1
|
Yan X, Yang C, Li B, Bian Y, Yu W, Zhu Y, Cheng B, Lyu Y, Shan A. Positive Charge-Concentrated Dimeric Lipopeptides with Enhanced Protease Resistance: A Potential Solution for Systemic Bacterial Infections. J Med Chem 2025. [PMID: 39742430 DOI: 10.1021/acs.jmedchem.4c01966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Antimicrobial peptides (AMPs) show potential as antibiotic alternatives for bacterial infections; nevertheless, the susceptibility to proteases limits their broader utilization. This study developed engineered lipopeptides using antienzymolysis modifications and cysteine (Cys)-dimerization strategy. As the key parameters for the functioning of AMPs, hydrophobicity and positive charges were concentrated within the peptide sequence by adjusting the intermolecular disulfide bond placement to study their distribution effects. Their centralization in the sequence induces a differential propensity of engineered lipopeptides toward bacterial membranes. Positive charge-concentrated dimeric lipopeptide (C-C10)C-C displayed strong resistance to various proteases, and demonstrated excellent stability and activity in vitro, effectively eliminating systemic bacterial infections in mice without eliciting in vivo toxicity. The bactericidal effects of (C-C10)C-C were achieved through a synergistic mechanism involving membrane cleavage and the inhibition of energy metabolism. In summary, these advances offered valuable insights into enhancing the protease resistance of AMPs and the potential for modifying peptide-based biomaterials through Cys-dimerization.
Collapse
Affiliation(s)
- Xi Yan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Chengyi Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Bo Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Yifeng Bian
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Weikang Yu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Yongjie Zhu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Baojing Cheng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Yinfeng Lyu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, People's Republic of China
| |
Collapse
|
2
|
Hu P, Chen P, Zhou G, Hu J, Chen S, Li Y, Yang Y, Ma J. Constructing two bifunctional tooth-targeting antimicrobial peptides for caries management: an in vitro study. Clin Oral Investig 2024; 29:36. [PMID: 39739049 DOI: 10.1007/s00784-024-06139-7] [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: 09/09/2024] [Accepted: 12/23/2024] [Indexed: 01/02/2025]
Abstract
OBJECTIVES Caries is a significant public health challenge. Herein, novel tooth-targeting antimicrobial peptides (HABPs@AMPs) were developed by combining the antimicrobial peptide DJK-5 with hydroxyapatite (HA) binding peptides, providing a potential new strategy for caries management. MATERIALS AND METHODS The minimal inhibitory concentration (MIC100) and minimal biofilm inhibitory concentration (MBIC100) values of HABPs@AMPs were determined via micro-broth dilution and crystal violet staining. The affinities of the peptides for HA were measured by mass depletion, and the abilities of peptides to inhibit Streptococcus mutans (S. mutans) biofilm formation and kill 3-day-old S. mutans biofilms were evaluated in HA disk and tooth slice biofilm models through confocal laser scanning microscopy. Biocompatibility with human gingival fibroblasts was evaluated via CCK8 assays. RESULTS The best performing peptides, DJK-5@SVA and SVA@DJK-5 exhibited MIC100 and MBIC100 values of 31.25 µg/mL, similar to DJK-5. DJK-5@linker2@YSL had the highest affinity for HA, followed by YSL@DJK-5, DJK-5@linker1@YSL, and DJK-5@SVA. Moreover, the biofilms on HABPs@DJK-5 coated surfaces had more dead bacteria by volume than those in the DJK-5 and SVA groups (p < 0.05). DJK-5@SVA outperformed SVA@DJK-5 and DJK-5 in killing 3-day-old S. mutans biofilms (p < 0.05). With the exception of established biofilms on tooth slices, DJK-5@SVA exhibited greater killing efficiency in the bottom half of the biofilms than in the top half. The CCK-8 assay results confirmed peptides' biocompatibility. CONCLUSIONS DJK-5@SVA with good affinity for HA, has excellent biocompatibility and efficacy against S. mutans biofilms. CLINICAL RELEVANCE HABPs@AMPs with effective inhibitory effects on the growth of S. mutans and biofilm formation, contributing to intraoral targeted application AMPs and providing a new strategy for caries management.
Collapse
Affiliation(s)
- Pei Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hang Kong Road, Wuhan, Hubei Province, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Pan Chen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hang Kong Road, Wuhan, Hubei Province, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Gengyu Zhou
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hang Kong Road, Wuhan, Hubei Province, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Jingyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hang Kong Road, Wuhan, Hubei Province, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Surong Chen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hang Kong Road, Wuhan, Hubei Province, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yingjie Li
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China
| | - Yan Yang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hang Kong Road, Wuhan, Hubei Province, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Jingzhi Ma
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei Province, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hang Kong Road, Wuhan, Hubei Province, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
3
|
Ye Z, Xu Z, Ouyang J, Shi W, Li S, Wang X, Lu B, Wang K, Wang Y. Improving the Stability and Anti-Infective Activity of Sea Turtle AMPs Using Multiple Structural Modification Strategies. J Med Chem 2024; 67:22104-22123. [PMID: 39636182 DOI: 10.1021/acs.jmedchem.4c02039] [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: 12/07/2024]
Abstract
Antimicrobial peptides (AMPs) are regarded as promising candidates for combating antimicrobial resistance. Previously we identified an AMP named Cm-CATH2 from the green sea turtle, which exhibited potent antibacterial activity and attractive potential in application. However, natural AMPs including Cm-CATH2 frequently suffer from structural instability and sensitivity to physiological conditions, limiting their effectiveness. Herein, we explored various strategies to enhance the efficacy and stability of Cm-CATH2, including peptide truncation, non-natural amino acid substitutions, disulfide bond-based cyclization, and stapled peptide techniques. The results demonstrated that the truncated NCM4 significantly improved the antimicrobial capability of Cm-CATH2 while also enhancing its anti-inflammatory and antibiofilm activities with minimal cytotoxicity. Further ornithine-substituted peptide oNCM markedly enhanced the stability of NCM4 without compromising its antimicrobial efficacy. This study successfully designed a lead peptide oNCM with significant development potential, while providing valuable insights into the advantages and limitations associated with diverse strategies for enhancing the stability of AMPs.
Collapse
Affiliation(s)
- Zifan Ye
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhouye Xu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jianhong Ouyang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wenzhuang Shi
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shuangyu Li
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xu Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Binjuan Lu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kang Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yipeng Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, China
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
4
|
Zhu X, Ren M, Zhang Z, Meng F, Li Z, Qin Y, Fang Y, Zhang M. Isolation and characterization of quinoa antimicrobial peptides and its effect on the microbial diversity of fresh apple juice. Food Chem 2024; 469:142536. [PMID: 39729667 DOI: 10.1016/j.foodchem.2024.142536] [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: 08/24/2024] [Revised: 11/30/2024] [Accepted: 12/14/2024] [Indexed: 12/29/2024]
Abstract
This study developed antimicrobial peptides (AMPs) from quinoa with high antibacterial activity and stability by mixed-bacteria fermentation. Furthermore, among 9 peptide fractions purified by membrane separation and chromatography, F1 could effectively inhibit the growth and propagation of bacterial microorganisms in apple juice. Subsequently, F1 identified LC-MS/MS as 95 peptides, molecular weights 494.25 Da to 1253.55 Da, notably, AGAAPE peptide (556.25 Da), negatively charged (-1), highly hydrophobic (50 %), with significant inhibitory effects on both Escherichia coli and Staphylococcus aureus (MIC 5 mg/mL). The antimicrobial mechanism of AGAAPE was determined to damage membrane through hydrogen-bond and hydrophobic interactions, resulting in leakage of intramembrane substances and inhibition of intracellular ATPase activity. Moreover, AGAAPE was pH resistant (pH 4-12), thermally stable (121 °C, 30 min), resistant to salt ion interference (Na+, Ca2+), and protease hydrolysis resistant (neutral protease, pepsin, trypsin). Overall, identifying AMPs from quinoa provides a promising new approach for fresh juice preservation.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Mengyao Ren
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhiwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Fanxing Meng
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zongda Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Yanan Qin
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Yan Fang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China.
| | - Minwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China.
| |
Collapse
|
5
|
Bauer M, Glowacka M, Kamysz W, Kleczkowska P. Marine Peptides: Potential Basic Structures for the Development of Hybrid Compounds as Multitarget Therapeutics for the Treatment of Multifactorial Diseases. Int J Mol Sci 2024; 25:12601. [PMID: 39684313 DOI: 10.3390/ijms252312601] [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: 10/27/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Marine-derived peptides display potent antihypertensive, antioxidant, analgesic and antimicrobial biological effects. Some of them have also been found to have anticancer activity via various mechanisms differing from those of continental organisms. This diversity of properties-together with the peptides' efficacy, which has been confirmed in several in vitro and in vivo studies-make these compounds attractive as functional ingredients in pharmacy, especially in regard to multitarget drugs known as hybrids. Given the possibilities offered by chimeric structures, it is expected that a hybridization strategy based on a marine-derived compound could result in a long-awaited success in the development of new effective compounds to combat a range of complex diseases. However, despite the fact that the biological activity of such new hybrids may exceed that of their parent compounds, there is still an urgent need to carefully determine their potential off-targets and thus possible clinically important side effects. Given the above, the aim of this paper is to provide information on compounds of marine origin with peptide structures and to verify the occurrence and usage of hybrid compounds built from these structures. Furthermore, the authors believe that information presented here will serve to increase public awareness of the new opportunities arising from the combination of hybridization strategies with marine molecules with known structures and biological properties, thereby accelerating the development of effective drug candidates.
Collapse
Affiliation(s)
- Marta Bauer
- Department of Analytical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Magdalena Glowacka
- Institute of Psychology and Human Sciences, WSEI Academy, 20-209 Lublin, Poland
| | - Wojciech Kamysz
- Department of Inorganic Chemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Patrycja Kleczkowska
- Institute of Psychology and Human Sciences, WSEI Academy, 20-209 Lublin, Poland
- Maria Sklodowska-Curie Medical Academy in Warsaw, 03-411 Warsaw, Poland
| |
Collapse
|
6
|
Canè C, Tammaro L, Duilio A, Di Somma A. Investigation of the Mechanism of Action of AMPs from Amphibians to Identify Bacterial Protein Targets for Therapeutic Applications. Antibiotics (Basel) 2024; 13:1076. [PMID: 39596769 PMCID: PMC11591259 DOI: 10.3390/antibiotics13111076] [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: 10/24/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Antimicrobial peptides (AMPs) from amphibians represent a promising source of novel antibacterial agents due to their potent and broad-spectrum antimicrobial activity, which positions them as valid alternatives to conventional antibiotics. This review provides a comprehensive analysis of the mechanisms through which amphibian-derived AMPs exert their effects against bacterial pathogens. We focus on the identification of bacterial protein targets implicated in the action of these peptides and on biological processes altered by the effect of AMPs. By examining recent advances in countering multidrug-resistant bacteria through multi-omics approaches, we elucidate how AMPs interact with bacterial membranes, enter bacterial cells, and target a specific protein. We discuss the implications of these interactions in developing targeted therapies and overcoming antibiotic resistance (ABR). This review aims to integrate the current knowledge on AMPs' mechanisms, identify gaps in our understanding, and propose future directions for research to harness amphibian AMPs in clinical applications.
Collapse
Affiliation(s)
- Carolina Canè
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy; (C.C.); (L.T.)
| | - Lidia Tammaro
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy; (C.C.); (L.T.)
| | - Angela Duilio
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cinthia 4, 80126 Napoli, Italy;
- National Institute of Biostructures and Biosystems (INBB), Via dei Carpegna 19, 00165 Roma, Italy
| | - Angela Di Somma
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cinthia 4, 80126 Napoli, Italy;
| |
Collapse
|
7
|
Yadav N, Chauhan VS. Advancements in peptide-based antimicrobials: A possible option for emerging drug-resistant infections. Adv Colloid Interface Sci 2024; 333:103282. [PMID: 39276418 DOI: 10.1016/j.cis.2024.103282] [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: 02/03/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024]
Abstract
In recent years, multidrug-resistant pathogenic microorganisms (MDROs) have emerged as a severe threat to human health, exhibiting robust resistance to traditional antibiotics. This has created a formidable challenge in modern medicine as we grapple with limited options to combat these resilient bacteria. Despite extensive efforts by scientists to develop new antibiotics targeting these pathogens, the quest for novel antibacterial molecules has become increasingly arduous. Fortunately, nature offers a potential solution in the form of cationic antimicrobial peptides (AMPs) and their synthetic counterparts. AMPs, naturally occurring peptides, have displayed promising efficacy in fighting bacterial infections by disrupting bacterial cell membranes, hindering their survival and reproduction. These peptides, along with their synthetic mimics, present an exciting alternative in combating antibiotic resistance. They hold the potential to emerge as a formidable tool against MDROs, offering hope for improved strategies to protect communities. Extensive research has explored the diversity, history, and structure-properties relationship of AMPs, investigating their amphiphilic nature for membrane disruption and mechanisms of action. However, despite their therapeutic promise, AMPs face several documented limitations. Among these challenges, poor pharmacokinetic properties stand out, impeding the attainment of therapeutic levels in the body. Additionally, some AMPs exhibit toxicity and susceptibility to protease cleavage, leading to a short half-life and reduced efficacy in animal models. These limitations pose obstacles in developing effective treatments based on AMPs. Furthermore, the high manufacturing costs associated with AMPs could significantly hinder their widespread use. In this review, we aim to present experimental and theoretical insights into different AMPs, focusing specifically on antibacterial peptides (ABPs). Our goal is to offer a concise overview of peptide-based drug candidates, drawing from a wide array of literature and peer-reviewed studies. We also explore recent advancements in AMP development and discuss the challenges researchers face in moving these molecules towards clinical trials. Our main objective is to offer a comprehensive overview of current AMP and ABP research to guide the development of more precise and effective therapies for bacterial infections.
Collapse
Affiliation(s)
- Nitin Yadav
- Gandhi Institute of Technology and Management, Gandhi Nagar, Rushikonda, Visakhapatnam, Andhra Pradesh 530045, India; Molecular Medicine, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India; Biotide Solutions LLP, B-23, Geetanjali Enclave, Malviya Nagar, New Delhi 110017, India.
| | - Virander S Chauhan
- Gandhi Institute of Technology and Management, Gandhi Nagar, Rushikonda, Visakhapatnam, Andhra Pradesh 530045, India; Molecular Medicine, International Centre for Genetic Engineering & Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India; Biotide Solutions LLP, B-23, Geetanjali Enclave, Malviya Nagar, New Delhi 110017, India.
| |
Collapse
|
8
|
Li X, Zhou S, Tae HS, Wang S, Li T, Cai W, Jiang T, Adams DJ, Yu R. N-Terminal Capping of the αO-Conotoxin Analogue GeX-2 Improves the Serum Stability and Selectivity toward the Human α9α10 Nicotinic Acetylcholine Receptor. J Med Chem 2024; 67:18400-18411. [PMID: 39361522 DOI: 10.1021/acs.jmedchem.4c01758] [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: 10/05/2024]
Abstract
α9α10 nicotinic acetylcholine receptors (nAChRs) are a promising nonopioid analgesic target, with α9α10 nAChR antagonists showing efficacy against chemotherapy-induced hyperalgesia and allodynia. GeX-2, a potent analgesic conotoxin antagonist of α9α10 nAChRs, has limited serum stability. This study improved GeX-2 stability by capping its N-terminal with fatty acids or polyethylene glycol chains, which enhanced its serum stability but eliminated activity at G protein-coupled γ-aminobutyric acid type B (GABAB) receptor-coupled CaV2.2 channels while preserving activity at α9α10 nAChRs. In vivo, α9α10 nAChRs antagonism alone did not alleviate neuropathic pain, highlighting the importance of GABAB receptor-coupled CaV2.2 channels in GeX-2's antinociceptive effects in the chronic constriction injury rat model. The GeX-2 analogue, with an N-terminal methyl group, showed improved activity and selectivity for α9α10 nAChRs, increased serum half-life, and strong analgesic effects in oxaliplatin-induced cold allodynia models. AlphaFold3 and molecular dynamics simulations provided insights into the binding modes and the effects of N-terminal capping, which informed future peptide therapeutic developments.
Collapse
Affiliation(s)
- Xiao Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Shenglu Zhou
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Shoushi Wang
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266042, China
| | - Tianmiao Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Wenqing Cai
- Shandong Academy of Pharmaceutical Sciences 989 Xinluo Street, Jinan 250101, China
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
9
|
Mitra S, Chen MT, Stedman F, Hernandez J, Kumble G, Kang X, Zhang C, Tang G, Daugherty I, Liu W, Ocloo J, Klucznik KR, Li AA, Heinrich F, Deslouches B, Tristram-Nagle S. How Unnatural Amino Acids in Antimicrobial Peptides Change Interactions with Lipid Model Membranes. J Phys Chem B 2024; 128:9772-9784. [PMID: 39328031 PMCID: PMC11472314 DOI: 10.1021/acs.jpcb.4c04152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
This study investigates the potential of antimicrobial peptides (AMPs) as alternatives to combat antibiotic resistance, with a focus on two AMPs containing unnatural amino acids (UAAs), E2-53R (16 AAs) and LE-54R (14 AAs). In both peptides, valine is replaced by norvaline (Nva), and tryptophan is replaced by 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic). Microbiological studies reveal their potent activity against both Gram-negative (G(-)) and Gram-positive (G(+)) bacteria without any toxicity to eukaryotic cells at test concentrations up to 32 μM. Circular dichroism (CD) spectroscopy indicates that these peptides maintain α-helical structures when interacting with G(-) and G(+) lipid model membranes (LMMs), a feature linked to their efficacy. X-ray diffuse scattering (XDS) demonstrates a softening of G(-), G(+) and eukaryotic (Euk33) LMMs and a nonmonotonic decrease in chain order as a potential determinant for bacterial membrane destabilization. Additionally, XDS finds a significant link between both peptides' interfacial location in G(-) and G(+) LMMs and their efficacy. Neutron reflectometry (NR) confirms the AMP locations determined using XDS. Lack of toxicity in eukaryotic cells may be related to their loss of α-helicity and their hydrocarbon location in Euk33 LMMs. Both AMPs with UAAs offer a novel strategy to wipe out antibiotic-resistant strains while maintaining human cells. These findings are compared with previously published data on E2-35, which consists of the natural amino acids arginine, tryptophan, and valine.
Collapse
Affiliation(s)
- Saheli Mitra
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Mei-Tung Chen
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Francisca Stedman
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Jedidiah Hernandez
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Grace Kumble
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Xi Kang
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Churan Zhang
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Grace Tang
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Ian Daugherty
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Wanqing Liu
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Jeremy Ocloo
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Kevin Raphael Klucznik
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Alexander Anzhi Li
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Frank Heinrich
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
- Center
for Neutron Research, National Institute
of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Berthony Deslouches
- Department
of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Stephanie Tristram-Nagle
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
10
|
Matthyssen T, Li W, Holden JA, Lenzo JC, Hadjigol S, O’Brien-Simpson NM. Dimerization and lysine substitution of melittin have differing effects on bacteria. Front Pharmacol 2024; 15:1443497. [PMID: 39434904 PMCID: PMC11492869 DOI: 10.3389/fphar.2024.1443497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/09/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Melittin is a potent antimicrobial peptide from bee venom that is effective against both Gram-positive and Gram-negative bacteria. However, it is extremely toxic to mammalian cells and, as yet, has no clinical use. Modifications to its amino acid sequence, cyclization, truncation, and dimerization have been attempted in order to reduce its toxicity whilst maintaining its antimicrobial activity. Methods In this study, we targeted the three lysine residues present in melittin and substituted them with lysine homologs containing shorter side chains (ornithine, Orn, diaminobutyric acid, Dab, and diaminopropanoic acid, Dap) and made both parallel and antiparallel melittin dimers to observe how lysine substitution and dimerization affects its activity and toxicity. The antibacterial activity of melittin and its analogs was tested against S. aureus (Gram-positive bacteria) and E. coli (Gram-negative bacteria), and cytotoxicity was tested against the mammalian cell lines HEK293 and H4IIE. Results Overall, dimerization and lysine substitution exhibited improved antimicrobial activity toward E. coli and limited improvement toward S. aureus. However, mammalian cell toxicity was only marginally reduced compared to native melittin. Interestingly, the parallel dimer was found to be marginally more active than the antiparallel dimer, indicating orientation maybe important for activity, although both dimers were less effective than the native and Lys-analog peptides toward S. aureus. Of the Lys substitutions, Dab and Dap improved melittin's activity toward E. coli. Discussion Dimerization and Lys substitution of melittin improved the antimicrobial activity toward Gram-negative bacteria but did not significantly improve its activity toward Gram-positive bacteria. Some analogs also displayed reduced toxicity toward HEK293 and H4IIE cells but overall remained toxic at bactericidal concentrations. Our data indicates that although highly antibacterial, melittin's toxicity is the major drawback in its potential use.
Collapse
Affiliation(s)
- Tamara Matthyssen
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - Wenyi Li
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - James A. Holden
- Melbourne Dental School, Centre for Oral Health Research, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - Jason C. Lenzo
- Western Australian Health Translation Network, Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Sara Hadjigol
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| | - Neil M. O’Brien-Simpson
- ACTV Research Group, Melbourne Dental School, Division of Basic and Clinical Oral Sciences, Royal Dental Hospital and The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
11
|
Bellavita R, Braccia S, Imbò LE, Grieco P, Galdiero S, D'Auria G, Falanga A, Falcigno L. Exploring Fe(III) coordination and membrane interaction of a siderophore-peptide conjugate: Enhancing synergistically the antimicrobial activity. J Inorg Biochem 2024; 259:112658. [PMID: 38964199 DOI: 10.1016/j.jinorgbio.2024.112658] [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: 04/26/2024] [Revised: 06/10/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Many microbes produce siderophores, which are extremely potent weapons capable of stealing iron ions from human tissues, fluids and cells and transferring them into bacteria through their appropriate porins. We have recently designed a multi-block molecule, each block having a dedicated role. The first component is an antimicrobial peptide, whose good effectiveness against some bacterial strains was gradually improved through interactive sequence modifications. Connected to this block is a flexible bio-band, also optimized in length, which terminates in a hydroxyamide unit, a strong metal binder. Thus, the whole molecule brings together two pieces that work synergistically to fight infection. To understand if the peptide unit, although modified with a long tail, preserves the structure and therefore the antimicrobial activity, and to characterize the mechanism of interaction with bio-membrane models mimicking Gram-negative membranes, we performed a set of fluorescence-based experiments and circular dichroism studies, which further supported our design of a combination of two different entities working synergistically. The chelating activity and iron(III) binding of the peptide was confirmed by iron(III) paramagnetic NMR analyses, and through a competitive assay with ethylenediamine-tetra acetic acid by ultraviolet-visible spectroscopy. The complexation parameters, the Michaelis constant K, and the number of sites n, evaluated with spectrophotometric techniques are confirmed by Fe(III) paramagnetic NMR analyses here reported. In conclusion, we showed that the coupling of antimicrobial capabilities with iron-trapping capabilities works well in the treatment of infectious diseases caused by Gram-negative pathogens.
Collapse
Affiliation(s)
- Rosa Bellavita
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Simone Braccia
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Lorenzo Emiliano Imbò
- Department of Agricultural Science, University of Naples 'Federico II', Via Università 100, Portici, 80055 Portici, Italy
| | - Paolo Grieco
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Stefania Galdiero
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Gabriella D'Auria
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| | - Annarita Falanga
- Department of Agricultural Science, University of Naples 'Federico II', Via Università 100, Portici, 80055 Portici, Italy.
| | - Lucia Falcigno
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Via Domenico Montesano 49, 80131 Naples, Italy
| |
Collapse
|
12
|
Peng H, Martens S, Uvyn A, Chen Y, Zhong Z, Louage B, De Geest BG. Exploration of Solid Phase Peptoid Synthesis for the Design of Trifunctional Hapten-Lipid-TLR7/8 Agonist Antibody-Recruiting Oligomers That Combine Innate Effector with Innate Activation Function. ACS APPLIED BIO MATERIALS 2024. [PMID: 39288003 DOI: 10.1021/acsabm.4c00825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The strategic engagement of innate immunity is a promising avenue for cancer treatment. Antibody-recruiting molecules (ARMs) direct endogenous antibodies to target tumor sites, eliciting innate immune effector killing responses. In this study, we report the synthesis of ARMs by employing solid-phase peptoid synthesis to construct three libraries of antibody-recruiting oligomers. Using dinitrophenyl (DNP) as a model hapten and alkyl lipid chains for cell surface anchoring, we tailored oligomers with variations in valency and spatial configuration. Among these, an oligomer design featuring DNP connected to the oligomer backbone through an extended PEG linker and flanked by two lipid motifs emerged as the most effective in antibody recruitment in vitro. This oligomer was further functionalized to include an imidazoquinoline, creating a trifunctional hapten-lipid-TLR7/8 agonist oligomer, and a parallel variant was conjugated with rhodamine, resulting in a trifunctional hapten-lipid-dye oligomer. Upon intratumorally administration in a murine model, these oligomers induced localized immune activation within tumors. Subsequent ex vivo analysis of single-cell suspensions from excised tumors confirmed the enhanced binding of anti-DNP antibodies. These findings underscore the potential of custom-designed ARMs in orchestrating precise immune-mediated tumor targeting and highlight the adaptability of solid-phase synthesis in oligomer design for the design of multifunctional antibody recruiting molecules.
Collapse
Affiliation(s)
- Haixia Peng
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Steven Martens
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Annemiek Uvyn
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Zifu Zhong
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Benoit Louage
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| |
Collapse
|
13
|
Richardson JD, Van Lehn RC. Free Energy Analysis of Peptide-Induced Pore Formation in Lipid Membranes by Bridging Atomistic and Coarse-Grained Simulations. J Phys Chem B 2024; 128:8737-8752. [PMID: 39207202 DOI: 10.1021/acs.jpcb.4c03276] [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: 09/04/2024]
Abstract
Antimicrobial peptides (AMPs) are attractive materials for combating the antimicrobial resistance crisis because they can kill target microbes by directly disrupting cell membranes. Although thousands of AMPs have been discovered, their molecular mechanisms of action are still poorly understood. One broad mechanism for membrane disruption is the formation of membrane-spanning hydrophilic pores which can be stabilized by AMPs. In this study, we use molecular dynamics simulations to investigate the thermodynamics of pore formation in model single-component lipid membranes in the presence of one of three AMPs: aurein 1.2, melittin and magainin 2. To overcome the general challenge of modeling long time scale membrane-related behaviors, including AMP binding, clustering, and pore formation, we develop a generalizable methodology for sampling AMP-induced pore formation. This approach involves the long equilibration of peptides around a pore created with a nucleation collective variable by performing coarse-grained simulations, then backmapping equilibrated AMP-membrane configurations to all-atom resolution. We then perform all-atom simulations to resolve free energy profiles for pore formation while accurately modeling the interplay of lipid-peptide-solvent interactions that dictate pore formation free energies. Using this approach, we quantify free energy barriers for pore formation without direct biases on peptides or whole lipids, allowing us to investigate mechanisms of pore formation for these 3 AMPs that are a consequence of unbiased peptide diffusion and clustering. Further analysis of simulation trajectories then relates variations in pore lining by AMPs, AMP-induced lipid disruptions, and salt bridges between AMPs to the observed pore formation free energies and corresponding mechanisms. This methodology and mechanistic analysis have the potential to generalize beyond the AMPs in this study to improve our understanding of pore formation by AMPs and related antimicrobial materials.
Collapse
Affiliation(s)
- Joshua D Richardson
- Department of Chemical and Biological Engineering, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| | - Reid C Van Lehn
- Department of Chemical and Biological Engineering, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
- Department of Chemistry, University of Wisconsin─Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
14
|
Lawrence N, Handley TNG, de Veer SJ, Harding MD, Andraszek A, Hall L, Raven KD, Duffy S, Avery VM, Craik DJ, Malins LR, McMorran BJ. Enhancing the Intrinsic Antiplasmodial Activity and Improving the Stability and Selectivity of a Tunable Peptide Scaffold Derived from Human Platelet Factor 4. ACS Infect Dis 2024; 10:2899-2912. [PMID: 39087267 PMCID: PMC11320574 DOI: 10.1021/acsinfecdis.4c00276] [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: 04/09/2024] [Revised: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
The control of malaria, a disease caused by Plasmodium parasites that kills over half a million people every year, is threatened by the continual emergence and spread of drug resistance. Therefore, new molecules with different mechanisms of action are needed in the antimalarial drug development pipeline. Peptides developed from host defense molecules are gaining traction as anti-infectives due to theood of inducing drug resistance. Human platelet factor 4 (PF4) has intrinsic activity against P. falciparum, and a macrocyclic helix-loop-helix peptide derived from its active domain recapitulates this activity. In this study, we used a stepwise approach to optimize first-generation PF4-derived internalization peptides (PDIPs) by producing analogues with substitutions to charged and hydrophobic amino acid residues or with modifications to terminal residues including backbone cyclization. We evaluated the in vitro activity of PDIP analogues against P. falciparum compared to their overall helical structure, resistance to breakdown by serum proteases, selective binding to negatively charged membranes, and hemolytic activity. Next, we combined antiplasmodial potency-enhancing substitutions that retained favorable membrane and cell-selective properties onto the most stable scaffold to produce a backbone cyclic PDIP analogue with four-fold improved activity against P. falciparum compared to first-generation peptides. These studies demonstrate the ability to modify PDIP to select for and combine desirable properties and further validate the suitability of this unique peptide scaffold for developing a new molecule class that is distinct from existing antimalarial drugs.
Collapse
Affiliation(s)
- Nicole Lawrence
- Institute
for Molecular Bioscience and Australian Research Council Centre of
Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Thomas N. G. Handley
- Department
of Radiopharmaceutical Sciences, Cancer Imaging, The Peter MacCallum Cancer Centre, Victoria 3000, Australia
- Sir
Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
| | - Simon J. de Veer
- Institute
for Molecular Bioscience and Australian Research Council Centre of
Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Maxim D. Harding
- Institute
for Molecular Bioscience and Australian Research Council Centre of
Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alicja Andraszek
- Institute
for Molecular Bioscience and Australian Research Council Centre of
Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Lachlan Hall
- Institute
for Molecular Bioscience and Australian Research Council Centre of
Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Karoline D. Raven
- The
John Curtin School of Medical Research, College of Health and Medicine, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Sandra Duffy
- Discovery
Biology, Centre for Cellular Phenomics, School of Environment and
Science, Griffith University, Nathan, Queensland 4111, Australia
| | - Vicky M. Avery
- Discovery
Biology, Centre for Cellular Phenomics, School of Environment and
Science, Griffith University, Nathan, Queensland 4111, Australia
| | - David J. Craik
- Institute
for Molecular Bioscience and Australian Research Council Centre of
Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Lara R. Malins
- Research
School of Chemistry and Australian Research Council Centre of Excellence
for Innovations in Peptide and Protein Science, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Brendan J. McMorran
- The
John Curtin School of Medical Research, College of Health and Medicine, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| |
Collapse
|
15
|
Lauwerys L, Beroske L, Solania A, Vangestel C, Miranda A, Van Giel N, Adhikari K, Lambeir AM, Wyffels L, Wolan D, Van der Veken P, Elvas F. Development of caspase-3-selective activity-based probes for PET imaging of apoptosis. EJNMMI Radiopharm Chem 2024; 9:58. [PMID: 39117920 PMCID: PMC11310375 DOI: 10.1186/s41181-024-00291-x] [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: 06/26/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND The cysteine-aspartic acid protease caspase-3 is recognized as the main executioner of apoptosis in cells responding to specific extrinsic and intrinsic stimuli. Caspase-3 represents an interesting biomarker to evaluate treatment response, as many cancer therapies exert their effect by inducing tumour cell death. Previously developed caspase-3 PET tracers were unable to reach routine clinical use due to low tumour uptake or lack of target selectivity, which are two important requirements for effective treatment response evaluation in cancer patients. Therefore, the goal of this study was to develop and preclinically evaluate novel caspase-3-selective activity-based probes (ABPs) for apoptosis imaging. RESULTS A library of caspase-3-selective ABPs was developed for tumour apoptosis detection. In a first attempt, the inhibitor Ac-DW3-KE (Ac-3Pal-Asp-βhLeu-Phe-Asp-KE) was 18F-labelled on the N-terminus to generate a radiotracer that was incapable of adequately detecting an increase in apoptosis in vivo. The inability to effectively detect active caspase-3 in vivo was likely attributable to slow binding, as demonstrated with in vitro inhibition kinetics. Hence, a second generation of caspase-3 selective ABPs was developed based on the Ac-ATS010-KE (Ac-3Pal-Asp-Phe(F5)-Phe-Asp-KE) with greatly improved binding kinetics over Ac-DW3-KE. Our probes based on Ac-ATS010-KE were made by modifying the N-terminus with 6 different linkers. All the linker modifications had limited effect on the binding kinetics, target selectivity, and pharmacokinetic profile in healthy mice. In an in vitro apoptosis model, the least hydrophilic tracer [18F]MICA-316 showed an increased uptake in apoptotic cells in comparison to the control group. Finally, [18F]MICA-316 was tested in an in vivo colorectal cancer model, where it showed a limited tumour uptake and was unable to discriminate treated tumours from the untreated group, despite demonstrating that the radiotracer was able to bind caspase-3 in complex mixtures in vitro. In contrast, the phosphatidylethanolamine (PE)-binding radiotracer [99mTc]Tc-duramycin was able to recognize the increased cell death in the disease model, making it the best performing treatment response assessment tracer developed thus far. CONCLUSIONS In conclusion, a novel library of caspase-3-binding PET tracers retaining similar binding kinetics as the original inhibitor was developed. The most promising tracer, [18F]MICA-316, showed an increase uptake in an in vitro apoptosis model and was able to selectively bind caspase-3 in apoptotic tumour cells. In order to distinguish therapy-responsive from non-responsive tumours, the next generation of caspase-3-selective ABPs will be developed with higher tumour accumulation and in vivo stability.
Collapse
Affiliation(s)
- Louis Lauwerys
- Molecular Imaging and Radiology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Lucas Beroske
- Molecular Imaging and Radiology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Angelo Solania
- Departments of Molecular Medicine and Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Christel Vangestel
- Molecular Imaging and Radiology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Alan Miranda
- Molecular Imaging and Radiology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Nele Van Giel
- Molecular Imaging and Radiology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Karuna Adhikari
- Molecular Imaging and Radiology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Anne-Marie Lambeir
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Leonie Wyffels
- Molecular Imaging and Radiology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium
| | - Dennis Wolan
- Departments of Molecular Medicine and Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Filipe Elvas
- Molecular Imaging and Radiology, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Antwerp, Belgium.
| |
Collapse
|
16
|
Desgagné M, Désilets A, Ferková S, Lepage M, Perreault O, Joushomme A, Lemieux G, Guerrab W, Froehlich U, Comeau C, Sarret P, Leduc R, Boudreault PL. Rational In Silico Design of Selective TMPRSS6 Peptidomimetic Inhibitors via Exploitation of the S2 Subpocket. J Med Chem 2024; 67:12969-12983. [PMID: 39028865 PMCID: PMC11321340 DOI: 10.1021/acs.jmedchem.4c00922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/21/2024]
Abstract
TMPRSS6 is a potential therapeutic target for the treatment of iron overload due to its role in regulating levels of hepcidin. Although potent TMPRSS6 inhibitors have been previously developed, their lack of specificity requires optimization to avoid potential side effects before pursuing preclinical development with in vivo models. Here, using computer-aided drug design based on a TMPRSS6 homology model, we reveal that the S2 position of TMPRSS6 offers a potential avenue to achieve selectivity against other members of the TTSP family. Accordingly, we synthesized novel peptidomimetic molecules containing lipophilic amino acids at the P2 position to exploit this unexplored pocket. This enabled us to identify TMPRSS6-selective small molecules with low nanomolar affinity. Finally, pharmacokinetic parameters were determined, and a compound was found to be potent in cellulo toward its primary target while retaining TTSP-subtype selectivity and showing no signs of alteration in in vitro TEER experiments.
Collapse
Affiliation(s)
- Michael Desgagné
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Antoine Désilets
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Sára Ferková
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Matthieu Lepage
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Olivier Perreault
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Alexandre Joushomme
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Gabriel Lemieux
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Walid Guerrab
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Ulrike Froehlich
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Christian Comeau
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Philippe Sarret
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Richard Leduc
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology-Physiology,
Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
17
|
Camargo L, Gering I, Mastalipour M, Kraemer-Schulien V, Bujnicki T, Willbold D, Coronado MA, Eberle RJ. A Snake Venom Peptide and Its Derivatives Prevent Aβ 42 Aggregation and Eliminate Toxic Aβ 42 Aggregates In Vitro. ACS Chem Neurosci 2024; 15:2600-2611. [PMID: 38957957 PMCID: PMC11258689 DOI: 10.1021/acschemneuro.4c00089] [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: 02/07/2024] [Revised: 05/28/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
Over a century has passed since Alois Alzheimer first described Alzheimer's disease (AD), and since then, researchers have made significant strides in understanding its pathology. One key feature of AD is the presence of amyloid-β (Aβ) peptides, which form amyloid plaques, and therefore, it is a primary target for treatment studies. Naturally occurring peptides have garnered attention for their potential pharmacological benefits, particularly in the central nervous system. In this study, nine peptide derivatives of Crotamine, a polypeptide from Crotalus durissus terrificus Rattlesnake venom, as well as one d-enantiomer, were evaluated for their ability to modulate Aβ42 aggregation through various assays such as ThT, QIAD, SPR, and sFIDA. All tested peptides were able to decrease Aβ42 aggregation and eliminate Aβ42 aggregates. Additionally, all of the peptides showed an affinity for Aβ42. This study is the first to describe the potential of crotamine derivative peptides against Aβ42 aggregation and to identify a promising d-peptide that could be used as an effective pharmacological tool against AD in the future.
Collapse
Affiliation(s)
- Luana
Cristina Camargo
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Ian Gering
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Mohammadamin Mastalipour
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Victoria Kraemer-Schulien
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Tuyen Bujnicki
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
| | - Dieter Willbold
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Mônika A. Coronado
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Raphael J. Eberle
- Institute
of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich 52428, Germany
- Faculty
of Mathematics and Natural Sciences, Institute of Physical Biology, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| |
Collapse
|
18
|
Loffredo M, Casciaro B, Bellavita R, Troiano C, Brancaccio D, Cappiello F, Merlino F, Galdiero S, Fabrizi G, Grieco P, Stella L, Carotenuto A, Mangoni ML. Strategic Single-Residue Substitution in the Antimicrobial Peptide Esc(1-21) Confers Activity against Staphylococcus aureus, Including Drug-Resistant and Biofilm Phenotype. ACS Infect Dis 2024; 10:2403-2418. [PMID: 38848266 PMCID: PMC11250030 DOI: 10.1021/acsinfecdis.4c00130] [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: 02/19/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024]
Abstract
Staphylococcus aureus, a bacterium resistant to multiple drugs, is a significant cause of illness and death worldwide. Antimicrobial peptides (AMPs) provide an excellent potential strategy to cope with this threat. Recently, we characterized a derivative of the frog-skin AMP esculentin-1a, Esc(1-21) (1) that is endowed with potent activity against Gram-negative bacteria but poor efficacy against Gram-positive strains. In this study, three analogues of peptide 1 were designed by replacing Gly8 with α-aminoisobutyric acid (Aib), Pro, and dPro (2-4, respectively). The single substitution Gly8 → Aib8 in peptide 2 makes it active against the planktonic form of Gram-positive bacterial strains, especially Staphylococcus aureus, including multidrug-resistant clinical isolates, with an improved biostability without resulting in cytotoxicity to mammalian cells. Moreover, peptide 2 showed a higher antibiofilm activity than peptide 1 against both reference and clinical isolates of S. aureus. Peptide 2 was also able to induce rapid bacterial killing, suggesting a membrane-perturbing mechanism of action. Structural analysis of the most active peptide 2 evidenced that the improved biological activity of peptide 2 is the consequence of a combination of higher biostability, higher α helical content, and ability to reduce membrane fluidity and to adopt a distorted helix, bent in correspondence of Aib8. Overall, this study has shown how a strategic single amino acid substitution is sufficient to enlarge the spectrum of activity of the original peptide 1, and improve its biological properties for therapeutic purposes, thus paving the way to optimize AMPs for the development of new broad-spectrum anti-infective agents.
Collapse
Affiliation(s)
- Maria
Rosa Loffredo
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Bruno Casciaro
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Rosa Bellavita
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Cassandra Troiano
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133 Rome, Italy
| | - Diego Brancaccio
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Floriana Cappiello
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Francesco Merlino
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Stefania Galdiero
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Giancarlo Fabrizi
- Department
of Chemistry and Technology of Drugs, “Department of Excellence
2018−2022”, Sapienza University
of Rome, 00185 Rome, Italy
| | - Paolo Grieco
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Lorenzo Stella
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133 Rome, Italy
| | - Alfonso Carotenuto
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Maria Luisa Mangoni
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| |
Collapse
|
19
|
Selvaraj SP, Lin KH, Lin WC, You MF, Li TL, Chen JY. Rejuvenation of Meropenem by Conjugation with Tilapia Piscidin-4 Peptide Targeting NDM-1 Escherichia coli. ACS OMEGA 2024; 9:29756-29764. [PMID: 39005813 PMCID: PMC11238198 DOI: 10.1021/acsomega.4c03352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/13/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024]
Abstract
Gram-negative pathogens that produce β-lactamases pose a serious public health threat as they can render β-lactam antibiotics inactive via hydrolysis. This action contributes to the waning effectiveness of clinical antibiotics and creates an urgent need for new antimicrobials. Antimicrobial peptides (AMPs) exhibiting multimodal functions serve as a potential source in spite of a few limitations. Thus, the conjugation of conventional antibiotics with AMPs may be an effective strategy to leverage the advantages of each component. In this study, we conjugated meropenem to the AMP Tilapia piscidin 4 (TP4) using a typical coupling reaction. The conjugate was characterized by using HPLC-MS, HR-MS, and MS-MS fragmentation analysis. It was then evaluated in terms of antibacterial potency, hemolysis, and cytotoxicity toward RAW264.7 and CCD-966SK cell lines. The conjugation of meropenem with TP4 significantly reduced the cytotoxicity compared to TP4. Conjugation of unprotected TP4 with meropenem resulted in cross-linking at the N-terminal and lysine sites. The structural activity relationship of the two isomers of the TP4-meropenem conjugate was investigated. Both the isomers showed notable antibacterial activities against NDM-1 Escherichia coli and reduced red blood cell hemolysis as compared to TP4. Lysine conjugate (TP4-K-Mero) showed lesser hemolysis than the N-terminal conjugate (TP4-N-Mero). Molecular modeling further revealed that the conjugates can bind to lipopolysaccharides and inhibit NDM-1 β-lactamase. Together, these data show that conjugation of antibiotics with AMP can be a feasible approach to increase the therapeutic profile and effectively target multidrug-resistant pathogens. Furthermore, antibiotic conjugation at different AMP sites tends to show unique biological properties.
Collapse
Affiliation(s)
- Sanjay Prasad Selvaraj
- Molecular and Biological Agricultural Science Program, Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
| | - Kuan-Hung Lin
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wen-Chun Lin
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Rd, Jiaushi, Ilan 262, Taiwan
| | - Ming-Feng You
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Rd, Jiaushi, Ilan 262, Taiwan
| | - Tsung-Lin Li
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung City 402, Taiwan
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Rd, Jiaushi, Ilan 262, Taiwan
- The iEGG and Animal Biotechnology Center and the Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
20
|
Groover KE, Randall JR, Davies BW. Development of a Selective and Stable Antimicrobial Peptide. ACS Infect Dis 2024; 10:2151-2160. [PMID: 38712889 PMCID: PMC11185160 DOI: 10.1021/acsinfecdis.4c00142] [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: 02/23/2024] [Revised: 04/17/2024] [Accepted: 04/26/2024] [Indexed: 05/08/2024]
Abstract
Antimicrobial peptides (AMPs) are presented as potential scaffolds for antibiotic development due to their desirable qualities including broad-spectrum activity, rapid action, and general lack of susceptibility to current resistance mechanisms. However, they often lose antibacterial activity under physiological conditions and/or display mammalian cell toxicity, which limits their potential use. Identification of AMPs that overcome these barriers will help develop rules for how this antibacterial class can be developed to treat infection. Here we describe the development of our novel synthetic AMP, from discovery through in vivo application. Our evolved AMP, DTr18-dab, has broad-spectrum antibacterial activity and is nonhemolytic. It is active against planktonic bacteria and biofilm, is unaffected by colistin resistance, and importantly is active in both human serum and a Galleria mellonella infection model. Several modifications, including the incorporation of noncanonical amino acids, were used to arrive at this robust sequence. We observed that the impact on antibacterial activity with noncanonical amino acids was dependent on assay conditions and therefore not entirely predictable. Overall, our results demonstrate how a relatively weak lead can be developed into a robust AMP with qualities important for potential therapeutic translation.
Collapse
Affiliation(s)
- Kyra E. Groover
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Justin R. Randall
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Bryan W. Davies
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
- John
Ring LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
21
|
Enninful GN, Kuppusamy R, Tiburu EK, Kumar N, Willcox MDP. Non-canonical amino acid bioincorporation into antimicrobial peptides and its challenges. J Pept Sci 2024; 30:e3560. [PMID: 38262069 DOI: 10.1002/psc.3560] [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: 06/06/2023] [Revised: 10/01/2023] [Accepted: 11/14/2023] [Indexed: 01/25/2024]
Abstract
The rise of antimicrobial resistance and multi-drug resistant pathogens has necessitated explorations for novel antibiotic agents as the discovery of conventional antibiotics is becoming economically less viable and technically more challenging for biopharma. Antimicrobial peptides (AMPs) have emerged as a promising alternative because of their particular mode of action, broad spectrum and difficulty that microbes have in becoming resistant to them. The AMPs bacitracin, gramicidin, polymyxins and daptomycin are currently used clinically. However, their susceptibility to proteolytic degradation, toxicity profile, and complexities in large-scale manufacture have hindered their development. To improve their proteolytic stability, methods such as integrating non-canonical amino acids (ncAAs) into their peptide sequence have been adopted, which also improves their potency and spectrum of action. The benefits of ncAA incorporation have been made possible by solid-phase peptide synthesis. However, this method is not always suitable for commercial production of AMPs because of poor yield, scale-up difficulties, and its non-'green' nature. Bioincorporation of ncAA as a method of integration is an emerging field geared towards tackling the challenges of solid-phase synthesis as a green, cheaper, and scalable alternative for commercialisation of AMPs. This review focusses on the bioincorporation of ncAAs; some challenges associated with the methods are outlined, and notes are given on how to overcome these challenges. The review focusses particularly on addressing two key challenges: AMP cytotoxicity towards microbial cell factories and the uptake of ncAAs that are unfavourable to them. Overcoming these challenges will draw us closer to a greater yield and an environmentally friendly and sustainable approach to make AMPs more druggable.
Collapse
Affiliation(s)
| | - Rajesh Kuppusamy
- University of New South Wales, Kensington, New South Wales, Australia
| | | | - Naresh Kumar
- University of New South Wales, Kensington, New South Wales, Australia
| | - Mark D P Willcox
- University of New South Wales, Kensington, New South Wales, Australia
| |
Collapse
|
22
|
Alves PM, Barrias CC, Gomes P, Martins MCL. How can biomaterial-conjugated antimicrobial peptides fight bacteria and be protected from degradation? Acta Biomater 2024; 181:98-116. [PMID: 38697382 DOI: 10.1016/j.actbio.2024.04.043] [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: 01/12/2024] [Revised: 03/19/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
The emergence of antibiotic-resistant bacteria is a serious threat to public health. Antimicrobial peptides (AMP) are a powerful alternative to antibiotics due to their low propensity to induce bacterial resistance. However, cytotoxicity and short half-lives have limited their clinical translation. To overcome these problems, AMP conjugation has gained relevance in the biomaterials field. Nevertheless, few studies describe the influence of conjugation on enzymatic protection, mechanism of action and antimicrobial efficacy. This review addresses this gap by providing a detailed comparison between conjugated and soluble AMP. Additionally, commonly employed chemical reactions and factors to consider when promoting AMP conjugation are reviewed. The overall results suggested that AMP conjugated onto biomaterials are specifically protected from degradation by trypsin and/or pepsin. However, sometimes, their antimicrobial efficacy was reduced. Due to limited conformational freedom in conjugated AMP, compared to their soluble forms, they appear to act initially by creating small protuberances on bacterial membranes that may lead to the alteration of membrane potential and/or formation of holes, triggering cell death. Overall, AMP conjugation onto biomaterials is a promising strategy to fight infection, particularly associated to the use of medical devices. Nonetheless, some details need to be addressed before conjugated AMP reach clinical practice. STATEMENT OF SIGNIFICANCE: Covalent conjugation of antimicrobial peptides (AMP) has been one of the most widely used strategies by bioengineers, in an attempt to not only protect AMP from proteolytic degradation, but also to prolong their residence time at the target tissue. However, an explanation for the mode of action of conjugated AMP is still lacking. This review extensively gathers works on AMP conjugation and puts forward a mechanism of action for AMP when conjugated onto biomaterials. The implications of AMP conjugation on antimicrobial activity, cytotoxicity and resistance to proteases are all discussed. A thorough review of commonly employed chemical reactions for this conjugation is also provided. Finally, details that need to be addressed for conjugated AMP to reach clinical practice are discussed.
Collapse
Affiliation(s)
- Pedro M Alves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal
| | - Cristina C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Paula Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal
| | - M Cristina L Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal.
| |
Collapse
|
23
|
Kravchenko SV, Domnin PA, Grishin SY, Zakhareva AP, Zakharova AA, Mustaeva LG, Gorbunova EY, Kobyakova MI, Surin AK, Poshvina DV, Fadeev RS, Azev VN, Ostroumova OS, Ermolaeva SA, Galzitskaya OV. Optimizing Antimicrobial Peptide Design: Integration of Cell-Penetrating Peptides, Amyloidogenic Fragments, and Amino Acid Residue Modifications. Int J Mol Sci 2024; 25:6030. [PMID: 38892216 PMCID: PMC11173194 DOI: 10.3390/ijms25116030] [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: 04/27/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The escalating threat of multidrug-resistant pathogens necessitates innovative approaches to combat infectious diseases. In this study, we examined peptides R23FS*, V31KS*, and R44KS*, which were engineered to include an amyloidogenic fragment sourced from the S1 protein of S. aureus, along with one or two cell-penetrating peptide (CPP) components. We assessed the antimicrobial efficacy of these peptides in a liquid medium against various strains of both Gram-positive bacteria, including S. aureus (209P and 129B strains), MRSA (SA 180 and ATCC 43300 strains), and B. cereus (strain IP 5832), and Gram-negative bacteria such as P. aeruginosa (ATCC 28753 and 2943 strains) and E. coli (MG1655 and K12 strains). Peptides R23FS*, V31KS*, and R44KS* exhibited antimicrobial activity comparable to gentamicin and meropenem against all tested bacteria at concentrations ranging from 24 to 48 μM. The peptides showed a stronger antimicrobial effect against B. cereus. Notably, peptide R44KS* displayed high efficacy compared to peptides R23FS* and V31KS*, particularly evident at lower concentrations, resulting in significant inhibition of bacterial growth. Furthermore, modified peptides V31KS* and R44KS* demonstrated enhanced inhibitory effects on bacterial growth across different strains compared to their unmodified counterparts V31KS and R44KS. These results highlight the potential of integrating cell-penetrating peptides, amyloidogenic fragments, and amino acid residue modifications to advance the innovation in the field of antimicrobial peptides, thereby increasing their effectiveness against a broad spectrum of pathogens.
Collapse
Affiliation(s)
- Sergey V. Kravchenko
- Institute of Environmental and Agricultural Biology (X-BIO), Tyumen State University, 625003 Tyumen, Russia; (S.V.K.); (A.P.Z.); (D.V.P.)
| | - Pavel A. Domnin
- Biology Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia;
- Gamaleya Research Centre of Epidemiology and Microbiology, 123098 Moscow, Russia;
| | - Sergei Y. Grishin
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (S.Y.G.); (A.K.S.)
| | - Alena P. Zakhareva
- Institute of Environmental and Agricultural Biology (X-BIO), Tyumen State University, 625003 Tyumen, Russia; (S.V.K.); (A.P.Z.); (D.V.P.)
| | - Anastasiia A. Zakharova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (A.A.Z.); (O.S.O.)
| | - Leila G. Mustaeva
- The Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.M.); (E.Y.G.); (V.N.A.)
| | - Elena Y. Gorbunova
- The Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.M.); (E.Y.G.); (V.N.A.)
| | - Margarita I. Kobyakova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (M.I.K.); (R.S.F.)
- Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630060 Novosibirsk, Russia
| | - Alexey K. Surin
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (S.Y.G.); (A.K.S.)
- The Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.M.); (E.Y.G.); (V.N.A.)
- State Research Center for Applied Microbiology and Biotechnology, 142279 Obolensk, Russia
| | - Darya V. Poshvina
- Institute of Environmental and Agricultural Biology (X-BIO), Tyumen State University, 625003 Tyumen, Russia; (S.V.K.); (A.P.Z.); (D.V.P.)
| | - Roman S. Fadeev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (M.I.K.); (R.S.F.)
| | - Viacheslav N. Azev
- The Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia; (L.G.M.); (E.Y.G.); (V.N.A.)
| | - Olga S. Ostroumova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (A.A.Z.); (O.S.O.)
| | | | - Oxana V. Galzitskaya
- Gamaleya Research Centre of Epidemiology and Microbiology, 123098 Moscow, Russia;
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia; (S.Y.G.); (A.K.S.)
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Russia; (M.I.K.); (R.S.F.)
| |
Collapse
|
24
|
Sigal M, Matsumoto S, Beattie A, Katoh T, Suga H. Engineering tRNAs for the Ribosomal Translation of Non-proteinogenic Monomers. Chem Rev 2024; 124:6444-6500. [PMID: 38688034 PMCID: PMC11122139 DOI: 10.1021/acs.chemrev.3c00894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/21/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Ribosome-dependent protein biosynthesis is an essential cellular process mediated by transfer RNAs (tRNAs). Generally, ribosomally synthesized proteins are limited to the 22 proteinogenic amino acids (pAAs: 20 l-α-amino acids present in the standard genetic code, selenocysteine, and pyrrolysine). However, engineering tRNAs for the ribosomal incorporation of non-proteinogenic monomers (npMs) as building blocks has led to the creation of unique polypeptides with broad applications in cellular biology, material science, spectroscopy, and pharmaceuticals. Ribosomal polymerization of these engineered polypeptides presents a variety of challenges for biochemists, as translation efficiency and fidelity is often insufficient when employing npMs. In this Review, we will focus on the methodologies for engineering tRNAs to overcome these issues and explore recent advances both in vitro and in vivo. These efforts include increasing orthogonality, recruiting essential translation factors, and creation of expanded genetic codes. After our review on the biochemical optimizations of tRNAs, we provide examples of their use in genetic code manipulation, with a focus on the in vitro discovery of bioactive macrocyclic peptides containing npMs. Finally, an analysis of the current state of tRNA engineering is presented, along with existing challenges and future perspectives for the field.
Collapse
Affiliation(s)
- Maxwell Sigal
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Satomi Matsumoto
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Adam Beattie
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takayuki Katoh
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroaki Suga
- Department of Chemistry,
Graduate School of Science, The University
of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
25
|
Guha S, Cristy SA, Buda De Cesare G, Cruz MR, Lorenz MC, Garsin DA. Optimization of the antifungal properties of the bacterial peptide EntV by variant analysis. mBio 2024; 15:e0057024. [PMID: 38587425 PMCID: PMC11077972 DOI: 10.1128/mbio.00570-24] [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: 02/29/2024] [Accepted: 03/13/2024] [Indexed: 04/09/2024] Open
Abstract
Fungal resistance to commonly used medicines is a growing public health threat, and there is a dire need to develop new classes of antifungals. We previously described a peptide produced by Enterococcus faecalis, EntV, that restricts Candida albicans to a benign form rather than having direct fungicidal activity. Moreover, we showed that one 12-amino acid (aa) alpha helix of this peptide retained full activity, with partial activity down to the 10aa alpha helix. Using these peptides as a starting point, the current investigation sought to identify the critical features necessary for antifungal activity and to screen for new variants with enhanced activity using both biofilm and C. elegans infection assays. First, the short peptides were screened for residues with critical activity by generating alanine substitutions. Based on this information, we used synthetic molecular evolution (SME) to rationally vary the specific residues of the 10aa variant in combination to generate a library that was screened to identify variants with more potent antifungal activity than the parent template. Five gain-of-function peptides were identified. Additionally, chemical modifications to the peptides to increase stability, including substitutions of D-amino acids and hydrocarbon stapling, were investigated. The most promising peptides were additionally tested in mouse models of oropharyngeal and systemic candidiasis where their efficacy in preventing infection was demonstrated. The expectation is that these discoveries will contribute to the development of new therapeutics in the fight against antimicrobial resistant fungi. IMPORTANCE Since the early 1980s, the incidence of disseminated life-threatening fungal infections has been on the rise. Worldwide, Candida and Cryptococcus species are among the most common agents causing these infections. Simultaneously, with this rise of clinical incidence, there has also been an increased prevalence of antifungal resistance, making treatment of these infections very difficult. For example, there are now strains of Candida auris that are resistant to all three classes of currently used antifungal drugs. In this study, we report on a strategy that allows for the development of novel antifungal agents by using synthetic molecular evolution. These discoveries demonstrate that the enhancement of antifungal activity from naturally occurring peptides is possible and can result in clinically relevant agents that have efficacy in multiple in vivo models as well as the potential for broad-spectrum activity.
Collapse
Affiliation(s)
- Shantanu Guha
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shane A. Cristy
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Giuseppe Buda De Cesare
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Melissa R. Cruz
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Michael C. Lorenz
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Danielle A. Garsin
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
26
|
Porras-Dominguez J, Lothier J, Limami AM, Tcherkez G. d-amino acids metabolism reflects the evolutionary origin of higher plants and their adaptation to the environment. PLANT, CELL & ENVIRONMENT 2024; 47:1503-1512. [PMID: 38251436 DOI: 10.1111/pce.14826] [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: 06/22/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
d-amino acids are the d stereoisomers of the common l-amino acids found in proteins. Over the past two decades, the occurrence of d-amino acids in plants has been reported and circumstantial evidence for a role in various processes, including interaction with soil microorganisms or interference with cellular signalling, has been provided. However, examples are not numerous and d-amino acids can also be detrimental, some of them inhibiting growth and development. Thus, the persistence of d-amino acid metabolism in plants is rather surprising, and the evolutionary origins of d-amino acid metabolism are currently unclear. Systemic analysis of sequences associated with d-amino acid metabolism enzymes shows that they are not simply inherited from cyanobacterial metabolism. In fact, the history of plant d-amino acid metabolism enzymes likely involves multiple steps, cellular compartments, gene transfers and losses. Regardless of evolutionary steps, enzymes of d-amino acid metabolism, such as d-amino acid transferases or racemases, have been retained by higher plants and have not simply been eliminated, so it is likely that they fulfil important metabolic roles such as serine, folate or plastid peptidoglycan metabolism. We suggest that d-amino acid metabolism may have been critical to support metabolic functions required during the evolution of land plants.
Collapse
Affiliation(s)
- Jaime Porras-Dominguez
- Institut de Recherche en Horticulture et Semences, INRAe, Université d'Angers, Beaucouzé, France
| | - Jérémy Lothier
- Institut de Recherche en Horticulture et Semences, INRAe, Université d'Angers, Beaucouzé, France
| | - Anis M Limami
- Institut de Recherche en Horticulture et Semences, INRAe, Université d'Angers, Beaucouzé, France
| | - Guillaume Tcherkez
- Institut de Recherche en Horticulture et Semences, INRAe, Université d'Angers, Beaucouzé, France
- Research School of Biology, Australian National University, Canberra, Australia
| |
Collapse
|
27
|
Cresti L, Cappello G, Pini A. Antimicrobial Peptides towards Clinical Application-A Long History to Be Concluded. Int J Mol Sci 2024; 25:4870. [PMID: 38732089 PMCID: PMC11084544 DOI: 10.3390/ijms25094870] [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: 03/27/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Antimicrobial peptides (AMPs) are molecules with an amphipathic structure that enables them to interact with bacterial membranes. This interaction can lead to membrane crossing and disruption with pore formation, culminating in cell death. They are produced naturally in various organisms, including humans, animals, plants and microorganisms. In higher animals, they are part of the innate immune system, where they counteract infection by bacteria, fungi, viruses and parasites. AMPs can also be designed de novo by bioinformatic approaches or selected from combinatorial libraries, and then produced by chemical or recombinant procedures. Since their discovery, AMPs have aroused interest as potential antibiotics, although few have reached the market due to stability limits or toxicity. Here, we describe the development phase and a number of clinical trials of antimicrobial peptides. We also provide an update on AMPs in the pharmaceutical industry and an overall view of their therapeutic market. Modifications to peptide structures to improve stability in vivo and bioavailability are also described.
Collapse
Affiliation(s)
- Laura Cresti
- Medical Biotechnology Department, University of Siena, Via A Moro 2, 53100 Siena, Italy; (G.C.); (A.P.)
| | - Giovanni Cappello
- Medical Biotechnology Department, University of Siena, Via A Moro 2, 53100 Siena, Italy; (G.C.); (A.P.)
| | - Alessandro Pini
- Medical Biotechnology Department, University of Siena, Via A Moro 2, 53100 Siena, Italy; (G.C.); (A.P.)
- SetLance srl, Via Fiorentina 1, 53100 Siena, Italy
- Laboratory of Clinical Pathology, Santa Maria alle Scotte University Hospital, 53100 Siena, Italy
| |
Collapse
|
28
|
Yang P, Mao W, Zhang J, Yang Y, Zhang F, Ouyang X, Li B, Wu X, Ba Z, Ran K, Tian Y, Liu H, Zhang Y, Gou S, Zhong C, Ni J. A novel antimicrobial peptide with broad-spectrum and exceptional stability derived from the natural peptide Brevicidine. Eur J Med Chem 2024; 269:116337. [PMID: 38537511 DOI: 10.1016/j.ejmech.2024.116337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 04/07/2024]
Abstract
The global issue of antibiotic resistance is increasingly severe, highlighting the urgent necessity for the development of new antibiotics. Brevicidine, a natural cyclic lipopeptide, exhibits remarkable antimicrobial activity against Gram-negative bacteria. In this study, a comprehensive structure-activity relationship of Brevicidine was investigated through 20 newly synthesized cyclic lipopeptide analogs, resulting in the identification of an optimal linear analog 22. The sequence of analog 22 consisted of five d-amino acids and four non-natural amino acid 2,5-diaminovaleric acid (Orn) and conjugated with decanoic acid at N-terminal. Compared to Brevicidine, analog 22 was easier to synthesize, and exerted broad spectrum antimicrobial activity and excellent stability (t1/2 = 40.98 h). Additionally, analog 22 demonstrated a rapid bactericidal effect by permeating non-specifically through the bacterial membranes, thereby minimizing the likelihood of inducing resistance. Moreover, it exhibited remarkable efficacy in combating bacterial biofilms and reversing bacterial resistance to conventional antibiotics. Furthermore, it effectively suppressed the growth of bacteria in vital organs of mice infected with S. aureus ATCC 25923. In conclusion, analog 22 may represent a potential antimicrobial peptide for further optimization.
Collapse
Affiliation(s)
- Ping Yang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Wenbo Mao
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Jingying Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Yinyin Yang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Fangyan Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Xu Ouyang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Beibei Li
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Xiaoyan Wu
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Zufang Ba
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Kaixin Ran
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Yali Tian
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China
| | - Hui Liu
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Yun Zhang
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Sanhu Gou
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Chao Zhong
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Jingman Ni
- Institute of Pharmaceutics, School of Pharmacy, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou, 730000, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
29
|
Tan T, Hou Y, Shi J, Wang B, Zhang Y. Biostable hydrogels consisting of hybrid β-sheet fibrils assembled by a pair of enantiomeric peptides. Mater Today Bio 2024; 25:100961. [PMID: 38304341 PMCID: PMC10831280 DOI: 10.1016/j.mtbio.2024.100961] [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: 10/16/2023] [Revised: 12/22/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
The assembly of chiral peptides facilitates the formation of diverse supramolecular structures with unique physicochemical and biological properties. However, the effects of chirality on peptide assembly and resulting hydrogel properties remain underexplored. In this study, we systematically investigated the assembly propensity, morphology, and biostability of mixture of a pair of enantiomeric peptides LELCLALFLF (ECF-5) and DEDCDADFDF (ecf-5) at various ratios. Results indicate the development of β-sheet fibrils, ultimately leading to the formation of self-supporting hybrid hydrogels. The hydrogel formed at a ratio of 1:1 exhibits a significantly lower storage modulus (G') than of the ratios of 0:1, 1:3, 3:1 and 1:0 (nD/nL; same below). Kink-separated fragments of approximately 100 nm in length predominate at ratios of 1:3 and 3:1, compared with the smooth fibrils at other ratios, probably attributed to an alternating arrangement of the co-assembled and self-assembled peptide fragments. The introduction of ecf-5 to the hybrid hydrogels improves resistance to proteolytic digestion and maintains commendable biocompatibility in both MIN6 and HUVECs cells. These findings provide valuable insights into the development of hydrogels with tailored properties, positing them potential scaffolds for 3D cell culture and tissue engineering.
Collapse
Affiliation(s)
- Tingyuan Tan
- Research Institute of Interdisciplinary Sciences & School of Materials Science and Engineering, Dongguan University of Technology, Dongguan, 523808, China
| | - Yangqian Hou
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Jiali Shi
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Biao Wang
- Research Institute of Interdisciplinary Sciences & School of Materials Science and Engineering, Dongguan University of Technology, Dongguan, 523808, China
| | - Yi Zhang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| |
Collapse
|
30
|
Sara M, Yasir M, Kalaiselvan P, Hui A, Kuppusamy R, Kumar N, Chakraborty S, Yu TT, Wong EHH, Molchanova N, Jenssen H, Lin JS, Barron AE, Willcox M. The activity of antimicrobial peptoids against multidrug-resistant ocular pathogens. Cont Lens Anterior Eye 2024; 47:102124. [PMID: 38341309 PMCID: PMC11024869 DOI: 10.1016/j.clae.2024.102124] [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: 07/27/2023] [Revised: 01/11/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Ocular infections caused by antibiotic-resistant pathogens can result in partial or complete vision loss. The development of pan-resistant microbial strains poses a significant challenge for clinicians as there are limited antimicrobial options available. Synthetic peptoids, which are sequence-specific oligo-N-substituted glycines, offer potential as alternative antimicrobial agents to target multidrug-resistant bacteria. METHODS The antimicrobial activity of synthesised peptoids against multidrug-resistant (MDR) ocular pathogens was evaluated using the microbroth dilution method. Hemolytic propensity was assessed using mammalian erythrocytes. Peptoids were also incubated with proteolytic enzymes, after which their minimum inhibitory activity against bacteria was re-evaluated. RESULTS Several alkylated and brominated peptoids showed good inhibitory activity against multidrug-resistant Pseudomonas aeruginosa strains at concentrations of ≤15 μg mL-1 (≤12 µM). Similarly, most brominated compounds inhibited the growth of methicillin-resistant Staphylococcus aureus at 1.9 to 15 μg mL-1 (12 µM). The N-terminally alkylated peptoids caused less toxicity to erythrocytes. The peptoid denoted as TM5 had a high therapeutic index, being non-toxic to either erythrocytes or corneal epithelial cells, even at 15 to 22 times its MIC. Additionally, the peptoids were resistant to protease activity. CONCLUSIONS Peptoids studied here demonstrated potent activity against various multidrug-resistant ocular pathogens. Their properties make them promising candidates for controlling vision-related morbidity associated with eye infections by antibiotic-resistant strains.
Collapse
Affiliation(s)
- Manjulatha Sara
- School of Optometry and Vision Science, UNSW Sydney, Australia.
| | - Muhammad Yasir
- School of Optometry and Vision Science, UNSW Sydney, Australia
| | | | - Alex Hui
- School of Optometry and Vision Science, UNSW Sydney, Australia; Centre for Ocular Research and Education, University of Waterloo, Canada
| | - Rajesh Kuppusamy
- School of Optometry and Vision Science, UNSW Sydney, Australia; School of Chemistry, UNSW Sydney, Australia
| | | | | | - Tsz Tin Yu
- School of Chemistry, UNSW Sydney, Australia
| | | | - Natalia Molchanova
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 4720, USA
| | - Håvard Jenssen
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| | - Jennifer S Lin
- Department of Bioengineering, School of Medicine & School of Engineering, Stanford University, Stanford, CA 9430, USA
| | - Annelise E Barron
- Department of Bioengineering, School of Medicine & School of Engineering, Stanford University, Stanford, CA 9430, USA
| | - Mark Willcox
- School of Optometry and Vision Science, UNSW Sydney, Australia.
| |
Collapse
|
31
|
Karapetian M, Alimbarashvili E, Vishnepolsky B, Gabrielian A, Rosenthal A, Hurt DE, Tartakovsky M, Mchedlishvili M, Arsenadze D, Pirtskhalava M, Zaalishvili G. Evaluation of the synergistic potential and mechanisms of action for de novo designed cationic antimicrobial peptides. Heliyon 2024; 10:e27852. [PMID: 38560672 PMCID: PMC10979160 DOI: 10.1016/j.heliyon.2024.e27852] [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: 06/23/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Antimicrobial peptides (AMPs) have emerged as promising candidates in combating antimicrobial resistance - a growing issue in healthcare. However, to develop AMPs into effective therapeutics, a thorough analysis and extensive investigations are essential. In this study, we employed an in silico approach to design cationic AMPs de novo, followed by their experimental testing. The antibacterial potential of de novo designed cationic AMPs, along with their synergistic properties in combination with conventional antibiotics was examined. Furthermore, the effects of bacterial inoculum density and metabolic state on the antibacterial activity of AMPs were evaluated. Finally, the impact of several potent AMPs on E. coli cell envelope and genomic DNA integrity was determined. Collectively, this comprehensive analysis provides insights into the unique characteristics of cationic AMPs.
Collapse
Affiliation(s)
- Margarita Karapetian
- Laboratory of Chromatin Biology, Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 240 David Aghmashenebeli Alley, 0159, Tbilisi, Georgia
| | - Evgenia Alimbarashvili
- Laboratory of Chromatin Biology, Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 240 David Aghmashenebeli Alley, 0159, Tbilisi, Georgia
- Ivane Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Boris Vishnepolsky
- Ivane Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Andrei Gabrielian
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alex Rosenthal
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Darrell E. Hurt
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael Tartakovsky
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mariam Mchedlishvili
- Laboratory of Chromatin Biology, Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 240 David Aghmashenebeli Alley, 0159, Tbilisi, Georgia
| | - Davit Arsenadze
- Laboratory of Chromatin Biology, Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 240 David Aghmashenebeli Alley, 0159, Tbilisi, Georgia
| | - Malak Pirtskhalava
- Ivane Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Giorgi Zaalishvili
- Laboratory of Chromatin Biology, Institute of Cellular and Molecular Biology, Agricultural University of Georgia, 240 David Aghmashenebeli Alley, 0159, Tbilisi, Georgia
- Ivane Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| |
Collapse
|
32
|
Gao X, Kaluarachchi H, Zhang Y, Hwang S, Hannoush RN. A phage-displayed disulfide constrained peptide discovery platform yields novel human plasma protein binders. PLoS One 2024; 19:e0299804. [PMID: 38547072 PMCID: PMC10977726 DOI: 10.1371/journal.pone.0299804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/15/2024] [Indexed: 04/02/2024] Open
Abstract
Disulfide constrained peptides (DCPs) show great potential as templates for drug discovery. They are characterized by conserved cysteine residues that form intramolecular disulfide bonds. Taking advantage of phage display technology, we designed and generated twenty-six DCP phage libraries with enriched molecular diversity to enable the discovery of ligands against disease-causing proteins of interest. The libraries were designed based on five DCP scaffolds, namely Momordica charantia 1 (Mch1), gurmarin, Asteropsin-A, antimicrobial peptide-1 (AMP-1), and potato carboxypeptidase inhibitor (CPI). We also report optimized workflows for screening and producing synthetic and recombinant DCPs. Examples of novel DCP binders identified against various protein targets are presented, including human IgG Fc, serum albumin, vascular endothelial growth factor-A (VEGF-A) and platelet-derived growth factor (PDGF). We identified DCPs against human IgG Fc and serum albumin with sub-micromolar affinity from primary panning campaigns, providing alternative tools for potential half-life extension of peptides and small protein therapeutics. Overall, the molecular diversity of the DCP scaffolds included in the designed libraries, coupled with their distinct biochemical and biophysical properties, enables efficient and robust identification of de novo binders to drug targets of therapeutic relevance.
Collapse
Affiliation(s)
- Xinxin Gao
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Peptide Therapeutics, Genentech, South San Francisco, California, United States of America
| | - Harini Kaluarachchi
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
| | - Yingnan Zhang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Biological Chemistry, Genentech, South San Francisco, California, United States of America
| | - Sunhee Hwang
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
- Department of Peptide Therapeutics, Genentech, South San Francisco, California, United States of America
| | - Rami N Hannoush
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, California, United States of America
| |
Collapse
|
33
|
Ding XX, Ren BQ, Li BT, Pang ZJ, Xu YJ, Dong L. Pd(II)-Catalyzed β-C(sp 3)-H Alkynylation of Alanine in Di- and Tripeptides with Asn as an Endogenous Directing Group. J Org Chem 2024; 89:3390-3402. [PMID: 38377557 DOI: 10.1021/acs.joc.3c02823] [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: 02/22/2024]
Abstract
The introduction of alkyne moieties into peptides remains in demand as it represents a promising approach for further structural diversification of peptides. Herein, we describe the Pd(II)-catalyzed C(sp3)-H alkynylation of Ala-Asn-embedded di- and tripeptides using Asn as the endogenous lead group. In addition, a key building block for the glycopeptide Tyc4PG-14 and Tyc4PG-15 was produced by our methodology.
Collapse
Affiliation(s)
- Xing-Xing Ding
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- College of Chemistry and Material Science, Sichuan Normal University, Chengdu, Sichuan 610066, China
| | - Bo-Quan Ren
- College of Chemistry and Material Science, Sichuan Normal University, Chengdu, Sichuan 610066, China
| | - Bing-Tong Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhao-Jiong Pang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yan-Jun Xu
- College of Chemistry and Material Science, Sichuan Normal University, Chengdu, Sichuan 610066, China
| | - Lin Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
34
|
Zhao L, Li L, Hu M, Fang Y, Dong N, Shan A. Heterologous expression of the novel dimeric antimicrobial peptide LIG in Pichia pastoris. J Biotechnol 2024; 381:19-26. [PMID: 38181981 DOI: 10.1016/j.jbiotec.2023.12.015] [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: 08/08/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
The antimicrobial peptide (AMP) LI is a fusion product of antimicrobial peptide LL37 produced by human neutrophils and Indolicidin secreted by bovine neutrophils. LI retained the antimicrobial activity of the parental peptides and showed high cell selectivity. In this study, the flexible linker Gly-Ser-Gly (G-S-G) was used to ligate LI into dimeric LIG, and constructed the Pichia pastoris (P. pastoris) expression vector pPIC9K-6×His-3×FLAG-LIG. The total protein expression of P. pastoris GS115 reached the highest level (189.6 mg/L) after 96 h induction with 3 % methanol at the initial pH value of 7.0. Finally, 5.9 mg/L of recombinant LIG (rLIG) was obtained after enterokinase digestion and purification. The rLIG had high antimicrobial activity and low hemolytic activity. Compared with monomer LI, GSG linked dimeric LIG, which had no significant change in antimicrobial activity and had good salt ions stability. In this study, the dimeric antimicrobial peptide LIG was successfully expressed, which provided a new idea for the expression of AMPs in the P. pastoris expression system, and had important significance for the application of AMPs.
Collapse
Affiliation(s)
- Lu Zhao
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Ling Li
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Mingyang Hu
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Yuxin Fang
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| | - Na Dong
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China.
| | - Anshan Shan
- Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technology, Northeast Agricultural University, Harbin, PR China
| |
Collapse
|
35
|
Sarkar T, Ghosh S, Sundaravadivelu PK, Pandit G, Debnath S, Thummer RP, Satpati P, Chatterjee S. Mechanism of Protease Resistance of D-Amino Acid Residue Containing Cationic Antimicrobial Heptapeptides. ACS Infect Dis 2024; 10:562-581. [PMID: 38294842 DOI: 10.1021/acsinfecdis.3c00491] [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: 02/01/2024]
Abstract
Antimicrobial peptides (AMPs) have been an alternate promising class of therapeutics in combating global antibiotic resistance threat. However, the short half-life of AMPs, owing to protease degradability, is one of the major bottlenecks in its commercial success. In this study, we have developed all-D-amino acid containing small cationic peptides P4C and P5C, which are completely protease-resistant, noncytotoxic, nonhemolytic, and potent against the ESKAPE pathogens in comparison to their L analogues. MD simulations suggested marginal improvement in the peptide-binding affinity to the membrane-mimetic SDS micelle (∼ 1 kcal/mol) in response to L → D conversion, corroborating the marginal improvement in the antimicrobial activity. However, L → D chirality conversion severely compromised the peptide:protease (trypsin) binding affinity (≥10 kcal/mol). The relative distance between the scissile peptide carbonyl and the catalytic triad of the protease (H57, D102, and S195) was found to be significantly altered in the D-peptide:protease complex (inactive conformation) relative to the active L-peptide:protease complex. Thus, the poor binding affinity between D-peptides and the protease, resulting in the inactive complex formation, explained their experimentally observed proteolytic stability. This mechanistic insight might be extended to the proteolytic stability of the D-peptides in general and stimulate the rational design of protease-resistant AMPs.
Collapse
Affiliation(s)
- Tanumoy Sarkar
- Department of Chemistry, Indian Institute of Technology, Guwahati, Guwahati, Assam 781039, India
| | - Suvankar Ghosh
- Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, Guwahati, Assam 781039, India
| | | | - Gopal Pandit
- Department of Chemistry, Indian Institute of Technology, Guwahati, Guwahati, Assam 781039, India
| | - Swapna Debnath
- Department of Chemistry, Indian Institute of Technology, Guwahati, Guwahati, Assam 781039, India
| | - Rajkumar P Thummer
- Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, Guwahati, Assam 781039, India
| | - Priyadarshi Satpati
- Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, Guwahati, Assam 781039, India
| | - Sunanda Chatterjee
- Department of Chemistry, Indian Institute of Technology, Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
36
|
Behera LM, Ghosh M, Gupta PK, Rana S. A rationally engineered small antimicrobial peptide with potent antibacterial activity. J Cell Biochem 2024; 125:e30503. [PMID: 37992185 DOI: 10.1002/jcb.30503] [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: 09/06/2023] [Revised: 11/01/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023]
Abstract
Antimicrobial resistance (AMR) is a silent pandemic declared by the WHO that requires urgent attention in the post-COVID world. AMR is a critical public health concern worldwide, potentially affecting people at different stages of life, including the veterinary and agriculture industries. Notably, very few new-age antimicrobial agents are in the current developmental pipeline. Thus, the design, discovery, and development of new antimicrobial agents are required to address the menace of AMR. Antimicrobial peptides (AMPs) are an important class of antimicrobial agents for combating AMR due to their broad-spectrum activity and ability to evade AMR through a multimodal mechanism of action. However, molecular size, aggregability, proteolytic degradation, cytotoxicity, and hemolysis activity significantly limit the clinical application of natural AMPs. The de novo design and engineering of a short synthetic amphipathic AMP (≤16 aa, Mol. Wt. ≤ 2 kDa) with an unusual architecture comprised of coded and noncoded amino acids (NCAAs) is presented here, which demonstrates potent antibacterial activity against a few selected bacterial strains mentioned in the WHO priority list. The designer AMP is conformationally ordered in solution and effectively permeabilizes the outer and inner membranes, leading to bacterial growth inhibition and death. Additionally, the peptide is resistant to proteolysis and has negligible cytotoxicity and hemolysis activity up to 150 μM toward cultured human cell lines and erythrocytes. The designer AMP is unique and appears to be a potent therapeutic candidate, which can be subsequently subjected to preclinical studies to explicitly understand and address the menace of AMR.
Collapse
Affiliation(s)
- Lalita Mohan Behera
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Manaswini Ghosh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Pulkit Kr Gupta
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| |
Collapse
|
37
|
Maharani R, Muhajir MI, Dirgantara JM, Hardianto A, Mayanti T, Harneti D, Nurlelasari, Farabi K, Hidayat AT, Supratman U, Siahaan T. Synthesis and anticancer evaluation of [d-Ala]-nocardiotide A. RSC Adv 2024; 14:4097-4104. [PMID: 38292272 PMCID: PMC10825734 DOI: 10.1039/d4ra00025k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 02/01/2024] Open
Abstract
Cancer is currently one of the biggest causes of death in the world. Like some microorganisms, cancer cells also develop resistance to various chemotherapy drugs and are termed multidrug resistant (MDR). In this regard, there is a need to develop new alternative anticancer agents. Anticancer peptides (ACPs) with high selectivity and high cell penetration ability are a promising candidate, as well as they are easy to modify. A cyclohexapeptide called nocardiotide A was isolated from the marine sponge Callyspongia sp., which is cytotoxic towards several cancer cells such as MM, 1S, HeLa, and CT26 cells. Previously, nocardiotide A was synthesized with a very low yield owing to its challenging cyclization process. In this study, we synthesized [d-Ala]-nocardiotide A as a derivative of nocardiotide A using a combination of solid phase peptide synthesis (SPPS) and liquid phase peptide synthesis (LPPS). The synthesis was carried out by selecting a d-alanine residue at the C-terminus to give a desired cyclic peptide product with a yield of 31% after purification. The purified [d-Ala]-nocardiotide A was characterized using HR-ToF MS and 1H and 13C-NMR spectroscopy to validate the desired product. The anticancer activity of the peptide was determined against HeLa cancer cell lines with an IC50 value of 52 μM compared to the parent nocardiotide A with an IC50 value of 59 μM. In the future, we aim to mutate various l-amino acids in nocardiotide A to d-amino acids to prepare nocardiotide A derivatives with a higher activity to kill cancer cells with higher membrane permeation. In addition, the mechanism of action of nocardiotide A and its derivatives will be evaluated.
Collapse
Affiliation(s)
- Rani Maharani
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
- Central Laboratory, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
- Centre of Natural Products and Synthesis Studies, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
| | - Muhamad Imam Muhajir
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
| | - Jelang Muhammad Dirgantara
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
- Department of Chemistry, Graduate School of Science, Osaka University Toyonaka Osaka 560-0043 Japan
| | - Ari Hardianto
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
| | - Tri Mayanti
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
- Centre of Natural Products and Synthesis Studies, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
| | - Desi Harneti
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
- Centre of Natural Products and Synthesis Studies, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
| | - Nurlelasari
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
- Centre of Natural Products and Synthesis Studies, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
| | - Kindi Farabi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
- Central Laboratory, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
- Centre of Natural Products and Synthesis Studies, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
| | - Ace Tatang Hidayat
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
- Central Laboratory, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
- Centre of Natural Products and Synthesis Studies, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
| | - Unang Supratman
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor West Java Indonesia
- Central Laboratory, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
- Centre of Natural Products and Synthesis Studies, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jalan Raya Bandung-Sumedang KM 21 Jatinangor 45363 West Java Indonesia
| | - Teruna Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas 2095 Constant Avenue Lawrence Kansas 66047 USA
| |
Collapse
|
38
|
Gordon AT, Hosten EC, van Vuuren S, Ogunlaja AS. Copper(II)-photocatalyzed Hydrocarboxylation of Schiff bases with CO 2: antimicrobial evaluation and in silico studies of Schiff bases and unnatural α-amino acids. J Biomol Struct Dyn 2024:1-14. [PMID: 38192072 DOI: 10.1080/07391102.2024.2301765] [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: 08/11/2023] [Accepted: 12/30/2023] [Indexed: 01/10/2024]
Abstract
We synthesized and characterized two copper(II) complexes: [CuL2Cl]Cl and [CuL'2Cl]Cl, where L = 2,2'-bipyridine and L' = 4,4'-dimethyl-2,2'-bipyridine. We evaluated their photocatalytic hydrocarboxylation properties on a series of synthesized Schiff bases (SBs): (E)-1-(4-((5-bromo-2-hydroxybenzylidene)amino)phenyl)ethanone (SB1), (E)-N-(4-(dimethylamino)benzylidene)benzo[d]thiazol-2-amine (SB2), (E)-4-Bromo-2-((thiazol-2-ylimino)methyl)phenol (SB3), and (E)-4-((5-bromo-2-hydroxybenzylidene)amino)-1,5-dimethyl-2-phenyl-1H-pyrazol-3(2H)-one (SB4). Under mild photocatalytic reaction conditions (room temperature, 1 atm CO2, 30-watt Blue LED light), the derivatives of α-amino acids UAA1-4 were obtained with yields ranging from 5% to 44%. Experimental results demonstrated that [CuL2Cl]Cl exhibited superior photocatalytic efficiency compared to [CuL'2Cl]Cl, attributed to favourable electronic properties. In silico studies revealed strong binding strengths with E. faecalis DHFR (4M7U) for docked Schiff bases (SB) and unnatural α-amino acids (UAAs). In vitro studies further demonstrated significant antimicrobial and antifungal activity for SB2, SB3, and SB4, while none of the synthesized UAAs exhibited such properties, primarily due to the electronic and binding properties of these molecules.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Allen T Gordon
- Department of Chemistry, Nelson Mandela University, Port Elizabeth, South Africa
| | - Eric C Hosten
- Department of Chemistry, Nelson Mandela University, Port Elizabeth, South Africa
| | - Sandy van Vuuren
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, Parktown, South Africa
| | - Adeniyi S Ogunlaja
- Department of Chemistry, Nelson Mandela University, Port Elizabeth, South Africa
| |
Collapse
|
39
|
Behnke M, Holick CT, Vollrath A, Schubert S, Schubert US. Knowledge-Based Design of Multifunctional Polymeric Nanoparticles. Handb Exp Pharmacol 2024; 284:3-26. [PMID: 37017790 DOI: 10.1007/164_2023_649] [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: 04/06/2023]
Abstract
Conventional drug delivery systems (DDS) today still face several drawbacks and obstacles. High total doses of active pharmaceutical ingredients (API) are often difficult or impossible to deliver due to poor solubility of the API or undesired clearance from the body caused by strong interactions with plasma proteins. In addition, high doses lead to a high overall body burden, in particular if they cannot be delivered specifically to the target site. Therefore, modern DDS must not only be able to deliver a dose into the body, but should also overcome the hurdles mentioned above as examples. One of these promising devices are polymeric nanoparticles, which can encapsulate a wide range of APIs despite having different physicochemical properties. Most importantly, polymeric nanoparticles are tunable to obtain tailored systems for each application. This can already be achieved via the starting material, the polymer, by incorporating, e.g., functional groups. This enables the particle properties to be influenced not only specifically in terms of their interactions with APIs, but also in terms of their general properties such as size, degradability, and surface properties. In particular, the combination of size, shape, and surface modification allows polymeric nanoparticles to be used not only as a simple drug delivery device, but also to achieve targeting. This chapter discusses to what extent polymers can be designed to form defined nanoparticles and how their properties affect their performance.
Collapse
Affiliation(s)
- Mira Behnke
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Caroline T Holick
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Antje Vollrath
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Stephanie Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, Germany.
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
40
|
Baindara P, Mandal SM. Gut-Antimicrobial Peptides: Synergistic Co-Evolution with Antibiotics to Combat Multi-Antibiotic Resistance. Antibiotics (Basel) 2023; 12:1732. [PMID: 38136766 PMCID: PMC10740742 DOI: 10.3390/antibiotics12121732] [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: 11/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Due to huge diversity and dynamic competition, the human gut microbiome produces a diverse array of antimicrobial peptides (AMPs) that play an important role in human health. The gut microbiome has an important role in maintaining gut homeostasis by the AMPs and by interacting with other human organs via established connections such as the gut-lung, and gut-brain axis. Additionally, gut AMPs play a synergistic role with other gut microbiota and antimicrobials to maintain gut homeostasis by fighting against multi-antibiotic resistance (MAR) bacteria. Further, conventional antibiotics intake creates a synergistic evolutionary pressure for gut AMPs, where antibiotics and gut AMPs fight synergistically against MAR. Overall, gut AMPs are evolving under a complex and highly synergistic co-evolutionary pressure created by the various interactions between gut microbiota, gut AMPs, and antibiotics; however, the complete mechanism is not well understood. The current review explores the synergistic action of gut AMPs and antibiotics along with possibilities to fight against MAR bacteria.
Collapse
Affiliation(s)
- Piyush Baindara
- Radiation Oncology, NextGen Precision Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Santi M. Mandal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India;
| |
Collapse
|
41
|
Chang DH, Lee MR, Wang N, Lynn DM, Palecek SP. Establishing Quantifiable Guidelines for Antimicrobial α/β-Peptide Design: A Partial Least-Squares Approach to Improve Antimicrobial Activity and Reduce Mammalian Cell Toxicity. ACS Infect Dis 2023; 9:2632-2651. [PMID: 38014670 PMCID: PMC10807133 DOI: 10.1021/acsinfecdis.3c00468] [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] [Indexed: 11/29/2023]
Abstract
Antimicrobial peptides (AMPs) are promising candidates to combat pathogens that are resistant to conventional antimicrobial drugs because they operate through mechanisms that involve membrane disruption. However, the use of AMPs in clinical settings has been limited, at least in part, by their susceptibility to proteolytic degradation and their lack of selectivity toward pathogenic microbes vs mammalian cells. We recently reported on the design of α- and β-peptide oligomers structurally templated upon the naturally occurring α-helical AMP aurein 1.2. These α/β-peptide oligomers are more proteolytically stable than aurein 1.2 and have several other attributes that render them attractive as alternatives to conventional AMPs. This study describes the influence of peptide physicochemical properties on the broad-spectrum activity of aurein 1.2-based α/β-peptide mimics against nine bacterial, fungal, and mammalian cell lines. We used a partial least-squares regression (PLSR)-supervised machine learning model to quantify and visualize relationships between experimentally determined physicochemical properties (e.g., hydrophobicity, charge, and helicity) and experimentally measured cell-type-specific activities of 21 peptides in a 149-member α/β-peptide library. Using this approach, we identified several peptides that were predicted to exhibit enhanced broad-spectrum selectivity, a measure that evaluates antimicrobial activity relative to mammalian cell toxicity compared to aurein 1.2. Experimental validation demonstrated high model predictive performance, and characterization of compounds with the highest broad-spectrum selectivity revealed peptide hydrophobicity, helicity, and helical rigidity to be strong predictors of broad-spectrum selectivity. The most selective peptide identified from the model prediction has more than a 13-fold improvement in broad-spectrum selectivity than that of aurein 1.2, demonstrating the ability of using PLSR models to identify quantitative structure-function relationships for nonstandard amino acid-containing peptides. Overall, this work establishes quantifiable guidelines for the rational design of helical antimicrobial α/β-peptides and identifies promising new α/β-peptides with significantly reduced mammalian toxicities and improved antifungal and antibacterial activities relative to aurein 1.2.
Collapse
Affiliation(s)
- Douglas H. Chang
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
| | - Myung-Ryul Lee
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
| | - Nathan Wang
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
| | - David M. Lynn
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706, USA
| | - Sean P. Palecek
- Department of Chemical & Biological Engineering, University of Wisconsin–Madison, 1415 Engineering Dr., Madison, WI 53706, USA
| |
Collapse
|
42
|
Bonvin E, Personne H, Paschoud T, Reusser J, Gan BH, Luscher A, Köhler T, van Delden C, Reymond JL. Antimicrobial Peptide-Peptoid Hybrids with and without Membrane Disruption. ACS Infect Dis 2023; 9:2593-2606. [PMID: 38062792 PMCID: PMC10714400 DOI: 10.1021/acsinfecdis.3c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023]
Abstract
Among synthetic analogues of antimicrobial peptides (AMPs) under investigation to address antimicrobial resistance, peptoids (N-alkylated oligoglycines) have been reported to act both by membrane disruption and on intracellular targets. Here we gradually introduced peptoid units into the membrane-disruptive undecapeptide KKLLKLLKLLL to test a possible transition toward intracellular targeting. We found that selected hybrids containing up to five peptoid units retained the parent AMP's α-helical folding, membrane disruption, and antimicrobial effects against Gram-negative bacteria including multidrug-resistant (MDR) strains of Pseudomonas aeruginosa and Klebsiella pneumoniae while showing reduced hemolysis and cell toxicities. Furthermore, some hybrids containing as few as three peptoid units as well as the full peptoid lost folding, membrane disruption, hemolysis, and cytotoxicity but displayed strong antibacterial activity under dilute medium conditions typical for proline-rich antimicrobial peptides (PrAMPs), pointing to intracellular targeting. These findings parallel previous reports that partially helical amphiphilic peptoids are privileged oligomers for antibiotic development.
Collapse
Affiliation(s)
- Etienne Bonvin
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Hippolyte Personne
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Thierry Paschoud
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Jérémie Reusser
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Bee-Ha Gan
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Alexandre Luscher
- Department
of Microbiology and Molecular Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Service of
Infectious Diseases, University Hospital
of Geneva, CH-1211 Geneva, Switzerland
| | - Thilo Köhler
- Department
of Microbiology and Molecular Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Service of
Infectious Diseases, University Hospital
of Geneva, CH-1211 Geneva, Switzerland
| | - Christian van Delden
- Department
of Microbiology and Molecular Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Service of
Infectious Diseases, University Hospital
of Geneva, CH-1211 Geneva, Switzerland
| | - Jean-Louis Reymond
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| |
Collapse
|
43
|
Myšková A, Sýkora D, Kuneš J, Maletínská L. Lipidization as a tool toward peptide therapeutics. Drug Deliv 2023; 30:2284685. [PMID: 38010881 PMCID: PMC10987053 DOI: 10.1080/10717544.2023.2284685] [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: 08/06/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023] Open
Abstract
Peptides, as potential therapeutics continue to gain importance in the search for active substances for the treatment of numerous human diseases, some of which are, to this day, incurable. As potential therapeutic drugs, peptides have many favorable chemical and pharmacological properties, starting with their great diversity, through their high affinity for binding to all sort of natural receptors, and ending with the various pathways of their breakdown, which produces nothing but amino acids that are nontoxic to the body. Despite these and other advantages, however, they also have their pitfalls. One of these disadvantages is the very low stability of natural peptides. They have a short half-life and tend to be cleared from the organism very quickly. Their instability in the gastrointestinal tract, makes it impossible to administer peptidic drugs orally. To achieve the best pharmacologic effect, it is desirable to look for ways of modifying peptides that enable the use of these substances as pharmaceuticals. There are many ways to modify peptides. Herein we summarize the approaches that are currently in use, including lipidization, PEGylation, glycosylation and others, focusing on lipidization. We describe how individual types of lipidization are achieved and describe their advantages and drawbacks. Peptide modifications are performed with the goal of reaching a longer half-life, reducing immunogenicity and improving bioavailability. In the case of neuropeptides, lipidization aids their activity in the central nervous system after the peripheral administration. At the end of our review, we summarize all lipidized peptide-based drugs that are currently on the market.
Collapse
Affiliation(s)
- Aneta Myšková
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Prague, Czech Republic
- Institute of Organic Chemistry and Biochemistry, Academy of Science of the Czech Republic, Prague, Czech Republic
| | - David Sýkora
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, Academy of Science of the Czech Republic, Prague, Czech Republic
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, Academy of Science of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
44
|
Abdulbagi M, Di B, Li B. Resolving D-Amino Acid Containing Peptides Using Ion Mobility-Mass Spectrometry: Challenges and Recent Developments. Crit Rev Anal Chem 2023:1-10. [PMID: 37975700 DOI: 10.1080/10408347.2023.2282510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Peptides and proteins having D-amino acids in their sequences are now believed to be widespread among different living organisms. Their significance is attributed to the diverse functions of these molecules, such as having a certain pathological implication or enhancing biological activity. Indeed, some peptide molecules with D-amino acids in their structure have already found their way to clinical use such as the antibacterial gramicidin and the antidiabetic nateglinide. Ion mobility mass spectrometry (IM-MS) added an additional dimension of separation as it depends on ions mobility in the space, which is dependent on their shapes, and the shape depends on the orientation of atoms. Thus, D-amino acids containing peptides (DAACPs) will have different mobility and collision cross-section values than those with L-amino acids. Eventually, this will lead to baseline separation of the two peptides. Additionally, ion mobility can precisely locate the position of D-amino acids by analyzing the difference in the arrival times of the fragment ions. The importance of DAACPs, as well as the difficulties in discovering them, were addressed in this review. Similarly, we emphasized how recent developments in IM-MS have improved their detection and analysis. Consequently, the LC-IM-MS/MS platform appears to be promising in isomeric mixture analysis.
Collapse
Affiliation(s)
- Mohamed Abdulbagi
- Center Key Laboratory on Protein Chemistry and Structural Biology, China Pharmaceutical University, Nanjing, China
| | - Bin Di
- Center Key Laboratory on Protein Chemistry and Structural Biology, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Bo Li
- Center Key Laboratory on Protein Chemistry and Structural Biology, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
45
|
Ki MR, Kim SH, Park TI, Pack SP. Self-Entrapment of Antimicrobial Peptides in Silica Particles for Stable and Effective Antimicrobial Peptide Delivery System. Int J Mol Sci 2023; 24:16423. [PMID: 38003614 PMCID: PMC10671715 DOI: 10.3390/ijms242216423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Antimicrobial peptides (AMPs) have emerged as a promising solution to tackle bacterial infections and combat antibiotic resistance. However, their vulnerability to protease degradation and toxicity towards mammalian cells has hindered their clinical application. To overcome these challenges, our study aims to develop a method to enhance the stability and safety of AMPs applicable to effective drug-device combination products. The KR12 antimicrobial peptide was chosen, and in order to further enhance its delivery and efficacy the human immunodeficiency virus TAT protein-derived cell-penetrating peptide (CPP) was fused to form CPP-KR12. A new product, CPP-KR12@Si, was developed by forming silica particles with self-entrapped CPP-KR12 peptide using biomimetic silica precipitability because of its cationic nature. Peptide delivery from CPP-KR12@Si to bacteria and cells was observed at a slightly delivered rate, with improved stability against trypsin treatment and a reduction in cytotoxicity compared to CPP-KR12. Finally, the antimicrobial potential of the CPP-KR12@Si/bone graft substitute (BGS) combination product was demonstrated. CPP-KR12 is coated in the form of submicron-sized particles on the surface of the BGS. Self-entrapped AMP in silica nanoparticles is a safe and effective AMP delivery method that will be useful for developing a drug-device combination product for tissue regeneration.
Collapse
Affiliation(s)
- Mi-Ran Ki
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea
- Institute of Industrial Technology, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea
| | - Sung Ho Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea
| | - Tae In Park
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong-ro 2511, Sejong 30019, Republic of Korea
| |
Collapse
|
46
|
Tripathi AK, Singh J, Trivedi R, Ranade P. Shaping the Future of Antimicrobial Therapy: Harnessing the Power of Antimicrobial Peptides in Biomedical Applications. J Funct Biomater 2023; 14:539. [PMID: 37998108 PMCID: PMC10672284 DOI: 10.3390/jfb14110539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023] Open
Abstract
Antimicrobial peptides (AMPs) have emerged as a promising class of bioactive molecules with the potential to combat infections associated with medical implants and biomaterials. This review article aims to provide a comprehensive analysis of the role of antimicrobial peptides in medical implants and biomaterials, along with their diverse clinical applications. The incorporation of AMPs into various medical implants and biomaterials has shown immense potential in mitigating biofilm formation and preventing implant-related infections. We review the latest advancements in biomedical sciences and discuss the AMPs that were immobilized successfully to enhance their efficacy and stability within the implant environment. We also highlight successful examples of AMP coatings for the treatment of surgical site infections (SSIs), contact lenses, dental applications, AMP-incorporated bone grafts, urinary tract infections (UTIs), medical implants, etc. Additionally, we discuss the potential challenges and prospects of AMPs in medical implants, such as effectiveness, instability and implant-related complications. We also discuss strategies that can be employed to overcome the limitations of AMP-coated biomaterials for prolonged longevity in clinical settings.
Collapse
Affiliation(s)
- Amit Kumar Tripathi
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.T.); (P.R.)
| | - Jyotsana Singh
- Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Rucha Trivedi
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.T.); (P.R.)
| | - Payal Ranade
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.T.); (P.R.)
| |
Collapse
|
47
|
Sahsuvar S, Guner R, Gok O, Can O. Development and pharmaceutical investigation of novel cervical cancer-targeting and redox-responsive melittin conjugates. Sci Rep 2023; 13:18225. [PMID: 37880286 PMCID: PMC10600185 DOI: 10.1038/s41598-023-45537-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: 07/18/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023] Open
Abstract
Cervical cancer has recently become one of the most prevalent cancers among women throughout the world. Traditional cancer therapies generate side effects due to off-target toxicity. Thus, novel cancer medications coupled with suitable drug delivery systems are required to improve cancer therapies. Melittin peptide has a high affinity to disrupt cancer cells. In this study, we designed targeted and redox-responsive Melittin conjugates for cervical cancer and then tested them in vitro. Folic acid and squamous cell carcinoma-specific peptide (CKQNLAEG) were used as targeting agents to design various conjugates. Our findings indicate that both anticancer conjugates were effective against different cancer cell lines, including MCF-7, C33A, and HeLa. Moreover, these conjugates were found to have antioxidant and antibacterial effects as well as reduced hemolytic activity. The CM-Target (N-terminus cysteine modified-Melittin-targeting peptide-functionalized conjugate) has become more stable and acted specifically against squamous cell carcinoma, whereas folic acid (FA)-containing conjugates acted efficiently against all cancer types studied, especially for breast cancer. According to our results, these anticancer conjugates may be possible anticancer drug candidates that have fewer adverse effects.
Collapse
Affiliation(s)
- Seray Sahsuvar
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Rabia Guner
- Department of Biomedical Engineering, Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ozgul Gok
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Icerenkoy, Kayisdagi Cd., Atasehir, 34752, Istanbul, Turkey.
| | - Ozge Can
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Icerenkoy, Kayisdagi Cd., Atasehir, 34752, Istanbul, Turkey.
| |
Collapse
|
48
|
Carrera-Aubesart A, Gallo M, Defaus S, Todorovski T, Andreu D. Topoisomeric Membrane-Active Peptides: A Review of the Last Two Decades. Pharmaceutics 2023; 15:2451. [PMID: 37896211 PMCID: PMC10610229 DOI: 10.3390/pharmaceutics15102451] [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: 08/30/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
In recent decades, bioactive peptides have been gaining recognition in various biomedical areas, such as intracellular drug delivery (cell-penetrating peptides, CPPs) or anti-infective action (antimicrobial peptides, AMPs), closely associated to their distinct mode of interaction with biological membranes. Exploiting the interaction of membrane-active peptides with diverse targets (healthy, tumoral, bacterial or parasitic cell membranes) is opening encouraging prospects for peptides in therapeutics. However, ordinary peptides formed by L-amino acids are easily decomposed by proteases in biological fluids. One way to sidestep this limitation is to use topoisomers, namely versions of the peptide made up of D-amino acids in either canonic (enantio) or inverted (retroenantio) sequence. Rearranging peptide sequences in this fashion provides a certain degree of native structure mimicry that, in appropriate contexts, may deliver desirable biological activity while avoiding protease degradation. In this review, we will focus on recent accounts of membrane-active topoisomeric peptides with therapeutic applications as CPP drug delivery vectors, or as antimicrobial and anticancer candidates. We will also discuss the most common modes of interaction of these peptides with their membrane targets.
Collapse
Affiliation(s)
- Adam Carrera-Aubesart
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
| | - Maria Gallo
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
| | - Sira Defaus
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
| | - Toni Todorovski
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
- Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| | - David Andreu
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (A.C.-A.); (M.G.); (S.D.); (T.T.)
| |
Collapse
|
49
|
Meinberger D, Drexelius MG, Grabeck J, Hermes G, Roth A, Elezagic D, Neundorf I, Streichert T, Klatt AR. Modified CLEC3A-Derived Antimicrobial Peptides Lead to Enhanced Antimicrobial Activity against Drug-Resistant Bacteria. Antibiotics (Basel) 2023; 12:1532. [PMID: 37887233 PMCID: PMC10604565 DOI: 10.3390/antibiotics12101532] [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: 09/11/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
Antimicrobial peptides (AMPs) represent a promising alternative to conventional antibiotics. Sequence changes can significantly improve the therapeutic properties of antimicrobial peptides. In our study, we apply different sequence modifications to enhance the performance of the CLEC3A-derived AMPs HT-16 and HT-47. We truncated their sequences, inserting a triple-glycine linker, adding an N-terminal tryptophan residue, and generating a D-amino acid variant, resulting in the generation of seven new peptides. We investigated their antimicrobial activity against gram-positive and gram-negative bacteria, their cytotoxicity to murine cells, and the biostability of the modified peptides in serum. We identified a novel antimicrobial peptide, WRK-30, with enhanced antimicrobial potency against S. aureus and MRSA. Additionally, WRK-30 was less cytotoxic to eukaryotic cells, allowing its application in higher concentrations in an in vivo setting. In conclusion, we identified a novel CLEC3A-derived antimicrobial peptide WRK-30 with significantly improved therapeutic properties and the potential to widen the repertoire of conventional antibiotics.
Collapse
Affiliation(s)
- Denise Meinberger
- Institute for Clinical Chemistry, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Marco G. Drexelius
- Institute for Biochemistry, Department of Chemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Zuelpicher Str. 47a, 50674 Cologne, Germany
- Center for Molecular Biosciences, University of Cologne, Zuelpicher Str. 47a, 50674 Cologne, Germany
| | - Joshua Grabeck
- Institute for Biochemistry, Department of Chemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Zuelpicher Str. 47a, 50674 Cologne, Germany
- Center for Molecular Biosciences, University of Cologne, Zuelpicher Str. 47a, 50674 Cologne, Germany
| | - Gabriele Hermes
- Institute for Clinical Chemistry, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Annika Roth
- Institute for Clinical Chemistry, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Dzemal Elezagic
- Institute for Clinical Chemistry, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Ines Neundorf
- Institute for Biochemistry, Department of Chemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Zuelpicher Str. 47a, 50674 Cologne, Germany
- Center for Molecular Biosciences, University of Cologne, Zuelpicher Str. 47a, 50674 Cologne, Germany
| | - Thomas Streichert
- Institute for Clinical Chemistry, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Andreas R. Klatt
- Institute for Clinical Chemistry, Medical Faculty, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
50
|
Yang L, Luo M, Liu Z, Li Y, Lin Z, Geng S, Wang Y. BamA-targeted antimicrobial peptide design for enhanced efficacy and reduced toxicity. Amino Acids 2023; 55:1317-1331. [PMID: 37670010 DOI: 10.1007/s00726-023-03307-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/18/2023] [Indexed: 09/07/2023]
Abstract
The emergence of drug-resistant superbugs has necessitated a pressing need for innovative antibiotics. Antimicrobial peptides (AMPs) have demonstrated broad-spectrum antibacterial activity, reduced susceptibility to resistance, and immunomodulatory effects, rendering them promising for combating drug-resistant microorganisms. This study employed computational simulation methods to screen and design AMPs specifically targeting ESKAPE pathogens. Particularly, AMPs were rationally designed to target the BamA and obtain novel antimicrobial peptide sequences. The designed AMPs were assessed for their antibacterial activities, mechanisms, and stability. Molecular docking and dynamics simulations demonstrated the interaction of both designed AMPs, 11pep and D-11pep, with the β1, β9, β15, and β16 chains of BamA, resulting in misfolding of outer membrane proteins and antibacterial effects. Subsequent antibacterial investigations confirmed the broad-spectrum activity of both 11pep and D-11pep, with D-11pep demonstrating higher potency against resistant Gram-negative bacteria. D-11pep exhibited MICs of 16, 8, and 32 μg/mL against carbapenem-resistant Escherichia coli, carbapenem-resistant Pseudomonas aeruginosa, and multi-drug-resistant Acinetobacter baumannii, respectively, with a concomitant lower resistance induction. Mechanism of action studies confirmed that peptides could disrupt the bacterial outer membrane, aligning with the findings of molecular dynamics simulations. Additionally, D-11pep demonstrated superior stability and reduced toxicity in comparison to 11pep. The findings of this study underscore the efficacy of rational AMP design that targets BamA, along with the utilization of D-amino acid replacements as a strategy for developing AMPs against drug-resistant bacteria.
Collapse
Affiliation(s)
- Li Yang
- Pharmacy and Bioengineering of Technology, Chongqing University of Technology, Chongqing, 400054, China
| | - Minghe Luo
- Pharmacy and Bioengineering of Technology, Chongqing University of Technology, Chongqing, 400054, China
- Chongqing Municipal Key Laboratory of Institutions of Higher Education of Target Based Drug Screening and Activity Evaluation, Chongqing, 400054, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing, 400054, China
| | - Zhou Liu
- Pharmacy and Bioengineering of Technology, Chongqing University of Technology, Chongqing, 400054, China
| | - Yuepeng Li
- Pharmacy and Bioengineering of Technology, Chongqing University of Technology, Chongqing, 400054, China
| | - Zhihua Lin
- Pharmacy and Bioengineering of Technology, Chongqing University of Technology, Chongqing, 400054, China
- Chongqing Municipal Key Laboratory of Institutions of Higher Education of Target Based Drug Screening and Activity Evaluation, Chongqing, 400054, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing, 400054, China
| | - Shan Geng
- The People's Hospital of Dazu, Chongqing, 402360, China
| | - Yuanqiang Wang
- Pharmacy and Bioengineering of Technology, Chongqing University of Technology, Chongqing, 400054, China.
- Chongqing Municipal Key Laboratory of Institutions of Higher Education of Target Based Drug Screening and Activity Evaluation, Chongqing, 400054, China.
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing, 400054, China.
| |
Collapse
|