1
|
Talarico L, Clemente I, Gennari A, Gabbricci G, Pepi S, Leone G, Bonechi C, Rossi C, Mattioli SL, Detta N, Magnani A. Physiochemical Characterization of Lipidic Nanoformulations Encapsulating the Antifungal Drug Natamycin. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:726. [PMID: 38668220 PMCID: PMC11053702 DOI: 10.3390/nano14080726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/09/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Natamycin is a tetraene polyene that exploits its antifungal properties by irreversibly binding components of fungal cell walls, blocking the growth of infections. However, topical ocular treatments with natamycin require frequent application due to the low ability of this molecule to permeate the ocular membrane. This limitation has limited the use of natamycin as an antimycotic drug, despite it being one of the most powerful known antimycotic agents. In this work, different lipidic nanoformulations consisting of transethosomes or lipid nanoparticles containing natamycin are proposed as carriers for optical topical administration. Size, stability and zeta potential were characterized via dynamic light scattering, the supramolecular structure was investigated via small- and wide-angle X-ray scattering and 1H-NMR, and the encapsulation efficiencies of the four proposed formulations were determined via HPLC-DAD.
Collapse
Affiliation(s)
- Luigi Talarico
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
- National Interuniversity Consortium of Material Science and Technology (INSTM), Siena Research Unit, Via G. Giusti 9, 50121 Firenze, Italy
- Center for Colloids and Surface Science (CSGI), Siena Research Group, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Ilaria Clemente
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
- Center for Colloids and Surface Science (CSGI), Siena Research Group, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Alessandro Gennari
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
| | - Giulia Gabbricci
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
- Center for Colloids and Surface Science (CSGI), Siena Research Group, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Simone Pepi
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
- National Interuniversity Consortium of Material Science and Technology (INSTM), Siena Research Unit, Via G. Giusti 9, 50121 Firenze, Italy
- Center for Colloids and Surface Science (CSGI), Siena Research Group, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Gemma Leone
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
- National Interuniversity Consortium of Material Science and Technology (INSTM), Siena Research Unit, Via G. Giusti 9, 50121 Firenze, Italy
- Center for Colloids and Surface Science (CSGI), Siena Research Group, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Claudia Bonechi
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
- Center for Colloids and Surface Science (CSGI), Siena Research Group, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Claudio Rossi
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
- Center for Colloids and Surface Science (CSGI), Siena Research Group, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Simone Luca Mattioli
- Dompé Farmaceutici S.p.A, Via Campo di Pile SNC, 67100 L’Aquila, Italy; (S.L.M.); (N.D.)
| | - Nicola Detta
- Dompé Farmaceutici S.p.A, Via Campo di Pile SNC, 67100 L’Aquila, Italy; (S.L.M.); (N.D.)
| | - Agnese Magnani
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.T.); (I.C.); (A.G.); (G.G.); (S.P.); (G.L.); (C.B.); (C.R.)
- National Interuniversity Consortium of Material Science and Technology (INSTM), Siena Research Unit, Via G. Giusti 9, 50121 Firenze, Italy
- Center for Colloids and Surface Science (CSGI), Siena Research Group, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
2
|
Pattini VC, Polaquini CR, Lemes TH, Brizzotti-Mazuchi NS, Sardi JDCO, Paziani MH, Kress MRVZ, de Almeida MTG, Regasini LO. Antifungal activity of 3,3'-dimethoxycurcumin (DMC) against dermatophytes and Candida species. Lett Appl Microbiol 2024; 77:ovae019. [PMID: 38499446 DOI: 10.1093/lambio/ovae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/24/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
Dermatomycosis is an infection with global impacts caused especially by dermatophytes and Candida species. Current antifungal therapies involve drugs that face fungal resistance barriers. This clinical context emphasizes the need to discover new antifungal agents. Herein, the antifungal potential of 10 curcumin analogs was evaluated against four Candida and four dermatophyte species. The most active compound, 3,3'-dimethoxycurcumin, exhibited minimum inhibitory concentration values ranging from 1.9‒62.5 to 15.6‒62.5 µg ml-1 against dermatophytes and Candida species, respectively. According to the checkerboard method, the association between DMC and terbinafine demonstrated a synergistic effect against Trichophyton mentagrophytes and Epidermophyton floccosum. Ergosterol binding test indicated DMC forms a complex with ergosterol of Candida albicans, C. krusei, and C. tropicalis. However, results from the sorbitol protection assay indicated that DMC had no effect on the cell walls of Candida species. The in vivo toxicity, using Galleria mellonella larvae, indicated no toxic effect of DMC. Altogether, curcumin analog DMC was a promising antifungal agent with a promising ability to act against Candida and dermatophyte species.
Collapse
Affiliation(s)
- Veridianna Camilo Pattini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Carlos Roberto Polaquini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Thiago Henrique Lemes
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, São Paulo 15054-000, Brazil
| | | | | | - Mário Henrique Paziani
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 05508-000, Brazil
| | - Marcia Regina von Zeska Kress
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 05508-000, Brazil
| | | | - Luis Octávio Regasini
- Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University, São José do Rio Preto, São Paulo 15054-000, Brazil
| |
Collapse
|
3
|
Zhong X, Yang J, Liu H, Yang Z, Luo P. Potential lipid-based strategies of amphotericin B designed for oral administration in clinical application. Drug Deliv 2023; 30:2161671. [PMID: 36601799 PMCID: PMC9828648 DOI: 10.1080/10717544.2022.2161671] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Amphotericin B (AmB) is regarded as a first-line therapy against life-threatening invasive fungal infections. Due to its poor oral bioavailability, AmB is restricted to intravenous administration in clinical practice. As science continues to move forward, two lipid-based formulations are successfully developed for oral AmB administration, currently undergoing phase I clinical trials. Encouragingly, lipid-AmB conjugates with emulsions also exhibit a better bioavailability, which may be another strategy to design oral AmB formulation in clinical practice. Thus, this review mainly focused on the two lipid-based formulations in clinical trials, and discussed the potential perspectives of AmB-lipid conjugation-loaded nanocochleates and emulsions.
Collapse
Affiliation(s)
- Xiaoming Zhong
- Department of Oncology Radiotherapy, Jiangxi Cancer Hospital, Nanchang, China
| | - Jianqiong Yang
- Department of Clinical Medicine Research Center, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China;
| | - Hongyan Liu
- Department of Pharmacy, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Zhiwen Yang
- Department of Pharmacy, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Ping Luo
- Department of Breast surgery, Nanchang Third Hospital, Nanchang, China,CONTACT Ping Luo Department of Breast surgery, Nanchang Third Hospital, Nanchang, China
| |
Collapse
|
4
|
Chen Y, Gao Y, Yuan M, Zheng Z, Yin J. Anti- Candida albicans Effects and Mechanisms of Theasaponin E1 and Assamsaponin A. Int J Mol Sci 2023; 24:ijms24119350. [PMID: 37298302 DOI: 10.3390/ijms24119350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/27/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Candida albicans is an opportunistic human fungal pathogen, and its drug resistance is becoming a serious problem. Camellia sinensis seed saponins showed inhibitory effects on resistant Candida albicans strains, but the active components and mechanisms are unclear. In this study, the effects and mechanisms of two Camellia sinensis seed saponin monomers, theasaponin E1 (TE1) and assamsaponin A (ASA), on a resistant Candida albicans strain (ATCC 10231) were explored. The minimum inhibitory concentration and minimum fungicidal concentration of TE1 and ASA were equivalent. The time-kill curves showed that the fungicidal efficiency of ASA was higher than that of TE1. TE1 and ASA significantly increased the cell membrane permeability and disrupted the cell membrane integrity of C. albicans cells, probably by interacting with membrane-bound sterols. Moreover, TE1 and ASA induced the accumulation of intracellular ROS and decreased the mitochondrial membrane potential. Transcriptome and qRT-PCR analyses revealed that the differentially expressed genes were concentrated in the cell wall, plasma membrane, glycolysis, and ergosterol synthesis pathways. In conclusion, the antifungal mechanisms of TE1 and ASA included the interference with the biosynthesis of ergosterol in fungal cell membranes, damage to the mitochondria, and the regulation of energy metabolism and lipid metabolism. Tea seed saponins have the potential to be novel anti-Candida albicans agents.
Collapse
Affiliation(s)
- Yuhong Chen
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute of Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China
- Graduate School of Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Ying Gao
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute of Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China
| | - Mingan Yuan
- Jinhua Academy of Agricultural Science, Jinhua 321000, China
| | - Zhaisheng Zheng
- Jinhua Academy of Agricultural Science, Jinhua 321000, China
| | - Junfeng Yin
- Key Laboratory of Tea Biology and Resources Utilization, Tea Research Institute of Chinese Academy of Agricultural Sciences, Ministry of Agriculture, 9 South Meiling Road, Hangzhou 310008, China
| |
Collapse
|
5
|
Alfadhli A, Romanaggi C, Barklis RL, Barklis E. Analysis of HIV-1 envelope cytoplasmic tail effects on viral replication. Virology 2023; 579:54-66. [PMID: 36603533 PMCID: PMC10003682 DOI: 10.1016/j.virol.2022.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
Trimers of the HIV-1 envelope (Env) protein perform receptor binding and virus-cell fusion functions during the virus life cycle. The cytoplasmic tail (CT) of Env forms an unusual baseplate structure, and is palmitoylated, rich in arginines, carries trafficking motifs, binds cholesterol, and interacts with host proteins. To dissect CT activities, we examined a panel of Env variants, including CT truncations, mutations, and an extension. We found that whereas all variants could replicate in permissive cells, viruses with CT truncations or baseplate mutations were defective in restrictive cells. We also identified a determinant in HIV-1 amphotericin sensitivity, and characterized variants that escape amphotericin inhibition via viral protease-mediated CT cleavage. Results additionally showed that full-length, his tagged Env can oligomerize and be co-assembled with CT truncations that delete portions of the baseplate, host protein binding sites, and trafficking signals. Our observations illuminate novel aspects of HIV-1 CT structure, interactions, and functions.
Collapse
Affiliation(s)
- Ayna Alfadhli
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR, USA
| | - CeAnn Romanaggi
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR, USA
| | - Robin Lid Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR, USA
| | - Eric Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR, USA.
| |
Collapse
|
6
|
Punia A, Choudhary P, Sharma N, Dahiya S, Gulia P, Chhillar AK. Therapeutic Approaches for Combating Aspergillus Associated Infection. Curr Drug Targets 2022; 23:1465-1488. [PMID: 35748549 DOI: 10.2174/1389450123666220623164548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 01/25/2023]
Abstract
Now-a-days fungal infection emerges as a significant problem to healthcare management systems due to high frequency of associated morbidity, mortality toxicity, drug-drug interactions, and resistance of the antifungal agents. Aspergillus is the most common mold that cause infection in immunocompromised hosts. It's a hyaline mold that is cosmopolitan and ubiquitous in nature. Aspergillus infects around 10 million population each year with a mortality rate of 30-90%. Clinically available antifungal formulations are restricted to four classes (i.e., polyene, triazole, echinocandin, and allylamine), and each of them have their own limitations associated with the activity spectrum, the emergence of resistance, and toxicity. Consequently, novel antifungal agents with modified and altered chemical structures are required to combat these invasive fungal infections. To overcome these limitations, there is an urgent need for new antifungal agents that can act as potent drugs in near future. Currently, some compounds have shown effective antifungal activity. In this review article, we have discussed all potential antifungal therapies that contain old antifungal drugs, combination therapies, and recent novel antifungal formulations, with a focus on the Aspergillus associated infections.
Collapse
Affiliation(s)
- Aruna Punia
- Department of Biotechnology, Maharishi Dayanand University, Rohtak, Haryana 124001, India
| | - Pooja Choudhary
- Department of Biotechnology, Maharishi Dayanand University, Rohtak, Haryana 124001, India
| | - Namita Sharma
- Department of Biotechnology, Maharishi Dayanand University, Rohtak, Haryana 124001, India
| | - Sweety Dahiya
- Department of Biotechnology, Maharishi Dayanand University, Rohtak, Haryana 124001, India
| | - Prity Gulia
- Department of Biotechnology, Maharishi Dayanand University, Rohtak, Haryana 124001, India
| | - Anil K Chhillar
- Department of Biotechnology, Maharishi Dayanand University, Rohtak, Haryana 124001, India
| |
Collapse
|
7
|
Szomek M, Reinholdt P, Walther HL, Scheidt HA, Müller P, Obermaier S, Poolman B, Kongsted J, Wüstner D. Natamycin sequesters ergosterol and interferes with substrate transport by the lysine transporter Lyp1 from yeast. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184012. [PMID: 35914570 DOI: 10.1016/j.bbamem.2022.184012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/30/2022] [Accepted: 07/21/2022] [Indexed: 06/15/2023]
Abstract
Natamycin is a polyene macrolide, widely employed to treat fungal keratitis and other yeast infections as well as to protect food products against fungal molds. In contrast to other polyene macrolides, such as nystatin or amphotericin B, natamycin does not form pores in yeast membranes, and its mode of action is not well understood. Here, we have employed a variety of spectroscopic methods, computational modeling, and membrane reconstitution to study the molecular interactions of natamycin underlying its antifungal activity. We find that natamycin forms aggregates in an aqueous solution with strongly altered optical properties compared to monomeric natamycin. Interaction of natamycin with model membranes results in a concentration-dependent fluorescence increase which is more pronounced for ergosterol- compared to cholesterol-containing membranes up to 20 mol% sterol. Evidence for formation of specific ergosterol-natamycin complexes in the bilayer is provided. Using nuclear magnetic resonance (NMR) and electron spin resonance (ESR) spectroscopy, we find that natamycin sequesters sterols, thereby interfering with their well-known ability to order acyl chains in lipid bilayers. This effect is more pronounced for membranes containing the sterol of fungi, ergosterol, compared to those containing mammalian cholesterol. Natamycin interferes with ergosterol-dependent transport of lysine by the yeast transporter Lyp1, which we propose to be due to the sequestering of ergosterol, a mechanism that also affects other plasma membrane proteins. Our results provide a mechanistic explanation for the selective antifungal activity of natamycin, which can set the stage for rational design of novel polyenes in the future.
Collapse
Affiliation(s)
- Maria Szomek
- Department of Biochemistry and Molecular Biology, PhyLife, Physical Life Sciences, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Peter Reinholdt
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Hanna-Loisa Walther
- Department of Biochemistry and Molecular Biology, PhyLife, Physical Life Sciences, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Holger A Scheidt
- Institute for Medical Physics and Biophysics, University of Leipzig, Härtelstr. 16-18, 04107 Leipzig, Germany
| | - Peter Müller
- Department of Biology, Humboldt University Berlin, Invalidenstr. 43, 10115 Berlin, Germany
| | - Sebastian Obermaier
- Department of Biochemistry, University of Groningen, Nijenborgh 4, 9747 Groningen, the Netherlands
| | - Bert Poolman
- Department of Biochemistry, University of Groningen, Nijenborgh 4, 9747 Groningen, the Netherlands
| | - Jacob Kongsted
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Daniel Wüstner
- Department of Biochemistry and Molecular Biology, PhyLife, Physical Life Sciences, University of Southern Denmark, DK-5230 Odense M, Denmark.
| |
Collapse
|
8
|
Goode A, Yeh V, Bonev BB. Interactions of polymyxin B with lipopolysaccharide-containing membranes. Faraday Discuss 2021; 232:317-329. [PMID: 34550139 PMCID: PMC8704168 DOI: 10.1039/d1fd00036e] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bacterial resistance to antibiotics constantly remodels the battlefront between infections and antibiotic therapy. Polymyxin B, a cationic peptide with an anti-Gram-negative spectrum of activity is re-entering use as a last resort measure and as an adjuvant. We use fluorescence dequenching to investigate the role of the rough chemotype bacterial lipopolysaccharide from E. coli BL21 as a molecular facilitator of membrane disruption by LPS. The minimal polymyxin B/lipid ratio required for leakage onset increased from 5.9 × 10−4 to 1.9 × 10−7 in the presence of rLPS. We confirm polymyxin B activity against E. coli BL21 by the agar diffusion method and determined a MIC of 291 μg ml−1. Changes in lipid membrane stability and dynamics in response to polymyxin and the role of LPS are investigated by 31P NMR and high resolution 31P MAS NMR relaxation is used to monitor selective molecular interactions between polymyxin B and rLPS within bilayer lipid membranes. We observe a strong facilitating effect from rLPS on the membrane lytic properties of polymyxin B and a specific, pyrophosphate-mediated process of molecular recognition of LPS by polymyxin B. Polymyxin B uses bacterial LPS as docking receptor to cross the outer membrane.![]()
Collapse
Affiliation(s)
- Alice Goode
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | - Vivien Yeh
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | - Boyan B Bonev
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| |
Collapse
|
9
|
Guo X, Zhang J, Li X, Xiao E, Lange JD, Rienstra CM, Burke MD, Mitchell DA. Sterol Sponge Mechanism Is Conserved for Glycosylated Polyene Macrolides. ACS CENTRAL SCIENCE 2021; 7:781-791. [PMID: 34079896 PMCID: PMC8161476 DOI: 10.1021/acscentsci.1c00148] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Indexed: 05/07/2023]
Abstract
Amphotericin-like glycosylated polyene macrolides (GPMs) are a clinically and industrially important family of natural products, but the mechanisms by which they exert their extraordinary biological activities have remained unclear for more than half a century. Amphotericin B exerts fungicidal action primarily via self-assembly into an extramembranous sponge that rapidly extracts ergosterol from fungal membranes, but it has remained unclear whether this mechanism is applicable to other GPMs. Using a highly conserved polyene-hemiketal region of GPMs that we hypothesized to represent a conserved ergosterol-binding domain, we bioinformatically mapped the entirety of the GPM sequence-function space and expanded the number of GPM biosynthetic gene clusters (BGCs) by 10-fold. We further leveraged bioinformatic predictions and tetrazine-based reactivity screening targeting the electron-rich polyene region of GPMs to discover a first-in-class methyltetraene- and diepoxide-containing GPM, kineosporicin, and to assign BGCs to many new producers of previously reported members. Leveraging a range of structurally diverse known and newly discovered GPMs, we found that the sterol sponge mechanism of fungicidal action is conserved.
Collapse
Affiliation(s)
- Xiaorui Guo
- Department
of Chemistry, Roger Adams Laboratory, University
of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
| | - Jiabao Zhang
- Department
of Chemistry, Roger Adams Laboratory, University
of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, 1206 W. Gregory Avenue, Urbana, Illinois 61801, United States
| | - Xinyi Li
- Department
of Biochemistry, Roger Adams Laboratory, University of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
| | - Emily Xiao
- Department
of Chemistry, Roger Adams Laboratory, University
of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
| | - Justin D. Lange
- Department
of Chemistry, Roger Adams Laboratory, University
of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, 1206 W. Gregory Avenue, Urbana, Illinois 61801, United States
| | - Chad M. Rienstra
- Department
of Biochemistry and National Magnetic Resonance Facility at Madison, DeLuca Biochemistry Laboratories, 433 Babcock Drive, Madison, Wisconsin 53706, United States
| | - Martin D. Burke
- Department
of Chemistry, Roger Adams Laboratory, University
of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, 1206 W. Gregory Avenue, Urbana, Illinois 61801, United States
- Department
of Biochemistry, Roger Adams Laboratory, University of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
- Beckman
Institute for Advanced Science and Technology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801 United States
| | - Douglas A. Mitchell
- Department
of Chemistry, Roger Adams Laboratory, University
of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, 1206 W. Gregory Avenue, Urbana, Illinois 61801, United States
- Department
of Microbiology, University of Illinois
at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
10
|
The Antifungal Mechanism of Amphotericin B Elucidated in Ergosterol and Cholesterol-Containing Membranes Using Neutron Reflectometry. NANOMATERIALS 2020; 10:nano10122439. [PMID: 33291326 PMCID: PMC7762259 DOI: 10.3390/nano10122439] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/25/2022]
Abstract
We have characterized and compared the structures of ergosterol- and cholesterol-containing 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) membranes before and after interaction with the amphiphilic antifungal drug amphotericin B (AmB) using neutron reflection. AmB inserts into both pure POPC and sterol-containing membranes in the lipid chain region and does not significantly perturb the structure of pure POPC membranes. By selective per-deuteration of the lipids/sterols, we show that AmB extracts ergosterol but not cholesterol from the bilayers and inserts to a much higher degree in the cholesterol-containing membranes. Ergosterol extraction by AmB is accompanied by membrane thinning. Our results provide new insights into the mechanism and antifungal effect of AmB in these simple models of fungal and mammalian membranes and help understand the molecular origin of its selectivity and toxic side effects.
Collapse
|
11
|
Mishra A, Bano M, Bisen AC, Verma S, Sanap SN, Kishor R, Shukla P, Bhatta RS. Topical corneal targeted sustained release amphotericin B liposomal formulation for the treatment of fungal keratitis and its PK-PD evaluation. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
12
|
Jednačak T, Mikulandra I, Novak P. Advanced Methods for Studying Structure and Interactions of Macrolide Antibiotics. Int J Mol Sci 2020; 21:E7799. [PMID: 33096889 PMCID: PMC7589898 DOI: 10.3390/ijms21207799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 11/17/2022] Open
Abstract
Macrolide antibiotics are macrocyclic compounds that are clinically used and prescribed for the treatment of upper and lower respiratory tract infections. They inhibit the synthesis of bacterial proteins by reversible binding to the 23S rRNA at or near the peptidyl transferase center. However, their excellent antibacterial profile was largely compromised by the emergence of bacterial resistance. Today, fighting resistance to antibiotics is one of the greatest challenges in medicinal chemistry. Considering various physicochemical properties of macrolides, understanding their structure and interactions with macromolecular targets is crucial for the design of new antibiotics efficient against resistant pathogens. The solid-state structures of some macrolide-ribosome complexes have recently been solved, throwing new light on the macrolide binding mechanisms. On the other hand, a combination of NMR spectroscopy and molecular modeling calculations can be applied to study free and bound conformations in solution. In this article, a description of advanced physicochemical methods for elucidating the structure and interactions of macrolide antibiotics in solid state and solution will be provided, and their principal advantages and drawbacks will be discussed.
Collapse
Affiliation(s)
- Tomislav Jednačak
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia;
| | | | - Predrag Novak
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, HR-10000 Zagreb, Croatia;
| |
Collapse
|
13
|
SERAFINI KFC, ALENCAR ER, RIBEIRO JL, FERREIRA MDA. Influence of the salt concentration on action mechanisms of natamycin against microorganisms of importance in food manufacture. FOOD SCIENCE AND TECHNOLOGY 2020. [DOI: 10.1590/fst.33018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
14
|
Ledwaba MB, Ndumnego OC, Matle I, Gelaw AK, Van Heerden H. Investigating selective media for optimal isolation of Brucella spp. in South Africa. Onderstepoort J Vet Res 2020; 87:e1-e9. [PMID: 32129638 PMCID: PMC7136694 DOI: 10.4102/ojvr.v87i1.1792] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 01/22/2020] [Accepted: 10/01/2019] [Indexed: 01/22/2023] Open
Abstract
Bovine brucellosis in South Africa is caused mainly by Brucella abortus biovar (bv.) 1 and less frequently by B. abortus bv. 2. Bacterial isolation is regarded as the gold standard for diagnosis of Brucella species; however, it is not very sensitive. The aim of this study was to determine the selective medium with optimum antibiotic composition that will allow the growth of Brucella species (spp.) while inhibiting moulds, yeast and most, if not all, Gram-negative contaminants in South Africa. In the controlled experiment, modified Agrifood Research and Technology Center of Aragon (CITA) medium (mCITA) seemed to be the optimum selective medium for isolation of Brucella spp. as compared with Farrell's medium (FM) and modified Thayer Martin (mTM), while FM inhibited the growth of most fungal and bacterial contaminants. Mean comparison between the three media used to culture B. abortus resulted in lower mean difference ranging from 0 to 2.33. In case of Brucella ovis, high mean difference was obtained when comparing FM with mCITA (10.33) and mTM (12). However, the mean differences of 0.67 and 1.67 were obtained when comparing mCITA and mTM media used to, respectively, culture pasteurised and raw milk spiked with B. ovis. Further optimisation at the Agricultural Research Council - Onderstepoort Veterinary Research Institute resulted in a comparable performance between FM and mCITA; however, mCITA allowed optimal growth of the fastidious B. ovis, which is generally inhibited on FM. Generally, mCITA seemed to be the optimum selective medium for isolation of Brucella spp., while FM inhibits the growth of most fungal and bacterial contaminants. Thus, veterinary laboratories can use mCITA and/or FM but should take into consideration the detection of factious Brucella isolated in the country or region.
Collapse
Affiliation(s)
- Maphuti B Ledwaba
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria.
| | | | | | | | | |
Collapse
|
15
|
Abstract
Aspergillus fumigatus is a saprotrophic fungus; its primary habitat is the soil. In its ecological niche, the fungus has learned how to adapt and proliferate in hostile environments. This capacity has helped the fungus to resist and survive against human host defenses and, further, to be responsible for one of the most devastating lung infections in terms of morbidity and mortality. In this review, we will provide (i) a description of the biological cycle of A. fumigatus; (ii) a historical perspective of the spectrum of aspergillus disease and the current epidemiological status of these infections; (iii) an analysis of the modes of immune response against Aspergillus in immunocompetent and immunocompromised patients; (iv) an understanding of the pathways responsible for fungal virulence and their host molecular targets, with a specific focus on the cell wall; (v) the current status of the diagnosis of different clinical syndromes; and (vi) an overview of the available antifungal armamentarium and the therapeutic strategies in the clinical context. In addition, the emergence of new concepts, such as nutritional immunity and the integration and rewiring of multiple fungal metabolic activities occurring during lung invasion, has helped us to redefine the opportunistic pathogenesis of A. fumigatus.
Collapse
Affiliation(s)
- Jean-Paul Latgé
- School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Georgios Chamilos
- School of Medicine, University of Crete, Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Crete, Greece
| |
Collapse
|