1
|
Dubkara H, Lal J, Saxena D, Akhir A, Maitra R, Chopra S, Reddy DN. Discovery of a potent ornithine-modified gramicidin S analogue against drug-resistant Staphylococcus aureus and Enterococcus faecalis with minimal red blood cell toxicity. Eur J Med Chem 2025; 292:117654. [PMID: 40288118 DOI: 10.1016/j.ejmech.2025.117654] [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: 01/24/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
The high haemolytic toxicity of Gramicidin S restricts its therapeutic use to topical applications. Given the growing need for new antibiotics and drawing inspiration from the cyclic structure and druggable characteristics of Gramicidin S, we have synthesized 15 ornithine (Orn) modified analogous peptides systematically and investigated their antimicrobial activity and cytotoxicity. Results revealed that mono- ornithine residue replaced with tryptophan (11) and arginine (12) peptides showed improved activity against multidrug resistant bacterial strains of Staphylococcus aureus and Enterococcus faecalis (MIC 4-8 μg/mL) compared with comparators vancomycin (MIC >64 μg/mL), levofloxacin (MIC 32-64 μg/mL) and meropenem (MIC 8-64 μg/mL). Cytotoxicity data demonstrated that peptide 11 (HC50 = 112.1 μg/mL) and 12 (HC50 = 186 μg/mL) exhibited greatly reduced haemolytic activity, as compared with Gramicidin S (HC50 = 35.13 μg/mL). The concentration-dependent time-kill kinetic assay resulted the active peptide 12 represents better bactericidal effect compared with 11 and vancomycin. Scanning electron microscope analysis shows that GS and the modified peptide 12 disrupt the bacterial cell surface, causing damage and leading to bacterial cell death. 2D NOESY data of 12 showed that the arginine residue side-chain guanidinium ion and tryptophan indole form a cation-π interaction. This interaction between arginine and tryptophan stabilizes the β-sheet conformation, selectively targets bacterial membranes, hence exhibiting reduced red blood cell toxicity. The overall study suggests that the peptide 12 may be further developed as an antibiotic for systematic use against infections caused due to S. aureus.
Collapse
Affiliation(s)
- Harshita Dubkara
- Division of Medicinal and Process Chemistry, CSIR-CDRI, Lucknow, 226031, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, UP, 201002, India
| | - Jhajan Lal
- Division of Medicinal and Process Chemistry, CSIR-CDRI, Lucknow, 226031, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, UP, 201002, India
| | - Deepanshi Saxena
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Abdul Akhir
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Rahul Maitra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sidharth Chopra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, UP, 201002, India.
| | - Damodara N Reddy
- Division of Medicinal and Process Chemistry, CSIR-CDRI, Lucknow, 226031, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, UP, 201002, India.
| |
Collapse
|
2
|
Schouten G, Paulussen F, Grossmann TN, Bitter W, van Ulsen P. Membrane Modification and Adaptation of Metabolism by Acinetobacter baumannii Prompts Resistance to Antimicrobial Activity of Outer Membrane Perturbing Peptide L8. J Mol Biol 2025; 437:169135. [PMID: 40221130 DOI: 10.1016/j.jmb.2025.169135] [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: 12/06/2024] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/14/2025]
Abstract
Multidrug resistant (MDR) Acinetobacter baumannii has emerged as one of the most concerning nosocomial pathogens worldwide. One approach to target MDR A. baumannii is treatment with synergistic combinations of outer membrane-permeabilizing antimicrobial peptides (AMP) and antibiotics that otherwise only act against Gram-positive bacteria. Resistance against AMPs is rarely observed, especially when administered in combination with other drugs. Recently, we described the synergistic antimicrobial activity of AMPs L8 and L8S1 with rifampicin against a clinical isolate of A. baumannii. In the current work we explore the mechanisms of action of these peptides. We demonstrate that L8 and L8S1 perturb the cell envelope of A. baumannii. Moreover, we show that resistance against peptide L8 could be acquired in vitro either by increasing the amount of PE lipid on the surface or by increasing biofilm formation. Interestingly, the resistance to the antimicrobial activity of the peptides did not affect membrane perturbation or synergistic activity of the peptides with rifampicin, suggesting a dual mechanism of action for these peptides.
Collapse
Affiliation(s)
- Gina Schouten
- Medical Microbiology and Infection Control (MMI), Amsterdam UMC Location Vumc, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands
| | - Felix Paulussen
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Tom N Grossmann
- Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Wilbert Bitter
- Medical Microbiology and Infection Control (MMI), Amsterdam UMC Location Vumc, De Boelelaan 1108, 1081 HZ Amsterdam, the Netherlands; Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands; Molecular Microbiology, A-life, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - Peter van Ulsen
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands; Molecular Microbiology, A-life, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Lobka M, Siekierska I, Chyży P, Burmistrz M, Macyszyn J, Grzela R, Wojciechowska M, Trylska J. Design, synthesis and evaluation of lysine- and leucine-rich hydrocarbon-stapled peptides as antibacterial agents. Eur J Med Chem 2025; 290:117445. [PMID: 40101449 DOI: 10.1016/j.ejmech.2025.117445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/06/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
To address the challenge of antimicrobial resistance, we investigated new antibacterial peptides based on lysine- and leucine-rich sequences. We stabilised their membrane-active secondary structures by applying hydrocarbon stapling at sequence positions i and i+4. Stapling improved peptide structural stability in both aqueous and lipid environments, regardless of the staple position. It also enhanced antibacterial efficiency against both gram-negative and gram-positive bacteria, including antibiotic-resistant strains, with minimum inhibitory concentrations (MICs) of 2-4 μM (2.5-5.5 μg/mL). The stapled peptides showed increased resistance to enzymatic degradation, particularly with staples incorporated near the N-terminus, and were not haemolytic or cytotoxic at their MICs. Molecular dynamics simulations revealed how stapling aids in (i) stabilising the membrane-active secondary structure of amphipathic peptides and (ii) accelerating their membrane insertion. Our results provide insight into peptide design for antimicrobial use. We show that hydrocarbon stapling of lysine- and leucine-rich short sequences may offer a pathway towards more stable and effective antibacterial agents.
Collapse
Affiliation(s)
- Małgorzata Lobka
- University of Warsaw, Centre of New Technologies, S. Banacha 2c, 02-097, Warsaw, Poland
| | - Izabela Siekierska
- University of Warsaw, Centre of New Technologies, S. Banacha 2c, 02-097, Warsaw, Poland
| | - Piotr Chyży
- University of Warsaw, Centre of New Technologies, S. Banacha 2c, 02-097, Warsaw, Poland
| | - Michał Burmistrz
- University of Warsaw, Centre of New Technologies, S. Banacha 2c, 02-097, Warsaw, Poland
| | - Julia Macyszyn
- University of Warsaw, Centre of New Technologies, S. Banacha 2c, 02-097, Warsaw, Poland
| | - Renata Grzela
- University of Warsaw, Centre of New Technologies, S. Banacha 2c, 02-097, Warsaw, Poland; University of Warsaw, Faculty of Physics, Institute of Experimental Physics, Division of Biophysics, 02-093, Warsaw, Poland
| | - Monika Wojciechowska
- University of Warsaw, Centre of New Technologies, S. Banacha 2c, 02-097, Warsaw, Poland
| | - Joanna Trylska
- University of Warsaw, Centre of New Technologies, S. Banacha 2c, 02-097, Warsaw, Poland.
| |
Collapse
|
4
|
Zhao T, Zha Y, Jiang S, Wang R, Song Y, Li L, Lyu J, Hu W, Zhang D, Wu S, Zhang Y. Temporin-GHa derived peptide shows inhibitory efficacy against Cutibacterium acnes and alleviates inflammatory reactions in acne vulgaris. Int Immunopharmacol 2025; 157:114756. [PMID: 40318277 DOI: 10.1016/j.intimp.2025.114756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 04/15/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Acne vulgaris is a widespread chronic inflammatory skin disease that is mainly caused by Cutibacterium acnes infection and subsequent inflammation. Temporin-GHaR4G7R (GHaR4G7R) was derived from Temporin-GHa of frog Hylarana guentheri. Its antibacterial performance against C. acnes and potential mechanism against acne vulgaris in vitro and in vivo were evaluated. In vitro tests demonstrated that GHaR4G7R displayed potent bactericidal effects against C. acnes, with both the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of 3.1 μM. It exerted antibacterial effect by disrupting the bacterial membranes, accompanied by low propensity for developing drug resistance. GHaR4G7R significantly inhibited the formation of early biofilms and eliminated established biofilms of C. acnes by decreasing exopolysaccharide synthesis. Meanwhile, GHaR4G7R demonstrated an anti-inflammatory effect on HaCaT cells challenged with heat-inactivated C. acnes by significantly down-regulating the expression of TLR2/NF-κB/MAPK pathway-associated proteins by qRT-PCR, immunofluorescence, and Western blot assays. As expected, GHaR4G7R significantly decreased the bacterial colonization in rat ear model induced by C. acnes, and relieved the auricular swelling and inflammatory cell infiltration through inhibiting the TLR2/NF-κB/MAPK signaling pathway, leading to down-regulation of the inflammatory cytokine expression and alleviation of the skin inflammation in vivo. The research indicates that GHaR4G7R might be a potential alternative for developing novel strategies for treating acne vulgaris.
Collapse
Affiliation(s)
- Ting Zhao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Yanmei Zha
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou, China; College of Life Sciences, Hainan University, Haikou, China
| | - Shangjun Jiang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Rong Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Yanting Song
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Lushuang Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Junchen Lyu
- Chinese Communist Youth League Committee, Hainan University, Haikou, China
| | - Wenting Hu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou, China
| | - Daqi Zhang
- Department of Neurology, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shuang Wu
- Department of Dermatology, The First Hospital of Hohhot, Hohhot, China
| | - Yingxia Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou, China.
| |
Collapse
|
5
|
Ismael M, Juliah K, Edwin M. Antimicrobial and Potent Anti-Biofilm Properties of Rationally Designed α-Helix Antimicrobial Peptides. J Pept Sci 2025; 31:e70027. [PMID: 40326595 DOI: 10.1002/psc.70027] [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: 03/11/2025] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
The antimicrobial resistance (AMR) crisis represents a significant global threat. Unlike traditional antibiotics, antimicrobial peptides offer a promising pathway because of their primary mechanisms. This study aimed to evaluate and rationally design novel AMPs based on tobacco nectar's AMP (Pep 6) to combat antibiotic resistance issues. Substitution and truncation of some amino acids were applied. Four peptides, KF19, KF16, LK16, and LR16, were designed with enhanced net charge hydrophobicity. They were evaluated for their in vitro antibacterial activity. However, only promising AMPs were further evaluated for their hemolytic activity, time-killing kinetics, mode of action, and anti-biofilm properties. The results showed that only KF19 and LR16 have potent activity against Staphylococcus aureus ATCC25923 and resistant isolates with MIC values from 7.81 to 15.62 μg/mL. Hemolysis ratios were 2.38% and 2.24% at 125 μg/mL for KF19 and LR16, respectively. Both peptides were able to kill S. aureus ATCC25923 within 2 h. SEM results showed their ability to target the cell membrane. Both peptides destroyed the S. aureus biofilms significantly at 62.5 and 125 μg/mL (**p < 0.01, ***p < 0.001, ****p < 0.0001). This study supported rational design in developing new antibacterial agents and demonstrated the therapeutic potency of novel peptides that could solve the resistance issues.
Collapse
Affiliation(s)
- Motasim Ismael
- Department of Molecular Biology and Biotechnology, Pan African University Institute for Basic Sciences Technology and Innovation (PAUSTI), Nairobi, Kenya
| | - Khayeli Juliah
- Department of Zoology, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Madivoli Edwin
- Department of Chemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
- Department of Physics and Biophysics, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
6
|
Li Z, Shui Y, Wang H, Li S, Deng B, Zhang W, Gao S, Zhao L. In Vitro and In Vivo Anti-Candida albicans Activity of a Scorpion-Derived Peptide. Probiotics Antimicrob Proteins 2025; 17:1615-1623. [PMID: 38372937 DOI: 10.1007/s12602-024-10233-3] [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] [Accepted: 02/02/2024] [Indexed: 02/20/2024]
Abstract
The increasing infection and drug resistance frequency has encouraged the exploration of new and effective anti-Candida albicans agents. In this study, CT-K3K7, a scorpion antimicrobial peptide derivative, effectively inhibit the growth of C. albicans. CT-K3K7 killed C. albicans cells in a dose-dependent manner, mainly by damaging the plasma membrane. CT-K3K7 could also disrupt the nucleus and interact with nucleic acid. Moreover, CT-K3K7 induced C. albicans cells necrosis via a reactive oxygen species (ROS)-related pathway. Furthermore, CT-K3K7 inhibited the hyphal and biofilm formation of C. albicans. In the mouse skin subcutaneous infection model, CT-K3K7 significantly prevented skin abscess formation and reduced the number of C. albicans cells recovered from the infection area. Taken together, CT-K3K7 has the potential to be a therapeutic for C. albicans skin infections.
Collapse
Affiliation(s)
- Zhongjie Li
- Institute of Genetics and Developmental Biology, Translational Medicine Institute, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710049, China.
- Microbial Pathogen and Anti-Infection Research Group, School of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471000, China.
- Henan Provincial Key Laboratory of Microbiota and Esophageal Cancer Prevention and Control, Henan University of Science and Technology, Luoyang, 471000, China.
| | - Yingbin Shui
- Microbial Pathogen and Anti-Infection Research Group, School of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471000, China
| | - Huayi Wang
- Microbial Pathogen and Anti-Infection Research Group, School of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471000, China
| | - Shasha Li
- Microbial Pathogen and Anti-Infection Research Group, School of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471000, China
| | - Bo Deng
- Microbial Pathogen and Anti-Infection Research Group, School of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471000, China
| | - Wenlu Zhang
- Microbial Pathogen and Anti-Infection Research Group, School of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471000, China
| | - Shegan Gao
- Henan Provincial Key Laboratory of Microbiota and Esophageal Cancer Prevention and Control, Henan University of Science and Technology, Luoyang, 471000, China.
- The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471000, China.
| | - Lingyu Zhao
- Institute of Genetics and Developmental Biology, Translational Medicine Institute, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
7
|
Simonovic D, Dey H, Johansen N, Anderssen T, Hansen IKØ, Devold H, Vasskog T, Blencke HM, Øyen FJ, Aarag Fredheim EG, Haug T, Strøm MB. Antimicrobial Activity of Short Analogues of the Marine Peptide EeCentrocin 1: Synthesis of Lipopeptides and Head-to-Tail Cyclic Peptides and Mechanism of Action Studies. J Pept Sci 2025; 31:e70025. [PMID: 40295171 DOI: 10.1002/psc.70025] [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: 09/26/2024] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
We have synthesised a series of 12-residue analogues of a previously reported lead peptide (P6) developed from the heavy chain of the marine peptide EeCentrocin 1, isolated from the sea urchin Echinus esculentus. We optimised the lead peptide by increasing its net positive charge, its lipophilicity through N-terminal fatty acid acylation or incorporation of a Trp residue, and by synthesising head-to-tail cyclic peptides under pseudo-high-dilution conditions. All peptides were screened for antimicrobial and antifungal activity, and toxicity was determined against human red blood cells. The two most potent peptide analogues were the linear peptide P6-W6R8 and its head-to-tail cyclic analogue cP6-W6R8 displaying minimum inhibitory concentrations of 0.4-6.6 μM against Gram-positive and Gram-negative bacteria and 6.2-13 μM against fungi. All peptides showed low haemolytic activity except for two of the lipopeptides, in which haemolytic activity correlated with increasing acyl chain length. Mode of action studies using bacterial biosensor strains revealed a membrane disruptive effect of both the linear and the cyclic peptide. Overall, the results of our study demonstrated that relatively simple structural modifications could be successfully employed in the development of potent antimicrobial lead peptides derived from marine natural products.
Collapse
Affiliation(s)
- Danijela Simonovic
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Hymonti Dey
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Natascha Johansen
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Trude Anderssen
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ida K Ø Hansen
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Hege Devold
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Terje Vasskog
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| | - Hans-Matti Blencke
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Frode Jacobsen Øyen
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | | | - Tor Haug
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT The Arctic University of Norway, Tromsø, Norway
| | - Morten B Strøm
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
8
|
Yao Y, Zhang D, Fan H, Wu T, Su Y, Bin Y. Prediction of Chemically Modified Antimicrobial Peptides and Their Sub-functional Activities Using Hybrid Features. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10575-6. [PMID: 40397268 DOI: 10.1007/s12602-025-10575-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Antimicrobial peptides (AMPs) demonstrate a broad spectrum of activities against various pathogens, thereby offering a promising strategy to mitigate the urgent challenge of antimicrobial resistance. Recent studies indicate that chemically modified AMPs (cmAMPs), which contain chemically modified amino acids, have the potential to alleviate the adverse effects commonly associated with conventional AMPs. Nevertheless, there remains a notable deficiency in computational methods specifically designed for the analysis and prediction of cmAMPs and their sub-function predictions. In this study, we proposed a two-layer model, termed as iCMAMP, aimed for the identification of cmAMPs and their sub-functional activities. The first layer, referred to as iCMAMP-1L, integrates three categories encompassing seven distinct groups of features, in conjunction with an ensemble method designed at enhancing predictive accuracy for cmAMPs. This ensemble approach effectively extracts relevant insights from a heterogeneous array of features sets while addressing potential dimensionality challenges. On the test dataset, iCMAMP-1L achieved an ACC of 0.934 and an MCC of 0.868, representing improvements of 3.4% and 6.8%, respectively, over AntiMPmod, which is the sole existing method for predicting cmAMPs. A comparative analysis between cmAMPs and their corresponding AMPs revealed that chemical modifications can significantly reduce hemolysis and toxicity associated with AMPs, while the functional characteristics of the peptides are primarily determined by their sequences. The second layer of our model, designated as iCMAMP-2L, employed a multi-label classification approach to predict the sub-functional activities of cmAMPs, with a specific focus on the dipeptide composition-based features. On the test dataset, iCMAMP-2L achieved an Accuracy of 0.390 and an Absolute true of 0.621. The data and Python code used in the iCMAMP model are available at https://github.com/swicher123/iCMAMP/tree/master .
Collapse
Affiliation(s)
- Yujie Yao
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, Anhui, China
| | - Daijun Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, Anhui, China
| | - Henghui Fan
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, Anhui, China
| | - Ting Wu
- Department of Infectious Diseases & Anhui Province Key Laboratory of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
- Institute of Bacterial Resistance & Anhui Center for Surveillance of Bacterial Resistance, Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Yansen Su
- School of Artificial Intelligence, Anhui University, Hefei, 230601, Anhui, China.
| | - Yannan Bin
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, Anhui, China.
| |
Collapse
|
9
|
Schöpf C, Knapp M, Scheler J, Coraça-Huber DC, Romanelli A, Ladurner P, Seybold AC, Binder U, Würzner R, Marx F. The antibacterial activity and therapeutic potential of the amphibian-derived peptide TB_KKG6K. mSphere 2025:e0101624. [PMID: 40387366 DOI: 10.1128/msphere.01016-24] [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: 12/06/2024] [Accepted: 04/13/2025] [Indexed: 05/20/2025] Open
Abstract
Antimicrobial peptides (AMPs) have great potential to be developed as topical treatments for microbial infections of the skin, including those caused by the gram-positive human pathogen Staphylococcus aureus. Among the AMPs, temporin B (TB) is of particular interest. This 13-amino-acid-long cationic peptide is secreted by the granular glands of the European frog Rana temporaria and represents a primary line of defense against invading pathogens. The objective of this study was to investigate the antibacterial efficacy and the mode of action of the synthetic TB analog, TB_KKG6K, in a drug-resistant clinical isolate of S. aureus and assess the peptide's tolerance and curative potential in an in vitro infection model using three-dimensional human epidermis equivalents (HEEs). The results revealed a high bactericidal efficacy of TB_KKG6K at low micromolar concentrations. The peptide perturbed the bacterial cell membrane integrity by permeabilization and depolarization. TB_KKG6K showed no toxicity in the invertebrate mini-host model Galleria mellonella and a high level of tolerance when topically applied in HEEs. Importantly, the therapeutic potential of TB_KKG6K was confirmed in HEEs infected with S. aureus. The topical application of TB_KKG6K significantly reduced the bacterial load and lowered the pro-inflammatory response in the infected HEEs. These findings reinforce the antibacterial potential and therapeutic efficacy of TB_KKG6K against S. aureus infection, particularly in the context of a cutaneous infection.IMPORTANCEThe emergence of multidrug-resistant bacteria has rendered the exploration of novel therapeutic treatment strategies a pivotal area of research. Among the most promising candidates are amphibian-derived antimicrobial peptides (AMPs), which are ideal for the development of novel drugs due to their multifaceted mode of action. Extensive studies have been conducted on these peptides over the last decade, resulting in the development of temporin B (TB) peptide analogs that have undergone modifications to their primary sequence. These modified analogs have demonstrated enhanced antibacterial and antifungal efficacy, while exhibiting reduced hemolytic activity. TB_KKG6K has the potential to be a promising candidate for topical treatments due to its small size and high antimicrobial activity against pathogens of the human skin. In particular, it demonstrated efficacy against Staphylococcus aureus, a skin commensal that can become an opportunistic pathogen, causing a range of infections from minor skin infections to life-threatening diseases such as bacteremia and sepsis.
Collapse
Affiliation(s)
- Cristina Schöpf
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Knapp
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Jakob Scheler
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Débora C Coraça-Huber
- Research Laboratory for Implant Associated Infections (BIOFILM LAB), Experimental Orthopaedics, University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Peter Ladurner
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Anna C Seybold
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Ulrike Binder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Reinhard Würzner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
10
|
Isa MA, Kappo AP. Machine learning-driven discovery of antimicrobial peptides targeting the GAPDH-TPI protein-protein interaction in Schistosoma mansoni for novel antischistosomal therapeutics. Comput Biol Chem 2025; 118:108501. [PMID: 40373611 DOI: 10.1016/j.compbiolchem.2025.108501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/24/2025] [Accepted: 05/06/2025] [Indexed: 05/17/2025]
Abstract
Schistosomiasis, caused by Schistosoma mansoni, remains a significant public health burden, particularly in endemic regions with limited access to effective treatment. The emergence of resistance to praziquantel necessitates the urgent discovery of novel therapeutic targets. This study explores the potential of antimicrobial peptides (AMPs) as inhibitors of the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and triose phosphate isomerase (TPI) protein-protein interaction (PPI) in S. mansoni, a crucial glycolytic pathway component essential for parasite survival. A machine learning-driven approach was employed to filter 3306 AMPs from the Antimicrobial Peptide Database (APD) based on physicochemical properties and predicted binding affinities. Eighteen peptides were selected based on desirable physicochemical attributes and further subjected to molecular docking using HADDOCK 2.4. The results identified AP02590 (-103.5 ± 2.7 kcal/mol) and AP02754 (-87.8 ± 1.0 kcal/mol) as the most promising inhibitors, exhibiting strong binding affinities and stable complex formation compared to the native GAPDH-TPI complex (-77.8 ± 17.2 kcal/mol). Molecular dynamics (MD) simulations confirmed the stability of these complexes, with lower root mean square deviation (RMSD) values (AP02590: ∼2.5 Å, AP02754: ∼3.0 Å) and reduced root mean square fluctuation (RMSF) of key interacting residues. Radius of gyration (Rg) analysis further indicated compact structural stability. MMGBSA analysis validated these findings, showing favourable binding free energies for AP02590 (-50.80 ± 0.90 kcal/mol) and AP02754 (-46.31 ± 0.83 kcal/mol), reinforcing their potential as lead compounds for antischistosomal drug development. These findings provide a foundation for further experimental validation of peptide-based inhibitors targeting metabolic pathways in S. mansoni.
Collapse
Affiliation(s)
- Mustafa Alhaji Isa
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg 2006, South Africa.
| | - Abidemi Paul Kappo
- Molecular Biophysics and Structural Biology (MBSB) Group, Department of Biochemistry, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg 2006, South Africa
| |
Collapse
|
11
|
Tian L, Wang T, Luan L, Meng Z, Han J, Zhao C, Xu Y, Zeng C, Ye W, Jiang S, Zhang L, Yin J, Meng Q, Li S. Terminally Symmetric β-Turn Peptides for Multidrug-Resistant Bacterial Infections. J Med Chem 2025; 68:9341-9356. [PMID: 40254841 DOI: 10.1021/acs.jmedchem.4c03057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Antimicrobial peptides (AMPs) are considered promising agents to solve the problem of antibiotic resistance due to their unique membrane-disruption mechanism. In this research, de novo terminally symmetric β-turn AMPs were designed by combining the β-turn sequences derived from Tritrpticin with alternately arranged cationic and hydrophobic amino acid sequences. The structure-activity relationship of the peptides was studied. Among the designed peptides, P-07 (KIKIKPWWWPKIKIK-NH2) exhibited potent antimicrobial activity against all the tested bacterial strains, showing the highest bacterial selectivity, relatively low cytotoxicity, high bactericidal efficiency, and low potential to induce bacterial resistance. The antimicrobial mechanisms of P-07 involving membrane-disruption and lipopolysaccharide-binding were proven. Moreover, the in vivo studies confirmed the wound-healing ability of P-07 using a mice bacteria-infected full-thickness wound model. Taken together, P-07 showed great promise in the treatment of multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Long Tian
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Taoran Wang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Liang Luan
- Department of Laboratory Medical Center, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Zhao Meng
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jiaqi Han
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Chunhui Zhao
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yijie Xu
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Chunlan Zeng
- 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
| | - Shuyuan Jiang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Li Zhang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Jiye Yin
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Qingbin Meng
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Song Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
12
|
Seghetti F, Ocello R, Bisi A, Masetti M, Gobbi S, Falchi F, Gentilomi GA, Bonvicini F, Belluti F. Alkyl Tail Variation on Chalcone-Based Quaternary Pyridinium Salts as Rule-of-Thumb for Antimicrobial Activity. Arch Pharm (Weinheim) 2025; 358:e70003. [PMID: 40350787 PMCID: PMC12066977 DOI: 10.1002/ardp.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/29/2025] [Accepted: 04/13/2025] [Indexed: 05/14/2025]
Abstract
Aiming at developing a new class of quaternary pyridinium salts, the lead compound 1, characterized by a pyridine-3-yl chalcone framework, was rationally modified by inserting alkyl functions varying from 6 to 18 carbon units. Among the set, some valuable lead compounds were identified. Derivatives 4-6 were primarily active against Staphylococcus aureus and Candida albicans, respectively (MIC = 1.56 and 3.125 μM). In comparison, analogs 4 and 5 showed significant activities against Escherichia coli (MIC = 6.25 μM). Interestingly, the antimicrobial property of compounds 4-6, as well as their antibiofilm activity, occurred at lower concentrations than their cyto- and erythrocyte toxicities, thus ensuring a favorable safety profile. Structure-activity relationship analysis highlighted the critical role of the alkyl tail length in the antimicrobial activity, and optimal results were observed for moieties ranging from 10 to 14 carbon units. Molecular dynamics studies performed on 2 and 5 by modeling them on Gram-positive and Gram-negative membranes showed that the derivatives, upon diffusing across periodic boundary conditions, were able to intercalate into the microbial membranes. The difference in diffusion rates provides useful information to support the diverse antimicrobial potencies of the newly designed quaternary pyridinium salt.
Collapse
Affiliation(s)
- Francesca Seghetti
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
| | - Riccardo Ocello
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
- Computational and Chemical BiologyItalian Institute of Technology IITGenoaItaly
| | - Alessandra Bisi
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
| | - Matteo Masetti
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
| | - Silvia Gobbi
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
| | - Federico Falchi
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
- Computational and Chemical BiologyItalian Institute of Technology IITGenoaItaly
| | - Giovanna Angela Gentilomi
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
- Microbiology UnitIRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
| | - Francesca Bonvicini
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
| | - Federica Belluti
- Department of Pharmacy and BiotechnologyAlma Mater Studiorum‐University of BolognaBolognaItaly
| |
Collapse
|
13
|
Zhao H, Sun S, Ding X, Zhang Y, Li B, Wang S, Guo G, Zhang J. Activity and Safety Optimization of Mesoricin: A Dual-Domain Antifungal Peptide from Mesorhizobium sp. J Med Chem 2025; 68:8226-8243. [PMID: 40198836 DOI: 10.1021/acs.jmedchem.4c02917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Cryptococcus neoformans infections pose a significant global health threat. This study introduces mesoricin, a novel dual-domain antimicrobial peptide (AMP) scaffold derived from Mesorhizobium sp. identified using an in silico quantitative antifungal activity index (AFI). The peptide structure comprises an α-helix domain, which disrupts microbial membranes but exhibits highly hemolytic activity, and a β-sheet domain, which targets intracellular energy metabolism and resilient pathways. Rational design through α-helix domain removal and AFI-guided mutations yielded a mesoricin variant with enhanced antifungal activity and reduced cytotoxicity. The optimized mesoricin exhibited broad-spectrum antifungal activity against various Cryptococcus and Candida species (MIC 8-16 μg/mL) while maintaining high biosafety (IC50 > 128 μg/mL against human cell lines). Particularly, the variant demonstrated enhanced fungicidal effects at sub-MIC levels and superior biofilm control capabilities compared to the prototype peptide. These findings highlight mesoricins as a promising scaffold for AMP development targeting Cryptococcus infections.
Collapse
Affiliation(s)
- Hongwei Zhao
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education/Translational Medicine Research Center/Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Simei Sun
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education/Translational Medicine Research Center/Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Xiang Ding
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education/Translational Medicine Research Center/Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Yiling Zhang
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education/Translational Medicine Research Center/Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Boyan Li
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education/Translational Medicine Research Center/Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Shuyu Wang
- Cancer Molecular Diagnostics Core, Tianjin Medical University, Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin 300060, China
| | - Guo Guo
- The Key and Characteristic Laboratory of Modern Pathogen Biology/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Jin Zhang
- School of Public Health/Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education/Translational Medicine Research Center/Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| |
Collapse
|
14
|
Deschner F, Mostert D, Daniel JM, Voltz A, Schneider DC, Khangholi N, Bartel J, Pessanha de Carvalho L, Brauer M, Gorelik TE, Kleeberg C, Risch T, Haeckl FPJ, Herraiz Benítez L, Andreas A, Kany AM, Jézéquel G, Hofer W, Müsken M, Held J, Bischoff M, Seemann R, Brötz-Oesterhelt H, Schneider T, Sieber S, Müller R, Herrmann J. Natural products chlorotonils exert a complex antibacterial mechanism and address multiple targets. Cell Chem Biol 2025; 32:586-602.e15. [PMID: 40203831 DOI: 10.1016/j.chembiol.2025.03.005] [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: 08/21/2024] [Revised: 12/12/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
Antimicrobial resistance is a threat to human health rendering current first-line antibiotics ineffective. New agents overcoming resistance mechanisms are urgently needed to guarantee successful treatment of human disease in the future. Chlorotonils, a natural product class with yet unknown mode of action, were shown to have broad-spectrum activity against multi-resistant Gram-positive bacteria and the malaria parasite Plasmodium falciparum, with promising activity and safety in murine infection models. Here, we report that chlorotonils can target the cell membrane, cell wall, and protein biosynthesis. They can be characterized by a rapid onset of action via interference with ion homeostasis leading to membrane depolarization, however, without inducing severe barrier failure or cellular lysis. Further characterization confirmed binding of chlorotonils to bacterial membrane lipids eventually leading to uncontrolled potassium transport. Additionally, we identified functional inhibition of the peptidoglycan biosynthesis protein YbjG and methionine aminopeptidase MetAP as secondary targets of chlorotonils.
Collapse
Affiliation(s)
- Felix Deschner
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Dietrich Mostert
- Center for Functional Protein Assemblies, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Jan-Martin Daniel
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, 53127 Bonn, Germany
| | - Alexander Voltz
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Dana Carina Schneider
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany
| | - Navid Khangholi
- Experimental Physics and Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| | - Jürgen Bartel
- Department of Microbial Proteomics, Institute of Microbiology, University of Greifswald, 17489 Greifswald, Germany
| | | | - Madita Brauer
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, 17489 Greifswald, Germany
| | - Tatiana E Gorelik
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Institute of Inorganic and Analytical Chemistry, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany; Ernst Ruska-Centre for Microscopy and Spectroscopy with Electrons, Forschungszentrum Jülich GmbH, Jülich 52428, Germany
| | - Christian Kleeberg
- Institute for Inorganic and Analytical Chemistry, Technical University of Braunschweig, 38106 Braunschweig, Germany
| | - Timo Risch
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - F P Jake Haeckl
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Laura Herraiz Benítez
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Anastasia Andreas
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Andreas Martin Kany
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Gwenaëlle Jézéquel
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Walter Hofer
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Mathias Müsken
- Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Jana Held
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute of Tropical Medicine, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Markus Bischoff
- Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Institute for Medical Microbiology and Hygiene, Saarland University, 66421 Homburg, Germany
| | - Ralf Seemann
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany; Cluster or Excellence "Controlling Microbes to Fight Infections", Tübingen, Germany
| | - Tanja Schneider
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, 53127 Bonn, Germany
| | - Stephan Sieber
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Center for Functional Protein Assemblies, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Rolf Müller
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Jennifer Herrmann
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany.
| |
Collapse
|
15
|
Golonka I, Łukasiewicz IW, Sebastiańczyk A, Greber KE, Sawicki W, Musiał W. The Role of Temperature and Subphase Components in Shaping Selected Physicochemical Properties of the Phosphatidylinositol Monolayer. Int J Mol Sci 2025; 26:3472. [PMID: 40331933 PMCID: PMC12026758 DOI: 10.3390/ijms26083472] [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: 03/08/2025] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Acne vulgaris is one of the most common skin diseases, and its development is closely linked to the overgrowth of the bacterium Cutibacterium acnes. More than half of the strains of this bacterium are resistant to antibiotics, which has prompted scientists to look for alternatives, such as antibacterial peptides, that can replace traditional drugs. Due to its antioxidant properties, ascorbic acid may be a promising ally in the treatment of acne. The aim of our study was to evaluate the effect of peptide (KWK)2-KWWW-NH2(P5) in the presence of ascorbic acid (AA) and its derivative (3-O-ethyl-L-ascorbic acid, EAA) on the stability and organization of phosphatidylinositol monolayers (PI) at temperatures of 25-35 °C. This study showed that the monolayers were in the expanded liquid state (35.28-49.95 mN/m) or in the transition between the expanded liquid and condensed phases (51.50-57.49 mN/m). Compression and decompression isotherms indicated the highest flexibility of the PI + P5 system, where the compression reversibility coefficient of isotherm values ranged from 80.59% to 97.77% and increased for each loop with increasing temperature. At 35 °C, the surface pressure of the monolayer in the PI + P5, PI + P5 + AA and PI + P5 + EAA systems changed less with time.
Collapse
Affiliation(s)
- Iwona Golonka
- Department of Physical Chemistry and Biophysics, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (I.G.); (I.W.Ł.); (A.S.)
| | - Izabela W. Łukasiewicz
- Department of Physical Chemistry and Biophysics, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (I.G.); (I.W.Ł.); (A.S.)
| | - Aleksandra Sebastiańczyk
- Department of Physical Chemistry and Biophysics, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (I.G.); (I.W.Ł.); (A.S.)
| | - Katarzyna E. Greber
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (K.E.G.); (W.S.)
| | - Wiesław Sawicki
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (K.E.G.); (W.S.)
| | - Witold Musiał
- Department of Physical Chemistry and Biophysics, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (I.G.); (I.W.Ł.); (A.S.)
| |
Collapse
|
16
|
Zou J, Wang J, Gao L, Xue W, Zhu J, Zhang Y, Gou S, Liu H, Zhong C, Ni J. Ultra-short lipopeptides containing d-amino acid exhibiting excellent stability and antibacterial activity against gram-positive bacteria. Eur J Med Chem 2025; 287:117341. [PMID: 39908797 DOI: 10.1016/j.ejmech.2025.117341] [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: 12/11/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
As novel antibacterial agents, antimicrobial peptides (AMPs) possess broad-spectrum antibacterial activity and low drug resistance, holding significant development potential. Nevertheless, the stability of AMPs significantly restricts their application. In light of this, we synthesized a series of ultra-short lipopeptides using d-amino acid substitution to enhance the stability of ultra-short lipopeptide C12-RRW-NH2 that was selected from our previous research while maintaining its antibacterial activity against gram-positive bacteria. Amongst, the ultra-short lipopeptide Lip7 (C12-rrw-NH2) with full d-amino acid demonstrated outstanding stability in protease, serum, and salt ion environments. It exerted excellent antibacterial activity against gram-positive bacteria, especially against methicillin-resistant Staphylococcus aureus (MRSA). Meanwhile, Lip7 presented a low propensity to develop bacterial resistance with potential for combination therapy with conventional antibiotics. Studies on its antibacterial mechanism revealed that Lip7 could rapidly depolarize the bacterial cytoplasmic membrane, disrupt the integrity of the bacterial membrane, lead to leakage of nucleic acid and protein, promote the generation of reactive oxygen species, and ultimately result in bacterial death. Additionally, Lip7 also exhibited therapeutic potential in both local and systemic MRSA-infected mice models with better safety in vivo. These findings highlighted that Lip7 is an ideal novel antibacterial alternative to offer guiding schemes for developing high-stability antimicrobial peptides to fight multidrug-resistant gram-bacteria.
Collapse
Affiliation(s)
- Jing Zou
- 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
| | - Jiahui Wang
- 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
| | - Luyang Gao
- 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
| | - Wenjing Xue
- 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
| | - Jingyi Zhu
- 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
| | - 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
| | - 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
| | - 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
|
17
|
Zhao C, Zhao T, Liu Y, Xia X, Li X. Role of ygaD in mediating polymyxin B resistance in Bacillus subtilis via efflux mechanisms. Microb Pathog 2025; 201:107345. [PMID: 39924090 DOI: 10.1016/j.micpath.2025.107345] [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: 09/13/2024] [Revised: 01/02/2025] [Accepted: 01/27/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVE The aim of this study is to analyze the resistance function and mechanism of the putative ATP-binding cassette (ABC) transporter YgaD against polymyxin B in Bacillus subtilis. METHODS The interaction between the YgaD protein and the antimicrobial peptide polymyxin B was initially assessed using molecular docking and molecular dynamics simulation. Subsequently, resistance assays and intracellular polymyxin B content measurements were conducted on Bacillus subtilis with a knockout of the ygaD gene and on Escherichia coli with heterologous expression of the ygaD gene to validate the resistance function mediated by the YgaD protein and deduce its potential resistance mechanism. RESULTS The results demonstrated that the YgaD protein could form stable complexes with polymyxin B and facilitated its efflux from bacterial cells, thereby reducing its intracellular accumulation and conferring resistance to polymyxin B. CONCLUSION Our study revealed that YgaD regulates polymyxin B resistance in Bacillus subtilis through an efflux mechanism. These findings contribute to the understanding of microbial resistance mechanisms against antimicrobial peptides and provide a theoretical basis for the future design and development of antimicrobial drugs.
Collapse
Affiliation(s)
- Chongyi Zhao
- Department of Gynecology, The First People's Hospital of Yunnan Province, (the Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan Province, China
| | - Ting Zhao
- Department of Gynecology, The First People's Hospital of Yunnan Province, (the Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan Province, China
| | - Ying Liu
- Medical School, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, Yunnan Province, China.
| | - Xiao Li
- Department of Gynecology, The First People's Hospital of Yunnan Province, (the Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan Province, China.
| |
Collapse
|
18
|
Fernandez-Sánchez F, Flores-Ávila J, García HS, Mixcoha E, Balleza D. Molecular dynamics study of the helix-to-disorder transition in short antimicrobial peptides from Urodacus yaschenkoi. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2025:10.1007/s00249-025-01740-4. [PMID: 40137971 DOI: 10.1007/s00249-025-01740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/27/2024] [Accepted: 01/14/2025] [Indexed: 03/29/2025]
Abstract
The bioactivity of the short antimicrobial peptides (ssAMPs) UyCT1, CT2, CT3, CT5, Uy17, Uy192, and Uy234 from the scorpion Urodacus yaschenkoi has been well-characterized. The antagonistic effect reported in those studies on some clinical isolates of pathogenic bacteria, including Staphylococcus aureus, Klebsiella pneumoniae, and Escherichia coli was studied with an in silico approach to contrast their bioactivity in molecular terms. The peptides were modeled by generating high-quality structures with AlphaFold2, properly validated, and subjected to dynamic simulations in aqueous systems with the Gromos 43a1 and Charmm 36 force fields. Our analysis indicates that the degree of helicity of these peptides is closely linked to their composition and several physicochemical factors such as the hydrophobicity index, electrostatic potential, intrinsic flexibility, and dipole moment. We also found interesting parallels between the degree of order mentioned and the potency of each peptide with previously studied bacterial strains, specifically S. aureus. We analyzed in more detail of two specific peptides, UyCT1 and UyCT2, whose sequences are almost identical, except for the presence of a G-cap in the former. This subtle difference has a decisive impact on the conformational dynamics of these peptides, making the UyCT2 peptide more prone to disorder and the UyCT1 peptide more stable through the formation of multiple H-bonds. This analysis, based on an exhaustive characterization of the physicochemical properties of these ssAMPs, together with the determination of their conformational dynamics and the correlation with experimental data, could be the basis for the design and optimization of new drugs based on natural peptides found in scorpion venoms.
Collapse
Affiliation(s)
- Flora Fernandez-Sánchez
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico
| | - Jenny Flores-Ávila
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico
| | - Hugo S García
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico
| | - Edgar Mixcoha
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico
| | - Daniel Balleza
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico.
| |
Collapse
|
19
|
Aguilar S, Moreira D, Pereira Lourenço AL, Wilke N, Crosio MA, Vasconcelos A, Barbosa EA, Bispo ECI, Saldanha-Araujo F, Ramada MHS, Escobar FM, Torres CV, Leite JRSA, Marani MM. Enhancing Antimicrobial Peptides from Frog Skin: A Rational Approach. Biomolecules 2025; 15:449. [PMID: 40149984 PMCID: PMC11939955 DOI: 10.3390/biom15030449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Antimicrobial resistance is a global health threat, which has been worsened by the slow development of new antibiotics. The rational design of natural-derived antimicrobial peptides (AMPs) offers a promising alternative for enhancing the efficacy of AMPs and accelerating drug discovery. This paper describes the rational design of improved peptide derivatives starting from hylin-Pul3, a peptide previously isolated from the frog Boana pulchella, by optimizing its hydrophobicity, cationicity, and amphipathicity. In silico screening identified six promising candidates: dHP3-31, dHP3-50, dHP3-50.137, dHP3-50.190, dHP3-84, and dHP3-84.39. These derivatives exhibited enhanced activity against Gram-negative bacteria, emphasizing the role of cationicity and the strategic arginine incorporation. Hemolytic assays revealed the derivatives' improved selectivity, particularly for the derivatives with "imperfect amphipathicity". In fibroblast assays, dHP3-84 was well-tolerated, while dHP3-84.39 promoted cell proliferation. Antioxidant assays (ABTS assays) highlighted the Trp-containing derivatives' (dHP3-50.137, dHP3-31) significant activity. The lipid membrane interaction studies showed that hylin-Pul3 disrupts membranes directly, while dHP3-84.39, dHP3-50, and dHP3-50.137 promote vesicle aggregation. Conversely, dHP3-84 did not induce membrane disruption or aggregation, suggesting an intracellular mode of action. Machine learning models were effective in predicting bioactivity, as these predicted AMPs showed enhanced selectivity and potency. Among them, dHP3-84 demonstrated broad-spectrum potential. These findings highlight the value of rational design, in silico screening, and structure-activity studies in optimizing AMPs for therapeutic applications.
Collapse
Affiliation(s)
- Silvana Aguilar
- IPEEC-CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, Puerto Madryn U9120ACD, Argentina;
| | - Daniel Moreira
- Research Center in Morphology and Applied Immunology, NuPMIA, Faculty of Medicine, University of Brasília, UnB, Brasília 70910-900, DF, Brazil; (D.M.); (A.V.); (E.A.B.); (J.R.S.A.L.)
| | - Ana Laura Pereira Lourenço
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Taguatinga 71966-700, DF, Brazil; (A.L.P.L.); (M.H.S.R.)
| | - Natalia Wilke
- Facultad de Ciencias Químicas, Departamento de Química Biológica Ranwel Caputto, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (N.W.); (M.A.C.)
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - Matías A. Crosio
- Facultad de Ciencias Químicas, Departamento de Química Biológica Ranwel Caputto, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina; (N.W.); (M.A.C.)
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - Andreanne Vasconcelos
- Research Center in Morphology and Applied Immunology, NuPMIA, Faculty of Medicine, University of Brasília, UnB, Brasília 70910-900, DF, Brazil; (D.M.); (A.V.); (E.A.B.); (J.R.S.A.L.)
- University Center of the Federal District, UDF, Brasília 70390-045, DF, Brazil
| | - Eder Alves Barbosa
- Research Center in Morphology and Applied Immunology, NuPMIA, Faculty of Medicine, University of Brasília, UnB, Brasília 70910-900, DF, Brazil; (D.M.); (A.V.); (E.A.B.); (J.R.S.A.L.)
- Laboratory of Synthesis and Analysis of Biomolecules, LSAB, Institute of Chemistry, IQ, University of Brasília, UnB, Brasília 70910-900, DF, Brazil
| | - Elizabete C. I. Bispo
- Laboratory of Hematology and Stem Cells, Faculty of Health Sciences, University of Brasília, UnB, Brasília 70910-900, DF, Brazil; (E.C.I.B.); (F.S.-A.)
| | - Felipe Saldanha-Araujo
- Laboratory of Hematology and Stem Cells, Faculty of Health Sciences, University of Brasília, UnB, Brasília 70910-900, DF, Brazil; (E.C.I.B.); (F.S.-A.)
| | - Marcelo H. S. Ramada
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Taguatinga 71966-700, DF, Brazil; (A.L.P.L.); (M.H.S.R.)
| | - Franco M. Escobar
- Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Ruta 36, Km 601, Río Cuarto 5800, Argentina; (F.M.E.); (C.V.T.)
| | - Cristina V. Torres
- Departamento de Microbiología e Inmunología, Universidad Nacional de Río Cuarto, Ruta 36, Km 601, Río Cuarto 5800, Argentina; (F.M.E.); (C.V.T.)
| | - José R. S. A. Leite
- Research Center in Morphology and Applied Immunology, NuPMIA, Faculty of Medicine, University of Brasília, UnB, Brasília 70910-900, DF, Brazil; (D.M.); (A.V.); (E.A.B.); (J.R.S.A.L.)
| | - Mariela M. Marani
- IPEEC-CONICET, Consejo Nacional de Investigaciones Científicas y Técnicas, Puerto Madryn U9120ACD, Argentina;
| |
Collapse
|
20
|
Su SS, An CN, Lin GJ, Li HQ, Yin F, Li XL, Wang KR. Amphiphilic lysine-based glycopeptides exert antibacterial effects on Pseudomonas aeruginosa. J Mater Chem B 2025; 13:3945-3951. [PMID: 40017413 DOI: 10.1039/d4tb02771j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
The development of new antibiotics with novel antimicrobial mechanisms and strategies has attracted considerable interest. Herein, three amphiphilic lysine-based glycopeptides (SA-l-Gal, OA-l-Gal and LOA-l-Gal) were developed, and their antibacterial activity and inhibition effects on biofilm formation on P. aeruginosa were studied. SA-l-Gal with a stearic acid group modification exhibited better antibacterial effects than the other lysine-based glycopeptides by damaging the bacterial membrane. Furthermore, SA-l-Gal could effectively reduce inflammation factor expression, enhance the formation of new blood vessels, and promote the healing of P. aeruginosa-infected mouse wounds. This result provides insights into the development of glycomimetic drugs against P. aeruginosa infection.
Collapse
Affiliation(s)
- Shan-Shan Su
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Chao-Na An
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Gao-Juan Lin
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Hai-Qing Li
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Fangqian Yin
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
- College of Chemical Engineering & Material, Hebei Key Laboratory of Heterocyclic Compounds, Handan key laboratory of organic small molecule materials, Handan University, Handan 056005, P. R. China
| | - Xiao-Liu Li
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| |
Collapse
|
21
|
Zheng S, Tu Y, Li B, Qu G, Li A, Peng X, Li S, Shao C. Antimicrobial peptide biological activity, delivery systems and clinical translation status and challenges. J Transl Med 2025; 23:292. [PMID: 40055730 PMCID: PMC11887333 DOI: 10.1186/s12967-025-06321-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
Antibiotic resistance is currently one of the most significant threats to global public health and safety. And studies have found that over the next 25 years, 39 million people will die directly and 169 million indirectly due to antibiotic-resistant diseases. Consequently, the development of new types of antimicrobial drugs is urgently needed. Antimicrobial peptides (AMPs) constitute an essential component of the innate immune response in all organisms. They exhibit a distinctive mechanism of action that endows them with a broad spectrum of biological activities, including antimicrobial, antibiofilm, antiviral, and anti-inflammatory effects. However, AMPs also present certain limitations, such as cytotoxicity, susceptibility to protein hydrolysis, and poor pharmacokinetic properties, which have impeded their clinical application. The development of delivery systems can address these challenges by modifying AMP delivery and enabling precise, controlled release at the site of infection or disease. This review offers a comprehensive analysis of the mechanisms of action and biological advantages of AMPs. and systematically evaluate how emerging drug delivery systems, such as nanoparticles and hydrogels, enhance the stability and bioavailability of AMPs, discussing both their strengths and limitations. Moreover, unlike previous reviews, this review highlight the most recent clinically approved AMP-based drugs and those currently in development, emphasizing the key challenges in translating these drugs into clinical practice. With these perspectives, it is hoped that this review will provide some insights into overcoming translational barriers and advancing AMPs drugs into clinical practice.
Collapse
Affiliation(s)
- Sainan Zheng
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, 325604, People's Republic of China
| | - Yuhan Tu
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, 325604, People's Republic of China
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China
| | - Bin Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China
| | - Gaoer Qu
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, 325604, People's Republic of China
| | - Anqi Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China
| | - Xuemei Peng
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China
| | - Shijun Li
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035, People's Republic of China.
| | - Chuanfeng Shao
- Department of Pharmacy, Yueqing Third People's Hospital, Wenzhou, 325604, People's Republic of China.
| |
Collapse
|
22
|
Rogers NJ, Postings ML, Dixon AM, Moat J, Shreeve G, Stuart L, Waterfield NR, Scott P. Membrane lipid composition directs the cellular selectivity of antimicrobial metallohelices. RSC Med Chem 2025:d4md00973h. [PMID: 40110349 PMCID: PMC11917443 DOI: 10.1039/d4md00973h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025] Open
Abstract
Two enantiomeric pairs of iron(ii) metallohelices, available as water-soluble, stable, and optically pure bimetallic complexes, differ principally in the length of the central hydrophobic region between two cationic domains, and have distinct activity and cell selectivity profiles against Gram-positive and Gram-negative microbes. The effects of dose concentration and temperature on levels of intracellular accumulation in E. coli and S. aureus, studied via isotopic labelling, indicate that the metallohelices enter the microbial cells via passive diffusion, whereupon (as previously determined) they act on intracellular targets. Whilst the metallohelices with the shorter central hydrophobic regions accumulate less readily than those with the longer hydrophobic bridge in both E. coli and S. aureus cells when incubated at the same concentration, an order of magnitude less is actually required per cell to inhibit growth in E. coli, hence they are more active. Furthermore, these more Gram-negative active compounds (with the shorter central hydrophobic region) are less toxic towards human APRE-19 mammalian cells and equine red blood cells. We hypothesise that these cell selectivities originate from the membrane composition. Dynamic light scattering and zeta potential measurements demonstrate that the more lipophilic metallohelices interact more strongly with the membrane-mimetic vesicles, notably in the charge-neutral mammalian model; thus the selectivity is not simply a result of electrostatic effects. For the less lipophilic metallohelices we observe that the binding affinity with the E. coli model vesicles is greater than with S. aureus vesicles, despite the lower negative surface charge, and this corresponds with the cellular accumulation data and the measured MICs. Specifically, the presence of membrane phosphatidylethanolamine (POPE) significantly increases the binding affinity of these metallohelices, and we postulate that a high proportion of such conical, non-lamellar phospholipids is important for metallohelix transport across the membrane. The metallohelices with the shorter hydrophobic bridge studied have a balance of charge and lipophilicity which allows selective cell entry in E. coli over mammalian cells, while the more lipophilic metallohelices are membrane promiscuous and unselective.
Collapse
Affiliation(s)
- Nicola J Rogers
- Department of Chemistry, Hong Kong Baptist University Kowloon Tong Hong Kong
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - Miles L Postings
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - Ann M Dixon
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - John Moat
- School of Life Sciences, University of Warwick Gibbet Hill Campus Coventry UK
| | - Georgia Shreeve
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | - Louise Stuart
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| | | | - Peter Scott
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
| |
Collapse
|
23
|
Erriah P, Puan SL, Yahaya NM, Wan Ahmad Kamil WNI, Amin Nordin S, Muhamad A, Sabri S. Harnessing bacterial antimicrobial peptides: a comprehensive review on properties, mechanisms, applications, and challenges in combating antimicrobial resistance. J Appl Microbiol 2025; 136:lxae290. [PMID: 40036746 DOI: 10.1093/jambio/lxae290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 03/06/2025]
Abstract
Antimicrobial resistance (AMR) is a significant global health concern due to the persistence of pathogens and the emergence of resistance in bacterial infections. Bacterial-derived antimicrobial peptides (BAMPs) have emerged as a promising strategy to combat these challenges. Known for their diversity and multifaceted nature, BAMPs are notable bioactive agents that exhibit potent antimicrobial activities against various pathogens. This review explores the intricate properties and underlying mechanisms of BAMPs, emphasizing their diverse applications in addressing AMR. Additionally, the review investigates the mechanisms, analyses the challenges in utilizing BAMPs effectively, and examines their potential applications and associated deployment challenges providing comprehensive insights into how BAMPs can be harnessed to combat AMR across different domains. The significance of this review lies in highlighting the potential of BAMPs as transformative agents in combating AMR, offering sustainable and eco-friendly solutions to this pressing global health challenge.
Collapse
Affiliation(s)
- Pirasannah Erriah
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor, Malaysia
| | - Sheau Ling Puan
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor, Malaysia
| | - Normi Mohd Yahaya
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor, Malaysia
| | - Wan Nur Ismah Wan Ahmad Kamil
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor, Malaysia
| | - Syafinaz Amin Nordin
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor, Malaysia
| | - Azira Muhamad
- National Institutes of Biotechnology Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Suriana Sabri
- Enzyme and Microbial Technology Research Centre, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor, Malaysia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
24
|
Anahid M, Mahnam K, Saffar B. Improving the antimicrobial activity of RP9 peptide through theoretical and experimental investigation. Biochem Biophys Rep 2025; 41:101953. [PMID: 40034258 PMCID: PMC11872504 DOI: 10.1016/j.bbrep.2025.101953] [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: 11/12/2024] [Revised: 01/30/2025] [Accepted: 02/09/2025] [Indexed: 03/05/2025] Open
Abstract
Future threats to humanity may stem from the rise of antimicrobial resistance, which has compromised the effectiveness of existing antibiotics. Antimicrobial peptides possess the ability to directly eliminate pathogens and cancer cells, generally without the development of resistance. Among these peptides is RP9 (RGSALTHLP), derived from the white blood cells of crocodiles. In this research, three mutations were initially designed: LR-mut (RGSALTHLR), KR-mut (RGSAKTHLR), and WP-mut (RGSAWTHLP). The physicochemical characteristics of these peptides were assessed, revealing that KR-mut exhibited the most favorable biophysical properties. Subsequently, twenty molecular dynamics simulations were conducted for all peptides in pure water and at four different octanol concentrations (30 %, 50 %, 70 %, and 100 %) to evaluate their biophysical attributes. The findings from the 4000 ns molecular dynamics simulations revealed that the KR-mut exhibited reduced values of RMSD, the radius of gyration, solvent accessible surface area, and RMSF, while simultaneously showing an increased number of hydrogen bonds and interactions with water molecules. This peptide also showed the lowest free energy of solvation and the highest solubility across various octanol concentrations compared to the other peptides. The results obtained from the biophysical assessments and molecular dynamics simulations were consistent, resulting in the conclusion that KR-mut is expected to exhibit superior antibacterial activity compared to both the other mutated peptides and the wild type peptides. These theoretical findings were validated through experimental minimum inhibitory concentration (MIC) tests on gram-negative Escherichia coli and gram-positive Staphylococcus aureus. The outcomes of this study suggest that molecular dynamics simulations can effectively predict changes in the bactericidal efficacy of peptides at varying octanol concentrations, potentially enhancing the speed and efficiency of antimicrobial peptide design while reducing associated costs.
Collapse
Affiliation(s)
- Mahya Anahid
- Department of Genetics, Faculty of Science, Shahrekord University, Shahrekord, Iran
| | - Karim Mahnam
- Department of Biology, Faculty of Science, Shahrekord University, Shahrekord, Iran
- Biotechnology Research Institute, Shahrekord University, Shahrekord, Iran
| | - Behnaz Saffar
- Department of Genetics, Faculty of Science, Shahrekord University, Shahrekord, Iran
- Biotechnology Research Institute, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
25
|
Williams MD, Smith L. Streptococcus salivarius and Ligilactobacillus salivarius: Paragons of Probiotic Potential and Reservoirs of Novel Antimicrobials. Microorganisms 2025; 13:555. [PMID: 40142448 PMCID: PMC11944278 DOI: 10.3390/microorganisms13030555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
This review highlights several basic problems associated with bacterial drug resistance, including the decreasing efficacy of commercially available antimicrobials as well as the related problem of microbiome irregularity and dysbiosis. The article explains that this present situation is addressable through LAB species, such as Streptococcus salivarius and Ligilactobacillus salivarius, which are well established synthesizers of both broad- and narrow-spectrum antimicrobials. The sheer number of antimicrobials produced by LAB species and the breadth of their biological effects, both in terms of their bacteriostatic/bactericidal abilities and their immunomodulation, make them prime candidates for new probiotics and antibiotics. Given the ease with which several of the molecules can be biochemically engineered and the fact that many of these compounds target evolutionarily constrained target sites, it seems apparent that these compounds and their producing organisms ought to be looked at as the next generation of robust dual action symbiotic drugs.
Collapse
Affiliation(s)
| | - Leif Smith
- Department of Biology, Texas A&M University, College Station, TX 77843, USA;
- Antimicrobial Division, Sano Chemicals Inc., Bryan, TX 77808, USA
| |
Collapse
|
26
|
Yun S, Kang SH, Ryu J, Kim K, Lee KY, Lee JJ, Hong JY, Son GH. The Role of Beta-Defensin 2 in Preventing Preterm Birth with Chorioamnionitis: Insights into Inflammatory Responses and Epithelial Barrier Protection. Int J Mol Sci 2025; 26:2127. [PMID: 40076749 PMCID: PMC11900102 DOI: 10.3390/ijms26052127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Antimicrobial peptides, such as beta-defensin 2 (BD2), are vital in controlling infections and immune responses. In this study, we investigated the expression and role of BD2 in the amniotic membrane and human amniotic epithelial cells (hAECs) from patients with preterm birth and chorioamnionitis, focusing on its regulation of inflammatory cytokines and its protective effect on the epithelial barrier. Our results show increased BD2 expression in chorioamnionitis, and Lipopolysaccharide (LPS)-induced inflammation increased BD2 release from hAECs in a dose- and time-dependent manner. BD2 treatment effectively modulated the inflammatory response by reducing pro-inflammatory cytokines (IL-6, IL-1β) and enhancing the release of the anti-inflammatory cytokine IL-10. Additionally, BD2 helps preserve epithelial barrier integrity by restoring E-cadherin expression and reducing Snail expression in inflamed hAECs. In an LPS-induced preterm birth mouse model, BD2 treatment delayed preterm delivery and reduced inflammatory cytokine levels. These results suggest that BD2 plays a protective role in preventing preterm birth by regulating inflammation and maintaining epithelial barrier function, highlighting its therapeutic potential for inflammation-related preterm birth.
Collapse
Affiliation(s)
- Sangho Yun
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.); (S.-H.K.); (J.J.L.)
| | - Shin-Hae Kang
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.); (S.-H.K.); (J.J.L.)
| | - Jiwon Ryu
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea; (J.R.); (K.K.); (K.-Y.L.)
| | - Kyoungseon Kim
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea; (J.R.); (K.K.); (K.-Y.L.)
| | - Keun-Young Lee
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea; (J.R.); (K.K.); (K.-Y.L.)
| | - Jae Jun Lee
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.); (S.-H.K.); (J.J.L.)
- Departments of Anesthesiology and Pain Medicine, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Ji Young Hong
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.); (S.-H.K.); (J.J.L.)
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Sacred Heart Hospital, Hallym University Medical Center, Chuncheon 24253, Republic of Korea
| | - Ga-Hyun Son
- Institute of New Frontier Research Team, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea; (S.Y.); (S.-H.K.); (J.J.L.)
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea; (J.R.); (K.K.); (K.-Y.L.)
| |
Collapse
|
27
|
Anju MV, Archana K, Musthafa SM, Anooja VV, Athira PP, Neelima S, Dhaneesha M, Sajeevan TP, Singh ISB, Philip R. A Novel Hepcidin Isoform Jd-Hep from the Sin Croaker Johnius dussumieri (Cuvier, 1830): Recombinant Expression and Insights into the Antibacterial Property and Modes of Action. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2025; 27:52. [PMID: 39969620 DOI: 10.1007/s10126-025-10426-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Hepcidin is a cysteine-rich antimicrobial peptide that plays an important role in fish immunity. In the current study, we report a novel isoform of hepcidin (Jd-Hep) from Sin croaker, Johnius dussumieri, with an open reading frame (ORF) of 258 nucleotide bases that encodes 85 amino acids containing a signal peptide (24 amino acids), a prodomain (35 amino acids) and a biologically active mature peptide (26 amino acids). Phylogenetic tree analysis showed that J. dussumieri hepcidin belonged to the HAMP2 cluster of hepcidin. The tissue distribution showed that the expression of hepcidin was highest in the liver in wild-caught J. dussumieri. The mature peptide mJd-Hep was recombinantly expressed in a prokaryotic host, E. coli Rosetta-gami™B (DE3) pLysS cells, and the peptide was isolated and purified. The recombinant peptide, rJd-Hep, exhibited notable antibacterial activity against aquatic pathogens such as Aeromonas hydrophila, Vibrio parahaemolyticus, Vibrio harveyi, Vibrio alginolyticus, Vibrio proteolyticus, and Vibrio fluvialis. The mode of action of the peptide was proven to be membrane-based (pore formation and depolarization). The rJd-Hep was found to be non-hemolytic to hRBCs and non-cytotoxic to the mammalian cell line. The peptide showed 85% growth inhibition of cancer cell line, MCF-7. These findings expand our knowledge of the potential application of hepcidin in aquaculture as a therapeutic agent.
Collapse
Affiliation(s)
- M V Anju
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - K Archana
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - S Muhammed Musthafa
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - V V Anooja
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - P P Athira
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - S Neelima
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - M Dhaneesha
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, Kerala, 682016, India
- Department of Biophysics, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, 560029, India
| | - T P Sajeevan
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India
| | - I S Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, Kerala, 682016, India
| | - Rosamma Philip
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Fine Arts Avenue, Kochi, Kerala, 682016, India.
| |
Collapse
|
28
|
Saini S, Pal S, Sharma R. Decoding the Role of Antimicrobial Peptides in the Fight against Mycobacterium tuberculosis. ACS Infect Dis 2025; 11:350-365. [PMID: 39873328 DOI: 10.1021/acsinfecdis.4c00806] [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] [Indexed: 01/30/2025]
Abstract
Tuberculosis (TB), a leading infectious disease caused by the pathogen Mycobacterium tuberculosis, poses a significant treatment challenge due to its unique characteristics and resistance to existing drugs. The conventional treatment regimens, which are lengthy and involve multiple drugs, often result in poor patient adherence and subsequent drug resistance, particularly with multidrug-resistant (MDR) and extensively drug-resistant (XDR) strains. This highlights the urgent need for novel anti-TB therapies and new drug targets. Antimicrobial peptides (AMPs), which are natural host defense molecules present in all living organisms, offer a promising alternative to traditional small-molecule drugs. AMPs have several advantages, including their broad-spectrum activity and the potential to circumvent existing resistance mechanisms. However, their clinical application faces challenges such as stability, delivery, and potential toxicity. This review aims to provide essential information on AMPs, including their sources, classification, mode of action, induction within the host under stress, efficacy against M. tuberculosis, clinical status and hurdles to their use. It also highlights future research directions to address these challenges and advance the development of AMP-based therapies for TB.
Collapse
Affiliation(s)
- Sapna Saini
- Infectious Diseases Division, CSIR─Indian Institute of Integrative Medicine, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sunny Pal
- Infectious Diseases Division, CSIR─Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Rashmi Sharma
- Infectious Diseases Division, CSIR─Indian Institute of Integrative Medicine, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
29
|
Lou M, Ji S, Wu R, Zhu Y, Wu J, Zhang J. Microbial production systems and optimization strategies of antimicrobial peptides: a review. World J Microbiol Biotechnol 2025; 41:66. [PMID: 39920500 DOI: 10.1007/s11274-025-04278-x] [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: 11/13/2024] [Accepted: 01/26/2025] [Indexed: 02/09/2025]
Abstract
Antibiotic resistance has become a public safety issue of the twenty-first century, posing a growing threat and drawing increased attention. Compared to traditional antibiotics, antimicrobial peptides (AMPs), as naturally produced small peptides, can target multiple pathways within pathogens and render them less prone to developing resistance. This makes them promising alternatives to antibiotics. However, traditional chemical synthesis methods face challenges, such as high costs, low yields, and poor stability, limiting the large-scale industrial production of AMPs. Despite extensive research to improve AMP production efficiency, issues such as low yields and complex extraction processes continue to pose significant barriers to commercial application. Therefore, there is an urgent need for new biosynthesis strategies and optimization methods to enhance AMP production efficiency and quality. This review summarizes the sources, classification, mechanisms of action and recent advances in the microbial synthesis of AMPs. It also explores innovative production methods, including recombinant microbial expression systems, fusion tags, codon optimization, tandem multimer expression, and hybrid peptide expression. Furthermore, we review the applications of gene editing technologies and artificial intelligence in AMP production, providing new perspectives and strategies for efficient, large-scale AMP production.
Collapse
Affiliation(s)
- Mengxue Lou
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, People's Republic of China
| | - Shuaiqi Ji
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, People's Republic of China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang, 110866, People's Republic of China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, People's Republic of China
| | - Yi Zhu
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang, 110866, People's Republic of China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang, 110866, People's Republic of China.
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang, 110866, People's Republic of China.
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, 110866, People's Republic of China.
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou, Hainan, 570228, People's Republic of China.
| |
Collapse
|
30
|
Sceglovs A, Skadins I, Chitto M, Kroica J, Salma-Ancane K. Failure or future? Exploring alternative antibacterials: a comparative analysis of antibiotics and naturally derived biopolymers. Front Microbiol 2025; 16:1526250. [PMID: 39963493 PMCID: PMC11830819 DOI: 10.3389/fmicb.2025.1526250] [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: 11/11/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
The global crisis of antimicrobial resistance (AMR) is escalating due to the misuse and overuse of antibiotics, the slow development of new therapies, and the rise of multidrug-resistant (MDR) infections. Traditional antibiotic treatments face limitations, including the development of resistance, disruption of the microbiota, adverse side effects, and environmental impact, emphasizing the urgent need for innovative alternative antibacterial strategies. This review critically examines naturally derived biopolymers with intrinsic (essential feature) antibacterial properties as a sustainable, next-generation alternative to traditional antibiotics. These biopolymers may address bacterial resistance uniquely by disrupting bacterial membranes rather than cellular functions, potentially reducing microbiota interference. Through a comparative analysis of the mechanisms and applications of antibiotics and antibacterial naturally derived biopolymers, this review highlights the potential of such biopolymers to address AMR while supporting human and environmental health.
Collapse
Affiliation(s)
- Artemijs Sceglovs
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Ingus Skadins
- Department of Biology and Microbiology, Riga Stradins University, Riga, Latvia
| | | | - Juta Kroica
- Department of Biology and Microbiology, Riga Stradins University, Riga, Latvia
| | - Kristine Salma-Ancane
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
31
|
Mensah JO, Boakye A, Laryea M, Gasu EN, Borquaye LS. Molecular insights into the differential membrane targeting of maximin 1 in prokaryotic and eukaryotic cells. J Biomol Struct Dyn 2025; 43:1579-1592. [PMID: 38084788 DOI: 10.1080/07391102.2023.2292297] [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: 09/09/2023] [Accepted: 11/25/2023] [Indexed: 01/16/2025]
Abstract
Antimicrobial resistance is a pressing global health concern, underscoring the need for alternative treatments. Antimicrobial peptides (AMPs) have shown promise in this regard, with maximin 1 being a cationic, amphipathic AMP possessing antibacterial, antifungal, and antiviral activities with low hemolytic activity. In this study, we used molecular dynamics simulation to investigate the molecular basis for membrane selectivity of Maximin 1. By studying interactions between maximin 1 and different models of prokaryotic (anionic) and eukaryotic (zwitterionic) membranes, we found that Maximin 1 interacts more strongly with the prokaryotic membrane due to electrostatic attraction, while it weakly interacts with the zwitterionic eukaryotic membrane. Our simulations also revealed that Gly-1, Lys-5, Lys-11, Lys-15, and Lys-19 were identified to play a crucial role in the adsorption of maximin unto the prokaryotic membrane surface. The alpha-helical nature of the peptide, in addition to its amphipathic nature, was necessary for the adsorption of the peptide onto the surface of the prokaryotic membrane. Interestingly, the later transition of the alpha helix into a random coil was crucial in penetrating the prokaryotic membrane while hindering interactions with the eukaryotic membrane. Residues in the middle region of the peptide (residues 9-16) were also responsible for permeating the prokaryotic membrane over the eukaryotic membrane. These findings shed light on the peptide's selective targeting of bacterial membranes over human cell membranes and could inform the design of more effective AMPs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Aaron Boakye
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Michael Laryea
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Edward Ntim Gasu
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Lawrence Sheringham Borquaye
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Central Laboratory, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
32
|
Ali W, Chen Y, Wang Z, Yan K, Men Y, Li Z, Cai W, He Y, Qi J. Characterization of antimicrobial properties of TroH2A-29 peptide from golden pompano (Trachinotus ovatus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 163:105315. [PMID: 39805411 DOI: 10.1016/j.dci.2025.105315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/04/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025]
Abstract
Antimicrobial peptides (AMPs) are small, potent molecules that serve as a crucial first line of defense across a wide range of organisms, including fish. In this study, we investigated the antimicrobial properties of a novel peptide, spanning residues 52 to 80 of the full-length histone H2A protein, comprising a total of 29 amino acids. This peptide, designated as Histone H2A-29 (TroH2A-29), was derived from the golden pompano (Trachinotus ovatus) and evaluated for its activity against both Gram-positive bacteria, Lactococcus garvieae and Staphylococcus epidermidis, and Gram-negative bacteria, Vibrio alginolyticus and Vibrio harveyi. The expression of TroH2A in the intestines, liver, and gills of T. ovatus was significantly upregulated after bacterial infections with L. garvieae and V. harveyi. The highest expression levels were observed at 48 h post-infection in the intestines and at different time points in the liver and gills. TroH2A-29 exhibited a high hydrophobic ratio (51 %) and formed an α-helical structure, suggesting its potential as an antimicrobial agent. Notably, TroH2A-29 induced significant agglutination of all four bacterial species in the presence of Ca2⁺. TroH2A-29 demonstrated bactericidal effects against L. garvieae, V. harveyi, and V. alginolyticus, with a MIC50 of 60 μM. However, it showed no antibacterial activity against S. epidermidis. Transmission electron microscopy (TEM) revealed that TroH2A-29 caused morphological damage to the bacterial cells, including cell collapse in L. garvieae and shrinkage in V. alginolyticus and V. harveyi. No morphological changes were observed in S. epidermidis. Membrane permeability assays showed that TroH2A-29 increased membrane disruption in L. garvieae, V. harveyi, and V. alginolyticus, but had little effect on S. epidermidis. Additionally, TroH2A-29 caused membrane depolarization in all tested bacterial strains. These findings highlight the potential of TroH2A-29 as a novel antimicrobial peptide with selective bactericidal properties.
Collapse
Affiliation(s)
- Wajid Ali
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Ying Chen
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Zhuoyu Wang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Kai Yan
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Yu Men
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Zibin Li
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Wenxiu Cai
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Yan He
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China
| | - Jie Qi
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/ Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266003, Shandong, China.
| |
Collapse
|
33
|
Yu J, Chen G, Jin Y, Zhang M, Wu T. Research Progress of Bioactive Peptides in Improving Type II Diabetes. Foods 2025; 14:340. [PMID: 39941934 PMCID: PMC11817365 DOI: 10.3390/foods14030340] [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: 12/02/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Type II diabetes mellitus (T2DM) is a prevalent, long-standing metabolic condition marked by the body's reduced response to insulin and inadequate insulin production, impacting a significant portion of the global population. Research has demonstrated that bioactive peptides play a crucial role in reducing blood sugar levels, enhancing insulin sensitivity, balancing lipid metabolism, and combating inflammation. These peptides also contribute to the enhancement of pancreatic islet function, lowering systemic inflammation by influencing various molecular signaling pathways. This paper provides an overview of recent advancements and potential applications of bioactive peptides in addressing T2DM. It highlights the diverse impacts of bioactive peptides sourced from different origins in combating diabetes. This comprehensive review offers theoretical substantiation and novel insights to support the future clinical utilization and exploration of bioactive peptides for T2DM management.
Collapse
Affiliation(s)
- Jiaxin Yu
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China; (J.Y.); (G.C.); (Y.J.); (M.Z.)
| | - Guoxing Chen
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China; (J.Y.); (G.C.); (Y.J.); (M.Z.)
| | - Yan Jin
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China; (J.Y.); (G.C.); (Y.J.); (M.Z.)
| | - Min Zhang
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China; (J.Y.); (G.C.); (Y.J.); (M.Z.)
| | - Tao Wu
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China; (J.Y.); (G.C.); (Y.J.); (M.Z.)
- School of Ocean and Environment, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
34
|
Jensen A, Clarke EJ, Nugent Z, Paice E, Gringel I, Yamamoto K, Rocchigiani G, Peffers AJ, Cooper L, Peffers MJ. Inflammation and response to bacterial infection as potential drivers of equine odontoclastic tooth resorption and hypercementosis: A proteomics insight. Equine Vet J 2025. [PMID: 39777419 DOI: 10.1111/evj.14469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Equine dental diseases significantly impact a horse's overall health, performance and quality of life. They can result in secondary infections and digestive disturbances, potentially leading to colic. A recently described disease affecting the incisors of horses is equine odontoclastic tooth resorption and hypercementosis (EOTRH). Understanding EOTRH is crucial for early diagnosis, effective management and prevention of its severe consequences. OBJECTIVES To determine proteomic differences in incisor cementum in horses with and without clinical EOTRH. STUDY DESIGN Comparative and observational clinical study. METHODS Teeth were extracted (N = 5) and cementum was isolated using a diamond wire. Proteins were extracted using an optimised sequential workflow, and trypsin was digested for mass spectrometry. Protein identification and label-free quantification were undertaken. RESULTS In total 1149 unique proteins were detected in cementum across all samples. We identified four proteins exclusively in EOTRH-affected cementum. EOTRH samples showed a higher heterogeneity than healthy samples. In total, 54 proteins were increased in EOTRH, and 64 proteins were reduced (adjusted p-value <0.05). Inflammatory proteins, such as cathepsin G (p = 0.004), neutrophil elastase (p = 0.003), bactericidal permeability-increasing protein (p = 0.002), azurocidin (p = 0.003) and lactotransferrin (p = 0.002) were all increased in EOTRH. Pathway analysis revealed that antimicrobial peptides (Z score 2.65, p = 1.93E-09) and neutrophil degranulation (Z-score 1.89, p = 1.7E-04) were commonly up-regulated canonical pathways. MAIN LIMITATIONS The sample size was limited. Lack of age-matched healthy controls. CONCLUSION EOTRH leads to biochemical changes within the cementum proteome, which are important in explaining the physiological changes occurring in disease. Differentially abundant proteins may represent promising biomarkers for earlier disease detection and the establishment of a cell-based model could provide further insight into the role these proteins play in hypercementosis and resorption.
Collapse
Affiliation(s)
- Anders Jensen
- University of Liverpool, Institute of Life Course and Medical Sciences, William Henry Duncan Building, Liverpool, UK
| | - Emily J Clarke
- University of Liverpool, Institute of Life Course and Medical Sciences, William Henry Duncan Building, Liverpool, UK
| | - Zoe Nugent
- University of Liverpool, Institute of Life Course and Medical Sciences, William Henry Duncan Building, Liverpool, UK
| | - Emily Paice
- University of Liverpool, Institute of Life Course and Medical Sciences, William Henry Duncan Building, Liverpool, UK
| | - Iris Gringel
- University of Liverpool, Institute of Life Course and Medical Sciences, William Henry Duncan Building, Liverpool, UK
| | - Kazuhiro Yamamoto
- University of Liverpool, Institute of Life Course and Medical Sciences, William Henry Duncan Building, Liverpool, UK
| | - Guido Rocchigiani
- University of Liverpool, Institute of Infection, Veterinary and Ecological Sciences, Leahurst Campus, Neston, UK
| | | | - Lee Cooper
- University of Liverpool, Institute of Life Course and Medical Sciences, School of Dentistry, Liverpool, UK
| | - Mandy J Peffers
- University of Liverpool, Institute of Life Course and Medical Sciences, William Henry Duncan Building, Liverpool, UK
| |
Collapse
|
35
|
Lin CC, Chen CS. Bacterial proteome microarray technology in biomedical research. Trends Biotechnol 2025:S0167-7799(24)00361-5. [PMID: 39755450 DOI: 10.1016/j.tibtech.2024.12.001] [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: 06/11/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 01/06/2025]
Abstract
Bacterial proteome microarrays are high-throughput, adaptable tools that allow the simultaneous investigation of thousands of proteins from various bacterial species. These arrays are used to explore bacterial pathogenicity, pathogen-host interactions, and clinical diseases. Recent advancements have expanded their application to profiling human antibodies, identifying biomarkers for infectious and autoimmune diseases, and studying antimicrobial peptides (AMPs). This review highlights significant outcomes from recent studies, focusing on their diverse applications in biomedical research. Notable findings include the identification of novel antigens and diagnostic markers for gastrointestinal infections, autoimmune diseases, and mental health disorders. This technology promises to further elucidate the complex relationship between bacteria and their hosts, ultimately informing the development of new diagnostic, therapeutic, and preventive strategies.
Collapse
Affiliation(s)
- Chia-Chi Lin
- School of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Sheng Chen
- Department of Food Safety/Hygiene and Risk Management, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
36
|
Shen H, Li Y, Pi Q, Tian J, Xu X, Huang Z, Huang J, Pian C, Mao S. Unveiling novel antimicrobial peptides from the ruminant gastrointestinal microbiomes: A deep learning-driven approach yields an anti-MRSA candidate. J Adv Res 2025:S2090-1232(25)00005-0. [PMID: 39756573 DOI: 10.1016/j.jare.2025.01.005] [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: 10/12/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025] Open
Abstract
INTRODUCTION Antimicrobial peptides (AMPs) present a promising avenue to combat the growing threat of antibiotic resistance. The ruminant gastrointestinal microbiome serves as a unique ecosystem that offers untapped potential for AMP discovery. OBJECTIVES The aims of this study are to develop an effective methodology for the identification of novel AMPs from ruminant gastrointestinal microbiomes, followed by evaluating their antimicrobial efficacy and elucidating the mechanisms underlying their activity. METHODS We developed a deep learning-based model to identify AMP candidates from a dataset comprising 120 metagenomes and 10,373 metagenome-assembled genomes derived from the ruminant gastrointestinal tract. Both in vivo and in vitro experiments were performed to examine and validate the antimicrobial activities of the AMP candidates that were selected through bioinformatic analysis and subsequently synthesized chemically. Additionally, molecular dynamics simulations were conducted to explore the action mechanism of the most potent AMP candidate. RESULTS The deep learning model identified 27,192 potential secretory AMP candidates. Following bioinformatic analysis, 39 candidates were synthesized and tested. Remarkably, all synthesized peptides demonstrated antimicrobial activity against Staphylococcus aureus, with 79.5% showing effectiveness against multiple pathogens. Notably, Peptide 4, which exhibited the highest antimicrobial activity against methicillin-resistant Staphylococcus aureus (MRSA), confirmed this effect in a mouse model with wound infection, exhibiting a low propensity for resistance development and minimal cytotoxicity and hemolysis towards mammalian cells. Molecular dynamics simulations provided insights into the mechanism of Peptide 4, primarily its ability to disrupt bacterial cell membranes, leading to cell death. CONCLUSION This study highlights the power of combining deep learning with microbiome research to uncover novel therapeutic candidates, paving the way for the development of next-generation antimicrobials like Peptide 4 to combat the growing threat of MRSA would infections. It also underscores the value of utilizing ruminant microbial resources.
Collapse
Affiliation(s)
- Hong Shen
- Bioinformatics Center, Academy for Advanced Interdisciplinary Studies, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Yanru Li
- College of Agriculture, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Qingjie Pi
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Junru Tian
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Xianghan Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China
| | - Zan Huang
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Jinghu Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| | - Cong Pian
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, Jiangsu, China.
| | - Shengyong Mao
- Ruminant Nutrition and Feed Engineering Technology Research Center, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China; Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, National Center for International Research on Animal Gut Nutrition, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, Jiangsu, China.
| |
Collapse
|
37
|
Roque‐Borda CA, Primo LMDG, Medina‐Alarcón KP, Campos IC, Nascimento CDF, Saraiva MMS, Berchieri Junior A, Fusco‐Almeida AM, Mendes‐Giannini MJS, Perdigão J, Pavan FR, Albericio F. Antimicrobial Peptides: A Promising Alternative to Conventional Antimicrobials for Combating Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410893. [PMID: 39530703 PMCID: PMC11714181 DOI: 10.1002/advs.202410893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Polymicrobial biofilms adhere to surfaces and enhance pathogen resistance to conventional treatments, significantly contributing to chronic infections in the respiratory tract, oral cavity, chronic wounds, and on medical devices. This review examines antimicrobial peptides (AMPs) as a promising alternative to traditional antibiotics for treating biofilm-associated infections. AMPs, which can be produced as part of the innate immune response or synthesized therapeutically, have broad-spectrum antimicrobial activity, often disrupting microbial cell membranes and causing cell death. Many specifically target negatively charged bacterial membranes, unlike host cell membranes. Research shows AMPs effectively inhibit and disrupt polymicrobial biofilms and can enhance conventional antibiotics' efficacy. Preclinical and clinical research is advancing, with animal studies and clinical trials showing promise against multidrug-resistant bacteria and fungi. Numerous patents indicate increasing interest in AMPs. However, challenges such as peptide stability, potential cytotoxicity, and high production costs must be addressed. Ongoing research focuses on optimizing AMP structures, enhancing stability, and developing cost-effective production methods. In summary, AMPs offer a novel approach to combating biofilm-associated infections, with their unique mechanisms and synergistic potential with existing antibiotics positioning them as promising candidates for future treatments.
Collapse
Affiliation(s)
- Cesar Augusto Roque‐Borda
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
- Vicerrectorado de InvestigaciónUniversidad Católica de Santa MaríaArequipa04000Peru
| | - Laura Maria Duran Gleriani Primo
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Kaila Petronila Medina‐Alarcón
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Isabella C. Campos
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Camila de Fátima Nascimento
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Mauro M. S. Saraiva
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Ana Marisa Fusco‐Almeida
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Maria José Soares Mendes‐Giannini
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - João Perdigão
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
| | - Fernando Rogério Pavan
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Fernando Albericio
- Peptide Science LaboratorySchool of Chemistry and PhysicsUniversity of KwaZulu‐NatalDurban4001South Africa
- CIBER‐BBNNetworking Centre on BioengineeringBiomaterials and Nanomedicineand Department of Organic ChemistryUniversity of BarcelonaBarcelona08028Spain
| |
Collapse
|
38
|
Iksanova AM, Ojovan IM, Arzumanian VG, Vartanova NO, Poddubikov AV, Kolyganova TI, Konanykhina SY. The Sensitivity of Opportunistic Bacteria to Blood Serum Low-Molecular Fraction Containing Antimicrobial Peptides and Proteins. Bull Exp Biol Med 2025; 178:339-345. [PMID: 39945948 DOI: 10.1007/s10517-025-06333-x] [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: 06/20/2024] [Indexed: 02/28/2025]
Abstract
We studied the sensitivity of clinically significant (opportunistic) bacteria to blood serum fraction with a molecular weight <100 kDa containing antimicrobial peptides and proteins. The degree of damage to cytoplasmic membranes of bacterial cells was assessed by a spectrophotometric method. A total of 129 bacterial isolates belonging to 46 species, 19 genera, 13 families, 8 orders, 4 classes, and 3 types were studied. The mean sensitivity varied from -5.2 to 36.6%. The sensitivity depends on the structure of bacterial cell wall: higher sensitivity was demonstrated by taxonomic ranks related to gram-negative bacteria (median 19.1%) and lower values were observed in gram-positive bacteria (median 9.3%) (p<0.01). Among all orders, Lactobacilliales showed the lowest sensitivity (3.6%). Ranking of the studied species by their sensitivity showed that 7 of the first 15 species with sensitivity >19% are among to the leading causative agents of sepsis.
Collapse
Affiliation(s)
- A M Iksanova
- I. Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia.
| | - I M Ojovan
- I. Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| | - V G Arzumanian
- I. Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| | - N O Vartanova
- I. Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| | - A V Poddubikov
- I. Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| | - T I Kolyganova
- I. Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
- I. M. Sechenov First Moscow State Medical University (Sechenov University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - S Yu Konanykhina
- I. Mechnikov Research Institute for Vaccines and Sera, Moscow, Russia
| |
Collapse
|
39
|
Haddad H, da Franca Rodrigues KA, Othman H, Veras LMC, Rodrigues RRL, Ouahchi I, Ouni B, Zaϊri A. In vitro Antileishmanial Activity and In silico Molecular Modeling Studies of Novel Analogs of Dermaseptins S4 and B2. Curr Pharm Biotechnol 2025; 26:276-288. [PMID: 39257149 DOI: 10.2174/0113892010296038240427050421] [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: 02/09/2024] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Leishmaniasis is responsible for approximately 65,000 annual deaths. Various Leishmania species are the predominant cause of visceral, cutaneous, or mucocutaneous leishmaniasis, affecting millions worldwide. The lack of a vaccine, emergence of resistance, and undesirable side effects caused by antileishmanial medications have prompted researchers to look for novel therapeutic approaches to treat this disease. Antimicrobial peptides (AMPs) offer an alternative for promoting the discovery of new drugs. METHODS In this study, we detail the synthesis process and investigate the antileishmanial activity against Leishmania (Viannia) braziliensis for peptides belonging to the dermaseptin (DS) family and their synthetic analogs. The MTT assay was performed to investigate the cytotoxicity of these peptides on the murine macrophage cell line RAW 264.7. Subsequently, we performed molecular modeling analysis to explore the structure-function correlation of the derivatives interacting with the parasitic membrane. RESULTS All examined derivatives displayed concentration-dependent antileishmanial effect at low concentrations. Their effectiveness varied according to the peptide's proprieties. Notably, peptides with higher levels of charge demonstrated the most pronounced activities. Cytotoxicity assays showed that all the tested peptides were not cytotoxic compared to the tested conventional drug. The structure-function relationships demonstrated that the charged N-terminus could be responsible for the antileishmanial effect observed on promastigotes. CONCLUSION Collectively, these results propose that dermaseptins (DS) might offer potential as promising candidates for the development of effective antileishmanial therapies.
Collapse
Affiliation(s)
- Houda Haddad
- BIOLIVAL Laboratory, Higher Institute of Biotechnology of Monastir ISBM, University of Monastir, 5000 Monastir, Tunisia
- Biochemistry Department, Faculty of Medicine, University of Sousse, 4002 Sousse, Tunisia
| | | | - Houcemeddine Othman
- Laboratory of Cytogenetics and Reproductive Biology, CHU Farhat Hached, 4000 Sousse, Tunisia
| | - Leiz Maria Costa Veras
- Biodiversity and Biotechnology Research Center, BIOTEC, Federal University of Piauí, Parnaíba, PI, Brazil
| | - Raiza Raianne Luz Rodrigues
- Laboratory of Infectious Diseases, Ladic, Campus Ministro Reis Velloso, Federal University of Delta do Parnaíba, 64202-020, Brazil
| | - Ines Ouahchi
- Biodiversity Cytogenetics, Molecular Genetics and Reproductive Biology, Farhat Hached University Hospital, 4000 Sousse, Tunisia
| | - Bouraoui Ouni
- Pharmacology department, Faculty of Medicine, University of Sousse, 4002 Sousse, Tunisia
| | - Amira Zaϊri
- Biochemistry Department, Faculty of Medicine, University of Sousse, 4002 Sousse, Tunisia
| |
Collapse
|
40
|
Mojgani N, Bagheri M, Ashique S, Islam A, Moharrami M, Modirrousta H, Hussain A. Honeybee defense mechanisms: Role of honeybee gut microbiota and antimicrobial peptides in maintaining colony health and preventing diseases. Microb Pathog 2025; 198:107161. [PMID: 39603566 DOI: 10.1016/j.micpath.2024.107161] [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: 08/12/2024] [Revised: 10/20/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Honeybees play a vital role in pollination and the maintenance of ecosystem biodiversity, making their health and well-being crucial for agriculture and environmental sustainability. Bee health is modulated by symbiotic microorganisms colonizing the gut in balanced proportions. Studies have demonstrated that these beneficial bacteria have the capacity to enhance the immune system of honey bees, having substantial impact on regulating their immunological responses and hence aiding in defending against pathogenic illnesses. Another important aspect of honeybee health is their innate immune system that is related to their ability to synthesize antimicrobial peptides (AMP). AMPs, the small, cationic peptides are the humoral effector molecules that are synthesized in the hemolymph of the insects after being exposed to microbial infectious agents. A number of honeybee's gut microbiota especially Lactic Acid Bacteria (LAB), are known to regulate the production of several AMPs and hence are able to provide protection to these insects against a number of disease agents by modulating their innate immune response via induction of the AMPs genes. These AMPs mainly produced by adult workers are an important and integral part of an insect's immune response. Several AMPs namely apidaecins, abaecins, hymenoptaecins and defensins produced in the adult honeybee, hold the ability to control or prevent a number of diseases in these pollinator insects.
Collapse
Affiliation(s)
- Naheed Mojgani
- Razi Vaccine and Serum Research Institute- Agriculture Research, Education and Extension Organization (AREEO), Karaj, Iran.
| | - Masoumeh Bagheri
- Razi Vaccine and Serum Research Institute- Agriculture Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Sumel Ashique
- Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, 713212, West Bengal, India; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, 226026, Uttar Pradesh, India
| | - Mojtaba Moharrami
- Razi Vaccine and Serum Research Institute- Agriculture Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Hossein Modirrousta
- Razi Vaccine and Serum Research Institute- Agriculture Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Abrar Hussain
- H.E.J Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
41
|
Xu X, Yu YB. Role of antimicrobial peptides in gastrointestinal diseases: Recent advances. Shijie Huaren Xiaohua Zazhi 2024; 32:865-871. [DOI: 10.11569/wcjd.v32.i12.865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/26/2024] [Accepted: 10/21/2024] [Indexed: 12/28/2024] Open
Affiliation(s)
- Xia Xu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250000, Shandong Province, China
| | - Yan-Bo Yu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250000, Shandong Province, China
| |
Collapse
|
42
|
Gong Y, Xue Q, Li J, Zhang S. Antifungal peptides from living organisms. Front Microbiol 2024; 15:1511461. [PMID: 39741586 PMCID: PMC11685209 DOI: 10.3389/fmicb.2024.1511461] [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: 10/15/2024] [Accepted: 11/29/2024] [Indexed: 01/03/2025] Open
Abstract
In the post-COVID-19 era, people are increasingly concerned about microbial infections, including fungal infections that have risen in recent years. However, the currently available antifungal agents are rather limited. Worse still, the widespread use of the antifungal agents has caused the emergence of antifungal resistance in Candida, Cryptococcus, and Aspergillus species. Therefore, the development of novel antifungals is urgently needed. Antimicrobial peptides (AMPs), as components of the first-line defense of the host, are found to exhibit broad antimicrobial activity against bacteria, fungi, parasites, viruses, and protozoa. AMPs with antifungal activity are specifically referred to as antifungal peptides (AFPs). AFPs are currently regarded as the most promising alternative to conventional antifungal agents due to the fact that they are highly selective and less prone to facilitate the selection of drug resistance. In this review, we present an overview of the origin and classification of natural AFPs as well as their modes of action. Additionally, the production of natural, semisynthetic, and synthetic AFPs with a view to greater levels of exploitation is discussed. Finally, we evaluate the current and potential applications of AFPs in clinics and in the food industry.
Collapse
Affiliation(s)
- Yi Gong
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Qunhang Xue
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Jun Li
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Shicui Zhang
- Key Laboratory of Biological Resources and Ecology of Pamirs Plateau in Xinjiang Uygur Autonomous Region, College of Life and Geographic Sciences, Kashi University, Kashi, China
- Department of Marine Biology, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
43
|
Dzień E, Wątły J, Hecel A, Mikołajczyk A, Matera-Witkiewicz A, Adrover M, Barceló-Oliver M, Domínguez-Martín A, Rowińska-Żyrek M. Zn(II) coordination influences the secondary structure, but not antimicrobial activity of the N-terminal histatin 3 hydrolysis product. Dalton Trans 2024; 53:19202-19213. [PMID: 39508362 DOI: 10.1039/d4dt02274b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
The relationship between the coordination chemistry and antimicrobial activity of Zn(II) and Cu(II)-bound histatins, salivary antimicrobial peptides, remains enigmatic. We focus on metal complexes of histatin 3 and its two products of hydrolysis: histatin 4 and its N-terminal fragment (histatin 3-4). The thermodynamic stability of these complexes is quite expected - the binding of Cu(II) via the ATCUN motif results in the formation of very stable complexes. In histatin-Zn(II) complexes, the {2Nim} type of coordination dominates, with polymorphic binding sites observed for histatin 3-4 and 5-8, resulting in their low thermodynamic stability compared to the complexes of histatin 3, 4, 5 and 8 with Zn(II), in which we observe a {2Nim, O-} type of coordination. Histatin 3, 3-4 and 4 have greater activity against Gram-positive bacteria than against Gram-negative ones, and Cu(II) or Zn(II) binding can, in some cases, moderately increase the antimicrobial activity of the native histatin 3 and 4, but not the remaining 3-4 fragment. The most probable reason for the metal-enhanced antimicrobial activity is, in this case, a local change of charge, while the chemically fascinating metal binding induced structural changes do not result in a change of biological activity. Neither histatin 3-4, the N-terminal fragment of histatin 3, which remains in solution after cleavage, nor its metal complexes have any antimicrobial activity, but histatin 3-4 presents intriguing Zn(II)-induced structural behavior, changing its secondary structure, with a tendency to form an α-helix.
Collapse
Affiliation(s)
- Emilia Dzień
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, 50-383 Wrocław, Poland.
| | - Joanna Wątły
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, 50-383 Wrocław, Poland.
| | - Aleksandra Hecel
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, 50-383 Wrocław, Poland.
| | - Aleksandra Mikołajczyk
- Screening of Biological Activity Assays and Collection of Biological Material Laboratory, Wroclaw Medical Univeristy Biobank, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Agnieszka Matera-Witkiewicz
- Screening of Biological Activity Assays and Collection of Biological Material Laboratory, Wroclaw Medical Univeristy Biobank, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Miquel Adrover
- Department of Chemistry, University of Balearic Islands, Cra. de Valldemossa, km 7.7, 07122 Palma de Mallorca, Spain
- Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Spain
- Institut de Recerca en Ciències de la Salut (IdISBa), Spain
| | - Miquel Barceló-Oliver
- Department of Chemistry, University of Balearic Islands, Cra. de Valldemossa, km 7.7, 07122 Palma de Mallorca, Spain
| | - Alicia Domínguez-Martín
- Department of Inorganic Chemistry, Faculty of Pharmacy, University of Granada, E-18071 Granada, Spain
| | | |
Collapse
|
44
|
Ojah EO, Gneid H, Herschede SR, Busschaert N. Structure-Activity Relationships in Supramolecular Hosts Targeting Bacterial Phosphatidylethanolamine (PE) Lipids. Chemistry 2024; 30:e202402698. [PMID: 39231001 DOI: 10.1002/chem.202402698] [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: 07/16/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
The World Health Organization has described the antimicrobial resistance crisis as one of the top ten global public health threats. New antimicrobial agents that can fight infections caused by antimicrobial resistant pathogens are therefore needed. A potential strategy is the development of small molecules that can selectively interact with bacterial membranes (or membranes of other microbial pathogens), and thereby rapidly kill the bacteria. Here, we report the structure-activity relationship within a group of 22 compounds that were designed to bind the bacterial lipid phosphatidylethanolamine (PE). Liposome-based studies reveal that the lipophilicity of the compounds has the strongest effect on both the affinity and selectivity for PE. The best results were obtained for compounds with logP≈3.75, which showed a 5x-7x selectivity for bacterial PE lipids over human PC (phosphatidylcholine) lipids. Furthermore, these compounds also showed potent antibacterial activity against the Gram-positive bacterium B. cereus, with minimum inhibitory concentrations (MICs) below 10 μM, a concentration where they showed minimal hemolytic activity against human red blood cells. These results not only show the possibility of PE-binding small molecules to function as antibiotics, but also provide guidelines for the development of compounds targeting other types of biologically relevant membrane lipids.
Collapse
Affiliation(s)
- Emmanuel O Ojah
- Chemistry, Tulane University, New Orleans, LA, United States
| | - Hassan Gneid
- Chemistry, Tulane University, New Orleans, LA, United States
| | | | | |
Collapse
|
45
|
Islam T, Tamanna NT, Sagor MS, Zaki RM, Rabbee MF, Lackner M. Antimicrobial Peptides: A Promising Solution to the Rising Threat of Antibiotic Resistance. Pharmaceutics 2024; 16:1542. [PMID: 39771521 PMCID: PMC11728462 DOI: 10.3390/pharmaceutics16121542] [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: 11/02/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
The demand for developing novel antimicrobial drugs has increased due to the rapid appearance and global spread of antibiotic resistance. Antimicrobial peptides (AMPs) offer distinct advantages over traditional antibiotics, such as broad-range efficacy, a delayed evolution of resistance, and the capacity to enhance human immunity. AMPs are being developed as potential medicines, and current computational and experimental tools aim to facilitate their preclinical and clinical development. Structural and functional constraints as well as a more stringent regulatory framework have impeded clinical translation of AMPs as possible therapeutic agents. Although around four thousand AMPs have been identified so far, there are some limitations of using these AMPs in clinical trials due to their safety in the host and sometimes limitations in the biosynthesis or chemical synthesis of some AMPs. Overcoming these obstacles may help to open a new era of AMPs to combat superbugs without using synthetic antibiotics. This review describes the classification, mechanisms of action and immune modulation, advantages, difficulties, and opportunities of using AMPs against multidrug-resistant pathogens and highlights the need and priorities for creating targeted development strategies that take into account the most cutting-edge tools currently available. It also describes the barriers to using these AMPs in clinical trials.
Collapse
Affiliation(s)
- Tarequl Islam
- Department of Microbiology, Noakhali Science and Technology University, Sonapur, Noakhali 3814, Bangladesh;
| | - Noshin Tabassum Tamanna
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali 3814, Bangladesh;
| | - Md Shahjalal Sagor
- Department of Microbiology, Jagannath University, Dhaka 1100, Bangladesh;
| | - Randa Mohammed Zaki
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia;
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Muhammad Fazle Rabbee
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea
| | - Maximilian Lackner
- Department of Industrial Engineering, University of Applied Sciences Technikum Wien, Hoechstaedtplatz 6, 1200 Vienna, Austria
| |
Collapse
|
46
|
Gutierrez CDO, Almeida LHDO, Sardi JDCO, Almeida CV, de Oliveira CFR, Marchetto R, Crusca E, Buccini DF, Franco OL, Cardoso MH, Macedo MLR. Boosting the antibacterial potential of a linear encrypted peptide in a Kunitz-type inhibitor (ApTI) through physicochemical-guided approaches. Biochimie 2024; 227:161-171. [PMID: 39029576 DOI: 10.1016/j.biochi.2024.07.009] [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: 04/09/2024] [Revised: 06/26/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Bacterial resistance has become a serious public health problem in recent years, thus encouraging the search for new antimicrobial agents. Here, we report an antimicrobial peptide (AMP), called PEPAD, which was designed based on an encrypted peptide from a Kunitz-type plant peptidase inhibitor. PEPAD was capable of rapidly inhibiting and eliminating numerous bacterial species at micromolar concentrations (from 4μM to 10 μM), with direct membrane activity. It was also observed that the peptide can act synergistically with ciprofloxacin and showed no toxicity in the G. mellonella in vivo assay. Circular dichroism assays revealed that the peptide's secondary structure adopts different scaffolds depending on the environment in which it is inserted. In lipids mimicking bacterial cell membranes, PEPAD adopts a more stable α-helical structure, which is consistent with its membrane-associated mechanism of action. When in contact with lipids mimicking mammalian cells, PEPAD adopts a disordered structure, losing its function and suggesting cellular selectivity. Therefore, these findings make PEPAD a promising candidate for future antimicrobial therapies with low toxicity to the host.
Collapse
Affiliation(s)
- Camila de Oliveira Gutierrez
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Luís Henrique de Oliveira Almeida
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Janaina de Cássia Orlandi Sardi
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Claudiane Vilharroel Almeida
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Caio Fernando Ramalho de Oliveira
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso Do Sul, Campo Grande, MS, Brazil
| | - Reinaldo Marchetto
- Universidade Estadual Paulista (UNESP), Instituto de Química, Araraquara, São Paulo, Brazil
| | - Edson Crusca
- Universidade Estadual Paulista (UNESP), Instituto de Química, Araraquara, São Paulo, Brazil
| | | | - Octavio Luiz Franco
- S-Inova Biotech, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil; Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | - Marlon Henrique Cardoso
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso Do Sul, Campo Grande, MS, Brazil; S-Inova Biotech, Universidade Católica Dom Bosco, Campo Grande, MS, Brazil; Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil
| | - Maria Lígia Rodrigues Macedo
- Laboratório de Purificação de Proteínas e Suas Funções Biológicas, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal de Mato Grosso Do Sul, Campo Grande, MS, Brazil.
| |
Collapse
|
47
|
Hussaini IM, Sulaiman AN, Abubakar SC, Abdulazeez TM, Abdullahi MM, Sulaiman MA, Madika A, Bishir M, Muhammad A. Unveiling the arsenal against antibiotic resistance: Antibacterial peptides as broad-spectrum weapons targeting multidrug-resistant bacteria. THE MICROBE 2024; 5:100169. [DOI: 10.1016/j.microb.2024.100169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
48
|
Gao N, Fang C, Bai P, Wang J, Dong N, Shan A, Zhang L. De novo design of Na +-activated lipopeptides with selective antifungal activity: A promising strategy for antifungal drug discovery. Int J Biol Macromol 2024; 283:137894. [PMID: 39571872 DOI: 10.1016/j.ijbiomac.2024.137894] [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: 09/19/2024] [Revised: 11/03/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
In recent years, invasive fungal infections have posed a significant threat to human health, particularly due to the limited availability of effective antifungal medications. This study responds to the urgent need for powerful and selective antifungal agents by designing and synthesizing a series of lipopeptides with lipoylation at the N-terminus of the antimicrobial peptide I6. Compared to the parent peptide I6, lipopeptides exhibited selective antifungal efficacy in the presence of Na+. Among the variants tested, C8-I6 emerged as the most effective, with an average effective concentration of 5.3 μM against 12 different fungal species. C8-I6 combated fungal infections by disrupting both cytoplasmic and mitochondrial membranes, impairing the proton motive force, generating reactive oxygen species, and triggering apoptosis in fungal cells. Importantly, C8-I6 exhibited minimal hemolysis and cytotoxicity while effectively inhibiting fungal biofilm formation. In vivo experiments further validated the safety and therapeutic potential of C8-I6 in treating fungal skin infections. These findings underscore the significance of lipoylation in enhancing the efficacy of antimicrobial peptides, positioning C8-I6 as a promising candidate in fighting against drug-resistant fungal infections.
Collapse
Affiliation(s)
- Nan Gao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Chunyang Fang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Pengfei Bai
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiajun Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China.
| | - Na Dong
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China.
| | - Licong Zhang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| |
Collapse
|
49
|
Mir Drikvand R, Sohrabi SM, Sohrabi SS, Samiei K. Molecular Identification and Characterization of Hevein Antimicrobial Peptide Genes in Two-Row and Six-Row Cultivars of Barley (Hordeum vulgare L.). Biochem Genet 2024; 62:5092-5114. [PMID: 38386212 DOI: 10.1007/s10528-024-10695-8] [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: 09/29/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024]
Abstract
Heveins are one of the most important groups of plant antimicrobial peptides. So far, various roles in plant growth and development and in response to biotic and abiotic stresses have reported for heveins. The present study aimed to identify and characterize the hevein genes in two-row and six-row cultivars of barley. In total, thirteen hevein genes were identified in the genome of two-row and six-row cultivars of barley. The identified heveins were identical in two-row and six-row cultivars of barley and showed a high similarity with heveins from other plant species. The hevein coding sequences produced open reading frames (ORFs) ranged from 342 to 1002 bp. Most of the identified hevein genes were intronless, and the others had only one intron. The hevein ORFs produced proteins ranged from 113 to 333 amino acids. Search for conserved functional domains showed CBD and LYZ domains in barley heveins. All barley heveins comprised extracellular signal peptides ranged from 19 to 35 amino acids. The phylogenetic analysis divided barley heveins into two groups. The promoter analysis showed regulatory elements with different frequencies between two-row and six-row cultivars. These cis-acting elements included elements related to growth and development, hormone response, and environmental stresses. The expression analysis showed high expression level of heveins in root and reproductive organs of both two-row and six-row cultivars. The expression analysis also showed that barley heveins is induced by both biotic and abiotic stresses. The results of antimicrobial activity prediction showed the highest antimicrobial activity in CBD domain of barley heveins. The findings of the current study can improve our knowledge about the role of hevein genes in plant and can be used for future studies.
Collapse
Affiliation(s)
- Reza Mir Drikvand
- Department of Plant Genetics and Breeding, Islamic Azad University, Khorramabad Branch, Khorramabad, Iran.
| | - Seyyed Mohsen Sohrabi
- Department of Production Engineering and Plant Genetics, Faculty of Agriculture, Shahid Chamran University, Ahvaz, Iran
| | - Seyed Sajad Sohrabi
- Department of Production Engineering and Plant Genetics, Faculty of Agriculture, Lorestan University, Khorramabad, Iran
| | - Kamran Samiei
- Department of Plant Genetics and Breeding, Islamic Azad University, Khorramabad Branch, Khorramabad, Iran
| |
Collapse
|
50
|
Wang J, Zhang M, Li C, Liu M, Qi Y, Xie X, Zhou C, Ma L. A novel cathelicidin TS-CATH derived from Thamnophis sirtalis combats drug-resistant gram-negative bacteria in vitro and in vivo. Comput Struct Biotechnol J 2024; 23:2388-2406. [PMID: 38882682 PMCID: PMC11176561 DOI: 10.1016/j.csbj.2024.05.020] [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: 12/28/2023] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 06/18/2024] Open
Abstract
Antimicrobial peptides are promising therapeutic agents for treating drug-resistant bacterial disease due to their broad-spectrum antimicrobial activity and decreased susceptibility to evolutionary resistance. In this study, three novel cathelicidin antimicrobial peptides were identified from Thamnophis sirtalis, Balaenoptera musculus, and Lipotes vexillifer by protein database mining and sequence alignment and were subsequently named TS-CATH, BM-CATH, and LV-CATH, respectively. All three peptides exhibited satisfactory antibacterial activity and broad antibacterial spectra against clinically isolated E. coli, P. aeruginosa, K. pneumoniae, and A. baumannii in vitro. Among them, TS-CATH displayed the best antimicrobial/bactericidal activity, with a rapid elimination efficiency against the tested drug-resistant gram-negative bacteria within 20 min, and exhibited the lowest cytotoxicity toward mammalian cells. Furthermore, TS-CATH effectively enhanced the survival rate of mice with ceftazidime-resistant E. coli bacteremia and promoted wound healing in meropenem-resistant P. aeruginosa infection. These results were achieved through the eradication of bacterial growth in target organs and wounds, further inhibiting the systemic dissemination of bacteria and the inflammatory response. TS-CATH exhibited direct antimicrobial activity by damaging the inner and outer membranes, resulting in leakage of the bacterial contents at super-MICs. Moreover, TS-CATH disrupted the bacterial respiratory chain, which inhibited ATP synthesis and induced ROS formation, significantly contributing to its antibacterial efficacy at sub-MICs. Overall, TS-CATH has potential for use as an antibacterial agent.
Collapse
Affiliation(s)
- Jian Wang
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Meina Zhang
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Chao Li
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Mengyuan Liu
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yixin Qi
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xiaolin Xie
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Changlin Zhou
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Lingman Ma
- Institution of all authors: College of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| |
Collapse
|