1
|
Li Y, Li Y, Gao H, Liu J, Liang H. Edible thermosensitive chitosan/hydroxypropyl β-cyclodextrin hydrogel with natural licoricidin for enhancing oral health: Biofilm disruption and demineralization prevention. Int J Biol Macromol 2024; 282:136647. [PMID: 39423986 DOI: 10.1016/j.ijbiomac.2024.136647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Dental caries, a widespread and significantly detrimental health condition, is characterized by demineralization, pain, compromised tooth functionality, and various other adverse effects. Licoricidin (LC), a natural isoflavonoid, demonstrates potent antimicrobial properties for maintaining oral health. However, its practical application is significantly hindered by its limited water solubility and susceptibility to removal within the oral environment. To tackle this issue, we developed a delivery oral system by an edible thermosensitive chitosan- disodium beta-glycerol phosphate pentahydrate (CS/β-GP) hydrogel to load LC/Hydroxypropyl beta-cyclodextrin (HP-β-CD) inclusion complexes. These hydrogels (LC/HP-β-CD/CS/β-GP) could solidify rapidly at oral temperature and sustainably release LC, thereby preventing its rapid clearance from the oral cavity. We confirmed the significant antibacterial activity of this hydrogel against Streptococcus mutans and Staphylococcus aureus. Additionally, the HP-β-CD combination enhanced LC to penetrate bacterial biofilms and inhibit biofilm growth, leading to leakage of cellular proteins and DNA. Additionally, we studied the effect of LC/HP-β-CD/CS/β-GP on intracellular ROS levels and MMP, comprehensively exploring its antimicrobial mechanism. Furthermore, LC/HP-β-CD/CS/β-GP exhibited the ability to inhibit demineralization and demonstrated excellent biocompatibility. In summary, this study presented a safer approach to oral delivering bioactive substances, offering a promising strategy for enhanced oral health and safety.
Collapse
Affiliation(s)
- Yishan Li
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yaqian Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China
| | - Huiling Gao
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jianzhang Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China.
| | - Hao Liang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
2
|
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
|
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
|
Graff Reis J, Dai Prá I, Michelon W, Viancelli A, Piedrahita Marquez DG, Schmitz C, Maraschin M, Moura S, Thaís Silva I, de Oliveira Costa G, Tizziani T, Sandjo LP, Rodríguez-Lázaro D, Fongaro G. Characterization of Planktochlorella nurekis Extracts and Virucidal Activity against a Coronavirus Model, the Murine Coronavirus 3. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15823. [PMID: 36497896 PMCID: PMC9735810 DOI: 10.3390/ijerph192315823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Certain members of the Coronaviridae family have emerged as zoonotic agents and have recently caused severe respiratory diseases in humans and animals, such as SARS, MERS, and, more recently, COVID-19. Antivirals (drugs and antiseptics) capable of controlling viruses at the site of infection are scarce. Microalgae from the Chlorellaceae family are sources of bioactive compounds with antioxidant, antiviral, and antitumor activity. In the present study, we aimed to evaluate various extracts from Planktochlorella nurekis in vitro against murine coronavirus-3 (MHV-3), which is an essential human coronavirus surrogate for laboratory assays. Methanol, hexane, and dichloromethane extracts of P. nurekis were tested in cells infected with MHV-3, and characterized by UV-vis spectrophotometry, nuclear magnetic resonance (NMR) spectroscopy, ultraperformance liquid chromatography-mass spectrometry (UPLC-MS), and the application of chemometrics through principal component analysis (PCA). All the extracts were highly efficient against MHV-3 (more than a 6 Log unit reduction), regardless of the solvent used or the concentration of the extract, but the dichloromethane extract was the most effective. Chemical characterization by spectrophotometry and NMR, with the aid of statistical analysis, showed that polyphenols, carbohydrates, and isoprene derivatives, such as terpenes and carotenoids have a more significant impact on the virucidal potential. Compounds identified by UPLC-MS were mainly lipids and only found in the dichloromethane extract. These results open new biotechnological possibilities to explore the biomass of P. nurekis; it is a natural extract and shows low cytotoxicity and an excellent antiviral effect, with low production costs, highlighting a promising potential for development and implementation of therapies against coronaviruses, such as SARS-CoV-2.
Collapse
Affiliation(s)
- Jacqueline Graff Reis
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Isabella Dai Prá
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
- Department of Pharmaceutical Sciences, Federal University Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - William Michelon
- Mestrado Profissional em Engenharia Civil, Sanitária e Ambiental, Universidade do Contestado Concórdia, Concórdia 89520-000, SC, Brazil
| | - Aline Viancelli
- Mestrado Profissional em Engenharia Civil, Sanitária e Ambiental, Universidade do Contestado Concórdia, Concórdia 89520-000, SC, Brazil
| | | | - Caroline Schmitz
- Plant Morphogenesis and Biochemistry Laboratory, Federal University of Santa Catarina, Florianópolis 88034-000, SC, Brazil
| | - Marcelo Maraschin
- Plant Morphogenesis and Biochemistry Laboratory, Federal University of Santa Catarina, Florianópolis 88034-000, SC, Brazil
| | - Sidnei Moura
- LBIOP—Laboratory of Biotechnology of Natural and Synthetics Products, Technology Department, Biotechnology Institute, University of Caxias do Sul, Caxias do Sul 95070-560, RS, Brazil
| | - Izabella Thaís Silva
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
- Department of Pharmaceutical Sciences, Federal University Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Geovanna de Oliveira Costa
- Programa de Pós-Graduação em Química, Department of Chemistry, CFM, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Tiago Tizziani
- Programa de Pós-Graduação em Química, Department of Chemistry, CFM, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Louis P. Sandjo
- Programa de Pós-Graduação em Química, Department of Chemistry, CFM, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - David Rodríguez-Lázaro
- Microbiology Section, Faculty of Sciences, University of Burgos, 09001 Burgos, Spain
- Centre for Emerging pathogens and Global Health, University of Burgos, 09001 Burgos, Spain
| | - Gislaine Fongaro
- Laboratory of Applied Virology, Department of Microbiology, Immunology, and Parasitology, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| |
Collapse
|
5
|
Chen J, Han J, Zhang J, Li L, Zhang Z, Yang Y, Jiang Y. Rhodium/Amine Dual Catalytic System for Reassembling C≡C Bonds of Conjugated Alkynes with Cyclopropenes via Cutting/Insertion Cascade. ACS Catal 2022. [DOI: 10.1021/acscatal.2c03777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Jie Chen
- Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Jiabin Han
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Jian Zhang
- Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China
| | - Ling Li
- Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China
| | - Zhengyu Zhang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yanhui Yang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Yaojia Jiang
- Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| |
Collapse
|
6
|
Etayash H, Alford M, Akhoundsadegh N, Drayton M, Straus SK, Hancock REW. Multifunctional Antibiotic-Host Defense Peptide Conjugate Kills Bacteria, Eradicates Biofilms, and Modulates the Innate Immune Response. J Med Chem 2021; 64:16854-16863. [PMID: 34784220 DOI: 10.1021/acs.jmedchem.1c01712] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Effective anti-infective therapies are required to offset the rise in antibiotic resistance. A novel vancomycin-innate defense regulator conjugate (V-IDR1018) was constructed with multimodal functionality, including bacterial killing, biofilm eradication, and immune modulation. The conjugate killed bacteria within 30 min, exhibited potent activity against persister cells, and showed no susceptibility to antimicrobial resistance in tissue culture assays. Additionally, it stimulated the release of chemokine MCP-1 and anti-inflammatory cytokine IL-10 and suppressed pro-inflammatory IL-1β from lipopolysaccharide-stimulated white blood cells. The conjugate demonstrated ∼90% eradication efficacy when assessed against the MRSA biofilm formed on an organoid human skin equivalent. Similarly, when evaluated in a murine, high-density skin abscess infection model using MRSA or Staphylococcus epidermidis, the conjugate decreased dermonecrosis and reduced bacterial load. The exceptional in vitro and in vivo efficacy of the conjugate, in addition to its safety profile, makes it a valuable candidate to treat complex infectious diseases.
Collapse
Affiliation(s)
- Hashem Etayash
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver V6T 1Z4, British Columbia, Canada
| | - Morgan Alford
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver V6T 1Z4, British Columbia, Canada
| | - Noushin Akhoundsadegh
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver V6T 1Z4, British Columbia, Canada
| | - Matthew Drayton
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver V6T 1Z1, British Columbia, Canada
| | - Suzana K Straus
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver V6T 1Z1, British Columbia, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, 2259 Lower Mall Research Station, Vancouver V6T 1Z4, British Columbia, Canada
| |
Collapse
|
7
|
Demeritte A, Wuest WM. A look around the West Indies: The spices of life are secondary metabolites. Bioorg Med Chem 2020; 28:115792. [PMID: 33038665 PMCID: PMC7528826 DOI: 10.1016/j.bmc.2020.115792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 11/22/2022]
Abstract
Natural products possess a wide range of bioactivities with potential for therapeutic usage. While the distribution of these molecules can vary greatly there is some correlation that exists between the biodiversity of an environment and the uniqueness and concentration of natural products found in that region or area. The Caribbean and pan-Caribbean area is home to thousands of species of endemic fauna and flora providing huge potential for natural product discovery and by way, potential leads for drug development. This can especially be said for marine natural products as many of are rapidly diluted through diffusion once released and therefore are highly potent to achieve long reaching effects. This review seeks to highlight a small selection of marine natural products from the Caribbean region which possess antiproliferative, anti-inflammatory and antipathogenic properties while highlighting any synthetic efforts towards bioactive analogs.
Collapse
Affiliation(s)
- Adrian Demeritte
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, GA 30322, USA
| | - William M Wuest
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, GA 30322, USA.
| |
Collapse
|
8
|
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: 82] [Impact Index Per Article: 16.4] [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
|
9
|
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
|
10
|
Liu K, Huigens RW. Instructive Advances in Chemical Microbiology Inspired by Nature's Diverse Inventory of Molecules. ACS Infect Dis 2020; 6:541-562. [PMID: 31842540 PMCID: PMC7346871 DOI: 10.1021/acsinfecdis.9b00413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural product antibiotics have played an essential role in the treatment of bacterial infection in addition to serving as useful tools to explore the intricate biology of bacteria. Our current arsenal of antibiotics operate through the inhibition of well-defined bacterial targets critical for replication and growth. Pathogenic bacteria effectively utilize a diversity of mechanisms that lead to acquired resistance and/or innate tolerance toward antibiotic therapies, which can result in devastating consequences to human life. Several research groups have established innovative programs that work at the chemistry-biology interface to develop new molecules that aim to define and address concerns related to antibiotic resistance and tolerance. In this Review, we present recent progress by select research groups that highlight a diversity of integrated chemical biology and medicinal chemistry approaches aimed at the development and utilization of chemical tools that have led to promising new microbiological insights that may lead to significant clinical advances regarding the treatment of pathogenic bacteria.
Collapse
Affiliation(s)
- Ke Liu
- 1345 Center Drive, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Robert W. Huigens
- 1345 Center Drive, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
11
|
Ochoa C, Solinski AE, Nowlan M, Dekarske MM, Wuest WM, Kozlowski MC. A Bisphenolic Honokiol Analog Outcompetes Oral Antimicrobial Agent Cetylpyridinium Chloride via a Membrane-Associated Mechanism. ACS Infect Dis 2020; 6:74-79. [PMID: 31663323 DOI: 10.1021/acsinfecdis.9b00190] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Targeting Streptococcus mutans is the primary focus in reducing dental caries, one of the most common maladies in the world. Previously, our groups discovered a potent bactericidal biaryl compound that was inspired by the natural product honokiol. Herein, a structure activity relationship (SAR) study to ascertain structural motifs key to inhibition is outlined. Furthermore, mechanism studies show that bacterial membrane disruption is central to the bacterial growth inhibition. During this process, it was discovered that analog C2 demonstrated a 4-fold better therapeutic index compared to the commercially available antimicrobial cetylpyridinium chloride (CPC) making it a viable alternative for oral care.
Collapse
Affiliation(s)
- Cristian Ochoa
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - Amy E. Solinski
- Department of Chemistry and the Emory Antibiotic Resistance Center, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Marcus Nowlan
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - Madeline M. Dekarske
- Department of Chemistry and the Emory Antibiotic Resistance Center, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - William M. Wuest
- Department of Chemistry and the Emory Antibiotic Resistance Center, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Marisa C. Kozlowski
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
12
|
Greene JR, Merrett KL, Heyert AJ, Simmons LF, Migliori CM, Vogt KC, Castro RS, Phillips PD, Baker JL, Lindberg GE, Fox DT, Del Sesto RE, Koppisch AT. Scope and efficacy of the broad-spectrum topical antiseptic choline geranate. PLoS One 2019; 14:e0222211. [PMID: 31527873 PMCID: PMC6748422 DOI: 10.1371/journal.pone.0222211] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/23/2019] [Indexed: 11/18/2022] Open
Abstract
Choline geranate (also described as Choline And GEranic acid, or CAGE) has been developed as a novel biocompatible antiseptic material capable of penetrating skin and aiding the transdermal delivery of co-administered antibiotics. The antibacterial properties of CAGE were analyzed against 24 and 72 hour old biofilms of 11 clinically isolated ESKAPE pathogens (defined as Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumanii, Pseudomonas aeruginosa, and Enterobacter sp, respectively), including multidrug resistant (MDR) isolates. CAGE was observed to eradicate in vitro biofilms at concentrations as low as 3.56 mM (0.156% v:v) in as little as 2 hours, which represents both an improved potency and rate of biofilm eradication relative to that reported for most common standard-of-care topical antiseptics in current use. In vitro time-kill studies on 24 hour old Staphylococcus aureus biofilms indicate that CAGE exerts its antibacterial effect upon contact and a 0.1% v:v solution reduced biofilm viability by over three orders of magnitude (a 3log10 reduction) in 15 minutes. Furthermore, disruption of the protective layer of exopolymeric substances in mature biofilms of Staphylococcus aureus by CAGE (0.1% v:v) was observed in 120 minutes. Insight into the mechanism of action of CAGE was provided with molecular modeling studies alongside in vitro antibiofilm assays. The geranate ion and geranic acid components of CAGE are predicted to act in concert to integrate into bacterial membranes, affect membrane thinning and perturb membrane homeostasis. Taken together, our results show that CAGE demonstrates all properties required of an effective topical antiseptic and the data also provides insight into how its observed antibiofilm properties may manifest.
Collapse
Affiliation(s)
- Joshua R. Greene
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
| | - Kahla L. Merrett
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
| | - Alexanndra J. Heyert
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
| | - Lucas F. Simmons
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
| | - Camille M. Migliori
- Department of Chemistry, Dixie State University, St. George, UT, United States of America
| | - Kristen C. Vogt
- Department of Chemistry, The College of New Jersey, Ewing, NJ, United States of America
| | - Rebeca S. Castro
- Department of Chemistry, The College of New Jersey, Ewing, NJ, United States of America
| | - Paul D. Phillips
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
| | - Joseph L. Baker
- Department of Chemistry, The College of New Jersey, Ewing, NJ, United States of America
| | - Gerrick E. Lindberg
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
- Center for Materials Interfaces in Research and Application, Northern Arizona University, Flagstaff, AZ, United States of America
| | - David T. Fox
- Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - Rico E. Del Sesto
- Department of Chemistry, Dixie State University, St. George, UT, United States of America
| | - Andrew T. Koppisch
- Department of Chemistry, Northern Arizona University, Flagstaff, AZ, United States of America
- Center for Materials Interfaces in Research and Application, Northern Arizona University, Flagstaff, AZ, United States of America
| |
Collapse
|
13
|
Chen J, Guo P, Zhang J, Rong J, Sun W, Jiang Y, Loh T. Synthesis of Functionalized α‐Vinyl Aldehydes from Enaminones. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201906213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jie Chen
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Pan Guo
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Jianguo Zhang
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Jiaxin Rong
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Wangbin Sun
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Yaojia Jiang
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Teck‐Peng Loh
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
- Division of Chemistry and Biological ChemistrySchool of Physical and Mathematical SciencesNanyang Technological University Singapore 637616 Singapore
| |
Collapse
|
14
|
Chen J, Guo P, Zhang J, Rong J, Sun W, Jiang Y, Loh T. Synthesis of Functionalized α‐Vinyl Aldehydes from Enaminones. Angew Chem Int Ed Engl 2019; 58:12674-12679. [DOI: 10.1002/anie.201906213] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Indexed: 01/07/2023]
Affiliation(s)
- Jie Chen
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Pan Guo
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Jianguo Zhang
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Jiaxin Rong
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Wangbin Sun
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Yaojia Jiang
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
| | - Teck‐Peng Loh
- Institute of Advanced SynthesisSchool of Chemistry and Molecular EngineeringNanjing Tech University Nanjing 211816 China
- Division of Chemistry and Biological ChemistrySchool of Physical and Mathematical SciencesNanyang Technological University Singapore 637616 Singapore
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Herndon JW. The chemistry of the carbon-transition metal double and triple bond: Annual survey covering the year 2017. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
18
|
Lakemeyer M, Zhao W, Mandl FA, Hammann P, Sieber SA. Thinking Outside the Box-Novel Antibacterials To Tackle the Resistance Crisis. Angew Chem Int Ed Engl 2018; 57:14440-14475. [PMID: 29939462 DOI: 10.1002/anie.201804971] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Indexed: 12/13/2022]
Abstract
The public view on antibiotics as reliable medicines changed when reports about "resistant superbugs" appeared in the news. While reasons for this resistance development are easily spotted, solutions for re-establishing effective antibiotics are still in their infancy. This Review encompasses several aspects of the antibiotic development pipeline from very early strategies to mature drugs. An interdisciplinary overview is given of methods suitable for mining novel antibiotics and strategies discussed to unravel their modes of action. Select examples of antibiotics recently identified by using these platforms not only illustrate the efficiency of these measures, but also highlight promising clinical candidates with therapeutic potential. Furthermore, the concept of molecules that disarm pathogens by addressing gatekeepers of virulence will be covered. The Review concludes with an evaluation of antibacterials currently in clinical development. Overall, this Review aims to connect select innovative antimicrobial approaches to stimulate interdisciplinary partnerships between chemists from academia and industry.
Collapse
Affiliation(s)
- Markus Lakemeyer
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85747, Garching, Germany
| | - Weining Zhao
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85747, Garching, Germany
| | - Franziska A Mandl
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85747, Garching, Germany
| | - Peter Hammann
- R&D Therapeutic Area Infectious Diseases, Sanofi-Aventis (Deutschland) GmbH, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Stephan A Sieber
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85747, Garching, Germany
| |
Collapse
|
19
|
Lakemeyer M, Zhao W, Mandl FA, Hammann P, Sieber SA. Über bisherige Denkweisen hinaus - neue Wirkstoffe zur Überwindung der Antibiotika-Krise. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201804971] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Markus Lakemeyer
- Fakultät für Chemie; Lehrstuhl für Organische Chemie II, Center for Integrated Protein Science (CIPSM); Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Weining Zhao
- Fakultät für Chemie; Lehrstuhl für Organische Chemie II, Center for Integrated Protein Science (CIPSM); Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Franziska A. Mandl
- Fakultät für Chemie; Lehrstuhl für Organische Chemie II, Center for Integrated Protein Science (CIPSM); Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| | - Peter Hammann
- R&D Therapeutic Area Infectious Diseases; Sanofi-Aventis (Deutschland) GmbH; Industriepark Höchst 65926 Frankfurt am Main Deutschland
| | - Stephan A. Sieber
- Fakultät für Chemie; Lehrstuhl für Organische Chemie II, Center for Integrated Protein Science (CIPSM); Technische Universität München; Lichtenbergstraße 4 85747 Garching Deutschland
| |
Collapse
|
20
|
Steele AD, Ernouf G, Lee YE, Wuest WM. Diverted Total Synthesis of the Baulamycins and Analogues Reveals an Alternate Mechanism of Action. Org Lett 2018; 20:1126-1129. [PMID: 29388431 PMCID: PMC5869691 DOI: 10.1021/acs.orglett.8b00054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The baulamycins were identified as in vitro siderophore biosynthesis inhibitors. Diverted total synthesis was used to construct the natural products and eight strategic analogues, three of which had improved inhibitory activity. Biological testing then revealed that membrane damage is the predominant mode of action in Staphylococcus aureus cells.
Collapse
Affiliation(s)
- Andrew D. Steele
- Department of Chemistry, Emory University, 1515 Dickey Drive Atlanta, Georgia 30322, United States
| | - Guillaume Ernouf
- Department of Chemistry, Emory University, 1515 Dickey Drive Atlanta, Georgia 30322, United States
| | - Young Eun Lee
- Department of Chemistry, Temple University, 1901 North 13th Street Philadelphia, Pennsylvania 19122, United States
| | - William M. Wuest
- Department of Chemistry, Emory University, 1515 Dickey Drive Atlanta, Georgia 30322, United States
| |
Collapse
|
21
|
Varkhedkar R, Dogra S, Tiwari D, Hussain Y, Yadav PN, Pandey G. Discovery of Novel Muscarinic Receptor Modulators by Integrating a Natural Product Framework and a Bioactive Molecule. ChemMedChem 2018; 13:384-395. [DOI: 10.1002/cmdc.201800001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Rajesh Varkhedkar
- Molecular Synthesis Laboratory, Centre of Biomedical Research (CBMR); Sanjay Gandhi Postgraduate Institute of Medical Sciences Campus; Raebareli Road Lucknow 226014 India
| | - Shalini Dogra
- Division of Pharmacology; CSIR - Central Drug Research Institute (CDRI); Lucknow 226031 India
| | - Divya Tiwari
- Molecular Synthesis Laboratory, Centre of Biomedical Research (CBMR); Sanjay Gandhi Postgraduate Institute of Medical Sciences Campus; Raebareli Road Lucknow 226014 India
| | - Yusuf Hussain
- Division of Pharmacology; CSIR - Central Drug Research Institute (CDRI); Lucknow 226031 India
| | - Prem Narayan Yadav
- Division of Pharmacology; CSIR - Central Drug Research Institute (CDRI); Lucknow 226031 India
| | - Ganesh Pandey
- Molecular Synthesis Laboratory, Centre of Biomedical Research (CBMR); Sanjay Gandhi Postgraduate Institute of Medical Sciences Campus; Raebareli Road Lucknow 226014 India
| |
Collapse
|
22
|
Solinski AE, Ochoa C, Lee YE, Paniak T, Kozlowski MC, Wuest WM. Honokiol-Inspired Analogs as Inhibitors of Oral Bacteria. ACS Infect Dis 2018; 4:118-122. [PMID: 29236466 PMCID: PMC5869685 DOI: 10.1021/acsinfecdis.7b00178] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The oral microbiome is a complex ecological niche where both commensal and pathogenic bacteria coexist. Previous reports have cited that the plant isolate honokiol is a potent inhibitor of S. mutans biofilms. Herein we report a cross-coupling method that provides access to a concise library of honokiol-inspired analogs. Through this work we determined that the inhibitory activity of honokiol is highly dependent on the growth conditions. Further, we identify a series of analogs that display significant potency against oral bacteria leading to the discovery of a potent antimicrobial.
Collapse
Affiliation(s)
- Amy E. Solinski
- Department of Chemistry, 1515 Dickey Drive, Emory University, Atlanta, Georgia 30322, United States
| | - Cristian Ochoa
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Young Eun Lee
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Thomas Paniak
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Marisa C. Kozlowski
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - William M. Wuest
- Department of Chemistry, 1515 Dickey Drive, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
23
|
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
|
24
|
Abstract
Natural products have served as powerful therapeutics against pathogenic bacteria since the golden age of antibiotics of the mid-20th century. However, the increasing frequency of antibiotic-resistant infections clearly demonstrates that new antibiotics are critical for modern medicine. Because combinatorial approaches have not yielded effective drugs, we propose that the development of new antibiotics around proven natural scaffolds is the best short-term solution to the rising crisis of antibiotic resistance. We analyze herein synthetic approaches aiming to reengineer natural products into potent antibiotics. Furthermore, we discuss approaches in modulating quorum sensing and biofilm formation as a nonlethal method, as well as narrow-spectrum pathogen-specific antibiotics, which are of interest given new insights into the implications of disrupting the microbiome.
Collapse
Affiliation(s)
- Sean E. Rossiter
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Madison H. Fletcher
- Department of Chemistry, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - William M. Wuest
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
25
|
Saint-Auret S, Abdelkafi H, Le Nouen D, Bisseret P, Blanchard N. Synthetic strategies towards mycolactone A/B, an exotoxin secreted by Mycobacterium ulcerans. Org Chem Front 2017. [DOI: 10.1039/c7qo00608j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pitfalls and dead-ends pave the way to mycolactone A/B. This full account reports synthetic efforts towards this natural product that eventually culminated in a de novo total synthesis.
Collapse
Affiliation(s)
- Sarah Saint-Auret
- Université de Strasbourg
- CNRS
- Laboratoire de Chimie Moléculaire UMR 7509
- 67000 Strasbourg
- France
| | - Hajer Abdelkafi
- Université de Strasbourg
- CNRS
- Laboratoire de Chimie Moléculaire UMR 7509
- 67000 Strasbourg
- France
| | - Didier Le Nouen
- Université de Haute-Alsace
- Laboratoire de Chimie Organique et Bioorganique EA 4566
- 68093 Mulhouse Cedex
- France
| | - Philippe Bisseret
- Université de Strasbourg
- CNRS
- Laboratoire de Chimie Moléculaire UMR 7509
- 67000 Strasbourg
- France
| | - Nicolas Blanchard
- Université de Strasbourg
- CNRS
- Laboratoire de Chimie Moléculaire UMR 7509
- 67000 Strasbourg
- France
| |
Collapse
|
26
|
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
|