1
|
Zhang J, Luan L, Xu Y, Jiang S, Zhang W, Tian L, Ye W, Han J, Zhang C, Wang T, Meng Q. Development of novel broad-spectrum amphipathic antimicrobial peptides against multidrug-resistant bacteria through a rational combination strategy. J Adv Res 2025:S2090-1232(25)00048-7. [PMID: 39832719 DOI: 10.1016/j.jare.2025.01.029] [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: 11/06/2024] [Revised: 12/27/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION In recent years, cationic amphipathic antimicrobial peptides (AMPs) have shown great promise in combating antibiotic resistance on account of their distinctive membrane-disruptive mechanism. However, the clinical application of AMPs is restricted by their unsatisfactory stability and safety. Although attempts have been made to improve the stability and safety of AMPs, many of them are accompanied by a decline in their antimicrobial activity and bacterial selectivity. OBJECTIVES To develop AMPs with excellent and balanced antimicrobial activity, stability, and safety using a combination strategy. METHODS A series of sC184b-derived peptide analogues were designed by a combination strategy of subtly adjusting the charges, hydrophobic properties, and introducing specific unnatural amino acids in a well-balanced manner. The antimicrobial activity, cytotoxicity, hemolytic activity, stability, anti-biofilm activity, mechanism of action, synergistic effects, in vivo efficacy, and pharmacokinetics of the analogues were evaluated. RESULTS Among these analogues, P-α-02-B stood out for its broad-spectrum and potent antimicrobial activity, anti-biofilm activity, desirable bacterial selectivity, high plasma stability, and synergistic effect with antibiotic levofloxacin. P-α-02-B exhibited strong membrane disturbance effect, which could be explained by its rigid α-helical structure revealed by molecular dynamics simulations. More importantly, P-α-02-B showed favorable therapeutic efficacy in vivo, whether used alone or in combination with levofloxacin. CONCLUSION P-α-02-B is a promising antimicrobial agent for MDR bacterial infections, demonstrating the effectiveness of the combination strategy for AMP development.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; Key laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Liang Luan
- Department of Laboratory Medical Center, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Youdong Xu
- National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing 102206, China
| | - Shuyuan Jiang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; Key laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Wenpeng Zhang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Long Tian
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Weifeng Ye
- Center of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jiaqi Han
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Changhao Zhang
- Key laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China.
| | - Taoran Wang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Qingbing Meng
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
2
|
Song Y, Lv H, Xu L, Liu Z, Wang J, Fang T, Deng X, Zhou Y, Li D. In vitro and in vivo activities of scutellarein, a novel polyphosphate kinase 1 inhibitor against Acinetobacter baumannii infection. Microb Cell Fact 2024; 23:269. [PMID: 39379932 PMCID: PMC11462863 DOI: 10.1186/s12934-024-02540-9] [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: 06/22/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Inorganic polyphosphate (polyP)-targeted polyphosphate kinase 1 (PPK1) has attracted much attention by virtue of its importance in bacterial pathogenicity and persistence, as well as its exclusive presence in microorganisms. However, only very few drugs have been found to be efficacious in inhibiting the Acinetobacter baumannii (A. baumannii) PPK1 protein. RESULTS In this study, we identified Scutellarein (Scu), a potent PPK1 inhibitor that could significantly influence PPK1-regulated motility, biofilm formation, and bacterial persistence, which was further validated by the results of transcriptome analysis. Mechanistic explorations revealed that Scu achieved its enzyme inhibitory activity predominantly through direct engagement with the active center of PPK1. Moreover, the survival rate of Galleria mellonella larvae was increased by about 35% with 20 mg/kg of Scu treatment. The remarkable therapeutic benefits of Scu were also observed in the mouse pneumonia model, shown mainly by reduced bacterial colonization, pathological lesions, and inflammatory factors. CONCLUSION Our results revealed that Scu could attenuate the pathogenicity and persistence of A. baumannii by interfering with its important kinase PPK1.
Collapse
Affiliation(s)
- Yuping Song
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Hongfa Lv
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lei Xu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhiying Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfeng Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tianqi Fang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
- Jilin Mushuo Breeding Co., Ltd, Changchun, Jilin, 130052, China
| | - Xuming Deng
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yonglin Zhou
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, School of Life Sciences, Ningxia University, Yinchuan, China
| | - Dan Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
3
|
Hernández-Ortiz N, Sánchez-Murcia PA, Gil-Campillo C, Domenech M, Lucena-Agell D, Hortigüela R, Velázquez S, Camarasa MJ, Bustamante N, de Castro S, Menéndez M. Design, synthesis and structure-activity relationship (SAR) studies of an unusual class of non-cationic fatty amine-tripeptide conjugates as novel synthetic antimicrobial agents. Front Pharmacol 2024; 15:1428409. [PMID: 39156106 PMCID: PMC11329928 DOI: 10.3389/fphar.2024.1428409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/01/2024] [Indexed: 08/20/2024] Open
Abstract
Cationic ultrashort lipopeptides (USLPs) are promising antimicrobial candidates to combat multidrug-resistant bacteria. Using DICAMs, a newly synthesized family of tripeptides with net charges from -2 to +1 and a fatty amine conjugated to the C-terminus, we demonstrate that anionic and neutral zwitterionic USLPs can possess potent antimicrobial and membrane-disrupting activities against prevalent human pathogens such as Streptococcus pneumoniae and Streptococcus pyogenes. The strongest antimicrobials completely halt bacterial growth at low micromolar concentrations, reduce bacterial survival by several orders of magnitude, and may kill planktonic cells and biofilms. All of them comprise either an anionic or neutral zwitterionic peptide attached to a long fatty amine (16-18 carbon atoms) and show a preference for anionic lipid membranes enriched in phosphatidylglycerol (PG), which excludes electrostatic interactions as the main driving force for DICAM action. Hence, the hydrophobic contacts provided by the long aliphatic chains of their fatty amines are needed for DICAM's membrane insertion, while negative-charge shielding by salt counterions would reduce electrostatic repulsions. Additionally, we show that other components of the bacterial envelope, including the capsular polysaccharide, can influence the microbicidal activity of DICAMs. Several promising candidates with good-to-tolerable therapeutic ratios are identified as potential agents against S. pneumoniae and S. pyogenes. Structural characteristics that determine the preference for a specific pathogen or decrease DICAM toxicity have also been investigated.
Collapse
Affiliation(s)
- Noelia Hernández-Ortiz
- Instituto de Química-Física “Blas Cabrera” (IQF), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Pedro A. Sánchez-Murcia
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Laboratory of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Celia Gil-Campillo
- Instituto de Química-Física “Blas Cabrera” (IQF), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mirian Domenech
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Departamento Genética, Fisiología y Microbiología, Facultad Ciencias Biológicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Daniel Lucena-Agell
- Centro de Investigaciones Biológicas Margarita Salas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Rafael Hortigüela
- Centro de Investigaciones Biológicas Margarita Salas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Sonsoles Velázquez
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - María José Camarasa
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Noemí Bustamante
- Instituto de Química-Física “Blas Cabrera” (IQF), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Sonia de Castro
- Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Margarita Menéndez
- Instituto de Química-Física “Blas Cabrera” (IQF), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Wu X, Tian Y, Ran K, Yao J, Wang Y, Ouyang X, Mao W, Zhang J, Li B, Yang P, Ba Z, Liu H, Gou S, Zhong C, Zhang Y, Ni J. Rational design of a new short anticancer peptide with good potential for cancer treatment. Eur J Med Chem 2024; 273:116519. [PMID: 38795519 DOI: 10.1016/j.ejmech.2024.116519] [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: 12/29/2023] [Revised: 05/19/2024] [Accepted: 05/19/2024] [Indexed: 05/28/2024]
Abstract
Anticancer peptides (ACPs) have regarded as a new generation of promising antitumor drugs due to the unique mode of action. The main challenge is to develop potential anticancer peptides with satisfied antitumor activity and low toxicity. Here, a series of new α-helical anticancer peptides were designed and synthesized based on the regular repeat motif KLLK. The optimal peptides 14E and 14Aad were successfully derived from the new short α-helical peptide KL-8. Our results demonstrated that 14E and 14Aad had good antitumor activity and low toxicity, exhibiting excellent selectivity index. This result highlighted that the desirable modification position and appropriate hydrophobic side-chain structure of acidic amino acids played critical roles in regulating the antitumor activity/toxicity of new peptides. Further studies indicated that they could induce tumor cell death via the multiple actions of efficient membrane disruption and intracellular mechanisms, displaying apparent superiority in combination with PTX. In addition, the new peptides 14E and 14Aad showed excellent antitumor efficacy in vivo and low toxicity in mice compared to KL-8 and PTX. Particularly, 14Aad with the longer side chain at the 14th site exhibited the best therapeutic performance. In conclusion, our work provided a new avenue to develop promising anticancer peptides with good selectivity for tumor therapy.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Yali Tian
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Kaixin Ran
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Jia Yao
- The First Hospital, Lanzhou University, Lanzhou, 730000, PR China
| | - Yuxia Wang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Xu Ouyang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Wenbo Mao
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Jingying Zhang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Beibei Li
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Ping Yang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Zufang Ba
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Hui Liu
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Sanhu Gou
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Chao Zhong
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Yun Zhang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China.
| | - Jingman Ni
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou, 730000, PR China; Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
5
|
Decker T, Rautenbach M, Khan S, Khan W. Antibacterial efficacy and membrane mechanism of action of the Serratia-derived non-ionic lipopeptide, serrawettin W2-FL10. Microbiol Spectr 2024; 12:e0295223. [PMID: 38842361 PMCID: PMC11218446 DOI: 10.1128/spectrum.02952-23] [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: 07/26/2023] [Accepted: 05/14/2024] [Indexed: 06/07/2024] Open
Abstract
The study aimed to investigate the antibacterial activity, cytotoxicity, and mechanism of action of the non-ionic, cyclic lipopeptide, serrawettin W2-FL10 against Staphylococcus aureus. W2-FL10 exhibited potent activity against the Gram-positive bacteria S. aureus, Enterococcus faecalis, Enterococcus faecium, Listeria monocytogenes, and Bacillus subtilis, with minimum inhibitory concentration (MIC) values ranging from 6.3 to 31.3 μg/mL, while no activity was observed against Gram-negative bacteria. Broth microdilution assays showed that W2-FL10 interacted with key cell membrane components, such as lipid phosphatidyl glycerol and lipoteichoic acid of S. aureus. Upon membrane interaction, W2-FL10 dissipated membrane potential within 12 min and increased S. aureus membrane permeability within 28-40 min, albeit at slower rates and higher concentrations than the lytic peptide melittin. The observed membrane permeability, as detected with propidium iodide (PI), may be attributed to transmembrane pores/lesions, possibly dependent on dimer-driven lipopeptide oligomerization in the membrane. Scanning electron microscopy (SEM) imaging also visually confirmed the formation of lesions in the cell wall of one of the S. aureus strains, and cell damage within 1 h of exposure to W2-FL10, corroborating the rapid time-kill kinetics of the S. aureus strains. This bactericidal action against the S. aureus strains corresponded to membrane permeabilization by W2-FL10, indicating that self-promoted uptake into the cytosol may be part of the mode of action. Finally, this lipopeptide exhibited low to moderate cytotoxicity to the Chinese hamster ovarian (CHO) cell line in comparison to the control (emetine) with an optimal lipophilicity range (log D value of 2.5), signifying its potential as an antibiotic candidate. IMPORTANCE Antimicrobial resistance is a major public health concern, urgently requiring antibacterial compounds exhibiting low adverse health effects. In this study, a novel antibacterial lipopeptide analog is described, serrawettin W2-FL10 (derived from Serratia marcescens), with potent activity displayed against Staphylococcus aureus. Mechanistic studies revealed that W2-FL10 targets the cell membrane of S. aureus, causing depolarization and permeabilization because of transmembrane lesions/pores, resulting in the leakage of intracellular components, possible cytosolic uptake of W2-FL10, and ultimately cell death. This study provides the first insight into the mode of action of a non-ionic lipopeptide. The low to moderate cytotoxicity of W2-FL10 also highlights its application as a promising therapeutic agent for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Tanya Decker
- Water Health Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, Johannesburg, Gauteng, South Africa
| | - Marina Rautenbach
- BioPep Peptide Group, Department of Biochemistry, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Sehaam Khan
- Water Health Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, Johannesburg, Gauteng, South Africa
| | - Wesaal Khan
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
6
|
Panjla A, Kaul G, Shukla M, Akhir A, Tripathi S, Arora A, Chopra S, Verma S. Membrane-targeting, ultrashort lipopeptide acts as an antibiotic adjuvant and sensitizes MDR gram-negative pathogens toward narrow-spectrum antibiotics. Biomed Pharmacother 2024; 176:116810. [PMID: 38823276 DOI: 10.1016/j.biopha.2024.116810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
Globally, infections due to multi-drug resistant (MDR) Gram-negative bacterial (GNB) pathogens are on the rise, negatively impacting morbidity and mortality, necessitating urgent treatment alternatives. Herein, we report a detailed bio-evaluation of an ultrashort, cationic lipopeptide 'SVAP9I' that demonstrated potent antibiotic activity and acted as an adjuvant to potentiate existing antibiotic classes towards GNBs. Newly synthesized lipopeptides were screened against ESKAPE pathogens and cytotoxicity assays were performed to evaluate the selectivity index (SI). SVAP9I exhibited broad-spectrum antibacterial activity against critical MDR-GNB pathogens including members of Enterobacteriaceae (MIC 4-8 mg/L), with a favorable CC50 value of ≥100 mg/L and no detectable resistance even after 50th serial passage. It demonstrated fast concentration-dependent bactericidal action as determined via time-kill analysis and also retained full potency against polymyxin B-resistant E. coli, indicating distinct mode of action. SVAP9I targeted E. coli's outer and inner membranes by binding to LPS and phospholipids such as cardiolipin and phosphatidylglycerol. Membrane damage resulted in ROS generation, depleted intracellular ATP concentration and a concomitant increase in extracellular ATP. Checkerboard assays showed SVAP9I's synergism with narrow-spectrum antibiotics like vancomycin, fusidic acid and rifampicin, potentiating their efficacy against MDR-GNB pathogens, including carbapenem-resistant Acinetobacter baumannii (CRAB), a WHO critical priority pathogen. In a murine neutropenic thigh infection model, SVAP9I and rifampicin synergized to express excellent antibacterial efficacy against MDR-CRAB outcompeting polymyxin B. Taken together, SVAP9I's distinct membrane-targeting broad-spectrum action, lack of resistance and strong in vitro andin vivopotency in synergism with narrow spectrum antibiotics like rifampicin suggests its potential as a novel antibiotic adjuvant for the treatment of serious MDR-GNB infections.
Collapse
Affiliation(s)
- Apurva Panjla
- Department of Chemistry, IIT Kanpur, Uttar Pradesh 208016, India
| | - Grace Kaul
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manjulika Shukla
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, Uttar Pradesh 226031, India
| | - Abdul Akhir
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, Uttar Pradesh 226031, India
| | - Sarita Tripathi
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, Uttar Pradesh 226031, India
| | - Ashish Arora
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sidharth Chopra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Sandeep Verma
- Department of Chemistry, IIT Kanpur, Uttar Pradesh 208016, India; Mehta Family Center for Engineering in Medicine, Center for Nanoscience Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.
| |
Collapse
|
7
|
Yu C, E R, An Y, Guo X, Bao G, Li Y, Xie J, Sun W. Michael Addition Reaction between Dehydroalanines and Phosphites Enabled the Introduction of Phosphonates into Oligopeptides. Org Lett 2024. [PMID: 38780227 DOI: 10.1021/acs.orglett.4c01567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
A method for introducing a range of phosphonates into oligopeptides through a Michael addition reaction between dehydroalanine and phosphite is presented. The method offers a mild, cheap, and straightforward approach to peptide phosphorylation that has potential applications in chemical biology and medicinal chemistry. Moreover, the introduction of a phosphonate group into short antibacterial peptides is described to demonstrate its utility, leading to the discovery of phosphonated antibacterial peptides with potent broad-spectrum antibacterial activity.
Collapse
Affiliation(s)
- Changjun Yu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, P. R. China
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, P. R. China
| | - Ruiyao E
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yingying An
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, P. R. China
| | - Xiaomin Guo
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, P. R. China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yiping Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, P. R. China
| | - Junqiu Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, P. R. China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, P. R. China
| |
Collapse
|
8
|
Yang P, Mao W, Zhang J, Yang Y, Zhang F, Ouyang X, Li B, Wu X, Ba Z, Ran K, Tian Y, Liu H, Zhang Y, Gou S, Zhong C, Ni J. A novel antimicrobial peptide with broad-spectrum and exceptional stability derived from the natural peptide Brevicidine. Eur J Med Chem 2024; 269:116337. [PMID: 38537511 DOI: 10.1016/j.ejmech.2024.116337] [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: 12/13/2023] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 04/07/2024]
Abstract
The global issue of antibiotic resistance is increasingly severe, highlighting the urgent necessity for the development of new antibiotics. Brevicidine, a natural cyclic lipopeptide, exhibits remarkable antimicrobial activity against Gram-negative bacteria. In this study, a comprehensive structure-activity relationship of Brevicidine was investigated through 20 newly synthesized cyclic lipopeptide analogs, resulting in the identification of an optimal linear analog 22. The sequence of analog 22 consisted of five d-amino acids and four non-natural amino acid 2,5-diaminovaleric acid (Orn) and conjugated with decanoic acid at N-terminal. Compared to Brevicidine, analog 22 was easier to synthesize, and exerted broad spectrum antimicrobial activity and excellent stability (t1/2 = 40.98 h). Additionally, analog 22 demonstrated a rapid bactericidal effect by permeating non-specifically through the bacterial membranes, thereby minimizing the likelihood of inducing resistance. Moreover, it exhibited remarkable efficacy in combating bacterial biofilms and reversing bacterial resistance to conventional antibiotics. Furthermore, it effectively suppressed the growth of bacteria in vital organs of mice infected with S. aureus ATCC 25923. In conclusion, analog 22 may represent a potential antimicrobial peptide for further optimization.
Collapse
Affiliation(s)
- Ping Yang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Wenbo Mao
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Jingying Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Yinyin Yang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Fangyan Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Xu Ouyang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Beibei Li
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Xiaoyan Wu
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Zufang Ba
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Kaixin Ran
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Yali Tian
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Hui Liu
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Yun Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Sanhu Gou
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Chao Zhong
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Jingman Ni
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
9
|
Lu H, Wang C, Lu W, Li X, Wang G, Dong W, Wang X, Chen H, Tan C. Antibacterial efficacy and mechanism of Cyprinus carpio chemokine-derived L-10 against multidrug-resistant Escherichia coli infections. Int J Antimicrob Agents 2024; 63:107104. [PMID: 38325720 DOI: 10.1016/j.ijantimicag.2024.107104] [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: 11/13/2023] [Revised: 01/02/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVES Antimicrobial resistance has raised concerns regarding untreatable infections and poses a growing threat to public health. Rational design of new AMPs is an ideal solution to this threat. METHODS In this study, we designed, modified, and synthesised an excellent AMP, L-10, based on the original sequence of the Cyprinus carpio chemokine. All experimental data were presented as the mean ± standard deviation (SD), and the two-tailed unpaired T-test method was used to analyze all data. RESULTS L-10 exhibited excellent antibacterial activity with negligible toxicity and improved the efficacy of a broad class of antibiotics against MDR Gram-negative pathogens, including tetracycline, meropenem, levofloxacin, and rifampin. Mechanistic studies have suggested that L-10 targets the bacterial membrane components, LPS and PG, to disrupt bacterial membrane integrity, thereby exerting antibacterial effects and enhancing the efficacy of antibiotics. Moreover, in animal infection models, L-10 significantly increased the survival rate of infected animals and effectively reduced the tissue bacterial load and inflammatory factor levels. In addition to its direct antibacterial activity, L-10 dramatically reduced pulmonary pathological alterations in a mouse model of endotoxemia and suppressed LPS-induced proinflammatory cytokines in vitro and in vivo. Lastly, L-10 was successfully expressed in Pichia pastoris and maintained antimicrobial activity against MDR Gram-negative pathogens in vivo and in vitro. CONCLUSION Collectively, these results reveal the potential of L-10 as an ideal candidate against MDR bacterial infections and provide new insights into the design, development, and clinical application of AMPs.
Collapse
Affiliation(s)
- Hao Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Chenchen Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Wenjia Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Xiaodan Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Gaoyan Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Wenqi Dong
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China; Hubei Hongshan Laboratory, Wuhan, Hubei, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China.
| |
Collapse
|
10
|
Yang B, Li W, Mao Y, Zhao Y, Xue Y, Xu X, Zhao Y, Liu K. Study on antimicrobial activity of sturgeon skin mucus polypeptides (Rational Design, Self-Assembly and Application). Food Chem X 2024; 21:101236. [PMID: 38406763 PMCID: PMC10884804 DOI: 10.1016/j.fochx.2024.101236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024] Open
Abstract
Despite the favorable biocompatibility of natural antimicrobial peptides (AMPs), their scarcity limits their practical application. Through rational design, the activity of AMPs can be enhanced to expand their application. In this study, we selected a natural sturgeon epidermal mucus peptide, AP-16 (APATPAAPALLPLWLL), as the model molecule and studied its conformational regulation and antimicrobial activity through amino acid substitutions and N-terminal lipidation. The structural and morphological transitions of the peptide self-assemblies were investigated using circular dichroism and transmission electron microscopy. Following amino acid substitution, the conformation of AL-16 (AKATKAAKALLKLWLL) did not change. Following N-terminal alkylation, the C8-AL-16 and C12-AL-16 conformations changed from random coil to β-sheet or α-helix, and the self-assembly changed from nanofibers to nanospheres. AL-16, C8-AL-16, and C8-AL-16 presented significant antimicrobial activity against Pseudomonas and Shewanella at low concentrations. N-terminal alkylation effectively extended the shelf life of Litopenaeus vannamei. These results support the application of natural AMPs.
Collapse
Affiliation(s)
- Beining Yang
- Sanya Oceanographic Institution /College of Food Science and Engineering, Ocean University of China, Sanya/Qingdao, China
- State Key Laboratory of Marine Food Processing & Safety Control, Ocean University of China, Qingdao, Shandong, China
| | - Wei Li
- China Department of General Surgery, The District Hospital of Qingdao West Coast New Area, Qngdao, Shandong, China
| | - Yuxuan Mao
- National Engineering Research Center for Wheat and Corn Deep Processing, College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin, China
| | - Yuanhui Zhao
- Sanya Oceanographic Institution /College of Food Science and Engineering, Ocean University of China, Sanya/Qingdao, China
- State Key Laboratory of Marine Food Processing & Safety Control, Ocean University of China, Qingdao, Shandong, China
| | - Yong Xue
- Sanya Oceanographic Institution /College of Food Science and Engineering, Ocean University of China, Sanya/Qingdao, China
- State Key Laboratory of Marine Food Processing & Safety Control, Ocean University of China, Qingdao, Shandong, China
| | - Xinxing Xu
- State Key Laboratory of Marine Food Processing & Safety Control, Ocean University of China, Qingdao, Shandong, China
| | - Yilin Zhao
- Sanya Oceanographic Institution /College of Food Science and Engineering, Ocean University of China, Sanya/Qingdao, China
- State Key Laboratory of Marine Food Processing & Safety Control, Ocean University of China, Qingdao, Shandong, China
| | - Kang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, China
- National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Xiamen, China
| |
Collapse
|
11
|
Wang HG, Zhang HZ. Emodin Alcohols: Design, Synthesis, Biological Evaluation and Multitargeting Studies with DNA, RNA, and HSA. Curr Med Chem 2024; 31:2788-2808. [PMID: 37183459 DOI: 10.2174/0929867330666230512161856] [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: 09/30/2022] [Revised: 02/04/2023] [Accepted: 03/05/2023] [Indexed: 05/16/2023]
Abstract
OBJECTIVE A series of novel emodin alcohols were designed and prepared in an effort to overcome the increasing microorganism resistance. METHODS Novel emodin alcohols were prepared from commercial emodin and different nitrogen-containing heterocycles via different synthetic strategies, such as O-alkylation and N-alkylation. The antimicrobial activity of synthesized emodin compounds was evaluated in vitro by a two-fold serial dilution technique. The interaction of emodin compound 3d with biomolecule was researched using UV-vis spectroscopic method and fluorescence spectroscopy. RESULTS Emodin compound 3d containing 2-methyl-5-nitro imidazole ring showed relatively good antimicrobial activity. Notably, it exhibited equivalent activity against S. aureus in comparison to the reference drug norfloxacin (MIC = 4 μg/mL). The combination of strong active compound 3d with reference drugs showed better antimicrobial activity with less dosage and a broader antimicrobial spectrum than their separate use. Further research displayed that emodin compound 3d could intercalate into S. aureus DNA to form the 3d-DNA complex, which might correlate with the inhibitory activity. The hydrogen bonds were found between S. aureus DNA gyrase and strong active compound 3d during the docking research, which were in accordance with the spectral experiment results. The interaction with yeast RNA of compound 3d could also form a complex via hydrogen bonds. The hydrogen bonds were found to play a major role in the transportation of emodin compound 3d by human serum albumin (HSA), as confirmed by molecular simulation. CONCLUSION This work provides a promising starting point to optimize the structures of emodin derivatives as potent antimicrobial agents.
Collapse
Affiliation(s)
- Hai-Guang Wang
- School of Pharmacy, Linyi University, Linyi 276000, China
| | - Hui-Zhen Zhang
- School of Pharmacy, Linyi University, Linyi 276000, China
| |
Collapse
|
12
|
Selvaraj SP, Chen JY. Conjugation of antimicrobial peptides to enhance therapeutic efficacy. Eur J Med Chem 2023; 259:115680. [PMID: 37515922 DOI: 10.1016/j.ejmech.2023.115680] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/05/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
The growing prevalence of antimicrobial resistance (AMR) has brought with it a continual increase in the numbers of deaths from multidrug-resistant (MDR) infections. Since the current arsenal of antibiotics has become increasingly ineffective, there exists an urgent need for discovery and development of novel antimicrobials. Antimicrobial peptides (AMPs) are considered to be a promising class of molecules due to their broad-spectrum activities and low resistance rates compared with other types of antibiotics. Since AMPs also often play major roles in elevating the host immune response, the molecules may also be called "host defense peptides." Despite the great promise of AMPs, the majority remain unsuitable for clinical use due to issues of structural instability, degradation by proteases, and/or toxicity to host cells. Moreover, AMP activities in vivo can be influenced by many factors, such as interaction with blood and serum biomolecules, physiological salt concentrations or different pH values. To overcome these limitations, structural modifications can be made to the AMP. Among several modifications, physical and chemical conjugation of AMP to other biomolecules is widely considered an effective strategy. In this review, we discuss structural modification strategies related to conjugation of AMPs and their possible effects on mode of action. The conjugation of fatty acids, glycans, antibiotics, photosensitizers, polymers, nucleic acids, nanoparticles, and immobilization to biomaterials are highlighted.
Collapse
Affiliation(s)
- Sanjay Prasad Selvaraj
- Molecular and Biological Agricultural Science Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 11529, Taiwan; Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, 402, Taiwan
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Rd, Jiaushi, Ilan, 262, Taiwan; The iEGG and Animal Biotechnology Center and the Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
| |
Collapse
|
13
|
Pilz M, Cavelius P, Qoura F, Awad D, Brück T. Lipopeptides development in cosmetics and pharmaceutical applications: A comprehensive review. Biotechnol Adv 2023; 67:108210. [PMID: 37460047 DOI: 10.1016/j.biotechadv.2023.108210] [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: 01/26/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/25/2023]
Abstract
Lipopeptides are surface active, natural products of bacteria, fungi and green-blue algae origin, having diverse structures and functionalities. In analogy, a number of chemical synthesis techniques generated new designer lipopeptides with desirable features and functions. Lipopetides are self-assembly guided, supramolecular compounds which have the capacity of high-density presentation of the functional epitopes at the surface of the nanostructures. This feature contributes to their successful application in several industry sectors, including food, feed, personal care, and pharmaceutics. In this comprehensive review, the novel class of ribosomally synthesized lipopeptides is introduced alongside the more commonly occuring non-ribosomal lipopeptides. We highlight key representatives of the most researched as well as recently described lipopeptide families, with emphasis on structural features, self-assembly and associated functions. The common biological, chemical and hybrid production routes of lipopeptides, including prominent analogues and derivatives are also discussed. Furthermore, genetic engineering strategies aimed at increasing lipopeptide yields, diversity and biological activity are summarized and exemplified. With respect to application, this work mainly details the potential of lipopeptides in personal care and cosmetics industry as cleansing agents, moisturizer, anti-aging/anti-wrinkling, skin whitening and preservative agents as well as the pharmaceutical industry as anitimicrobial agents, vaccines, immunotherapy, and cancer drugs. Given that this review addresses human applications, we conclude on the topic of safety of lipopeptide formulations and their sustainable production.
Collapse
Affiliation(s)
- Melania Pilz
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Philipp Cavelius
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Farah Qoura
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany
| | - Dania Awad
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany.
| | - Thomas Brück
- Werner Siemens-Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), 85748 Garching, Germany.
| |
Collapse
|
14
|
Peng J, Liu X, Lu Q, Yuan L, Xu W, Zhang H, Zang H. Ultrashort lipo-tetrapeptide with potent antibacterial activity and local therapeutic effect against Staphylococcus aureus. Int J Antimicrob Agents 2023; 62:106916. [PMID: 37423581 DOI: 10.1016/j.ijantimicag.2023.106916] [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: 12/26/2022] [Revised: 05/24/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
OBJECTIVES Mastitis in dairy cows is a common infectious disease on dairy farms and a major danger to the dairy industry. The harmful bacteria with the greatest clinical isolation rate are Staphylococcus aureus. As a result, bacterial mastitis in dairy cows can lead to decreased milk output, quality, and costs. Traditional antibiotics are currently used to treat mastitis in dairy cows. Nonetheless, long-term usage of high doses of antibiotics increases the risk of the establishment of drug-resistant strains, and the problem of drug residues is becoming more prevalent. We investigated the antibacterial effects of varying molecular side chain length lipopeptides on Staphylococcus aureus ATCC25923 and GS1311 using five tetrapeptide ultrashort lipopeptides developed and synthesised in this study. METHODS To evaluate the application value of the synthesized lipopeptides in the prevention and treatment of mastitis, the lipopeptides with the best antibacterial action were chosen for safety testing and a mouse mastitis model treatment test. RESULTS Three of the lipopeptides produced have strong antibacterial properties. Within the drug's safe concentration range, C16KGGK has an excellent antibacterial action and can have a therapeutic influence on mastitis induced by Staphylococcus aureus infection in mice. CONCLUSION The findings of this study can be used to develop new antibacterial medications and their therapeutic application in the treatment of mastitis in dairy cows.
Collapse
Affiliation(s)
- Jie Peng
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, China.
| | - Xuming Liu
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, China.
| | - Qiangsheng Lu
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, China.
| | - Lvfeng Yuan
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, China.
| | - Wanyou Xu
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, China.
| | - Hecheng Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, China.
| | - Haoyue Zang
- College of Veterinary Medicine, Gansu Agricultural University, Gansu, China.
| |
Collapse
|
15
|
Lu W, Lu H, Wang C, Wang G, Dong W, Tan C. Effectors of the Type VI Secretion System Have the Potential to Be Modified into Antimicrobial Peptides. Microbiol Spectr 2023; 11:e0030823. [PMID: 37470717 PMCID: PMC10434152 DOI: 10.1128/spectrum.00308-23] [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: 01/19/2023] [Accepted: 04/26/2023] [Indexed: 07/21/2023] Open
Abstract
The use of antibiotics has led to the emergence of multidrug-resistant (MDR) bacteria, and there is an urgent need to find alternative treatments to alleviate this pressure. The type VI secretion system (T6SS) is a protein delivery system present in bacterial cells that secretes effectors that participate in bacterial virulence. Given the potential for the transformation of these effectors into antimicrobial peptides (AMPs), we designed T6SS effectors into AMPs that have a membrane-disrupting effect. These effectors kill bacteria by altering the membrane potential and increasing the intracellular reactive oxygen species (ROS) content. Moreover, AMPs also have a significant therapeutic effect both in vivo and in vitro. This finding suggests that it is possible to modify bacterial components of bacteria themselves to create compounds that fight bacteria. IMPORTANCE This study first identified and modified the T6SS effector into positively charged alpha-helical peptides. These peptides have good antibacterial and bactericidal effects on G+ bacteria and G- bacteria. This study broadens the source of AMPs and makes T6SS effectors more useful.
Collapse
Affiliation(s)
- Wenjia Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Hao Lu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Chenchen Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Gaoyan Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Wenqi Dong
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, China
| |
Collapse
|
16
|
Yang L, Chen C, Liang T, Hao L, Gu Q, Xu H, Zhao Y, Jiang L, Fan X. Disassembling ability of lipopeptide promotes the antibacterial activity. J Colloid Interface Sci 2023; 649:535-546. [PMID: 37356155 DOI: 10.1016/j.jcis.2023.05.168] [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: 10/26/2022] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/27/2023]
Abstract
Lipopeptides have become one of the most potent antibacterial agents, however, there is so far no consensus about the link between their physic-chemical properties and biological activity, in particular their inherent aggregation propensity and antibacterial potency. To this end, we here de novo design a series of lipopeptides (CnH(2n-1)O-(VVKK)2V-NH2), in which an alkyl chain is covalently attached onto the N-terminus of a short cationic peptide sequence with an alternating pattern of hydrophobic VV (Val) and positively charged KK (Lys) motifs. By varying the alkyl chain length (ortho-octanoic acid (C8), lauric acid (C12), and palmitic acid (C16)), the lipopeptides show distinct physicochemical properties and self-assembly behaviors, which have great effect on their antibacterial activities. C8H15O-(VVKK)2V-NH2, which contains the lowest hydrophobicity and surface activity has the lowest antibacterial activity. C12H23O-(VVKK)2V-NH2 and C16H31O-(VVKK)2V-NH2 both have high hydrophobicity and surface activity, and self-assembled into long nanofibers. However, the nanofibers formed by C12H23O-(VVKK)2V-NH2 disassembled by dilution, resulting in its high antibacterial activity via bacterial membrane disruption. Comparatively, the nanofibers formed by C16H31O-(VVKK)2V-NH2 were very stable, which can closely attach on bacterial surface but not permeate bacterial membrane, leading to its low antibacterial activity. Thus, the stability other than the morphologies of lipopeptides' nanostructures contribute to their antibacterial ability. Importantly, this study enhances our understanding of the antibacterial mechanisms of self-assembling lipopeptides that will be helpful in exploring their biomedical applications.
Collapse
Affiliation(s)
- Liuxin Yang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Cuixia Chen
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China.
| | - Tiantian Liang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Liyun Hao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Qilong Gu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Hai Xu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Yurong Zhao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Lixia Jiang
- Hospital of China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Xinglong Fan
- Department of Thoracic Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China.
| |
Collapse
|
17
|
Peng J, Lu Q, Yuan L, Zhang H. Synthetic Cationic Lipopeptide Can Effectively Treat Mouse Mastitis Caused by Staphylococcus aureus. Biomedicines 2023; 11:biomedicines11041188. [PMID: 37189805 DOI: 10.3390/biomedicines11041188] [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: 02/06/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Mastitis caused by Staphylococcus aureus (S. aureus) in dairy cows is one of the most common clinical diseases in dairy cattle. Unfortunately, traditional antibiotic treatment has resulted in the emergence of drug-resistant strains of bacteria, making this disease more difficult to treat. Therefore, novel lipopeptide antibiotics are becoming increasingly important in treating bacterial diseases, and developing novel antibiotics is critical in controlling mastitis in dairy cows. We designed and synthesized three cationic lipopeptides with palmitic acid, all with two positive charges and dextral amino acids. The lipopeptides' antibacterial activity against S. aureus was determined using MIC and scanning electron microscopy. The safety concentration range of lipopeptides for clinical usage was then estimated using the mouse erythrocyte hemolysis assay and CCK8 cytotoxicity. Finally, lipopeptides with high antibacterial activity and minimal cytotoxicity were selected for the treatment experiments regarding mastitis in mice. The observation of histopathological changes, bacterial tissue load and expression of inflammatory factors determined the therapeutic effects of lipopeptides on mastitis in mice. The results showed that all three lipopeptides displayed some antibacterial activity against S. aureus, with C16dKdK having a strong antibacterial impact and being able to treat the mastitis induced by S. aureus infection in mice within a safe concentration range. The findings of this study can be used as a starting point for the development of new medications for the treatment of mastitis in dairy cows.
Collapse
Affiliation(s)
- Jie Peng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Qiangsheng Lu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Lvfeng Yuan
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
- State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, China
| | - Hecheng Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
18
|
Jörgensen AM, Wibel R, Bernkop-Schnürch A. Biodegradable Cationic and Ionizable Cationic Lipids: A Roadmap for Safer Pharmaceutical Excipients. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206968. [PMID: 36610004 DOI: 10.1002/smll.202206968] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Cationic and ionizable cationic lipids are broadly applied as auxiliary agents, but their use is associated with adverse effects. If these excipients are rapidly degraded to endogenously occurring metabolites such as amino acids and fatty acids, their toxic potential can be minimized. So far, synthesized and evaluated biodegradable cationic and ionizable cationic lipids already showed promising results in terms of functionality and safety. Within this review, an overview about the different types of such biodegradable lipids, the available building blocks, their synthesis and cleavage by endogenous enzymes is provided. Moreover, the relationship between the structure of the lipids and their toxicity is described. Their application in drug delivery systems is critically discussed and placed in context with the lead compounds used in mRNA vaccines. Moreover, their use as preservatives is reviewed, guidance for their design is provided, and an outlook on future developments is given.
Collapse
Affiliation(s)
- Arne Matteo Jörgensen
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, Innsbruck, 6020, Austria
| | - Richard Wibel
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, Innsbruck, 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, Innsbruck, 6020, Austria
| |
Collapse
|
19
|
Li R, Wu J, He F, Xu Q, Yin K, Li S, Li W, Wei A, Zhang L, Zhang XH, Zhang B. Rational design, synthesis, antifungal evaluation and docking studies of antifungal peptide CGA-N12 analogues based on the target CtKRE9. Bioorg Chem 2023; 132:106355. [PMID: 36669359 DOI: 10.1016/j.bioorg.2023.106355] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/02/2022] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
Candida tropicalis is a major non-albicans species that causes invasive candidiasis. CGA-N12, an anti-Candida peptide found by our group, disrupted cell wall architecture by inhibiting the activity of the protein killer-resistant 9 (KRE9), a β-1,6-glucan synthase specific to Candida spp. and plants. Herein, a set of CGA-N12 analogues were rationally designed based on the interaction networks between CGA-N12 and C. tropicalis KRE9 (CtKRE9). Seven CGA-N12 analogues with significantly improved antifungal activity against C. tropicalis were screened by reducing the docking energy of CGA-N12 and CtKRE9 and increasing the number of positive charges on CGA-N12 based on a stable three-dimensional model of CtKRE9. CGA-N12 and its analogues exhibited antifungal activity against C. tropicalis and its persist cells; they also inhibited biofilm formation and eradicated preformed biofilms. Compared with fluconazole, they displayed higher activities against the growth of persister cells and more effective preformed biofilm eradication. Among them, CGA-N12-0801, CGA-N12-0902 and CGA-N12-1002 displayed much higher activity and anti-proteinase digestion stability than CGA-N12. Specifically, CGA-N12-0801 was the optimal analogue, with a minimum inhibitory concentration of 3.46 μg/mL and a therapeutic index of 158.07. The results of electronic microscopy observations and KRE9 activity inhibition assays showed that CGA-N12 and its analogues killed C. tropicalis by disrupting the architecture of the cell wall and the integrity of the cell membrane. In conclusion, for the first time, we provide a simple and reliable method for the rational design of antimicrobial peptides and ideal candidates for treating Candida infections that not effectively eliminated by azole drugs.
Collapse
Affiliation(s)
- Ruifang Li
- College of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China.
| | - Jiasha Wu
- College of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Fuyang He
- School of Artificial Intelligence and Big Data, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Qingpeng Xu
- College of Information Science and Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Kedong Yin
- College of Information Science and Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Shang Li
- College of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Weitong Li
- College of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Ao Wei
- College of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Lan Zhang
- College of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Xin-Hui Zhang
- College of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Beibei Zhang
- College of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China.
| |
Collapse
|
20
|
Secondary metabolic profiling of Serratia marcescens NP10 reveals new stephensiolides and glucosamine derivatives with bacterial membrane activity. Sci Rep 2023; 13:2360. [PMID: 36759548 PMCID: PMC9911388 DOI: 10.1038/s41598-023-28502-6] [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: 11/07/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Secondary metabolic profiling, using UPLC-MSE and molecular networking, revealed the secondary metabolites produced by Serratia marcescens NP10. The NP10 strain co-produced cyclic and open-ring stephensiolides (i.e., fatty acyl chain linked to Thr-Ser-Ser-Ile/Leu-Ile/Leu/Val) and glucosamine derivatives (i.e., fatty acyl chain linked to Val-glucose-butyric/oxo-hexanoic acid), with the structures of sixteen new stephensiolides (L-Y) and three new glucosamine derivatives (L-N) proposed. Genome mining identified sphA (stephensiolides) and gcd (glucosamine derivatives) gene clusters within Serratia genomes available on NBCI using antiSMASH, revealing specificity scores of the adenylation-domains within each module that corroborates MSE data. Of the nine RP-HPLC fractions, two stephensiolides and two glucosamine derivatives exhibited activity against Staphylococcus aureus (IC50 of 25-79 µg/mL). 1H NMR analysis confirmed the structure of the four active compounds as stephensiolide K, a novel analogue stephensiolide U, and glucosamine derivatives A and C. Stephensiolides K and U were found to cause membrane depolarisation and affect the membrane permeability of S. aureus, while glucosamine derivatives A and C primarily caused membrane depolarisation. New members of the stephensiolide and glucosamine derivative families were thus identified, and results obtained shed light on their antibacterial properties and mode of membrane activity.
Collapse
|
21
|
Cui Q, Yu HD, Xu QJ, Liu Y, Wang YT, Li PH, Kong LC, Zhang HP, Jiang XY, Giuliodori AM, Fabbretti A, He CG, Ma HX. Antibiotic synergist OM19r reverses aminoglycoside resistance in multidrug-resistant Escherichia coli. Front Microbiol 2023; 14:1144946. [PMID: 37143537 PMCID: PMC10151501 DOI: 10.3389/fmicb.2023.1144946] [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: 01/15/2023] [Accepted: 03/24/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction The continued emergence and spread of multidrug-resistant (MDR) bacterial pathogens require a new strategy to improve the efficacy of existing antibiotics. Proline-rich antimicrobial peptides (PrAMPs) could also be used as antibacterial synergists due to their unique mechanism of action. Methods Utilizing a series of experiments on membrane permeability, In vitro protein synthesis, In vitro transcription and mRNA translation, to further elucidate the synergistic mechanism of OM19r combined with gentamicin. Results A proline-rich antimicrobial peptide OM19r was identified in this study and its efficacy against Escherichia coli B2 (E. coli B2) was evaluated on multiple aspects. OM19r increased antibacterial activity of gentamicin against multidrug-resistance E. coli B2 by 64 folds, when used in combination with aminoglycoside antibiotics. Mechanistically, OM19r induced change of inner membrane permeability and inhibited translational elongation of protein synthesis by entering to E. coli B2 via intimal transporter SbmA. OM19r also facilitated the accumulation of intracellular reactive oxygen species (ROS). In animal models, OM19r significantly improved the efficacy of gentamicin against E. coli B2. Discussion Our study reveals that OM19r combined with GEN had a strong synergistic inhibitory effect against multi-drug resistant E. coli B2. OM19r and GEN inhibited translation elongation and initiation, respectively, and ultimately affected the normal protein synthesis of bacteria. These findings provide a potential therapeutic option against multidrug-resistant E. coli.
Collapse
Affiliation(s)
- Qi Cui
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Han-Dong Yu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Qi-Jun Xu
- Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Yue Liu
- Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Yu-Ting Wang
- Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Peng-Hui Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Ling-Cong Kong
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Hai-Peng Zhang
- Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, College of Life Sciences, Jilin Agricultural University, Changchun, China
| | - Xiu-Yun Jiang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Anna Maria Giuliodori
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Attilio Fabbretti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Cheng-Guang He
- Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, College of Life Sciences, Jilin Agricultural University, Changchun, China
- Cheng-Guang He,
| | - Hong-Xia Ma
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, College of Life Sciences, Jilin Agricultural University, Changchun, China
- *Correspondence: Hong-Xia Ma,
| |
Collapse
|
22
|
Muñoz L, Caparrós E, Albillos A, Francés R. The shaping of gut immunity in cirrhosis. Front Immunol 2023; 14:1139554. [PMID: 37122743 PMCID: PMC10141304 DOI: 10.3389/fimmu.2023.1139554] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Cirrhosis is the common end-stage of chronic liver diseases of different etiology. The altered bile acids metabolism in the cirrhotic liver and the increase in the blood-brain barrier permeability, along with the progressive dysbiosis of intestinal microbiota, contribute to gut immunity changes, from compromised antimicrobial host defense to pro-inflammatory adaptive responses. In turn, these changes elicit a disruption in the epithelial and gut vascular barriers, promoting the increased access of potential pathogenic microbial antigens to portal circulation, further aggravating liver disease. After summarizing the key aspects of gut immunity during homeostasis, this review is intended to update the contribution of liver and brain metabolites in shaping the intestinal immune status and, in turn, to understand how the loss of homeostasis in the gut-associated lymphoid tissue, as present in cirrhosis, cooperates in the advanced chronic liver disease progression. Finally, several therapeutic approaches targeting the intestinal homeostasis in cirrhosis are discussed.
Collapse
Affiliation(s)
- Leticia Muñoz
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Caparrós
- Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain
- Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Agustín Albillos
- Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- *Correspondence: Agustín Albillos, ; Rubén Frances,
| | - Rubén Francés
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
- Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain
- Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnologiía Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Elche, Spain
- *Correspondence: Agustín Albillos, ; Rubén Frances,
| |
Collapse
|
23
|
Zhang F, Yang P, Mao W, Zhong C, Zhang J, Chang L, Wu X, Liu H, Zhang Y, Gou S, Ni J. Short, mirror-symmetric antimicrobial peptides centered on "RRR" have broad-spectrum antibacterial activity with low drug resistance and toxicity. Acta Biomater 2022; 154:145-167. [PMID: 36241015 DOI: 10.1016/j.actbio.2022.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/31/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022]
Abstract
The increasingly severe bacterial resistance worldwide pushes people to discover and design potential antibacterial drugs unavoidably. In this work, a series of short, mirror-symmetric peptides were designed and successfully synthesized, centered on "RRR" and labeled with hydrophobic amino acids at both ends. Based on the structure-activity relationship analysis, LWWR (LWWRRRWWL-NH2) was screened as a desirable mirror-symmetric peptide for further study. As expected, LWWR displayed broad-spectrum antibacterial activity against the standard bacteria and antibiotic-resistant strains. Undoubtedly, the high stability of LWWR in a complex physiological environment was an essential guarantee to maximizing its antibacterial activity. Indeed, LWWR also exhibited a rapid bactericidal speed and a low tendency to develop bacterial resistance, based on the multiple actions of non-receptor-mediated membrane actions and intra-cellular mechanisms. Surprisingly, although LWWR showed similar in vivo antibacterial activity compared with Polymyxin B and Melittin, the in vivo safety of LWWR was far higher than that of them, so LWWR had better therapeutic potential. In summary, the desirable mirror-symmetric peptide LWWR was promised as a potential antibacterial agent to confront the antibiotics resistance crisis. STATEMENT OF SIGNIFICANCE: Witnessing the growing problem of antibiotic resistance, a series of short, mirror-symmetric peptides based on the symmetric center "RRR" and hydrophobic terminals were designed and synthesized in this study. Among, LWWR (LWWRRRWWL-NH2) presented broad-spectrum antibacterial activity both in vitro and in vivo due to its multiple mechanisms and good stability. Meanwhile, the low drug resistance and toxicity of LWWR also suggested its potential for clinical application. The findings of this study will provide some inspiration for the design and development of potential antibacterial agents, and contribute to the elimination of bacterial infections worldwide as soon as possible.
Collapse
Affiliation(s)
- Fangyan Zhang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Ping Yang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Wenbo Mao
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Chao Zhong
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Jingying Zhang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Linlin Chang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Xiaoyan Wu
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Hui Liu
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yun Zhang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Sanhu Gou
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Jingman Ni
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China.
| |
Collapse
|
24
|
Liu S, She P, Li Z, Li Y, Li L, Yang Y, Zhou L, Wu Y. Drug synergy discovery of tavaborole and aminoglycosides against Escherichia coli using high throughput screening. AMB Express 2022; 12:151. [PMID: 36454354 PMCID: PMC9715904 DOI: 10.1186/s13568-022-01488-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 12/05/2022] Open
Abstract
High incidences of urinary tract infection (UTI) of aminoglycosides-resistant E.coli causes a severe burden for public health. A new therapeutic strategy to ease this crisis is to repurpose non-antibacterial compounds to increase aminoglycosides sensibility against multidrug resistant E.coli pathogens. Based on high throughput screening technology, we profile the antimicrobial activity of tavaborole, a first antifungal benzoxaborole drug for onychomycosis treatment, and investigate the synergistic interaction between tavaborole and aminoglycosides, especially tobramycin and amikacin. Most importantly, by resistance accumulation assay, we found that, tavaborole not only slowed resistance occurrence of aminoglycosides, but also reduced invasiveness of E.coli in combination with tobramycin. Mechanistic studies preliminary explored that tavaborole and aminoglycosides lead to mistranslation, but would be still necessary to investigate more details for further research. In addition, tavaborole exhibited low systematic toxicity in vitro and in vivo, and enhanced aminoglycoside bactericidal activity in mice peritonitis model. Collectively, these results suggest the potential of tavaborole as a novel aminoglycosides adjuvant to tackle the clinically relevant drug resistant E. coli and encourages us to discover more benzoxaborole analogues for circumvention of recalcitrant infections.
Collapse
Affiliation(s)
- Shasha Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Zehao Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Yimin Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Linhui Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Yifan Yang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Linying Zhou
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, Hunan, China
| | - Yong Wu
- Department of Laboratory Medicine, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, Hunan, China.
| |
Collapse
|
25
|
C-terminal modification of a de novo designed antimicrobial peptide via capping of macrolactam rings. Bioorg Chem 2022; 130:106251. [DOI: 10.1016/j.bioorg.2022.106251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
|
26
|
Wang L, Liu H, Li X, Yao C. Assessment of New Strategies to Improve the Performance of Antimicrobial Peptides. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3691. [PMID: 36296881 PMCID: PMC9610275 DOI: 10.3390/nano12203691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023]
Abstract
In this research, we constructed a novel engineered tripeptide modified with lipoic acid (LA-RWR), followed by crosslinking of lipoic acid to form nanoparticles (c-LA-RWR). LA-RWR was also modified with phenethylamine (PEA) on the C-terminus to achieve better antibacterial activities. The as-prepared c-LA-RWR and LA-RWR-PEA were effective against E.coli, S.aureus, C.albicans, and methicillin-resistant Staphylococcus aureus, with minimum inhibitory concentration values ranging from 2 to 16 µg/mL, which greatly improved the performance of LA-RWR. Similar antibacterial activities were demonstrated in anti-biofilm activity; there was no matter on the biofilm that was already established or forming. Moreover, c-LA-RWR/LA-RWR-PEA remarkably induced cytoplasmic membrane depolarization and outer membrane permeabilization, resulting in varying degrees of damage to the bacterial morphology, which were consistent with the results obtained via electron microscopy. Thus, our results show that c-LA-RWR/LA-RWR-PEA exhibited excellent efficacy against a variety of microorganisms with good biosafety, providing new strategies by which to improve the performance of antimicrobial peptides.
Collapse
Affiliation(s)
| | | | | | - Chen Yao
- Correspondence: ; Tel.: +86-138-1386-1022
| |
Collapse
|
27
|
Polyphosphate Kinase Is Required for the Processes of Virulence and Persistence in Acinetobacter baumannii. Microbiol Spectr 2022; 10:e0123022. [PMID: 35867473 PMCID: PMC9430702 DOI: 10.1128/spectrum.01230-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Acinetobacter baumannii, one of the most successful bacteria causing severe nosocomial infection, was identified as a top-priority pathogen by the WHO. Thus, genetic manipulations to clarify the potential targets for fighting A. baumannii resistance and virulence are vital. Polyphosphate (polyP) kinase (PPK) is conserved in nearly all bacteria and is responsible for polyP formation, which is associated with bacterial pathogenicity and antibiotic resistance. In this study, ppk1-deficient (Δppk1::Apr), ppk1-complemented (Δppk1::Apr/PJL02-ppk1), and wild-type strains of A. baumannii ATCC 17978 were used to determine the influence of PPK1 on A. baumannii virulence and persistence mainly by polyP quantification, surface motility, biofilm formation, and bacterial persistence assays. Our work found that PPK1 is indispensable for polyP formation in vivo and that the motility of the PPK1-deficient strain was significantly impaired due to the lack of a pilus-like structure typically present compared with the complemented and wild-type strains. The deficiency of PPK1 also inhibited the biofilm formation of A. baumannii and decreased bacterial persistence under stimuli of high-concentration ampicillin (Amp) treatment, H2O2 stress, heat shock, and starvation stress. Furthermore, ppk1-deficient bacterium-infected mice showed a significantly reduced bacterial load and a decreased inflammatory response. However, complementation with PPK1 effectively rescued the impaired virulence and persistence of ppk1-deficient A. baumannii. In addition, metabonomic analysis revealed that PPK1 was associated with glycerophospholipid metabolism and fatty acid biosynthesis. Taken together, our results suggest that targeting PPK1 to control A. baumannii pathogenicity and persistence is a feasible strategy to fight this pathogen. IMPORTANCEA. baumannii was identified as a top-priority pathogen by the WHO due to its antibiotic resistance. Meanwhile, the pathogenicity of A. baumannii mediated by several vital virulence factors also cannot be ignored. Here, the role of PPK1 in A. baumannii was also explored. We found that the motility ability and biofilm formation of a PPK1-deficient strain were significantly impaired. Furthermore, PPK1 was essential for its persistence maintenance to resist stimuli of high-concentration Amp treatment, H2O2 stress, heat shock, and starvation stress. Metabonomic analysis revealed that PPK1 was associated with glycerophospholipid metabolism and fatty acid biosynthesis. In addition, ppk1-deficient bacterium-infected mice showed significantly reduced bacterial loads and a decreased inflammatory responses in vivo. Together, our results suggest that PPK1 is vital for A. baumannii pathogenicity and persistence.
Collapse
|
28
|
Ma L, Huang S, Xie H, Ma P, Jia B, Yao Y, Gao Y, Li W, Song J, Zhang W. Influence of chain length on the anticancer activity of the antimicrobial peptide CAMEL with fatty acid modification. Eur J Med Chem 2022; 239:114557. [PMID: 35759906 DOI: 10.1016/j.ejmech.2022.114557] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/09/2022] [Accepted: 06/18/2022] [Indexed: 01/10/2023]
Abstract
Antimicrobial peptides (AMPs) display promising potential in cancer therapy. Modification with fatty acids is a simple and effective approach to improve the activity of AMPs. In the present study, we investigated the effects of fatty acid chain lengths on the anticancer activity, self-assembly and mechanism of action of CAMEL (CM15, KWKLFKKIGAVLKVL-NH2), an amphipathic AMP with 15 amino acids. Conjugation of fatty acids could obviously improve the in vitro anticancer activity of CAMEL. Among the tested peptides, C12-CAMEL showed the highest anticancer activity, while C16-CAMEL killed cancer cells with the slowest kinetics. This may be related to the self-assembly of C12-CAMEL and C16-CAMEL, which could form spherical nanoparticles and tightened nanofibers, respectively. In addition, necrosis and necroptosis rather than apoptosis were the major mechanisms underlying the anticancer activity of CAMEL, C12-CAMEL and C16-CAMEL, implying that modification with fatty acids did not obviously alter the mechanism of action of CAMEL. Notably, C12-CAMEL, with high and rapid cell-killing activity, exhibited significantly stronger in vivo anticancer activity than CAMEL and C16-CAMEL. Overall, the present work suggests that the choice of a suitable fatty acid for structural modification is necessary for improving the anticancer activity of AMPs.
Collapse
Affiliation(s)
- Ling Ma
- The Institute of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Sujie Huang
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Huan Xie
- Department of Medical, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Panpan Ma
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Bo Jia
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yufan Yao
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yuxuan Gao
- The Institute of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Wenyuan Li
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jingjing Song
- The Institute of Pharmacology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Wei Zhang
- Institute of Physiology, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Veterinary Etiological Biology, College of Veterinary Medicine, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
29
|
Svenson J, Molchanova N, Schroeder CI. Antimicrobial Peptide Mimics for Clinical Use: Does Size Matter? Front Immunol 2022; 13:915368. [PMID: 35720375 PMCID: PMC9204644 DOI: 10.3389/fimmu.2022.915368] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
The search for efficient antimicrobial therapies that can alleviate suffering caused by infections from resistant bacteria is more urgent than ever before. Infections caused by multi-resistant pathogens represent a significant and increasing burden to healthcare and society and researcher are investigating new classes of bioactive compounds to slow down this development. Antimicrobial peptides from the innate immune system represent one promising class that offers a potential solution to the antibiotic resistance problem due to their mode of action on the microbial membranes. However, challenges associated with pharmacokinetics, bioavailability and off-target toxicity are slowing down the advancement and use of innate defensive peptides. Improving the therapeutic properties of these peptides is a strategy for reducing the clinical limitations and synthetic mimics of antimicrobial peptides are emerging as a promising class of molecules for a variety of antimicrobial applications. These compounds can be made significantly shorter while maintaining, or even improving antimicrobial properties, and several downsized synthetic mimics are now in clinical development for a range of infectious diseases. A variety of strategies can be employed to prepare these small compounds and this review describes the different compounds developed to date by adhering to a minimum pharmacophore based on an amphiphilic balance between cationic charge and hydrophobicity. These compounds can be made as small as dipeptides, circumventing the need for large compounds with elaborate three-dimensional structures to generate simplified and potent antimicrobial mimics for a range of medical applications. This review highlight key and recent development in the field of small antimicrobial peptide mimics as a promising class of antimicrobials, illustrating just how small you can go.
Collapse
Affiliation(s)
| | - Natalia Molchanova
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Christina I. Schroeder
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| |
Collapse
|
30
|
Li B, Ouyang X, Ba Z, Yang Y, Zhang J, Liu H, Zhang T, Zhang F, Zhang Y, Gou S, Ni J. Novel β-Hairpin Antimicrobial Peptides Containing the β-Turn Sequence of -RRRF- Having High Cell Selectivity and Low Incidence of Drug Resistance. J Med Chem 2022; 65:5625-5641. [DOI: 10.1021/acs.jmedchem.1c02140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Beibei Li
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Xu Ouyang
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Zufang Ba
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Yinyin Yang
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Jingying Zhang
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Hui Liu
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Tianyue Zhang
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Fangyan Zhang
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Yun Zhang
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Sanhu Gou
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Jingman Ni
- Institute of Pharmaceutics, School of Pharmacy and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
31
|
Dewangan RP, Verma DP, Verma NK, Gupta A, Pant G, Mitra K, Habib S, Ghosh JK. Spermine-Conjugated Short Proline-Rich Lipopeptides as Broad-Spectrum Intracellular Targeting Antibacterial Agents. J Med Chem 2022; 65:5433-5448. [PMID: 35297625 DOI: 10.1021/acs.jmedchem.1c01809] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Toward the design of new proline-rich peptidomimetics, a short peptide segment, present in several proline-rich antimicrobial peptides (AMPs), was selected. Fatty acids of varying lengths and spermine were conjugated at the N- and C-terminals of the peptide, respectively. Spermine-conjugated lipopeptides, C10-PR-Spn and C12-PR-Spn, exhibited minimum inhibitory concentrations within 1.5-6.2 μM against the tested pathogens including resistant bacteria and insignificant hemolytic activity against human red blood cells up to 100 μM concentrations and demonstrated resistance against trypsin digestion. C10-PR-Spn and C12-PR-Spn showed synergistic antimicrobial activity against multidrug-resistant methicillin-resistant Staphylococcus aureus with several tested antibiotics. These lipopeptides did not permeabilize bacterial membrane-mimetic lipid vesicles or damage the Escherichia coli membrane like the nonmembrane-lytic AMP, buforin-II. The results suggested that C10-PR-Spn and C12-PR-Spn could interact with the 70S ribosome of E. coli and inhibit its protein synthesis. C10-PR-Spn and C12-PR-Spn demonstrated superior clearance of bacteria from the spleen, liver, and kidneys of mice, infected with S. aureus ATCC 25923 compared to levofloxacin.
Collapse
Affiliation(s)
- Rikeshwer Prasad Dewangan
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Devesh Pratap Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Neeraj Kumar Verma
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Ankit Gupta
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Garima Pant
- Electron Microscopy Unit, SAIF Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Kalyan Mitra
- Electron Microscopy Unit, SAIF Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Saman Habib
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| | - Jimut Kanti Ghosh
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226031, India
| |
Collapse
|
32
|
Zhang F, Zhong C, Yao J, Zhang J, Zhang T, Li B, Gou S, Ni J. Antimicrobial peptides–antibiotics combination: An effective strategy targeting drug‐resistant Gram‐negative bacteria. Pept Sci (Hoboken) 2022. [DOI: 10.1002/pep2.24261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Fangyan Zhang
- School of Pharmacy Lanzhou University Lanzhou China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences Lanzhou University Lanzhou China
| | - Chao Zhong
- School of Pharmacy Lanzhou University Lanzhou China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences Lanzhou University Lanzhou China
| | - Jia Yao
- The First Hospital Lanzhou University Lanzhou China
| | - Jingying Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences Lanzhou University Lanzhou China
| | | | - Beibei Li
- School of Pharmacy Lanzhou University Lanzhou China
| | - Sanhu Gou
- School of Pharmacy Lanzhou University Lanzhou China
| | - Jingman Ni
- School of Pharmacy Lanzhou University Lanzhou China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences Lanzhou University Lanzhou China
- State Key Laboratory of Quality Research in Chinese Medicine Macau University of Science and Technology Taipa Macao China
| |
Collapse
|
33
|
Gong T, Fu J, Shi L, Chen X, Zong X. Antimicrobial Peptides in Gut Health: A Review. Front Nutr 2021; 8:751010. [PMID: 34660671 PMCID: PMC8514777 DOI: 10.3389/fnut.2021.751010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Animal antimicrobial peptides (AMPs), known as broad-spectrum and high-efficiency antibacterial activity, are important effector molecules in innate immune system. AMPs not only have antimicrobial, antiviral and antitumor effects but also exhibit important effects in vivo, such as anti-inflammatory response, recruiting immune cells, promoting epithelial damage repair, and promoting phagocytosis of bacteria. However, research on the application of AMPs is incomplete and controversial. This review mainly introduces the classification of AMPs, biological functions, as well as the mechanisms of action, expression rules, and nutrition regulation from three perspectives, aiming to provide important information for the application of AMPs.
Collapse
Affiliation(s)
- Tao Gong
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jie Fu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Lexuan Shi
- Guangzhou Dublin International College of Life Sciences and Technology, South China Agricultural University, Guangzhou, China
| | - Xin Chen
- School of Medicine, Foshan University, Foshan, China
| | - Xin Zong
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Animal Nutrition and Feed Science in Eastern China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
34
|
Gou S, Li B, Ouyang X, Ba Z, Zhong C, Zhang T, Chang L, Zhu Y, Zhang J, Zhu N, Zhang Y, Liu H, Ni J. Novel Broad-Spectrum Antimicrobial Peptide Derived from Anoplin and Its Activity on Bacterial Pneumonia in Mice. J Med Chem 2021; 64:11247-11266. [PMID: 34180670 DOI: 10.1021/acs.jmedchem.1c00614] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The emergence of multidrug-resistant bacteria has major issues for treating bacterial pneumonia. Currently, anoplin (GLLKRIKTLL-NH2) is a natural antimicrobial candidate derived from wasp venom. In this study, a series of new antimicrobial peptide (AMP) anoplin analogues were designed and synthesized. The relationship between their biological activities and their positive charge, hydrophobicity, amphipathicity, and secondary structure are described. The characteristic shared by these peptides is that positively charged amino acids and hydrophobic amino acids are severally arranged on the hydrophilic and hydrophobic surface of the α-helix to form a completely amphiphilic structure. To achieve ideal AMPs, below the range of the threshold of the cytotoxicity and hemolytic activity, their charges and hydrophobicity were increased as much. Among the new analogues, A-21 (KWWKKWKKWW-NH2) exhibited the greatest antimicrobial activity (geometric mean of minimum inhibitory concentrations = 4.76 μM) against all the tested bacterial strains, high bacterial cell selectivity in vitro, high effectiveness against bacterial pneumonia in mice infected with Klebsiella pneumoniae, and low toxicity in mice (LD50 = 82.01 mg/kg). A-21 exhibited a potent bacterial membrane-damaging mechanism and lipopolysaccharide-binding ability. These data provide evidence that A-21 is a promising antimicrobial candidate for the treatment of bacterial pneumonia.
Collapse
Affiliation(s)
- Sanhu Gou
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Beibei Li
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xu Ouyang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Zufang Ba
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Chao Zhong
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Tianyue Zhang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - LinLin Chang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Yuewen Zhu
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jingying Zhang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Ningyi Zhu
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yun Zhang
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Hui Liu
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jingman Ni
- Institute of Pharmaceutics, School of Pharmacy, Lanzhou University, Lanzhou 730000, China.,Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|