofilm Affinity-Based Protein Profiling Identifies the Streptococcal Hydrolase GbpB as the Target of a Carolacton-Inspired Chemical Probe. J Am Chem Soc 2024; 146:23449-23456. [PMID: 39133525 PMCID: PMC11345752 DOI: 10.1021/jacs.4c06658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
Natural products are important precursors for antibiotic drug design. These chemical scaffolds serve as synthetic inspiration for chemists who leverage their structures to develop novel antibacterials and chemical probes. We have previously studied carolacton, a natural product macrolactone fromSorangium cellulosum, and discovered a simplified derivative, A2, that maintained apparent biofilm inhibitory activity, although the biological target was unknown. Herein, we utilize affinity-based protein profiling (AfBPP) in situ during biofilm formation to identify the protein target using a photoexcitable cross-linking derivative of A2. From these studies, we identified glucan binding protein B (GbpB), a peptidoglycan hydrolase, as the primary target of A2. Further characterization of the interaction between A2 and GbpB, as well as PcsB, a closely related homologue from the more pathogenic S. pneumoniae, revealed binding to the catalytic CHAP (cysteine, histidine, aminopeptidase) domain. To the best of our knowledge, this is the first report of a small-molecule binder of a conserved and essential bacterial CHAP hydrolase, revealing its potential as an antibiotic target. This work also highlights A2 as a useful tool compound for streptococci and as an initial scaffold for the design of more potent CHAP binders.
Collapse
Affiliation(s)
- Amber
M. Scharnow
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Amy E. Solinski
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Sebastian Rowe
- Department
of Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Ines Drechsel
- Department
of Chemistry, Center for Functional Protein Assemblies, Technical University of Munich, Garching D-85747, Germany
| | - Hua Zhang
- Departments
of Pediatric Dentistry, Microbiology, Schools of Dentistry and Medicine, University of Alabama at Birmingham, Birmingham 35294, Alabama, United States
| | - Elana Shaw
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Julia E. Page
- Department
of Microbiology, Blavatnik Institute, Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - Hui Wu
- Departments
of Pediatric Dentistry, Microbiology, Schools of Dentistry and Medicine, University of Alabama at Birmingham, Birmingham 35294, Alabama, United States
| | - Stephan A. Sieber
- Department
of Chemistry, Center for Functional Protein Assemblies, Technical University of Munich, Garching D-85747, Germany
| | - William M. Wuest
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
2
|
Hussaini IM, Oyewole OA, Sulaiman MA, Dabban AI, Sulaiman AN, Tarek R. Microbial anti-biofilms: types and mechanism of action. Res Microbiol 2024; 175:104111. [PMID: 37844786 DOI: 10.1016/j.resmic.2023.104111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 10/18/2023]
Abstract
Biofilms have been recognized as a serious threat to public health as it protects microbes from antimicrobials, immune defence mechanisms, chemical treatments and nutritional stress. Biofilms are also a source of concern in industries and water treatment because their presence compromises the integrity of equipment. To overcome these problems, it is necessary to identify novel anti-biofilm compounds. Products of microorganisms have been identified as promising broad-spectrum anti-biofilm agents. These natural products include biosurfactants, antimicrobial peptides, enzymes and bioactive compounds. Anti-biofilm products of microbial origin are chemically diverse and possess a broad spectrum of activities against biofilms. The objective of this review is to give an overview of the different types of microbial anti-biofilm products and their mechanisms of action.
Collapse
Affiliation(s)
| | - Oluwafemi Adebayo Oyewole
- Department of Microbiology, School of Life Sciences, Federal University of Technology, Minna, Nigeria; African Center of Excellence for Mycotoxin and Food Safety, Federal University of Technology Minna, Nigeria.
| | | | | | - Asmau Nna Sulaiman
- Department of Microbiology, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Reham Tarek
- Department of Biotechnology, Cairo University, Egypt
| |
Collapse
|
3
|
Zhang H, Li B, Yang H, Tan Y, Tan X, Tang Y. Total Synthesis of Carolacton and Demethylcarolactons with Potent Antiviral Activity. Org Lett 2024; 26:370-375. [PMID: 38170945 DOI: 10.1021/acs.orglett.3c04038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Carolacton, a naturally occurring MTHFD1 inhibitor, exhibits potent inhibitory activity against various RNA viruses including SARS-CoV-2. Herein, we present a concise total synthesis of carolacton, featuring the Krische allylation, Marshall coupling, NHK coupling, and RCM reaction as key elements. Additionally, we have synthesized three simplified carolacton analogues, one of which, namely, 14-demethyl-carolacton, exhibited notable antiviral activity. The present work paves the way for further exploration of the therapeutic potential of carolacton and its analogues.
Collapse
Affiliation(s)
- Haoyu Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Bingsong Li
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Hongzhi Yang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Ya Tan
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Xu Tan
- Chinese Institutes for Medical Research, Beijing 100069, China
| | - Yefeng Tang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
4
|
Saggu SK, Nath A, Kumar S. Myxobacteria: biology and bioactive secondary metabolites. Res Microbiol 2023; 174:104079. [PMID: 37169232 DOI: 10.1016/j.resmic.2023.104079] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/22/2023] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
Myxobacteria are Gram-negative eubacteria and they thrive in a variety of habitats including soil rich in organic matter, rotting wood, animal dung and marine environment. Myxobacteria are a promising source of new compounds associated with diverse bioactive spectrum and unique mode of action. The genome information of myxobacteria has revealed many orphan biosynthetic pathways indicating that these bacteria can be the source of several novel natural products. In this review, we highlight the biology of myxobacteria with emphasis on their habitat, life cycle, isolation methods and enlist all the bioactive secondary metabolites purified till date and their mode of action.
Collapse
Affiliation(s)
- Sandeep Kaur Saggu
- Department of Biotechnology, Kanya Maha Vidyalaya, Jalandhar, Punjab, India - 144004.
| | - Amar Nath
- University Centre of Excellence in Research, Baba Farid University of Health Sciences, Faridkot, Punjab India 151203.
| | - Shiv Kumar
- Guru Gobind Singh Medical College, Baba Farid University of Health Sciences, Faridkot, Punjab India 151203.
| |
Collapse
|
5
|
Chen R, Du M, Liu C. Strategies for dispersion of cariogenic biofilms: applications and mechanisms. Front Microbiol 2022; 13:981203. [PMID: 36134140 PMCID: PMC9484479 DOI: 10.3389/fmicb.2022.981203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/11/2022] [Indexed: 11/05/2022] Open
Abstract
Bacteria residing within biofilms are more resistant to drugs than planktonic bacteria. They can thus play a significant role in the onset of chronic infections. Dispersion of biofilms is a promising avenue for the treatment of biofilm-associated diseases, such as dental caries. In this review, we summarize strategies for dispersion of cariogenic biofilms, including biofilm environment, signaling pathways, biological therapies, and nanovehicle-based adjuvant strategies. The mechanisms behind these strategies have been discussed from the components of oral biofilm. In the future, these strategies may provide great opportunities for the clinical treatment of dental diseases. Graphical Abstract.
Collapse
|
6
|
Sharma A, Kumar A, Babu V, Ali A, Katoch M. Myxobacteria from animal dung pellets collected from northwestern Himalayas: A new source of di-isobutyl phthalate. J Basic Microbiol 2021; 62:162-173. [PMID: 34923648 DOI: 10.1002/jobm.202100518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/03/2021] [Accepted: 12/03/2021] [Indexed: 11/10/2022]
Abstract
Myxobacteria have emerged as a rich manufacturer of a wide array of natural products captivating both the academic and drug discovery communities. Attempts to unearth novel bioactive, myxobacteria from unexploited habitats are far from exhaustion. This study reports the isolation of myxobacteria from dung pellets collected from various regions of northwestern Himalayas. The isolated myxobacteria were functionally characterized to evaluate their bioactive capability. Of all the isolates, ST/P/71 exhibited broad range activities such as anticancer against all the four human cancer cell lines with IC50 in range of 2.03-9.65 µg/ml, antimicrobial against all the tested human pathogens, also exhibiting biofilm inhibition with MBIC50 at 10.4 µg/ml against Salmonella typhimurium. Consequently, ST/P/71 was chosen for fermentation and isolation of bioactive secondary metabolite through semi-preparative HPLC. It yielded compound 1, characterized as di-isobutyl phthalate (DiBP) based on nuclear magnetic resonance (NMR) and mass data. DiBP exhibited promising cytotoxic activity against the lung cancer cell line (A549) at an IC50 values 3.09 µg/ml and biofilm inhibition activity against Bacillus subtilis and Salmonella typhimurium with MBIC50 2.703 and 9.263 µg/ml, respectively. ST/P/71 was identified as Myxococcus fulvus. Thus, M. fulvus ST/P/71 isolated from northwestern Himalayas is a new source of DiBP.
Collapse
Affiliation(s)
- Arushi Sharma
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Amit Kumar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,Quality Management and Instrumentation Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Vikash Babu
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Asif Ali
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
| | - Meenu Katoch
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
7
|
Shagufta, Ahmad I. An Update on Pharmacological Relevance and Chemical Synthesis of Natural Products and Derivatives with Anti SARS-CoV-2 Activity. ChemistrySelect 2021; 6:11502-11527. [PMID: 34909460 PMCID: PMC8661826 DOI: 10.1002/slct.202103301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/25/2021] [Indexed: 01/18/2023]
Abstract
Natural products recognized traditionally as a vital source of active constituents in pharmacotherapy. The COVID-19 infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is highly transmissible, pathogenic, and considered an ongoing global health emergency. The emergence of COVID-19 globally and the lack of adequate treatment brought attention towards herbal medicines, and scientists across the globe instigated the search for novel drugs from medicinal plants and natural products to tackle this deadly virus. The natural products rich in scaffold diversity and structural complexity are an excellent source for antiviral drug discovery. Recently the investigation of several natural products and their synthetic derivatives resulted in the identification of promising anti SARS-CoV-2 agents. This review article will highlight the pharmacological relevance and chemical synthesis of the recently discovered natural product and their synthetic analogs as SARS-CoV-2 inhibitors. The summarized information will pave the path for the natural product-based drug discovery of safe and potent antiviral agents, particularly against SARS-CoV-2.
Collapse
Affiliation(s)
- Shagufta
- Department of Mathematics and Natural SciencesSchool of Arts and SciencesAmerican University of Ras Al KhaimahRas Al Khaimah Road, P. O. Box10021Ras Al Khaimah, UAE
| | - Irshad Ahmad
- Department of Mathematics and Natural SciencesSchool of Arts and SciencesAmerican University of Ras Al KhaimahRas Al Khaimah Road, P. O. Box10021Ras Al Khaimah, UAE
| |
Collapse
|
8
|
Bhat MA, Mishra AK, Bhat MA, Banday MI, Bashir O, Rather IA, Rahman S, Shah AA, Jan AT. Myxobacteria as a Source of New Bioactive Compounds: A Perspective Study. Pharmaceutics 2021; 13:1265. [PMID: 34452226 PMCID: PMC8401837 DOI: 10.3390/pharmaceutics13081265] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/18/2022] Open
Abstract
Myxobacteria are unicellular, Gram-negative, soil-dwelling, gliding bacteria that belong to class δ-proteobacteria and order Myxococcales. They grow and proliferate by transverse fission under normal conditions, but form fruiting bodies which contain myxospores during unfavorable conditions. In view of the escalating problem of antibiotic resistance among disease-causing pathogens, it becomes mandatory to search for new antibiotics effective against such pathogens from natural sources. Among the different approaches, Myxobacteria, having a rich armor of secondary metabolites, preferably derivatives of polyketide synthases (PKSs) along with non-ribosomal peptide synthases (NRPSs) and their hybrids, are currently being explored as producers of new antibiotics. The Myxobacterial species are functionally characterized to assess their ability to produce antibacterial, antifungal, anticancer, antimalarial, immunosuppressive, cytotoxic and antioxidative bioactive compounds. In our study, we have found their compounds to be effective against a wide range of pathogens associated with the concurrence of different infectious diseases.
Collapse
Affiliation(s)
- Mudasir Ahmad Bhat
- Department of Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India;
| | | | - Mujtaba Aamir Bhat
- Department of Botany, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India;
| | - Mohammad Iqbal Banday
- Department of Microbiology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India;
| | - Ommer Bashir
- Department of School Education, Jammu 181205, Jammu and Kashmir, India;
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia;
| | - Safikur Rahman
- Department of Botany, MS College, BR Ambedkar Bihar University, Muzaffarpur 845401, Bihar, India;
| | - Ali Asghar Shah
- Department of Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India;
| | - Arif Tasleem Jan
- Department of Botany, Baba Ghulam Shah Badshah University, Rajouri 185234, Jammu and Kashmir, India;
| |
Collapse
|
9
|
Shrivastava A, Sharma RK. Myxobacteria and their products: current trends and future perspectives in industrial applications. Folia Microbiol (Praha) 2021; 66:483-507. [PMID: 34060028 DOI: 10.1007/s12223-021-00875-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 05/13/2021] [Indexed: 12/12/2022]
Abstract
Myxobacteria belong to a group of bacteria that are known for their well-developed communication system and synchronized or coordinated movement. This typical behavior of myxobacteria is mediated through secondary metabolites. They are capable of producing secondary metabolites belonging to several chemical classes with unique and wide spectrum of bioactivities. It is predominantly significant that myxobacteria specialize in mechanisms of action that are very rare with other producers. Most of the metabolites have been explored for their medical and pharmaceutical values while a lot of them are still unexplored. This review is an attempt to understand the role of potential metabolites produced by myxobacteria in different applications. Different myxobacterial metabolites have demonstrated antibacterial, antifungal, and antiviral properties along with cytotoxic activity against various cell lines. Beside their metabolites, these myxobacteria have also been discussed for better exploitation and implementation in different industrial sectors.
Collapse
Affiliation(s)
- Akansha Shrivastava
- Department of Biosciences, Manipal University Jaipur, Rajasthan, 303007, Jaipur, India
| | - Rakesh Kumar Sharma
- Department of Biosciences, Manipal University Jaipur, Rajasthan, 303007, Jaipur, India.
| |
Collapse
|
10
|
Yin W, Xu S, Wang Y, Zhang Y, Chou SH, Galperin MY, He J. Ways to control harmful biofilms: prevention, inhibition, and eradication. Crit Rev Microbiol 2020; 47:57-78. [PMID: 33356690 DOI: 10.1080/1040841x.2020.1842325] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Biofilms are complex microbial architectures that encase microbial cells in a matrix comprising self-produced extracellular polymeric substances. Microorganisms living in biofilms are much more resistant to hostile environments than their planktonic counterparts and exhibit enhanced resistance against the microbicides. From the human perspective, biofilms can be classified into beneficial, neutral, and harmful. Harmful biofilms impact food safety, cause plant and animal diseases, and threaten medical fields, making it urgent to develop effective and robust strategies to control harmful biofilms. In this review, we discuss various strategies to control biofilm formation on infected tissues, implants, and medical devices. We classify the current strategies into three main categories: (i) changing the properties of susceptible surfaces to prevent biofilm formation; (ii) regulating signalling pathways to inhibit biofilm formation; (iii) applying external forces to eradicate the biofilm. We hope this review would motivate the development of innovative and effective strategies for controlling harmful biofilms.
Collapse
Affiliation(s)
- Wen Yin
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Siyang Xu
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Yiting Wang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Yuling Zhang
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Shan-Ho Chou
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Michael Y Galperin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Jin He
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| |
Collapse
|
11
|
Melander RJ, Basak AK, Melander C. Natural products as inspiration for the development of bacterial antibiofilm agents. Nat Prod Rep 2020; 37:1454-1477. [PMID: 32608431 PMCID: PMC7677205 DOI: 10.1039/d0np00022a] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Natural products have historically been a rich source of diverse chemical matter with numerous biological activities, and have played an important role in drug discovery in many areas including infectious disease. Synthetic and medicinal chemistry have been, and continue to be, important tools to realize the potential of natural products as therapeutics and as chemical probes. The formation of biofilms by bacteria in an infection setting is a significant factor in the recalcitrance of many bacterial infections, conferring increased tolerance to many antibiotics and to the host immune response, and as yet there are no approved therapeutics for combatting biofilm-based bacterial infections. Small molecules that interfere with the ability of bacteria to form and maintain biofilms can overcome antibiotic tolerance conferred by the biofilm phenotype, and have the potential to form combination therapies with conventional antibiotics. Many natural products with anti-biofilm activity have been identified from plants, microbes, and marine life, including: elligic acid glycosides, hamamelitannin, carolacton, skyllamycins, promysalin, phenazines, bromoageliferin, flustramine C, meridianin D, and brominated furanones. Total synthesis and medicinal chemistry programs have facilitated structure confirmation, identification of critical structural motifs, better understanding of mechanistic pathways, and the development of more potent, more accessible, or more pharmacologically favorable derivatives of anti-biofilm natural products.
Collapse
Affiliation(s)
- Roberta J Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| | | | | |
Collapse
|
12
|
Li B, Pan T, Lin H, Zhou Y. The enhancing antibiofilm activity of curcumin on Streptococcus mutans strains from severe early childhood caries. BMC Microbiol 2020; 20:286. [PMID: 32938379 PMCID: PMC7493841 DOI: 10.1186/s12866-020-01975-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Streptococcus mutans (S. mutans) is one of the main cariogenic bacteria for caries. It was found that the clinical strains of S. mutans isolated from caries active population have stronger cariogenic ability than the isolates from caries-free (CF) people. Previous studies have found that curcumin can inhibit biofilm formation of S. mutans UA159. The objective of this study is to explore the antibiofilm effect of curcumin on the clinical isolates of S. mutans from severe early childhood caries(SECC). RESULTS The isolates from SECC group had more biomass than CF group (t = 4.296, P < 0.001). The acidogenicity and aciduricity of the strains from two groups showed no significant difference. After treatment with curcumin, the viability of biofilm was reduced to 61.865% ± 7.108% in SECC and to 84.059% ± 10.227% in CF group at 24 h (P < 0.05). The net reduction of live bacteria and total bacteria in the SECC group was significantly higher than that of the CF group (live bacteria t = 3.305, P = 0.016; total bacteria t = 2.378, P = 0.045) at 5 min. For 24 h, the net reduction of live bacteria and total bacteria in the SECC group was significantly higher than that of the CF group (live bacteria t = 3.305, P = 0.016; total bacteria t = 2.378, P = 0.045). The reduction of biofilm thickness reduced significantly in 5 min (t = 4.110, P = 0.015) and in 24 h (t = 3.453, P = 0.014). Long-term (24 h) curcumin treatment inhibited the amount of EPS in SECC group from (25.980 ± 1.156) μm3/μm2 to (20.136 ± 1.042) μm3/μm2, the difference was statistically significant (t = 7.510, P < 0.001). The gene of gtfC, gtfD, ftf, gbpB, fruA and srtA in the CF group and the gtfB, gtfC, gtfD, ftf, gbpB, srtA in SECC group were respectively reduced after 5 min curcumin treatment. After 24 h treatment, the gtfB, gtfC, gtfD, ftf, gbpB, fruA and srtA in both two groups were downregulation, all the differences were statistically significant. CONCLUSIONS Curcumin has antibiofilm activity on clinical strains of S. mutans, especially for those isolated from SECC.
Collapse
Affiliation(s)
- Bingchun Li
- Department of Preventive Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou, 510055, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Ting Pan
- Department of Preventive Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou, 510055, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Huancai Lin
- Department of Preventive Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou, 510055, China. .,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Yan Zhou
- Department of Preventive Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou, 510055, China. .,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
13
|
Kreth J, Merritt J, Pfeifer C, Khajotia S, Ferracane J. Interaction between the Oral Microbiome and Dental Composite Biomaterials: Where We Are and Where We Should Go. J Dent Res 2020; 99:1140-1149. [PMID: 32479134 PMCID: PMC7443996 DOI: 10.1177/0022034520927690] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Dental composites are routinely placed as part of tooth restoration procedures. The integrity of the restoration is constantly challenged by the metabolic activities of the oral microbiome. This activity directly contributes to a less-than-desirable half-life for the dental composite formulations currently in use. Therefore, many new antimicrobial dental composites are being developed to counteract the microbial challenge. To ensure that these materials will resist microbiome-derived degradation, the model systems used for testing antimicrobial activities should be relevant to the in vivo environment. Here, we summarize the key steps in oral microbial colonization that should be considered in clinically relevant model systems. Oral microbial colonization is a clearly defined developmental process that starts with the formation of the acquired salivary pellicle on the tooth surface, a conditioned film that provides the critical attachment sites for the initial colonizers. Further development includes the integration of additional species and the formation of a diverse, polymicrobial mature biofilm. Biofilm development is discussed in the context of dental composites, and recent research is highlighted regarding the effect of antimicrobial composites on the composition of the oral microbiome. Future challenges are addressed, including the potential of antimicrobial resistance development and how this could be counteracted by detailed studies of microbiome composition and gene expression on dental composites. Ultimately, progress in this area will require interdisciplinary approaches to effectively mitigate the inevitable challenges that arise as new experimental bioactive composites are evaluated for potential clinical efficacy. Success in this area could have the added benefit of inspiring other fields in medically relevant materials research, since microbial colonization of medical implants and devices is a ubiquitous problem in the field.
Collapse
Affiliation(s)
- J. Kreth
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - J. Merritt
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - C.S. Pfeifer
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - S. Khajotia
- Department of Restorative Sciences, College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - J.L. Ferracane
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
14
|
Wuest WM, Solinski AE. Collaboration in Natural Product Total Synthesis: Carolacton – A Decade of Discovery. Synlett 2020. [DOI: 10.1055/s-0040-1707244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Carolacton, a macrocyclic natural product with impressive anti-biofilm biological activity, has been a focus in multiple research groups for the past decade. Chemists and biologists, alike, have been interested in uncovering the mechanism of action and have made great strides towards this goal. Carolacton causes cellular defects in Streptococcus mutans biofilm, which leads to decreases in cellular viability. As biological targets have been uncovered, synthetic chemists have devised synthetic routes that have helped uncover the important chemical functionalities that lead to biological activity. Herein, we discuss our synthetic collaboration that galvanized an entire research program around the natural product carolacton.
Collapse
Affiliation(s)
- William M. Wuest
- Department of Chemistry, Emory University
- Emory Antibiotic Resistance Center, Emory University School of Medicine
| | | |
Collapse
|
15
|
Khan F, Oloketuyi SF, Kim YM. Diversity of Bacteria and Bacterial Products as Antibiofilm and Antiquorum Sensing Drugs Against Pathogenic Bacteria. Curr Drug Targets 2020; 20:1156-1179. [PMID: 31020938 DOI: 10.2174/1389450120666190423161249] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/25/2019] [Accepted: 04/12/2019] [Indexed: 12/14/2022]
Abstract
The increase in antibiotic resistance of pathogenic bacteria has led to the development of new therapeutic approaches to inhibit biofilm formation as well as interfere quorum sensing (QS) signaling systems. The QS system is a phenomenon in which pathogenic bacteria produce signaling molecules that are involved in cell to cell communication, production of virulence factors, biofilm maturation, and several other functions. In the natural environment, several non-pathogenic bacteria are present as mixed population along with pathogenic bacteria and they control the behavior of microbial community by producing secondary metabolites. Similarly, non-pathogenic bacteria also take advantages of the QS signaling molecule as a sole carbon source for their growth through catabolism with enzymes. Several enzymes are produced by bacteria which disrupt the biofilm architecture by degrading the composition of extracellular polymeric substances (EPS) such as exopolysaccharide, extracellular- DNA and protein. Thus, the interference of QS system by bacterial metabolic products and enzymatic catalysis, modification of the QS signaling molecules as well as enzymatic disruption of biofilm architecture have been considered as the alternative therapeutic approaches. This review article elaborates on the diversity of different bacterial species with respect to their metabolic products as well as enzymes and their molecular modes of action. The bacterial enzymes and metabolic products will open new and promising perspectives for the development of strategies against the pathogenic bacterial infections.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, South Korea
| | | | - Young-Mog Kim
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, South Korea.,Department of Food Science and Technology, Pukyong National University, Busan 48513, South Korea
| |
Collapse
|
16
|
Campbell M, Cho CY, Ho A, Huang JY, Martin B, Gilbert ES. 4-Ethoxybenzoic acid inhibits Staphylococcus aureus biofilm formation and potentiates biofilm sensitivity to vancomycin. Int J Antimicrob Agents 2020; 56:106086. [PMID: 32663508 DOI: 10.1016/j.ijantimicag.2020.106086] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/11/2020] [Accepted: 07/05/2020] [Indexed: 11/15/2022]
Abstract
The adverse health effects of Staphylococcus aureus biofilm infections coupled with an increased global prevalence of antibiotic resistance highlight the need for novel anti-pathogenic, anti-biofilm compounds. The authors recently determined that ethyl-4-ethoxybenzoic acid (EEB) had anti-pathogenic, anti-biofilm activity. Based on this finding, a structure-activity analysis was undertaken to identify more effective compounds. Microtitre crystal violet assays followed by plate counts were conducted to measure the dose-dependent anti-biofilm and antimicrobial activities of 13 phenolic compounds related to EEB. By displaying these characteristics on a two-component plot, 4-ethoxybenzoic acid (4EB) and methyl gallate were identified as two anti-pathogenic, anti-biofilm compounds of interest. To characterize their mechanisms of activity, their effects on cell hydrophobicity, hemolysis activity, membrane integrity, extracellular polymeric substance production and vancomycin sensitivity were examined. Both 4EB and methyl gallate inhibited up to 87% of biofilm formation with minimal impact on the viability of stationary-phase cells or bacterial growth. Combination treatments of 4EB and vancomycin decreased the viability of biofilm-dwelling cells by up to 85% compared with vancomycin alone, indicating a synergistic effect. Methyl gallate did not potentiate vancomycin. 4EB decreased the percentage of hydrophobic cells in culture from 78% to 49%, indicating that 4EB may prevent biofilm formation by altering cell membrane hydrophobicity. These findings suggest that 4EB has potential as an anti-pathogenic, anti-biofilm agent for the prevention of S. aureus biofilms, or as a treatment for established biofilms when combined with antibiotics.
Collapse
Affiliation(s)
- Mariya Campbell
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Chih-Yun Cho
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Andrew Ho
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Jye-Yu Huang
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Brooke Martin
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Eric S Gilbert
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
17
|
Campbell M, Fathi R, Cheng S, Ho A, Gilbert E. Rhamnus prinoides
(gesho) stem extract prevents co‐culture biofilm formation by
Streptococcus mutans
and
Candida albicans. Lett Appl Microbiol 2020; 71:294-302. [DOI: 10.1111/lam.13307] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 04/16/2020] [Accepted: 04/27/2020] [Indexed: 01/02/2023]
Affiliation(s)
- M. Campbell
- Department of Biology Georgia State University Atlanta GA USA
| | - R. Fathi
- Department of Biology Georgia State University Atlanta GA USA
| | - S.Y. Cheng
- Department of Biology Georgia State University Atlanta GA USA
| | - A. Ho
- Department of Biology Georgia State University Atlanta GA USA
| | - E.S. Gilbert
- Department of Biology Georgia State University Atlanta GA USA
| |
Collapse
|
18
|
Luo Z, Ang MJY, Chan SY, Yi Z, Goh YY, Yan S, Tao J, Liu K, Li X, Zhang H, Huang W, Liu X. Combating the Coronavirus Pandemic: Early Detection, Medical Treatment, and a Concerted Effort by the Global Community. RESEARCH (WASHINGTON, D.C.) 2020; 2020:6925296. [PMID: 32607499 PMCID: PMC7315394 DOI: 10.34133/2020/6925296] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 01/08/2023]
Abstract
The World Health Organization (WHO) has declared the outbreak of 2019 novel coronavirus, known as 2019-nCoV, a pandemic, as the coronavirus has now infected over 2.6 million people globally and caused more than 185,000 fatalities as of April 23, 2020. Coronavirus disease 2019 (COVID-19) causes a respiratory illness with symptoms such as dry cough, fever, sudden loss of smell, and, in more severe cases, difficulty breathing. To date, there is no specific vaccine or treatment proven effective against this viral disease. Early and accurate diagnosis of COVID-19 is thus critical to curbing its spread and improving health outcomes. Reverse transcription-polymerase chain reaction (RT-PCR) is commonly used to detect the presence of COVID-19. Other techniques, such as recombinase polymerase amplification (RPA), loop-mediated isothermal amplification (LAMP), clustered regularly interspaced short palindromic repeats (CRISPR), and microfluidics, have allowed better disease diagnosis. Here, as part of the effort to expand screening capacity, we review advances and challenges in the rapid detection of COVID-19 by targeting nucleic acids, antigens, or antibodies. We also summarize potential treatments and vaccines against COVID-19 and discuss ongoing clinical trials of interventions to reduce viral progression.
Collapse
Affiliation(s)
- Zichao Luo
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Melgious Jin Yan Ang
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, Singapore 117456, Singapore
| | - Siew Yin Chan
- Frontiers Science Center for Flexible Electronics & Shaanxi Institute of Flexible Electronics, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhigao Yi
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Yi Yiing Goh
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, Singapore 117456, Singapore
| | - Shuangqian Yan
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Jun Tao
- Sports Medical Centre, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Kai Liu
- State Key Laboratory of Rare Earth Resource Utilization, Chang Chun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xiaosong Li
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Chang Chun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics & Shaanxi Institute of Flexible Electronics, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory of Flexible Electronics & Institute of Advanced Materials, Nanjing Tech University, Nanjing 211816, China
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Fuzhou 350807, China
| |
Collapse
|
19
|
Wang QQ, Li MX, Li C, Gu XX, Zheng MZ, Chen LX, Li H. Natural Products and Derivatives Targeting at Cancer Energy Metabolism: A Potential Treatment Strategy. Curr Med Sci 2020; 40:205-217. [PMID: 32337682 DOI: 10.1007/s11596-020-2165-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/14/2020] [Indexed: 12/13/2022]
Abstract
In the 1920s, Dr Otto Warburg first suggested the significant difference in energy metabolism between malignant cancer cells and adjacent normal cells. Tumor cells mainly adopt the glycolysis as energy source to maintain tumor cell growth and biosynthesis under aerobic conditions. Investigation on energy metabolism pathway in cancer cells has aroused the interest of cancer researchers all around the world. In recent years, plentiful studies suggest that targeting the peculiar cancer energy metabolic pathways, including glycolysis, mitochondrial respiration, amino acid metabolism, and fatty acid oxidation may be an effective strategy to starve cancer cells by blocking essential nutrients. Natural products (NPs) are considered as the "treasure trove of small molecules drugs" and have played an extremely remarkable role in the discovery and development of anticancer drugs. And numerous NPs have been reported to act on cancer energy metabolism targets. Herein, a comprehensive overview about cancer energy metabolism targets and their natural-occurring inhibitors is prepared.
Collapse
Affiliation(s)
- Qi-Qi Wang
- Wuya College of Innovation, School of Pharmaceutical Engineering, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ming-Xue Li
- Wuya College of Innovation, School of Pharmaceutical Engineering, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chen Li
- Wuya College of Innovation, School of Pharmaceutical Engineering, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiao-Xia Gu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng-Zhu Zheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Li-Xia Chen
- Wuya College of Innovation, School of Pharmaceutical Engineering, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Hua Li
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
20
|
Campbell M, Zhao W, Fathi R, Mihreteab M, Gilbert ES. Rhamnus prinoides (gesho): A source of diverse anti-biofilm activity. JOURNAL OF ETHNOPHARMACOLOGY 2019; 241:111955. [PMID: 31102615 DOI: 10.1016/j.jep.2019.111955] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 04/25/2019] [Accepted: 05/10/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhamnus prinoides (gesho) is an evergreen shrub from East Africa traditionally used for the treatment of illnesses including atopic dermatitis, ear, nose and throat infections, pneumonia, arthritis, brucellosis, flu, indigestion and fatigue. AIM OF THE STUDY Several of the conditions for which gesho is traditionally used are associated with communities of surface-attached microorganisms, or biofilms. We hypothesized that gesho has anti-biofilm activity. The principal aim of this study was to evaluate gesho-associated anti-biofilm activity and identify active compounds. MATERIALS AND METHODS Lyophilized ethanol and aqueous extracts were prepared from dried Rhamnus prinoides stems and leaves. Biofilm inhibition was measured by crystal violet staining and subsequent viability assays were conducted on growth agar. Chemical fractionation, chemical testing, Fourier transform infrared spectroscopy (FTIR) and gas chromatography-mass spectrometry (GC-MS) were used to isolate and identify active compounds. RESULTS Leaf and stem ethanol extracts significantly inhibited Staphylococcus aureus, Bacillus subtilis and Streptococcus mutans biofilm formation up to 99.9% and reduced planktonic cell growth up to 10 log units relative to untreated controls. The anti-biofilm activity of the ethanol stem extracts was due to a biocidal or bacteriostatic mechanism while bacteriostatic or anti-pathogenic mechanisms were attributed to the leaf ethanol extract. Gesho extracts showed activity against all three species tested but the treatment efficacy and mechanism were species dependent. Chemical fractionation and activity screens of the leaf ethanol extract identified ethyl 4-ethoxybenzoate and 4-hydroxy 4-methyl pentanone to be compounds with anti-biofilm activity. Ethyl 4-ethoxybenzoate activity was potentiated by DMSO. Notably, concentrations of both compounds were identified where biofilm formation was prevented without inhibition of cell growth; i.e. anti-pathogenic characteristics were evident. CONCLUSION Gesho leaf ethanol extract contains chemicals with anti-biofilm and bactericidal activities. This work lends support to the traditional use of gesho for treating topical infections and warrants further investigation into Rhamnus prinoides as a source of antibacterial and anti-biofilm agents.
Collapse
Affiliation(s)
- Mariya Campbell
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| | - Weilun Zhao
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| | - Raghda Fathi
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| | | | - Eric S Gilbert
- Department of Biology, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
21
|
Tan X, Chung T, Chen Y, Macarisin D, LaBorde L, Kovac J. The occurrence of Listeria monocytogenes is associated with built environment microbiota in three tree fruit processing facilities. MICROBIOME 2019; 7:115. [PMID: 31431193 PMCID: PMC6702733 DOI: 10.1186/s40168-019-0726-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/29/2019] [Indexed: 05/24/2023]
Abstract
BACKGROUND Multistate foodborne disease outbreaks and recalls of apples and apple products contaminated with Listeria monocytogenes demonstrate the need for improved pathogen control in the apple supply chain. Apple processing facilities have been identified in the past as potential sources of persisting L. monocytogenes contamination. In this study, we sought to understand the composition of microbiota in built apple and other tree fruit processing environments and its association with the occurrence of the foodborne pathogen L. monocytogenes. RESULTS Analysis of 117 samples collected from three apple and other tree fruit packing facilities (F1, F2, and F3) showed that facility F2 had a significantly higher L. monocytogenes occurrence compared to F1 and F3 (p < 0.01). The microbiota in facility F2 was distinct compared to facilities F1 and F3 as supported by the mean Shannon index for bacterial and fungal alpha diversities that was significantly lower in F2, compared to F1 and F3 (p < 0.01). Microbiota in F2 was uniquely predominated by bacterial family Pseudomonadaceae and fungal family Dipodascaceae. CONCLUSIONS The composition and diversity of microbiota and mycobiota present in the investigated built food processing environments may be indicative of persistent contamination with L. monocytogenes. These findings support the need for further investigation of the role of the microbial communities in the persistence of L. monocytogenes to support the optimization of L. monocytogenes control strategies in the apple supply chain.
Collapse
Affiliation(s)
- Xiaoqing Tan
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA
- Microbiome Center, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Taejung Chung
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA
- Microbiome Center, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yi Chen
- Center for Food Safety and Applied Nutrition, Food and Drug Administration, College Park, MD, 20740, USA
| | - Dumitru Macarisin
- Center for Food Safety and Applied Nutrition, Food and Drug Administration, College Park, MD, 20740, USA
| | - Luke LaBorde
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Jasna Kovac
- Department of Food Science, The Pennsylvania State University, University Park, PA, 16802, USA.
- Microbiome Center, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
22
|
Solinski AE, Scharnow AM, Fraboni AJ, Wuest WM. Synthetic Simplification of Carolacton Enables Chemical Genetic Studies in Streptococcus mutans. ACS Infect Dis 2019; 5:1480-1486. [PMID: 31243986 PMCID: PMC7169375 DOI: 10.1021/acsinfecdis.9b00213] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Understanding the broader biological impact of carolacton, a macrolactone natural product, has been ongoing for the past decade. Multiple studies have shown connections to regulatory systems, acid tolerance mechanisms, biofilm formation, and recently folate dehydrogenase (FolD). Progress elucidating the cause of biofilm-specific activity in Streptococcus mutans has been limited due to low-throughput analyses of carolacton-treated cells. We disclose the discovery of a simplified carolacton-inspired analog that demonstrates inhibitory activity against S. mutans biofilm cells. This discovery permitted a proof of concept chemical genetic screen of S. mutans mutants identifying the carbon catabolite protein A signaling pathway as a putative target.
Collapse
Affiliation(s)
- Amy E. Solinski
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Amber M. Scharnow
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Americo J. Fraboni
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - William M. Wuest
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
- Emory Antibiotic Resistance Center, Emory University School of Medicine, 201 Dowman Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
23
|
Scharnow AM, Solinski AE, Wuest WM. Targeting S. mutans biofilms: a perspective on preventing dental caries. MEDCHEMCOMM 2019; 10:1057-1067. [PMID: 31391878 PMCID: PMC6644389 DOI: 10.1039/c9md00015a] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023]
Abstract
The prevalence of biofilm diseases, and dental caries in particular, have encouraged extensive research on S. mutans biofilms, including methods of preventing its formation. Numerous small molecules with specific anti-biofilm activity against this pathogen have been isolated and synthesized. Generally, these molecules can be characterized into three categories: sucrose-dependent anti-adhesion, sucrose-independent anti-adhesion and cellular signaling interference. This review aims to provide an overview of the current small molecule strategies used for targeting S. mutans biofilms, and a perspective of the future for the field.
Collapse
Affiliation(s)
- Amber M Scharnow
- Emory University , Chemistry Department , 1515 Dickey Dr , Atlanta , GA 30322 , USA .
| | - Amy E Solinski
- Emory University , Chemistry Department , 1515 Dickey Dr , Atlanta , GA 30322 , USA .
| | - William M Wuest
- Emory University , Chemistry Department , 1515 Dickey Dr , Atlanta , GA 30322 , USA .
| |
Collapse
|
24
|
Conrads G, Wendt LK, Hetrodt F, Deng ZL, Pieper D, Abdelbary MMH, Barg A, Wagner-Döbler I, Apel C. Deep sequencing of biofilm microbiomes on dental composite materials. J Oral Microbiol 2019; 11:1617013. [PMID: 31143408 PMCID: PMC6522937 DOI: 10.1080/20002297.2019.1617013] [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: 10/02/2018] [Revised: 04/16/2019] [Accepted: 05/06/2019] [Indexed: 12/31/2022] Open
Abstract
Background: The microbiome on dental composites has not been studied in detail before. It has not been conclusively clarified whether restorative materials influence the oral microbiome. Methods: We used Illumina Miseq next-generation sequencing of the 16S V1-V2 region to compare the colonisation patterns of bovine enamel (BE) and the composite materials Grandio Flow (GF) and Grandio Blocs (GB) after 48 h in vivo in 14 volunteers. Applying a new method to maintain the oral microbiome ex vivo for 48 h also, we compared the microbiome on GF alone and with the new antimicrobial substance carolacton (GF+C). Results: All in vitro biofilm communities showed a higher diversity and richness than those grown in vivo but the very different atmospheric conditions must be considered. Contrary to expectations, there were only a few significant differences between BE and the composite materials GB and GF either in vivo or in vitro: Oribacterium, Peptostreptococcaceae [XI][G-1] and Streptococcus mutans were more prevalent and Megasphaera, Prevotella oulorum, Veillonella atypica, V. parvula, Gemella morbillorum, and Fusobacterium periodonticum were less prevalent on BE than on composites. In vivo, such preferences were only significant for Granulicatella adiacens (more prevalent on BE) and Fusobacterium nucleatum subsp. animalis (more prevalent on composites). On DNA sequence level, there were no significant differences between the biofilm communities on GF and GF+C. Conclusion: We found that the oral microbiome showed an increased richness when grown on various composites compared to BE in vitro, but otherwise changed only slightly independent of the in vivo or in vitro condition. Our new ex vivo biofilm model might be useful for pre-clinical testing of preventive strategies.
Collapse
Affiliation(s)
- Georg Conrads
- Division of Oral Microbiology and Immunology, Department of Operative and Preventive Dentistry & Periodontology, RWTH Aachen University Hospital, Aachen, Germany
| | - Laura Katharina Wendt
- Division of Oral Microbiology and Immunology, Department of Operative and Preventive Dentistry & Periodontology, RWTH Aachen University Hospital, Aachen, Germany
| | - Franziska Hetrodt
- Division of Oral Microbiology and Immunology, Department of Operative and Preventive Dentistry & Periodontology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Biohybrid & Medical Textiles, Institute of Applied Medical Engineering, RWTH Aachen University, Aachen, Germany
| | - Zhi-Luo Deng
- Group Microbial Communication, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
| | - Dietmar Pieper
- Group Microbial Interactions and Processes, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
| | - Mohamed M H Abdelbary
- Division of Oral Microbiology and Immunology, Department of Operative and Preventive Dentistry & Periodontology, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Irene Wagner-Döbler
- Group Microbial Communication, Helmholtz Center for Infection Research (HZI), Braunschweig, Germany
| | - Christian Apel
- Department of Biohybrid & Medical Textiles, Institute of Applied Medical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
25
|
Hetrodt F, Lausch J, Meyer-Lueckel H, Conrads G, Apel C. Evaluation of Restorative Materials Containing Preventive Additives in a Secondary Caries Model in vitro. Caries Res 2019; 53:447-456. [DOI: 10.1159/000496401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/19/2018] [Indexed: 11/19/2022] Open
Abstract
The incorporation of antibacterial agents into dental restorative materials is a promising strategy for secondary caries prevention. Previously, Carolacton affected Streptococcus mutans biofilm formation on composite materials in vitro. The present study evaluated secondary caries formation adjacent to Carolacton-containing composites and conventional restorative materials using an artificial biofilm model. Standardized cavities were prepared in bovine dentin-enamel samples (n = 175) and restored with various dental materials (Tetric EvoCeram [T], GrandioSo composite without [G] and with Carolacton [GC], Grandio Flow without [F] and with Carolacton [FC], GrandioSo containing sodium fluoride [GNaF], and Ketac Fil [K]). After artificial aging, S. mutans was grown on the samples for 7 days. The investigation of gap sizes and secondary caries formation was performed using confocal laser scanning microscopy and transversal microradiography. Median gap size in enamel was 9.4 µm (interquartile range 7.9–12.7). Compared to all other groups significant differences in gap sizes could be observed for Ketac Fil (p < 0.001; Mann-Whitney test). Only GrandioSo composite containing 30% sodium fluoride and Ketac Fil showed significantly smaller lesion areas in enamel (p < 0.001; Mann-Whitney test) than all other groups which was confirmed by the mineral loss data (p < 0.001; Mann-Whitney test). Based on the present in vitro results, it seems that Carolacton-containing composite in the current formulation within the shown simplified monoculture biofilm model is not able to prevent caries formation compared to fluoride-releasing restorative materials.
Collapse
|
26
|
Abstract
Natural products (NPs) are important sources of clinical drugs due to their structural diversity and biological prevalidation. However, the structural complexity of NPs leads to synthetic difficulties, unfavorable pharmacokinetic profiles, and poor drug-likeness. Structural simplification by truncating unnecessary substructures is a powerful strategy for overcoming these limitations and improving the efficiency and success rate of NP-based drug development. Herein, we will provide a comprehensive review of the structural simplification of NPs with a focus on design strategies, case studies, and new technologies. In particular, a number of successful examples leading to marketed drugs or drug candidates will be discussed in detail to illustrate how structural simplification is applied in lead optimization of NPs.
Collapse
Affiliation(s)
- Shengzheng Wang
- Department of Medicinal Chemistry, School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai , 200433 , P.R. China.,Department of Medicinal Chemistry, School of Pharmacy , Fourth Military Medical University , 169 Changle West Road , Xi'an , 710032 , P.R. China
| | - Guoqiang Dong
- Department of Medicinal Chemistry, School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai , 200433 , P.R. China
| | - Chunquan Sheng
- Department of Medicinal Chemistry, School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai , 200433 , P.R. China
| |
Collapse
|
27
|
Fighting biofilms with lantibiotics and other groups of bacteriocins. NPJ Biofilms Microbiomes 2018; 4:9. [PMID: 29707229 PMCID: PMC5908865 DOI: 10.1038/s41522-018-0053-6] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/09/2018] [Accepted: 03/21/2018] [Indexed: 01/03/2023] Open
Abstract
Biofilms are sessile communities of bacteria typically embedded in an extracellular polymeric matrix. Bacterial cells embedded in biofilms are inherently recalcitrant to antimicrobials, compared to cells existing in a planktonic state, and are notoriously difficult to eradicate once formed. Avenues to tackle biofilms thus far have largely focussed on attempting to disrupt the initial stages of biofilm formation, including adhesion and maturation of the biofilm. Such an approach is advantageous as the concentrations required to inhibit formation of biofilms are generally much lower than removing a fully established biofilm. The crisis of antibiotic resistance in clinical settings worldwide has been further exacerbated by the ability of certain pathogenic bacteria to form biofilms. Perhaps the most notorious biofilm formers described from a clinical viewpoint have been methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus epidermidis, Pseudomonas aeruginosa, Gardnerella vaginalis and Streptococcus mutans, the latter of which is found in oral biofilms. Due to the dearth of novel antibiotics in recent decades, compounded by the increasing rate of emergence of resistance amongst pathogens with a propensity for biofilm formation, solutions are urgently required to mitigate these crises. Bacteriocins are a class of antimicrobial peptides, which are ribosomally synthesised and often are more potent than their antibiotic counterparts. Here, we review a selection of studies conducted with bacteriocins with the ultimate objective of inhibiting biofilms. Overall, a deeper understanding of the precise means by which a biofilm forms on a substrate as well as insights into the mechanisms by which bacteriocins inhibit biofilms is warranted.
Collapse
|
28
|
Li B, Li X, Lin H, Zhou Y. Curcumin as a Promising Antibacterial Agent: Effects on Metabolism and Biofilm Formation in S. mutans. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4508709. [PMID: 29682545 PMCID: PMC5851298 DOI: 10.1155/2018/4508709] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/16/2022]
Abstract
Streptococcus mutans (S. mutans) has been proved to be the main aetiological factor in dental caries. Curcumin, a natural product, has been shown to exhibit therapeutic antibacterial activity, suggesting that curcumin may be of clinical interest. The objective of this study is to evaluate the inhibitory effects of curcumin on metabolism and biofilm formation in S. mutans using a vitro biofilm model in an artificial oral environment. S. mutans biofilms were treated with varying concentrations of curcumin. The biofilm metabolism and biofilm biomass were assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and the crystal violet assay. Confocal laser scanning microscopy was used to analyse the composition and extracellular polysaccharide content of S. mutans biofilm after curcumin treatment. The biofilm structure was evaluated using a scanning electron microscope. The gene expression of virulence-related factors was assessed by real-time PCR. The antibiofilm effect of curcumin was compared with that of chlorhexidine. The sessile minimum inhibitory concentration (SMIC50%) of curcumin against S. mutans biofilm was 500 μM. Curcumin reduced the biofilm metabolism from 5 min to 24 h. Curcumin inhibited the quantity of live bacteria and total bacteria in both the short term (5 min) and the long term. Moreover, curcumin decreased the production of extracellular polysaccharide in the short term. The expression of genes related to extracellular polysaccharide synthesis, carbohydrate metabolism, adherence, and the two-component transduction system decreased after curcumin treatment. The chlorhexidine-treated group showed similar results. We speculate that curcumin has the capacity to be developed as an alternative agent with the potential to reduce the pathogenic traits of S. mutans biofilm.
Collapse
Affiliation(s)
- Bingchun Li
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Xinlong Li
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Huancai Lin
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Yan Zhou
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| |
Collapse
|
29
|
The natural product carolacton inhibits folate-dependent C1 metabolism by targeting FolD/MTHFD. Nat Commun 2017; 8:1529. [PMID: 29142318 PMCID: PMC5688156 DOI: 10.1038/s41467-017-01671-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/05/2017] [Indexed: 01/26/2023] Open
Abstract
The natural product carolacton is a macrolide keto-carboxylic acid produced by the myxobacterium Sorangium cellulosum, and was originally described as an antibacterial compound. Here we show that carolacton targets FolD, a key enzyme from the folate-dependent C1 metabolism. We characterize the interaction between bacterial FolD and carolacton biophysically, structurally and biochemically. Carolacton binds FolD with nanomolar affinity, and the crystal structure of the FolD–carolacton complex reveals the mode of binding. We show that the human FolD orthologs, MTHFD1 and MTHFD2, are also inhibited in the low nM range, and that micromolar concentrations of carolacton inhibit the growth of cancer cell lines. As mitochondrial MTHFD2 is known to be upregulated in cancer cells, it may be possible to use carolacton as an inhibitor tool compound to assess MTHFD2 as an anti-cancer target. The mechanisms behind the antibacterial activity of the natural product carolacton are unknown. Here, the authors show that carolacton is a potent inhibitor of FolD/MTHFD enzymes (involved in folate-dependent C1 metabolism in bacteria and humans) and inhibits the growth of cancer cell lines
Collapse
|
30
|
The Biofilm Inhibitor Carolacton Enters Gram-Negative Cells: Studies Using a TolC-Deficient Strain of Escherichia coli. mSphere 2017; 2:mSphere00375-17. [PMID: 28959742 PMCID: PMC5615136 DOI: 10.1128/mspheredirect.00375-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 08/26/2017] [Indexed: 12/18/2022] Open
Abstract
The emergence of pathogens resistant against most or all of the antibiotics currently used in human therapy is a global threat, and therefore the search for antimicrobials with novel targets and modes of action is of utmost importance. The myxobacterial secondary metabolite carolacton had previously been shown to inhibit biofilm formation and growth of streptococci. Here, we investigated if carolacton could act against Gram-negative bacteria, which are difficult targets because of their double-layered cytoplasmic envelope. We found that the model organism Escherichia coli is susceptible to carolacton, similar to the Gram-positive Streptococcus pneumoniae, if its multidrug efflux system AcrAB-TolC is either inactivated genetically, by disruption of the tolC gene, or physiologically by coadministering an efflux pump inhibitor. A carolacton epimer that has a different steric configuration at carbon atom 9 is completely inactive, suggesting that carolacton may interact with the same molecular target in both Gram-positive and Gram-negative bacteria. The myxobacterial secondary metabolite carolacton inhibits growth of Streptococcus pneumoniae and kills biofilm cells of the caries- and endocarditis-associated pathogen Streptococcus mutans at nanomolar concentrations. Here, we studied the response to carolacton of an Escherichia coli strain that lacked the outer membrane protein TolC. Whole-genome sequencing of the laboratory E. coli strain TolC revealed the integration of an insertion element, IS5, at the tolC locus and a close phylogenetic relationship to the ancient E. coli K-12. We demonstrated via transcriptome sequencing (RNA-seq) and determination of MIC values that carolacton penetrates the phospholipid bilayer of the Gram-negative cell envelope and inhibits growth of E. coli TolC at similar concentrations as for streptococci. This inhibition is completely lost for a C-9 (R) epimer of carolacton, a derivative with an inverted stereocenter at carbon atom 9 [(S) → (R)] as the sole difference from the native molecule, which is also inactive in S. pneumoniae and S. mutans, suggesting a specific interaction of native carolacton with a conserved cellular target present in bacterial phyla as distantly related as Firmicutes and Proteobacteria. The efflux pump inhibitor (EPI) phenylalanine arginine β-naphthylamide (PAβN), which specifically inhibits AcrAB-TolC, renders E. coli susceptible to carolacton. Our data indicate that carolacton has potential for use in antimicrobial chemotherapy against Gram-negative bacteria, as a single drug or in combination with EPIs. Strain E. coli TolC has been deposited at the DSMZ; together with the associated RNA-seq data and MIC values, it can be used as a reference during future screenings for novel bioactive compounds. IMPORTANCE The emergence of pathogens resistant against most or all of the antibiotics currently used in human therapy is a global threat, and therefore the search for antimicrobials with novel targets and modes of action is of utmost importance. The myxobacterial secondary metabolite carolacton had previously been shown to inhibit biofilm formation and growth of streptococci. Here, we investigated if carolacton could act against Gram-negative bacteria, which are difficult targets because of their double-layered cytoplasmic envelope. We found that the model organism Escherichia coli is susceptible to carolacton, similar to the Gram-positive Streptococcus pneumoniae, if its multidrug efflux system AcrAB-TolC is either inactivated genetically, by disruption of the tolC gene, or physiologically by coadministering an efflux pump inhibitor. A carolacton epimer that has a different steric configuration at carbon atom 9 is completely inactive, suggesting that carolacton may interact with the same molecular target in both Gram-positive and Gram-negative bacteria.
Collapse
|
31
|
Solinski AE, Koval AB, Brzozowski RS, Morrison KR, Fraboni AJ, Carson CE, Eshraghi AR, Zhou G, Quivey RG, Voelz VA, Buttaro BA, Wuest WM. Diverted Total Synthesis of Carolacton-Inspired Analogs Yields Three Distinct Phenotypes in Streptococcus mutans Biofilms. J Am Chem Soc 2017; 139:7188-7191. [PMID: 28502178 PMCID: PMC5891724 DOI: 10.1021/jacs.7b03879] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The oral microbiome is a dynamic environment inhabited by both commensals and pathogens. Among these is Streptococcus mutans, the causative agent of dental caries, the most prevalent childhood disease. Carolacton has remarkably specific activity against S. mutans, causing acid-mediated cell death during biofilm formation; however, its complex structure limits its utility. Herein, we report the diverted total synthesis and biological evaluation of a rationally designed library of simplified analogs that unveiled three unique biofilm phenotypes further validating the role of natural product synthesis in the discovery of new biological phenomena.
Collapse
Affiliation(s)
- Amy E. Solinski
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Alexander B. Koval
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Richard S. Brzozowski
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Kelly R. Morrison
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Americo J. Fraboni
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Carrie E. Carson
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Anisa R. Eshraghi
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Guangfeng Zhou
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Robert G. Quivey
- Center for Oral Biology, University of Rochester School of Medicine and Dentistry, Rochester New York 14642, United States
| | - Vincent A Voelz
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Bettina A. Buttaro
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - William M. Wuest
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
32
|
Brzozowski RS, Wuest WM. Twelve-membered macrolactones: privileged scaffolds for the development of new therapeutics. Chem Biol Drug Des 2017; 89:169-191. [PMID: 27153932 DOI: 10.1111/cbdd.12783] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/26/2016] [Accepted: 04/27/2016] [Indexed: 12/15/2022]
Abstract
Natural products commonly produced as secondary metabolites of various plants and micro-organisms represent a diverse chemical space of compounds. The diversity of natural products makes them an attractive target for interrogation by both chemists and biologists alike. Indeed, the study of 12-membered macrolactones has already led to the discovery of lead drug compounds and new biological targets, which has motivated the development of diverted total synthetic routes to libraries of analogs. This review explores the discovery, biological characterization, and synthesis of several 12-membered macrolactones, exploiting examples that underscore their importance in the drug discovery field. It is our hope that this review will motivate further interest in this class of natural products, a group of molecules that we think merit the classification of 'privileged scaffolds' within the medicinal chemistry community, to further investigate and develop novel compounds with promising bioactivity.
Collapse
Affiliation(s)
| | - William M Wuest
- Department of Chemistry, Temple University, Philadelphia, PA, USA
| |
Collapse
|
33
|
Abstract
A short and convergent strategy for the stereoselective total synthesis of biologically active natural product carolacton has been accomplished. Our synthesis highlights the Urpi acetal aldol, Crimmins aldol, Ireland-Claisen rearrangement, TiCl4-assisted aldol followed by β-hydroxy elimination to construct C7-C8 olefin, and ring-closing metathesis as the key steps for achieving the target molecule with an overall yield of 18.8%.
Collapse
Affiliation(s)
- Tapan Kumar Kuilya
- Department of Organic Chemistry, Indian Association for the Cultivation of Science , Jadavpur, Kolkata 700032, India
| | - Rajib Kumar Goswami
- Department of Organic Chemistry, Indian Association for the Cultivation of Science , Jadavpur, Kolkata 700032, India
| |
Collapse
|
34
|
Ammermann J, Schmidt T, Donner J, Reck M, Dalton M, Stumpp N, Stiesch M, Wagner-Döbler I, Kirschning A. The carolactam strategy is ineffective: synthesis and biological evaluation of carolactam. Org Biomol Chem 2017; 15:8553-8558. [DOI: 10.1039/c7ob02060k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The lactam analogue of carolacton was prepared which has lost biofilm inhibitory activity towards Streptococcus mutans.
Collapse
Affiliation(s)
- Jonas Ammermann
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ)
- Leibniz Universität Hannover
- 30167 Hannover
- Germany
| | - Thomas Schmidt
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ)
- Leibniz Universität Hannover
- 30167 Hannover
- Germany
| | - Jannik Donner
- Helmholtz-Center for Infection Research
- Department of Microbial Communication
- 38124 Braunschweig
- Germany
| | - Michael Reck
- Helmholtz-Center for Infection Research
- Department of Microbial Communication
- 38124 Braunschweig
- Germany
| | - Marly Dalton
- Department of Prosthetic Dentistry and Biomedical Materials Science
- Hannover Medical School
- 30625 Hannover
- Germany
| | - Nico Stumpp
- Department of Prosthetic Dentistry and Biomedical Materials Science
- Hannover Medical School
- 30625 Hannover
- Germany
| | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials Science
- Hannover Medical School
- 30625 Hannover
- Germany
| | - Irene Wagner-Döbler
- Helmholtz-Center for Infection Research
- Department of Microbial Communication
- 38124 Braunschweig
- Germany
| | - Andreas Kirschning
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ)
- Leibniz Universität Hannover
- 30167 Hannover
- Germany
| |
Collapse
|
35
|
Herrmann J, Fayad AA, Müller R. Natural products from myxobacteria: novel metabolites and bioactivities. Nat Prod Rep 2016; 34:135-160. [PMID: 27907217 DOI: 10.1039/c6np00106h] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Covering: 2011-July 2016Myxobacteria are a rich source for structurally diverse secondary metabolites with intriguing biological activities. Here we report on new natural products that were isolated from myxobacteria in the period of 2011 to July 2016. Some examples of recent advances on modes-of-action are also summarised along with a more detailed overview on five compound classes currently assessed in preclinical studies.
Collapse
Affiliation(s)
- J Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Department of Microbial Natural Products, Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany.
| | | | | |
Collapse
|
36
|
Affiliation(s)
- Silke C. Wenzel
- Saarland University; Department of Microbial Natural Products, Helmholtz-Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Pharmaceutical Biotechnology; Saarland University Campus, Building E8.1 66123 Saarbrücken Germany
| | - Rolf Müller
- Saarland University; Department of Microbial Natural Products, Helmholtz-Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Pharmaceutical Biotechnology; Saarland University Campus, Building E8.1 66123 Saarbrücken Germany
| |
Collapse
|
37
|
The biofilm inhibitor Carolacton inhibits planktonic growth of virulent pneumococci via a conserved target. Sci Rep 2016; 6:29677. [PMID: 27404808 PMCID: PMC4939601 DOI: 10.1038/srep29677] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/21/2016] [Indexed: 12/13/2022] Open
Abstract
New antibacterial compounds, preferentially exploiting novel cellular targets, are urgently needed to fight the increasing resistance of pathogens against conventional antibiotics. Here we demonstrate that Carolacton, a myxobacterial secondary metabolite previously shown to damage Streptococcus mutans biofilms, inhibits planktonic growth of Streptococcus pneumoniae TIGR4 and multidrug-resistant clinical isolates of serotype 19A at nanomolar concentrations. A Carolacton diastereomer is inactive in both streptococci, indicating a highly specific interaction with a conserved cellular target. S. mutans requires the eukaryotic-like serine/threonine protein kinase PknB and the cysteine metabolism regulator CysR for susceptibility to Carolacton, whereas their homologues are not needed in S. pneumoniae, suggesting a specific function for S. mutans biofilms only. A bactericidal effect of Carolacton was observed for S. pneumoniae TIGR4, with a reduction of cell numbers by 3 log units. The clinical pneumonia isolate Sp49 showed immediate growth arrest and cell lysis, suggesting a bacteriolytic effect of Carolacton. Carolacton treatment caused a reduction in membrane potential, but not membrane integrity, and transcriptome analysis revealed compensatory reactions of the cell. Our data show that Carolacton might have potential for treating pneumococcal infections.
Collapse
|
38
|
Bai L, Takagi S, Ando T, Yoneyama H, Ito K, Mizugai H, Isogai E. Antimicrobial activity of tea catechin against canine oral bacteria and the functional mechanisms. J Vet Med Sci 2016; 78:1439-1445. [PMID: 27246281 PMCID: PMC5059371 DOI: 10.1292/jvms.16-0198] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epigallocatechin gallate (EGCG) is the major polyphenolic compound of green tea.
Polyphenolic compounds were extracted from the leaf of Camellia sinensis
(Japanese green tea), and the minimum inhibitory concentration against canine oral
bacteria was measured. Subsequently, we investigated the inhibitory effects of
polyphenolic compounds and EGCG on the growth of canine oral bacteria. EGCG showed
antimicrobial activity against a model bacterium, Streptococcus mutans.
Our results indicate that EGCG can inhibit the growth and biofilm formation of S.
mutans and that EGCG does not interact with streptococcal lipoteichoic acid
(LTA). Furthermore, our findings suggest that EGCG interacts with other component(s) of
the bacterial membrane aside from streptococcal LTA to inhibit biofilm formation and
damage biofilms.
Collapse
Affiliation(s)
- Lanlan Bai
- Laboratory of Animal Microbiology, Department of Microbial Biotechnology, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori, Amamiya-machi Aoba-ku, Sendai, Miyagi 981-8555, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Reck M, Wagner-Döbler I. Carolacton Treatment Causes Delocalization of the Cell Division Proteins PknB and DivIVa in Streptococcus mutans in vivo. Front Microbiol 2016; 7:684. [PMID: 27242711 PMCID: PMC4862990 DOI: 10.3389/fmicb.2016.00684] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/26/2016] [Indexed: 12/20/2022] Open
Abstract
The small inhibitory molecule Carolacton has been shown to cause chain formation and bulging in Streptococci, suggesting a defect in cell division, but it is not known how cell division is impaired on a molecular level. Fluorescent fusion proteins have successfully been applied to visualize protein localization and dynamics in vivo and have revolutionized our understanding of cell wall growth, cell division, chromosome replication and segregation. However, in Streptococci the required vectors are largely lacking. We constructed vectors for chromosomal integration and inducible expression of fluorescent fusion proteins based on GFP+ in S. mutans. Their applicability was verified using four proteins with known localization in the cell. We then determined the effect of Carolacton on the subcellular localization of GFP+ fusions of the cell division protein DivIVa and the serine-threonine protein kinase PknB. Carolacton caused a significant delocalization of these proteins from midcell, in accordance with a previous study demonstrating the Carolacton insensitive phenotype of a pknB deletion strain. Carolacton treated cells displayed an elongated phenotype, increased septum formation and a severe defect in daughter cell separation. GFP+ fusions of two hypothetical proteins (SMU_503 and SMU_609), that had previously been shown to be the most strongly upregulated genes after Carolacton treatment, were found to be localized at the septum in midcell, indicating their role in cell division. These findings highlight the importance of PknB as a key regulator of cell division in streptococci and indicate a profound impact of Carolacton on the coordination between peripheral and septal cell wall growth. The established vector system represents a novel tool to study essential steps of cellular metabolism.
Collapse
Affiliation(s)
- Michael Reck
- Department of Microbiology, Microbial Communication, Helmholtz Centre for Infection Research Braunschweig, Germany
| | - Irene Wagner-Döbler
- Department of Microbiology, Microbial Communication, Helmholtz Centre for Infection Research Braunschweig, Germany
| |
Collapse
|
40
|
Ribeiro SM, Felício MR, Boas EV, Gonçalves S, Costa FF, Samy RP, Santos NC, Franco OL. New frontiers for anti-biofilm drug development. Pharmacol Ther 2016; 160:133-44. [DOI: 10.1016/j.pharmthera.2016.02.006] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Abstract
In the biofilm form, bacteria are more resistant to various antimicrobial treatments. Bacteria in a biofilm can also survive harsh conditions and withstand the host's immune system. Therefore, there is a need for new treatment options to treat biofilm-associated infections. Currently, research is focused on the development of antibiofilm agents that are nontoxic, as it is believed that such molecules will not lead to future drug resistance. In this review, we discuss recent discoveries of antibiofilm agents and different approaches to inhibit/disperse biofilms. These new antibiofilm agents, which contain moieties such as imidazole, phenols, indole, triazole, sulfide, furanone, bromopyrrole, peptides, etc. have the potential to disperse bacterial biofilms in vivo and could positively impact human medicine in the future.
Collapse
|
42
|
H. Kasper S, Hart R, Bergkvist M, A. Musah R, C. Cady N. Zein nanocapsules as a tool for surface passivation, drug delivery and biofilm prevention. AIMS Microbiol 2016. [DOI: 10.3934/microbiol.2016.4.422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
43
|
Sintim HO, Gürsoy UK. Biofilms as "Connectors" for Oral and Systems Medicine: A New Opportunity for Biomarkers, Molecular Targets, and Bacterial Eradication. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2015; 20:3-11. [PMID: 26583256 PMCID: PMC4739346 DOI: 10.1089/omi.2015.0146] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Oral health and systems medicine are intimately related but have remained, sadly, as isolated knowledge communities for decades. Are there veritable connector knowledge domains that can usefully link them together on the critical path to biomarker research and “one health”? In this context, it is noteworthy that bacteria form surface-attached communities on most biological surfaces, including the oral cavity. Biofilm-forming bacteria contribute to periodontal diseases and recent evidences point to roles of these bacteria in systemic diseases as well, with cardiovascular diseases, obesity, and cancer as notable examples. Interestingly, the combined mass of microorganisms such as bacteria are so large that when we combine all plants and animals on earth, the total biomass of bacteria is still bigger. They literally do colonize everywhere, not only soil and water but our skin, digestive tract, and even oral cavity are colonized by bacteria. Hence efforts to delineate biofilm formation mechanisms of oral bacteria and microorganisms and the development of small molecules to inhibit biofilm formation in the oral cavity is very timely for both diagnostics and therapeutics. Research on biofilms can benefit both oral and systems medicine. Here, we examine, review, and synthesize new knowledge on the current understanding of oral biofilm formation, the small molecule targets that can inhibit biofilm formation in the mouth. We suggest new directions for both oral and systems medicine, using various omics technologies such as SILAC and RNAseq, that could yield deeper insights, biomarkers, and molecular targets to design small molecules that selectively aim at eradication of pathogenic oral bacteria. Ultimately, devising new ways to control and eradicate bacteria in biofilms will open up novel diagnostic and therapeutic avenues for oral and systemic diseases alike.
Collapse
Affiliation(s)
- Herman O Sintim
- 1 Department of Chemistry and Biochemistry, University of Maryland , College Park, Maryland.,2 Department of Chemistry, Purdue University , West Lafayette, Indiana
| | - Ulvi Kahraman Gürsoy
- 3 Department of Periodontology, Institute of Dentistry, University of Turku , Turku, Finland
| |
Collapse
|
44
|
Reddy SV, Prasanna Kumar K, Ramakrishna KV, Sharma GV. Approaches towards the total synthesis of carolacton: synthesis of C1–C16 fragment. Tetrahedron Lett 2015. [DOI: 10.1016/j.tetlet.2015.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
45
|
Bem AE, Velikova N, Pellicer MT, Baarlen PV, Marina A, Wells JM. Bacterial histidine kinases as novel antibacterial drug targets. ACS Chem Biol 2015; 10:213-24. [PMID: 25436989 DOI: 10.1021/cb5007135] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bacterial histidine kinases (HKs) are promising targets for novel antibacterials. Bacterial HKs are part of bacterial two-component systems (TCSs), the main signal transduction pathways in bacteria, regulating various processes including virulence, secretion systems and antibiotic resistance. In this review, we discuss the biological importance of TCSs and bacterial HKs for the discovery of novel antibacterials, as well as published TCS and HK inhibitors that can be used as a starting point for structure-based approaches to develop novel antibacterials.
Collapse
Affiliation(s)
- Agnieszka E. Bem
- Host−Microbe
Interactomics, Wageningen University, De Elst 1, 6708 WD Wageningen, The Netherlands
| | - Nadya Velikova
- Instituto
de Biomedicina
de Valencia-Consejo Superior de Investigaciones Cientificas (IBV-CSIC), Jaume Roig 11, 46010-Valencia, Spain
| | - M. Teresa Pellicer
- R&D Department Interquim, Ferrer HealthTech, Joan Buscalla 10, 08137-Sant Cugat del Valles Barcelona, Spain
| | - Peter van Baarlen
- Host−Microbe
Interactomics, Wageningen University, De Elst 1, 6708 WD Wageningen, The Netherlands
| | - Alberto Marina
- Instituto
de Biomedicina
de Valencia-Consejo Superior de Investigaciones Cientificas (IBV-CSIC), Jaume Roig 11, 46010-Valencia, Spain
- Centro de Investigacion
Biomedica en Red de Enfermedades Raras (CIBER-ISCIII), Jaume Roig 11, 46010-Valencia, Spain
| | - Jerry M. Wells
- Host−Microbe
Interactomics, Wageningen University, De Elst 1, 6708 WD Wageningen, The Netherlands
| |
Collapse
|
46
|
Melander RJ, Melander C. Innovative strategies for combating biofilm-based infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 831:69-91. [PMID: 25384664 DOI: 10.1007/978-3-319-09782-4_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Roberta J Melander
- Department of Chemistry, North Carolina State University, Raleigh, NC, USA
| | | |
Collapse
|
47
|
Stumpp N, Premnath P, Schmidt T, Ammermann J, Dräger G, Reck M, Jansen R, Stiesch M, Wagner-Döbler I, Kirschning A. Synthesis of new carolacton derivatives and their activity against biofilms of oral bacteria. Org Biomol Chem 2015; 13:5765-74. [DOI: 10.1039/c5ob00460h] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Carolacton, a secondary metabolite isolated from the extracts ofSorangium cellulosum, causes membrane damage and cell death in biofilms of different oral bacteria.
Collapse
Affiliation(s)
- N. Stumpp
- Department of Prosthetic Dentistry and Biomedical Materials Science
- Hannover Medical School
- 30625 Hannover
- Germany
| | - P. Premnath
- Helmholtz-Center for Infection Research
- Department of Microbial Communication
- 38124 Braunschweig
- Germany
| | - T. Schmidt
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ)
- Leibniz Universität Hannover
- 30167 Hannover
- Germany
| | - J. Ammermann
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ)
- Leibniz Universität Hannover
- 30167 Hannover
- Germany
| | - G. Dräger
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ)
- Leibniz Universität Hannover
- 30167 Hannover
- Germany
| | - M. Reck
- Helmholtz-Center for Infection Research
- Department of Microbial Communication
- 38124 Braunschweig
- Germany
| | - R. Jansen
- Helmholtz-Center for Infection Research
- Department of Microbial Drugs
- 38124 Braunschweig
- Germany
| | - M. Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials Science
- Hannover Medical School
- 30625 Hannover
- Germany
| | - I. Wagner-Döbler
- Helmholtz-Center for Infection Research
- Department of Microbial Communication
- 38124 Braunschweig
- Germany
| | - A. Kirschning
- Institute of Organic Chemistry and Center of Biomolecular Drug Research (BMWZ)
- Leibniz Universität Hannover
- 30167 Hannover
- Germany
| |
Collapse
|
48
|
Fletcher MH, Jennings MC, Wuest WM. Draining the moat: disrupting bacterial biofilms with natural products. Tetrahedron 2014. [DOI: 10.1016/j.tet.2014.06.055] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
49
|
Abstract
Covering: up to the end of 2013. Myxobacteria produce a vast range of structurally diverse natural products with prominent biological activities. Here, we provide a detailed description and judge the potential of all antibiotically active myxobacterial compounds as lead structures, pointing out their particularities and, if known, their mode of action. Thus, the review provides an overview of the potential of specific compounds, suitable for future investigations and possible clinical applications.
Collapse
Affiliation(s)
- Till F Schäberle
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany.
| | | | | | | |
Collapse
|
50
|
Sudhakar P, Reck M, Wang W, He FQ, Wagner-Döbler I, Dobler IW, Zeng AP. Construction and verification of the transcriptional regulatory response network of Streptococcus mutans upon treatment with the biofilm inhibitor carolacton. BMC Genomics 2014; 15:362. [PMID: 24884510 PMCID: PMC4048456 DOI: 10.1186/1471-2164-15-362] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/17/2014] [Indexed: 11/26/2022] Open
Abstract
Background Carolacton is a newly identified secondary metabolite causing altered cell morphology and death of Streptococcus mutans biofilm cells. To unravel key regulators mediating these effects, the transcriptional regulatory response network of S. mutans biofilms upon carolacton treatment was constructed and analyzed. A systems biological approach integrating time-resolved transcriptomic data, reverse engineering, transcription factor binding sites, and experimental validation was carried out. Results The co-expression response network constructed from transcriptomic data using the reverse engineering algorithm called the Trend Correlation method consisted of 8284 gene pairs. The regulatory response network inferred by superimposing transcription factor binding site information into the co-expression network comprised 329 putative transcriptional regulatory interactions and could be classified into 27 sub-networks each co-regulated by a transcription factor. These sub-networks were significantly enriched with genes sharing common functions. The regulatory response network displayed global hierarchy and network motifs as observed in model organisms. The sub-networks modulated by the pyrimidine biosynthesis regulator PyrR, the glutamine synthetase repressor GlnR, the cysteine metabolism regulator CysR, global regulators CcpA and CodY and the two component system response regulators VicR and MbrC among others could putatively be related to the physiological effect of carolacton. The predicted interactions from the regulatory network between MbrC, known to be involved in cell envelope stress response, and the murMN-SMU_718c genes encoding peptidoglycan biosynthetic enzymes were experimentally confirmed using Electro Mobility Shift Assays. Furthermore, gene deletion mutants of five predicted key regulators from the response networks were constructed and their sensitivities towards carolacton were investigated. Deletion of cysR, the node having the highest connectivity among the regulators chosen from the regulatory network, resulted in a mutant which was insensitive to carolacton thus demonstrating not only the essentiality of cysR for the response of S. mutans biofilms to carolacton but also the relevance of the predicted network. Conclusion The network approach used in this study revealed important regulators and interactions as part of the response mechanisms of S. mutans biofilm cells to carolacton. It also opens a door for further studies into novel drug targets against streptococci. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-362) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | - Irene W Dobler
- Institute of Bioprocess and Biosystems Engineering, Hamburg University of Technology, 21073 Hamburg, Germany.
| | | |
Collapse
|