1
|
Bombelli A, Calligari P, Bocchinfuso G, Vincken JP, Abee T, den Besten HM, Stella L, Araya-Cloutier C. Mechanistic Insights into the Membrane Permeabilization Activity of Antimicrobial Prenylated Isoflavonoids: A Comparative Study of Glabridin, Wighteone, and Lupiwighteone. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6668-6677. [PMID: 40040444 PMCID: PMC11926876 DOI: 10.1021/acs.jafc.5c01688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
Prenylated isoflavonoids have shown remarkable antimicrobial activity. Previous studies showed that the antimicrobial compounds glabridin and wighteone induced membrane permeabilization in microbial cells. Other compounds, such as lupiwighteone, showed no antimicrobial activity. In this study, the permeabilization efficacy and interaction with lipid bilayers of glabridin, wighteone, and lupiwighteone were assessed in vitro and in silico using model membranes. Permeabilization of liposomes by glabridin and wighteone confirmed the lipid bilayer as the primary target. Notably, lupiwighteone did not induce the permeabilization of liposomes. Molecular dynamics (MD) simulations were used to study the interaction of these compounds with phospholipid membranes. The calculated potential of mean force profiles for the three molecules correlated with liposome permeabilization, indicating a favorable intercalation inside the lipid bilayer for wighteone, followed by glabridin, and an unfavorable intercalation for lupiwighteone. Additionally, MD simulations indicated that the location of glabridin and wighteone in the membrane was just below the head groups. Furthermore, this study underscored the importance of partitioning between polar and hydrophobic areas for prenylated isoflavonoids, which conceivably determines the membrane insertion and, subsequently, the antimicrobial activity. Overall, this study showed that interactions with and permeabilization of the lipid bilayer are key factors for the antimicrobial activity of these compounds.
Collapse
Affiliation(s)
- Alberto Bombelli
- Food
Microbiology, Wageningen University &
Research, AA Wageningen 6700, the Netherlands
- Food
Chemistry, Wageningen University & Research, AA Wageningen 6700, the Netherlands
| | - Paolo Calligari
- Department
of Chemical Science and Technologies, Tor
Vergata University of Rome, Rome 00133, Italy
| | - Gianfranco Bocchinfuso
- Department
of Chemical Science and Technologies, Tor
Vergata University of Rome, Rome 00133, Italy
| | - Jean-Paul Vincken
- Food
Chemistry, Wageningen University & Research, AA Wageningen 6700, the Netherlands
| | - Tjakko Abee
- Food
Microbiology, Wageningen University &
Research, AA Wageningen 6700, the Netherlands
| | - Heidy M.W. den Besten
- Food
Microbiology, Wageningen University &
Research, AA Wageningen 6700, the Netherlands
| | - Lorenzo Stella
- Department
of Chemical Science and Technologies, Tor
Vergata University of Rome, Rome 00133, Italy
| | - Carla Araya-Cloutier
- Food
Chemistry, Wageningen University & Research, AA Wageningen 6700, the Netherlands
| |
Collapse
|
2
|
Tong X, Fu X, Gong A, Yu G, Chen N, Chen B, Gu J, Liu Z. Effect of Luteolin on cadmium-inhibited bone growth via suppressing osteoclastogenesis in laying chickens. J Anim Sci 2025; 103:skaf033. [PMID: 39921628 PMCID: PMC11912829 DOI: 10.1093/jas/skaf033] [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: 11/18/2024] [Accepted: 02/07/2025] [Indexed: 02/10/2025] Open
Abstract
Luteolin (Lut) is a flavonoid derived from several plant sources. Cadmium (Cd) is a widespread environmental contaminant and potential toxin with detrimental effects on animal health. However, the effect of Lut on Cd-induced inhibition of bone growth in laying chickens remains unclear. This study investigates the effects of Lut on Cd-induced inhibition of bone growth in the femur and tibia of laying chickens. A total of sixty 1-d-old green-eggshell yellow feather laying chickens were randomly assigned to 4 groups after a 5-d acclimation period: basal diet (Con), cadmium chloride (CdCl2, Cd), Lut, and Lut + Cd. Bone microstructure, serum biomarkers of bone remodeling, the levels of Cd, calcium (Ca), phosphorus (P), and trace metal elements were assessed using the micro-computed tomography (Micro-CT), enzyme-linked immunosorbent assay (ELISA), and microwave digestion, respectively. Bone remodeling biomarkers, late endosomal/lysosomal adaptor and MAPK and mTOR activator 1 (LAMTOR1), as well as the phosphorylation of AMP-activated protein kinase α (AMPKα) and protein kinase B (Akt), were quantified using the qRT-PCR and western blot. The results indicated that Lut effectively mitigated Cd-induced bone mass loss compared to the Cd group, resulting in increased bone volume (BV), bone surface/BV (BS/BV), connectivity density (Conn.Dn), and the length and weight of the femur and tibia in laying chickens. Mechanistically, compared to the Cd group, Lut restored the ratio of osteoprotegerin (OPG)/receptor activator of NF-κB ligand (RANKL) in serum and bone tissue, enhanced the expression of bone morphogenetic protein-2 (BMP-2), runt-related transcription factor 2 (Runx2), osteocalcin (OCN), and Osterix (OSX), while reducing the levels of Ca, Cd, and alkaline phosphatase (ALP) activity, as well as the expression of osteopontin (OPN), c-Fos, osteoclast stimulatory-transmembrane protein (OC-STAMP), tartrate-resistant acid phosphatase, cathepsin K (CTSK), matrix metalloprotein-9 (MMP-9), LAMTOR1, and the phosphorylation of AMPKα and Akt. Therefore, Lut alleviates Cd-induced damage to the femur and tibia of chickens by promoting osteogenesis and inhibiting osteoclastogenesis, positioning Lut as a potential therapeutic plant extract for enhancing bone growth in laying chickens.
Collapse
Affiliation(s)
- Xishuai Tong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Xiaohui Fu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Anqing Gong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Gengsheng Yu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Naineng Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Bing Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Jianhong Gu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| | - Zongping Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of The Ministry of Education of China, Institute of Agricultural Science and Technology Development, College of Veterinary Medicine, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, P. R. China
| |
Collapse
|
3
|
Yarmolinsky L, Nakonechny F, Haddis T, Khalfin B, Dahan A, Ben-Shabat S. Natural Antimicrobial Compounds as Promising Preservatives: A Look at an Old Problem from New Perspectives. Molecules 2024; 29:5830. [PMID: 39769919 PMCID: PMC11728848 DOI: 10.3390/molecules29245830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/14/2025] Open
Abstract
Antimicrobial compounds of natural origin are of interest because of the large number of reports regarding the harmfulness of food preservatives. These natural products can be derived from plants, animal sources, microorganisms, algae, or mushrooms. The aim of this review is to consider known antimicrobials of natural origin and the mechanisms of their action, antimicrobial photodynamic technology, and ultrasound for disinfection. Plant extracts and their active compounds, chitosan and chitosan oligosaccharide, bioactive peptides, and essential oils are highly potent preservatives. It has been experimentally proven that they possess strong antibacterial capabilities against bacteria, yeast, and fungi, indicating the possibility of their use in the future to create preservatives for the pharmaceutical, agricultural, and food industries.
Collapse
Affiliation(s)
- Ludmila Yarmolinsky
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (L.Y.); (B.K.)
| | - Faina Nakonechny
- Department of Chemical Engineering, Ariel University, Ariel 4070000, Israel; (F.N.); (T.H.)
| | - Tigabu Haddis
- Department of Chemical Engineering, Ariel University, Ariel 4070000, Israel; (F.N.); (T.H.)
| | - Boris Khalfin
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (L.Y.); (B.K.)
| | - Arik Dahan
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (L.Y.); (B.K.)
| | - Shimon Ben-Shabat
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (L.Y.); (B.K.)
| |
Collapse
|
4
|
Gao Y, Zhu J, Zheng X, Lin G, Miao P. Luteolin Functionalized Zinc Oxide Nanoparticles for Cancer Therapy Based on Autophagy Activation and EMT Inhibition. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26363-26369. [PMID: 39589485 DOI: 10.1021/acs.langmuir.4c04194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Natural bioactive compounds have become increasingly important antitumor and antimicrobial agents due to their high therapeutic potential and low systemic toxicity. However, they also have significant drawbacks like nontumor targeting and low bioavailability, which limit their wide therapeutic applications. There is an urgent need for a safe and compatible nanoparticle-mediated drug delivery system for the efficient transport of drug candidates into tumor tissues. In this paper, amine-modified zinc oxide nanoparticles are synthesized to load luteolin. The functionalized nanoparticles exhibit good synergistic effects in terms of antitumor and antibacterial activities. They show significant induction of oxidative stress and antitumor effects, while no significant inhibitory effect on normal cells is observed, suggesting that the nanocomposites have good targeting of cancer cells. Autophagy activation and EMT inhibition effects are studied to explain the good antitumor performances. The prepared nanocomposites are demonstrated to be attractive candidates for cancer therapy.
Collapse
Affiliation(s)
- Yan Gao
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Jinwen Zhu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Xingye Zheng
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Guanghui Lin
- School of Biomedical Engineering, Sun Yat-Sen University, Shenzhen 518707, China
| | - Peng Miao
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
5
|
Martynyuk VA, Efimova SS, Malykhina AI, Ostroumova OS. The effects of plant flavones on the membrane boundary potential and lipid packing stress. Colloids Surf B Biointerfaces 2024; 245:114269. [PMID: 39341052 DOI: 10.1016/j.colsurfb.2024.114269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/12/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024]
Abstract
Here we have revealed the effects of different plant flavones on the physicochemical properties of model lipid membranes. We have demonstrated that baicalein increases the boundary potential of membranes composed of phosphatidylcholine, while wogonin does not affect it. Other flavones tested reduce membrane boundary potential, with this ability increasing among scutellarein, chrysin, apigenin, morin, fisetin, and luteolin. Molecular dynamics simulations demonstrate connection of alteration in boundary potential with the preferential orientation of intrinsic flavone dipole moments in membranes. We have also shown that flavones reduce the melting point of phosphatidylcholine, and this ability increases in the series of luteolin, morin, wogonin, scutellarein, apigenin, baicalein, chrysin, and fisetin. The introduction of baicalein, chrysin and fisetin also leads to a significant decrease in the sharpness of the lipid phase transition. We have hypothesized that the localization of flavones in the glycerol backbone or in the C1-C8 methylene region of lipid hydrocarbon chains leads to an increase in the area per lipid and, as a consequence, to an expansion of the lipid melting peak. Replacement of neutral phosphatidylcholine with negatively charged phosphatidylserine affects the membrane-modifying activity of flavones which given the externalization of phosphatidylserine on the surface of cancer cells may be crucial in the flavone anticancer effects.
Collapse
Affiliation(s)
- Vera A Martynyuk
- Institute of Cytology of Russian Academy of Sciences, ikhoretsky 4, Saint Petersburg 194064, Russian Federation
| | - Svetlana S Efimova
- Institute of Cytology of Russian Academy of Sciences, ikhoretsky 4, Saint Petersburg 194064, Russian Federation
| | - Anna I Malykhina
- Institute of Cytology of Russian Academy of Sciences, ikhoretsky 4, Saint Petersburg 194064, Russian Federation
| | - Olga S Ostroumova
- Institute of Cytology of Russian Academy of Sciences, ikhoretsky 4, Saint Petersburg 194064, Russian Federation.
| |
Collapse
|
6
|
Eloïse L, Petit L, Nominé Y, Heurtault B, Ben Hadj Kaddour I, Senger B, Rodon Fores J, Vrana NE, Barbault F, Lavalle P. The antibacterial properties of branched peptides based on poly(l-arginine): In vitro antibacterial evaluation and molecular dynamic simulations. Eur J Med Chem 2024; 268:116224. [PMID: 38387338 DOI: 10.1016/j.ejmech.2024.116224] [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: 12/08/2023] [Revised: 01/27/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024]
Abstract
The emergence of bacterial strains resistant to antibiotics is a major issue in the medical field. Antimicrobial peptides are widely studied as they do not generate as much resistant bacterial strains as conventional antibiotics and present a broad range of activity. Among them, the homopolypeptide poly(l-arginine) presents promising antibacterial properties, especially in the perspective of its use in biomaterials. Linear poly(l-arginine) has been extensively studied but the impact of its 3D structure remains unknown. In this study, the antibacterial properties of newly synthesized branched poly(l-arginine) peptides, belonging to the family of multiple antigenic peptides, are evaluated. First, in vitro activities of the peptides shows that branched poly(l-arginine) is more efficient than linear poly(l-arginine) containing the same number of arginine residues. Surprisingly, peptides with more arms and more residues are not the most effective. To better understand these unexpected results, interactions between these peptides and the membranes of Gram positive and Gram negative bacteria are simulated thanks to molecular dynamic. It is observed that the bacterial membrane is more distorted by the branched structure than by the linear one and by peptides containing smaller arms. This mechanism of action is in full agreement with in vitro results and suggest that our simulations form a robust model to evaluate peptide efficiency towards pathogenic bacteria.
Collapse
Affiliation(s)
- Lebaudy Eloïse
- Inserm UMR_S 1121, EMR 7003 CNRS, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, F67000, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| | - Lauriane Petit
- Inserm UMR_S 1121, EMR 7003 CNRS, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, F67000, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France; SPARTHA Medical, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Yves Nominé
- Institut de génétique et de biologie moléculaire et cellulaire, IGBMC, Illkirch, France
| | - Béatrice Heurtault
- Université de Strasbourg, Centre national de la recherche scientifique (CNRS), Laboratoire de Conception et Application de Molécules Bioactives UMR 7199, Faculté de Pharmacie, Illkirch, France
| | - Inès Ben Hadj Kaddour
- Inserm UMR_S 1121, EMR 7003 CNRS, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, F67000, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France; SPARTHA Medical, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Bernard Senger
- Inserm UMR_S 1121, EMR 7003 CNRS, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, F67000, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| | - Jennifer Rodon Fores
- Inserm UMR_S 1121, EMR 7003 CNRS, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, F67000, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| | - Nihal Engin Vrana
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France; SPARTHA Medical, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | | | - Philippe Lavalle
- Inserm UMR_S 1121, EMR 7003 CNRS, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, F67000, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France; SPARTHA Medical, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France.
| |
Collapse
|
7
|
Shang J, Yang J, Deng Q, Zhou M. Nano-scale drug delivery systems for luteolin: advancements and applications. J Mater Chem B 2023; 11:11198-11216. [PMID: 37986608 DOI: 10.1039/d3tb01753b] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Luteolin (Lu) is a naturally occurring flavonoid compound with a diverse array of pharmacological activities, including anti-tumor, anti-inflammatory, antibacterial, and neuroprotective properties. However, the therapeutic efficacy and clinical application of Lu are significantly hindered by inherent limitations, such as poor water solubility, short half-life, low bioavailability, and potential off-target toxicity. Recent studies have demonstrated that the utilization of nanocarriers presents a promising strategy to enhance the solubility of Lu, prolong its circulation time, and improve its targeting ability. Despite numerous reviews over the past few decades having focused on the source, pharmacological activities, and molecular mechanisms of Lu, there exists a conspicuous gap in the literature regarding a comprehensive review of Lu-loaded nanoformulations and their applications. To address this gap, we present an exhaustive overview of the advancements and applications of nano-scale drug delivery systems specifically designed for Lu. These platforms encompass micelles, nanocarrier-based systems, emulsified drug delivery systems, and vesicular drug delivery systems. We provide detailed insights into the synthetic materials, preparation methods, physicochemical properties, and significant outcomes associated with these nanoformulations. This systematic review will be particularly valuable to researchers seeking novel avenues in the field of nano-delivery strategies and exploring the potential clinical applications of Lu.
Collapse
Affiliation(s)
- Jinlu Shang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jing Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qinmin Deng
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
8
|
Izbicka E, Streeper RT. Mitigation of Insulin Resistance by Natural Products from a New Class of Molecules, Membrane-Active Immunomodulators. Pharmaceuticals (Basel) 2023; 16:913. [PMID: 37513825 PMCID: PMC10386479 DOI: 10.3390/ph16070913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Insulin resistance (IR), accompanied by an impaired cellular glucose uptake, characterizes diverse pathologies that include, but are not limited to, metabolic disease, prediabetes and type 2 diabetes. Chronic inflammation associated with deranged cellular signaling is thought to contribute to IR. The key molecular players in IR are plasma membrane proteins, including the insulin receptor and glucose transporter 4. Certain natural products, such as lipids, phenols, terpenes, antibiotics and alkaloids have beneficial effects on IR, yet their mode of action remains obscured. We hypothesized that these products belong to a novel class of bioactive molecules that we have named membrane-active immunomodulators (MAIMs). A representative MAIM, the naturally occurring medium chain fatty acid ester diethyl azelate (DEA), has been shown to increase the fluidity of cell plasma membranes with subsequent downstream effects on cellular signaling. DEA has also been shown to improve markers of IR, including blood glucose, insulin and lipid levels, in humans. The literature supports the notion that DEA and other natural MAIMs share similar mechanisms of action in improving IR. These findings shed a new light on the mechanism of IR mitigation using natural products, and may facilitate the discovery of other compounds with similar activities.
Collapse
|
9
|
Xue P, Sang R, Li N, Du S, Kong X, Tai M, Jiang Z, Chen Y. A new approach to overcoming antibiotic-resistant bacteria: Traditional Chinese medicine therapy based on the gut microbiota. Front Cell Infect Microbiol 2023; 13:1119037. [PMID: 37091671 PMCID: PMC10117969 DOI: 10.3389/fcimb.2023.1119037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/12/2023] [Indexed: 04/25/2023] Open
Abstract
With the irrational use of antibiotics and the increasing abuse of oral antibiotics, the drug resistance of gastrointestinal pathogens has become a prominent problem in clinical practice. Gut microbiota plays an important role in maintaining human health, and the change of microbiota also affects the activity of pathogenic bacteria. Interfering with antibiotic resistant bacteria by affecting gut microbiota has also become an important regulatory signal. In clinical application, due to the unique advantages of traditional Chinese medicine in sterilization and drug resistance, it is possible for traditional Chinese medicine to improve the gut microbial microenvironment. This review discusses the strategies of traditional Chinese medicine for the treatment of drug-resistant bacterial infections by changing the gut microenvironment, unlocking the interaction between microbiota and drug resistance of pathogenic bacteria.
Collapse
Affiliation(s)
- Peng Xue
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Rui Sang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Nan Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Siyuan Du
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xiuwen Kong
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Mingliang Tai
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Zhihao Jiang
- Center for Basic Medical Research, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Ying Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
- *Correspondence: Ying Chen,
| |
Collapse
|
10
|
Chen X, Wu X, Wang S. An optimized antimicrobial peptide analog acts as an antibiotic adjuvant to reverse methicillin-resistant Staphylococcus aureus. NPJ Sci Food 2022; 6:57. [PMID: 36509755 PMCID: PMC9744894 DOI: 10.1038/s41538-022-00171-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
The misuse of antibiotics in animal protein production has driven the emergence of a range of drug-resistant pathogens, which threaten existing public health security. Consequently, there is an urgent need to develop novel antimicrobials and new infection treatment options to address the challenges posed by the dramatic spread of antibiotic resistance. Piscidins, a class of fish-specific antimicrobial peptides (AMPs), are regarded as promising therapies for biomedical applications. Progress towards potential analogs from the piscidin family has been hampered by unenforceable structural optimization strategies. Here, we leverage a strategy of bioinformatics analysis combined with molecular dynamics (MD) simulation to identify specific functional hotspots in piscidins and rationally design related analogues. As expected, this approach yields a potent and non-toxic PIS-A-1 that can be used as an antibiotic adjuvant to reverse methicillin-resistant Staphylococcus aureus (MRSA) pathogens. Remarkably, the structural optimization scheme and application strategy proposed here will contribute richer therapeutic options for the safe production of animal protein.
Collapse
Affiliation(s)
- Xuan Chen
- grid.411604.60000 0001 0130 6528College of Chemical Engineering, Fuzhou University, Fuzhou, Fujian 350108 China ,grid.411604.60000 0001 0130 6528College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108 China
| | - Xiaoping Wu
- grid.411604.60000 0001 0130 6528College of Chemical Engineering, Fuzhou University, Fuzhou, Fujian 350108 China ,grid.411604.60000 0001 0130 6528College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108 China
| | - Shaoyun Wang
- grid.411604.60000 0001 0130 6528College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108 China
| |
Collapse
|
11
|
da Cruz Ramos Pires GH, Freire VT, Pereira RG, Amaral de Siqueira LJ, Umehara E, Lago JHG, Caseli L. Sakuranetin interacting with cell membranes models: Surface chemistry combined with molecular simulation. Colloids Surf B Biointerfaces 2022; 216:112546. [PMID: 35588685 DOI: 10.1016/j.colsurfb.2022.112546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 11/26/2022]
Abstract
Sakuranetin, a natural compound with activity in lipidic biointerfaces, was isolated from Baccharis retusa and studied with two models of lipid membranes: Langmuir monolayers and Molecular Simulation. For that, the mammalian lipid DPPC was chosen. Sakuranetin condensed the monolayers at high surface pressures, decreased the surface compressional modulus, reduced the molecular order of the acyl chains (diminution of all-trans/gauche conformers ratio), and increased the heterogeneity of the interface, forming aggregates. Molecular simulation data gave information on the bioactive compound's most favorable thermodynamic positions along the lipid monolayer, which was the lipid-air interface. These combined results lead to the conclusion that this lipophilic compound may interact with the lipidic layers, preferentially at the lipid-air interface, to minimize the free energy, and reaches this conformation disturbing the thermodynamic, structural, mechanical, rheological, and morphological properties of the well-packed DPPC monolayer.
Collapse
Affiliation(s)
| | - Vitor Torres Freire
- Department of Chemistry, Federal University of São Paulo, Diadema, SP, Brazil
| | | | | | - Eric Umehara
- Federal University of ABC, Santo André, SP, Brazil
| | | | - Luciano Caseli
- Department of Chemistry, Federal University of São Paulo, Diadema, SP, Brazil.
| |
Collapse
|
12
|
Molecular Basis for Luteolin as a Natural TatD DNase Inhibitor in Trueperella pyogenes. Int J Mol Sci 2022; 23:ijms23158374. [PMID: 35955509 PMCID: PMC9369154 DOI: 10.3390/ijms23158374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 02/07/2023] Open
Abstract
TatD960 and TatD825 are DNases that contribute to biofilm formation and virulence in Trueperella pyogenes (T. pyogenes). Luteolin is a natural flavonoid commonly found in plants that exhibits antimicrobial capacity. Our study aims to investigate the effects of luteolin on TatD DNases as a natural inhibitor. In this research, the expression of tatD genes and TatD proteins in T. pyogenes treated with luteolin was detected, and then the effect of luteolin on the hydrolysis of DNA by TatD DNases was analyzed using agarose gel electrophoresis. Moreover, the interactions between luteolin and TatD DNases were tested using surface plasmon resonance (SPR) assays and molecular docking analysis. After 1/2 MIC luteolin treatment, the transcription of tatD genes and expression of TatD proteins appeared to be reduced in 80–90% of T. pyogenes (n = 20). The gel assay revealed that luteolin can inhibit the activity of TatD DNases. The SPR assay showed that the KD values of luteolin to TatD960 and TatD825 were 6.268 × 10−6 M and 5.654 × 10−6 M, respectively. We found through molecular docking that hydrogen bonding is predominant in the interaction of luteolin and TatD DNases. Our data indicate that luteolin inhibited the ability of TatD DNases by decreasing their binding to DNA. The current study provides an insight into the development of luteolin as a DNase inhibitor in preventing biofilm formation and virulence in T. pyogenes.
Collapse
|
13
|
Tsai HY, Chen MY, Hsu C, Kuan KY, Chang CF, Wang CW, Hsu CP, Su NW. Luteolin Phosphate Derivatives Generated by Cultivating Bacillus subtilis var. Natto BCRC 80517 with Luteolin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:8738-8745. [PMID: 35795971 DOI: 10.1021/acs.jafc.2c03524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Luteolin (LUT), a plant-derived flavone, exhibits various bioactivities; however, the poor aqueous solubility hampers its applications. Here, we revealed bioconversion of LUT by Bacillus subtilis BCRC 80517, yielding three water-soluble phosphate conjugates. These derivatives were identified as luteolin 4'-O-phosphate (L4'P), luteolin 3'-O-phosphate (L3'P), and luteolin 7-O-phosphate (L7P) by LC-ESI-MS/MS and NMR. Besides, we found that Bacillus subtilis BCRC 80517 was able to convert different levels of LUT but showed a limited conversion rate. By observing bacterial morphology with transmission electron microscopy and confocal fluorescence microscopy, we found that LUT disrupted the bacterial membrane integrity, which explained the incomplete conversion. Additionally, we revealed a spontaneous intramolecular transesterification of L4'P to L3'P, the thermodynamically more stable form, under acidic conditions and proposed the possible mechanism involving a cyclic phosphate as the intermediate. This study provides insight into development of a potent structural modification strategy to enhance the solubility of LUT through biophosphorylation.
Collapse
Affiliation(s)
- Hsin-Ya Tsai
- Department of Agricultural Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Ming-Yu Chen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan
| | - Chen Hsu
- Department of Agricultural Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Kai-Yuan Kuan
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Chi-Fon Chang
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Che-Wei Wang
- Department of Agricultural Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Chao-Ping Hsu
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
- Physics Division, National Center for Theoretical Sciences, Taipei 106, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei 106, Taiwan
| | - Nan-Wei Su
- Department of Agricultural Chemistry, National Taiwan University, Taipei 106, Taiwan
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
14
|
Song L, Hu X, Ren X, Liu J, Liu X. Antibacterial Modes of Herbal Flavonoids Combat Resistant Bacteria. Front Pharmacol 2022; 13:873374. [PMID: 35847042 PMCID: PMC9278433 DOI: 10.3389/fphar.2022.873374] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/17/2022] [Indexed: 12/21/2022] Open
Abstract
The increasing dissemination of multidrug resistant (MDR) bacterial infections endangers global public health. How to develop effective antibacterial agents against resistant bacteria is becoming one of the most urgent demands to solve the drug resistance crisis. Traditional Chinese medicine (TCM) with multi-target antibacterial actions are emerging as an effective way to combat the antibacterial resistance. Based on the innovative concept of organic wholeness and syndrome differentiation, TCM use in antibacterial therapies is encouraging. Herein, advances on flavonoid compounds of heat-clearing Chinese medicine exhibit their potential for the therapy of resistant bacteria. In this review, we focus on the antibacterial modes of herbal flavonoids. Additionally, we overview the targets of flavonoid compounds and divide them into direct-acting antibacterial compounds (DACs) and host-acting antibacterial compounds (HACs) based on their modes of action. We also discuss the associated functional groups of flavonoid compounds and highlight recent pharmacological activities against diverse resistant bacteria to provide the candidate drugs for the clinical infection.
Collapse
Affiliation(s)
- Lianyu Song
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
| | - Xin Hu
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
| | - Xiaomin Ren
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
| | - Jing Liu
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
| | - Xiaoye Liu
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
- *Correspondence: Xiaoye Liu,
| |
Collapse
|
15
|
Luteolin Inhibits the Biofilm Formation and Cytotoxicity of Methicillin-Resistant Staphylococcus aureus via Decreasing Bacterial Toxin Synthesis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4476339. [PMID: 35586693 PMCID: PMC9110164 DOI: 10.1155/2022/4476339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/31/2022] [Accepted: 04/19/2022] [Indexed: 12/02/2022]
Abstract
Owing to the fact that luteolin has antibacterial activity against Staphylococcus aureus (S. aureus) and methicillin-resistant S. aureus (MRSA), its specific mechanism in MRSA is worthy of investigation, which is the focus of this study. Initially, the collected S. aureus strains were treated with luteolin. Then, the minimum inhibitory concentration (MIC) of luteolin against the S. aureus strains was measured by the broth microdilution. The growth curves, biofilm formation, and cytotoxicity of treated S. aureus were detected using a microplate reader. The live and dead bacteria were evaluated using confocal laser scanning microscopy, the bacterial morphology was observed using scanning electron microscopy, and the S. aureus colony-forming unit (CFU) numbers were assessed. The levels of alpha hemolysin (α-hemolysin), delta hemolysin (δ-hemolysin), and hlaA were detected via western blot and RT-PCR. The mortality of mouse model with S. aureus systemic infection was analyzed, and the levels of IL-6, IL-8, IL-10, and TNF-α were quantitated using ELISA. Concretely, the MIC of luteolin against MRSA N315 was 64 μg/mL. Luteolin at 16 μg/mL did not affect the growth of MRSA N315, but inhibited the biofilm formation and CFU, and promoted the morphological changes and death of MRSA N315. Luteolin decreased the cytotoxicity and the levels of α-hemolysin, δ-hemolysin, and hlaA in MRSA N315, elevated MRSA-reduced mice survival rate, and differentially modulated the inflammatory cytokine levels in MRSA-infected mice. Collectively, luteolin inhibits biofilm formation and cytotoxicity of MRSA via blocking the bacterial toxin synthesis.
Collapse
|
16
|
Lohan S, Mandal D, Choi W, Konshina AG, Tiwari RK, Efremov RG, Maslennikov I, Parang K. Small Amphiphilic Peptides: Activity Against a Broad Range of Drug-Resistant Bacteria and Structural Insight into Membranolytic Properties. J Med Chem 2022; 65:665-687. [PMID: 34978443 DOI: 10.1021/acs.jmedchem.1c01782] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report the synthesis and antibacterial activities of a series of amphiphilic membrane-active peptides composed, in part, of various nongenetically coded hydrophobic amino acids. The lead cyclic peptides, 8C and 9C, showed broad-spectrum activity against drug-resistant Gram-positive (minimum inhibitory concentration (MIC) = 1.5-6.2 μg/mL) and Gram-negative (MIC = 12.5-25 μg/mL) bacteria. The cytotoxicity study showed the predominant lethal action of the peptides against bacteria as compared with mammalian cells. A plasma stability study revealed approximately 2-fold higher stability of lead cyclic peptides as compared to their linear counterparts after 24 h of incubation. A calcein dye leakage experiment revealed the membranolytic effect of the cyclic peptides. Nuclear magnetic resonance spectroscopy and molecular dynamics simulation studies of the interaction of the peptides with the phospholipid bilayer provided a solid structural basis to explain the membranolytic action of the peptides with atomistic details. These results highlight the potential of newly designed amphiphilic peptides as the next generation of peptide-based antibiotics.
Collapse
Affiliation(s)
- Sandeep Lohan
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
- AJK Biopharmaceutical, 5270 California Avenue, Irvine, California 92617, United States
| | - Dindyal Mandal
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
- AJK Biopharmaceutical, 5270 California Avenue, Irvine, California 92617, United States
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar 751024, India
| | - Wonsuk Choi
- Structural Biology Research Center, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
| | - Anastasia G Konshina
- M.M. Shemyakin & Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street, 16/10, Moscow 117997, Russia
| | - Rakesh K Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
| | - Roman G Efremov
- M.M. Shemyakin & Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street, 16/10, Moscow 117997, Russia
- National Research University Higher School of Economics, Myasnitskaya ul. 20, Moscow 101000, Russia
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, 141701 Moscow, Oblast, Russia
| | - Innokentiy Maslennikov
- Structural Biology Research Center, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, 9401 Jeronimo Road, Irvine, California 92618, United States
| |
Collapse
|
17
|
Characterization and Quantification of Luteolin-Metal Complexes in Aqueous Extract of Lonicerae Japonicae Flos and Huangshan Wild Chrysanthemum. Int J Anal Chem 2021; 2021:6677437. [PMID: 33777144 PMCID: PMC7979300 DOI: 10.1155/2021/6677437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/08/2021] [Accepted: 03/01/2021] [Indexed: 11/30/2022] Open
Abstract
Luteolin is a flavonoid compound widely found in vegetables, fruits, and medicinal plants. In this study, the reaction conditions for luteolin and five metal ions (Ca2+, Mg2+, Zn2+, Fe3+, and Cu2+) to form complexes in hot water were optimized, which was at a molar ratio of 1 : 1 for luteolin and metal ions at 90°C in a volume of 20 mL for 2 h, and the ability of luteolin to form complexes with Cu2+ was the strongest. The DPPH scavenging test showed that luteolin exerted a dose-dependent effect on the clearance of free radicals; luteolin-Cu2+ complexes and luteolin-Fe3+ complexes accentuated the clearance of free radicals. Furthermore, we used high performance liquid chromatography (HPLC) to analyze luteolin in samples from two medicinal plants, obtained from the dissolution of aqueous extracts in two different solvents. The results showed that the peak areas for luteolin in the samples dissolved in 20% formic acid-methanol were significantly larger than those from the samples dissolved in methanol alone, with increases in the peak area being 135.6% (Lonicerae Japonicae Flos), and 161.16% (Huangshan wild chrysanthemum). The aforementioned results indicate that complexes formed from organic compounds and metal ions are present in the decoction of a plant.
Collapse
|
18
|
Hu J, Liu H, Xu P, Shang Y, Liu H. Investigation of Drug for Pulmonary Administration-Model Pulmonary Surfactant Monolayer Interactions Using Langmuir-Blodgett Monolayer and Molecular Dynamics Simulation: A Case Study of Ketoprofen. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:13452-13460. [PMID: 31524404 DOI: 10.1021/acs.langmuir.9b02412] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pulmonary administration is widely used for the treatment of lung diseases. The interaction between drug molecules and pulmonary surfactants affects the efficacy of the drug directly. The location and distribution of drug molecules in a model pulmonary surfactant monolayer under different surface pressures can provide vivid information on the interaction between drug molecules and pulmonary surfactants during the pulmonary administration. Ketoprofen is a nonsteroidal anti-inflammatory drug for pulmonary administration. The effect of ketoprofen molecules on the lipid monolayer containing 1,2-dipalmitoyl-sn-glycero-3-phosphatidylcholine (DPPC) and 1,2-dipalmitoyl-sn-glycero-3-phospho-rac-glycerol (DPPG) is studied by surface pressure (π)-area (A) isotherms and compressibility modulus (Cs-1)-surface pressure (π) isotherms. The location and distribution of ketoprofen molecules in a lipid monolayer under different surface pressures are explored by surface tension, density profile, radial distribution function (RDF), and the potential of mean force (PMF) simulated by molecular dynamics (MD) simulation. The introduction of ketoprofen molecules affects the properties of DPPC/DPPG monolayers and the location and distribution of ketoprofen molecules in monolayers with various surface pressures. The existence of ketoprofen molecules hinders the formation of liquid-condensed (LC) films and decreases the compressibility of DPPC/DPPG monolayers. The location and distribution of ketoprofen molecules in the lipid monolayer are affected by cation-π interaction between the choline group of lipids and the benzene ring of ketoprofen, the steric hindrance of the lipid head groups, and the hydrophobicity of ketoprofen molecule itself, comprehensively. The contact state of lipid head group with water is determined by surface pressure, which affects the interaction between drug molecules and lipids and further dominates the location and distribution of ketoprofen in the lipid monolayer. This work confirms that ketoprofen molecules can affect the property and the inner structure of DPPC/DPPG monolayers during breathing. Furthermore, the results obtained using a mixed monolayer containing two major pulmonary surfactants DPPC/DPPG and ketoprofen molecules will be helpful for the in-depth understanding of the mechanism of inhaled administration therapy.
Collapse
Affiliation(s)
- Jiajie Hu
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering , East China University of Science and Technology , Shanghai 200237 , China
| | - Hengjiang Liu
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering , East China University of Science and Technology , Shanghai 200237 , China
| | - Pu Xu
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering , East China University of Science and Technology , Shanghai 200237 , China
| | - Yazhuo Shang
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering , East China University of Science and Technology , Shanghai 200237 , China
| | - Honglai Liu
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering , East China University of Science and Technology , Shanghai 200237 , China
| |
Collapse
|
19
|
Penetration enhancement of menthol on quercetin through skin: insights from atomistic simulation. J Mol Model 2019; 25:235. [DOI: 10.1007/s00894-019-4135-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
|
20
|
Zhou Y, Huang L, Ji S, Hou S, Luo L, Li C, Liu M, Liu Y, Jiang L. Structural Basis for the Inhibition of the Autophosphorylation Activity of HK853 by Luteolin. Molecules 2019; 24:molecules24050933. [PMID: 30866470 PMCID: PMC6429454 DOI: 10.3390/molecules24050933] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 02/21/2019] [Accepted: 03/04/2019] [Indexed: 11/16/2022] Open
Abstract
The two-component system (TCS) is a significant signal transduction system for bacteria to adapt to complicated and variable environments, and thus has recently been regarded as a novel target for developing antibacterial agents. The natural product luteolin (Lut) can inhibit the autophosphorylation activity of the typical histidine kinase (HK) HK853 from Thermotoga maritime, but the inhibition mechanism is not known. Herein, we report on the binding mechanism of a typical flavone with HK853 by using solution NMR spectroscopy, isothermal titration calorimetry (ITC), and molecular docking. We show that luteolin inhibits the activity of HK853 by occupying the binding pocket of adenosine diphosphate (ADP) through hydrogen bonds and π-π stacking interaction structurally. Our results reveal a detailed mechanism for the inhibition of flavones and observe the conformational and dynamics changes of HK. These results should provide a feasible approach for antibacterial agent design from the view of the histidine kinases.
Collapse
Affiliation(s)
- Yuan Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
- Graduate University of Chinese Academy of Science, Beijing 100049, China.
| | - Liqun Huang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
- Graduate University of Chinese Academy of Science, Beijing 100049, China.
| | - Shixia Ji
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
- Graduate University of Chinese Academy of Science, Beijing 100049, China.
| | - Shi Hou
- Laboratory of Computer-Aided Drug Design and Discovery, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Liang Luo
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
- Graduate University of Chinese Academy of Science, Beijing 100049, China.
| | - Conggang Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Maili Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Yixiang Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Ling Jiang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center of Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
21
|
Microbial Biosynthesis of Antibacterial Chrysoeriol in Recombinant Escherichia coli and Bioactivity Assessment. Catalysts 2019. [DOI: 10.3390/catal9020112] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Various flavonoid derivatives including methoxylated flavones display remarkable biological activities. Chrysoeriol is a methoxylated flavone of great scientific interest because of its promising anti-microbial activities against various Gram-negative and Gram-positive bacteria. Sustainable production of such compounds is therefore of pronounced interest to biotechnologists in the pharmaceutical and nutraceutical industries. Here, we used a sugar O-methyltransferase enzyme from a spinosyn biosynthesis gene cluster of Saccharopolyspora spinosa to regioselectively produce chrysoeriol (15% conversion of luteolin; 30 µM) in a microbial host. The biosynthesized chrysoeriol was structurally characterized using high-resolution mass spectrometry and various nuclear magnetic resonance analyses. Moreover, the molecule was investigated against 17 superbugs, including thirteen Gram-positive and four Gram-negative pathogens, for anti-microbial effects. Chrysoeriol exhibited antimicrobial activity against nine pathogens in a disc diffusion assay at the concentration of 40 µg per disc. It has minimum inhibitory concentration (MIC) values of 1.25 µg/mL against a methicillin-resistant Staphylococcus aureus 3640 (MRSA) for which the parent luteolin has an MIC value of sixteen-fold higher concentration (i.e., 20 µg/mL). Similarly, chrysoeriol showed better anti-microbial activity (~1.7-fold lower MIC value) than luteolin against Proteus hauseri, a Gram-negative pathogen. In contrast, a luteolin 4′-O-methylated derivative, diosmetin, did not exhibit any anti-microbial activities against any tested pathogen.
Collapse
|