1
|
Nazir S, Khan AI, Maharjan R, Khan SN, Akram MA, Maresca M, Khan FA, Shaheen F. Synthesis of Temporin-SHa Retro Analogs with Lysine Addition/Substitution and Antibiotic Conjugation to Enhance Antibacterial, Antifungal, and Anticancer Activities. Antibiotics (Basel) 2024; 13:1213. [PMID: 39766603 PMCID: PMC11672801 DOI: 10.3390/antibiotics13121213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
In the face of rising the threat of resistant pathogens, antimicrobial peptides (AMPs) offer a viable alternative to the current challenge due to their broad-spectrum activity. This study focuses on enhancing the efficacy of temporin-SHa derived NST-2 peptide (1), which is known for its antimicrobial and anticancer activities. We synthesized new analogs of 1 using three strategies, i.e., retro analog preparation, lysine addition/substitution, and levofloxacin conjugation. Analogs were tested in terms of their antibacterial, antifungal, and anticancer activities. Analog 2, corresponding to retro analog of NST-2, was found to be more active but also more hemolytic, reducing its selectivity index and therapeutic potential. The addition of lysine (in analog 3) and lysine substitution (in analog 7) reduced the hemolytic effect resulting in safer peptides. Conjugation with levofloxacin on the lysine side chain (in analogs 4 and 5) decreased the hemolytic effect but unfortunately also the antimicrobial and anticancer activities of the analogs. Oppositely, conjugation with levofloxacin at the N-terminus of the peptide via the β-alanine linker (in analogs 6 and 8) increased their antimicrobial and anticancer activity but also their hemolytic effect, resulting in less safe/selective analogs. In conclusion, lysine addition/substitution and levofloxacin conjugation, at least at the N-terminal position through the β-alanine linker, were found to enhance the therapeutic potential of retro analogs of NST-2 whereas other modifications decreased the activity or increased the toxicity of the peptides.
Collapse
Affiliation(s)
- Shahzad Nazir
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; (S.N.); (A.I.K.); (R.M.); (S.N.K.); (M.A.A.); (F.-A.K.)
| | - Arif Iftikhar Khan
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; (S.N.); (A.I.K.); (R.M.); (S.N.K.); (M.A.A.); (F.-A.K.)
| | - Rukesh Maharjan
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; (S.N.); (A.I.K.); (R.M.); (S.N.K.); (M.A.A.); (F.-A.K.)
| | - Sadiq Noor Khan
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; (S.N.); (A.I.K.); (R.M.); (S.N.K.); (M.A.A.); (F.-A.K.)
| | - Muhammad Adnan Akram
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; (S.N.); (A.I.K.); (R.M.); (S.N.K.); (M.A.A.); (F.-A.K.)
| | - Marc Maresca
- Aix Marseille Univ, CNRS, Centrale Med, ISM2, 13013 Marseille, France
| | - Farooq-Ahmad Khan
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; (S.N.); (A.I.K.); (R.M.); (S.N.K.); (M.A.A.); (F.-A.K.)
| | - Farzana Shaheen
- Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; (S.N.); (A.I.K.); (R.M.); (S.N.K.); (M.A.A.); (F.-A.K.)
| |
Collapse
|
2
|
Mahto AK, Kanupriya, Kumari S, Yar MS, Dewangan RP. Hydrocarbon stapled temporin-L analogue as potential antibacterial and antiendotoxin agents with enhanced protease stability. Bioorg Chem 2024; 145:107239. [PMID: 38428282 DOI: 10.1016/j.bioorg.2024.107239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/06/2024] [Accepted: 02/22/2024] [Indexed: 03/03/2024]
Abstract
Antimicrobial resistance (AMR) is a serious global concern and a huge burden on the healthcare system. Antimicrobial peptides (AMPs) are considered as a solution of AMR due to their membrane-lytic and intracellular mode of action and therefore resistance development against AMPs is less frequent. One such AMPs, temporin-L (TL) is a 13-mer peptide reported as a potent and broad-spectrum antibacterial agent with significant immunomodulatory activity. However, TL is toxic to human erythrocytes at their antibacterial concentrations and therefore various analogues were synthesized with potent antimicrobial activity and lower hemolytic activity. In this work, we have selected a non-toxic engineered analogue of TL (eTL) and performed hydrocarbon stapling of amino acid residues at i to i + 4 positions at different part of sequence. The synthesized peptides were investigated against both the gram-positive and gram-negative bacteria as well as methicillin resistant S. aureus, its MIC was measured in the concentrations range of 0.9-15.2 µM. All analogues were found equal or better antibacterial as compared to parent peptide. Interestingly one analogue eTL [5-9] was found to be non-cytotoxic and stable in presence of the human serum. Mode of action studies revealed membrane depolarizing and disruptive mode of action with live MRSA. Further in vivo studies of antimicrobial against MRSA infection and anti-endotoxin activities in mice model revealed potential activity of the stapled peptide analogue. Overall, this reports on stapled analogue of the AMPs highlights an important strategy for the development of new antibacterial therapeutics against AMR.
Collapse
Affiliation(s)
- Aman Kumar Mahto
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard (Deemed to be University), New Delhi 110062, India
| | - Kanupriya
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard (Deemed to be University), New Delhi 110062, India
| | - Shalini Kumari
- CSIR-Institute of Genomics and Integrative Biology (IGIB), Sukhdev Vihar, Mathura Road, New Delhi 110025, India
| | - Mohammad Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard (Deemed to be University), New Delhi 110062, India
| | - Rikeshwer Prasad Dewangan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard (Deemed to be University), New Delhi 110062, India.
| |
Collapse
|
3
|
Acharya Y, Taneja KK, Haldar J. Dual functional therapeutics: mitigating bacterial infection and associated inflammation. RSC Med Chem 2023; 14:1410-1428. [PMID: 37593575 PMCID: PMC10429821 DOI: 10.1039/d3md00166k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/21/2023] [Indexed: 08/19/2023] Open
Abstract
The emergence of antimicrobial resistance, coupled with the occurrence of persistent systemic infections, has already complicated clinical therapy efforts. Moreover, infections are also accompanied by strong inflammatory responses, generated by the host's innate and adaptive immune systems. The closely intertwined relationship between bacterial infection and inflammation has multiple implications on the ability of antibacterial therapeutics to tackle infection and inflammation. Particularly, uncontrolled inflammatory responses to infection can lead to sepsis, a life-threatening physiological condition. In this review, we discuss dual-functional antibacterial therapeutics that have potential to be developed for treating inflammation associated with bacterial infections. Immense research is underway that aims to develop new therapeutic agents that, when administered, regulate the excess inflammatory response, i.e. they have immunomodulatory properties along with the desired antibacterial activity. The classes of antibiotics that have immunomodulatory function in addition to antibacterial activity have been reviewed. Host defense peptides and their synthetic mimics are amongst the most sought-after solutions to develop such dual-functional therapeutics. This review also highlights the important classes of peptidomimetics that exhibit both antibacterial and immunomodulatory properties.
Collapse
Affiliation(s)
- Yash Acharya
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| | - Kashish Kumar Taneja
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
- School of Advanced Materials, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Jakkur Bengaluru 560064 Karnataka India
| |
Collapse
|
4
|
Silva PSE, Guindo AS, Oliveira PHC, de Moraes LFRN, Boleti APDA, Ferreira MA, de Oliveira CFR, Macedo MLR, Rossato L, Simionatto S, Migliolo L. Evaluation of the Synthetic Multifunctional Peptide Hp-MAP3 Derivative of Temporin-PTa. Toxins (Basel) 2023; 15:42. [PMID: 36668862 PMCID: PMC9866994 DOI: 10.3390/toxins15010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 01/06/2023] Open
Abstract
In recent years, antimicrobial peptides isolated from amphibian toxins have gained attention as new multifunctional drugs interacting with different molecular targets. We aimed to rationally design a new peptide from temporin-PTa. Hp-MAP3 (NH2-LLKKVLALLKKVL-COOH), net charge (+4), hydrophobicity (0.69), the content of hydrophobic residues (69%), and hydrophobic moment (0.73). For the construction of the analog peptide, the physicochemical characteristics were reorganized into hydrophilic and hydrophobic residues with the addition of lysines and leucines. The minimum inhibitory concentration was 2.7 to 43 μM against the growth of Gram-negative and positive bacteria, and the potential for biofilm eradication was 173.2 μM. Within 20 min, the peptide Hp-MAP3 (10.8 μM) prompted 100% of the damage to E. coli cells. At 43.3 μM, eliminated 100% of S. aureus within 5 min. The effects against yeast species of the Candida genus ranged from 5.4 to 86.6 μM. Hp-MAP3 presents cytotoxic activity against tumor HeLa at a concentration of 21.6 μM with an IC50 of 10.4 µM. Furthermore, the peptide showed hemolytic activity against murine erythrocytes. Structural studies carried out by circular dichroism showed that Hp-MAP3, while in the presence of 50% trifluoroethanol or SDS, an α-helix secondary structure. Finally, Amphipathic Hp-MAP3 building an important model for the design of new multifunctional molecules.
Collapse
Affiliation(s)
- Patrícia Souza e Silva
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | - Alexya Sandim Guindo
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | - Pedro Henrique Cardoso Oliveira
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | | | - Ana Paula de Araújo Boleti
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | - Marcos Antonio Ferreira
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| | - Caio Fernando Ramalho de Oliveira
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Unidade de Tecnologia de Alimentos e da Saúde Pública, Universidade Federal de Mato Grosso do Sul, Campo Grande 79070-900, Mato Grosso do Sul, Brazil
| | - Maria Ligia Rodrigues Macedo
- Laboratório de Purificação de Proteínas e suas Funções Biológicas, Unidade de Tecnologia de Alimentos e da Saúde Pública, Universidade Federal de Mato Grosso do Sul, Campo Grande 79070-900, Mato Grosso do Sul, Brazil
| | - Luana Rossato
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados UFGD, Dourados 79825-070, Mato Grosso do Sul, Brazil
| | - Simone Simionatto
- Laboratório de Pesquisa em Ciências da Saúde, Universidade Federal da Grande Dourados UFGD, Dourados 79825-070, Mato Grosso do Sul, Brazil
| | - Ludovico Migliolo
- S-Inova Biotech, Postgraduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande 79117-900, Mato Grosso do Sul, Brazil
| |
Collapse
|
5
|
Zhu R, Guo R, Yu C, Tan X, Wei S, Song Y, Wang R, Li L, Xie X, Jiang W, Zhang Y. Arginine replacement of histidine on temporin-GHa enhances the antimicrobial and antibiofilm efficacy against Staphylococcus aureus. Biosci Biotechnol Biochem 2022; 87:63-73. [PMID: 36367541 DOI: 10.1093/bbb/zbac168] [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: 08/21/2022] [Accepted: 10/15/2022] [Indexed: 11/13/2022]
Abstract
Antimicrobial peptides (AMPs) show broad-spectrum microbicidal activity against bacteria, fungi, and viruses, and have been considered as one of the most promising candidates to overcome bacterial antimicrobial resistance. Structural modification of AMPs is an effective strategy to develop high-efficiency and low-toxicity antibacterial agents. A series of peptides GHaR6R, GHaR7R, GHaR8R, and GHaR9W with arginine replacement of histidine (His) derived from temporin-GHa of Hylarana guentheri were designed and synthesized. These derived peptides exhibit antibacterial activity against Staphylococcus aureus, and GHaR8R exerts bactericidal effect within 15 min at 4 × MIC (25 µm). The derived peptides caused rapid depolarization of bacteria, and the cell membrane damage was monitored using quartz crystal microbalance with dissipation assay, which suggests that they target cell membranes to exert antibacterial effects. The derived peptides can effectively eradicate mature biofilms of S. aureus. Taken together, the derived peptides are promising antibacterial agent candidates against S. aureus.
Collapse
Affiliation(s)
- Ruiying Zhu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Ran Guo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Chunmei Yu
- Administrative Law Enforcement Brigade of Agriculture, Ji'an, China
| | - Xiuchuan Tan
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | | | - Yanting Song
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Rong Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Lushuang Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Xi Xie
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Wenying Jiang
- Hainan University Affiliated Hospital, Haikou, China
| | - Yingxia Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| |
Collapse
|
6
|
Kumari T, Verma DP, Kuldeep J, Dhanabal VB, Verma NK, Sahai R, Tripathi AK, Saroj J, Ali M, Mitra K, Siddiqi MI, Bhattacharjya S, Ghosh JK. 10-Residue MyD88-Peptide Adopts β-Sheet Structure, Self-Assembles, Binds to Lipopolysaccharides, and Rescues Mice from Endotoxin-Mediated Lung-Infection and Death. ACS Chem Biol 2022; 17:3420-3434. [PMID: 36367958 DOI: 10.1021/acschembio.2c00569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Naturally occurring cationic antimicrobial peptides (AMPs) mostly adopt α-helical structures in bacterial membrane mimetic environments. To explore the design of novel β-sheet AMPs, we identified two short cationic amphipathic β-strand segments from the crystal structure of the innate immune protein, MyD88. Interestingly, of these, the 10-residue arginine-valine-rich synthetic MyD88-segment, KRCRRMVVVV (M3), exhibited β-sheet structure when bound to the outer membrane Gram-negative bacterial component, LPS. Isothermal titration calorimetric data showed that M3 bound to LPS with high affinity, and the interaction was hydrophobic in nature. Supporting these observations, computational studies indicated strong interactions of multiple and consecutive valine residues of M3 with the acyl chain of LPS. Moreover, M3 adopted nanosheet and nanofibrillar structure in 25% acetonitrile/water and isopropanol, respectively. M3 showed substantial antibacterial activities against both Gram-positive and Gram-negative bacteria which it appreciably retained in the presence of human serum and physiological salts. M3 was non-hemolytic against human red blood cells and non-cytotoxic to 3T3 cells up to 200 μM and to mice in vivo at a dose of 40 mg/kg. Furthermore, M3 neutralized LPS-induced pro-inflammatory responses in THP-1 cells and rat bone marrow-derived macrophages. Consequently, M3 attenuated LPS-mediated lung inflammation in mice and rescued them (80% survival at 10 mg/kg dose) against a lethal dose of LPS. The results demonstrate the identification of a 10-mer LPS-interacting, β-sheet peptide from MyD88 with the ability to form nanostructures and in vivo activity against LPS challenge in mice. The identified M3-template provides scope for designing novel bioactive peptides with β-sheet structures and self-assembling properties.
Collapse
Affiliation(s)
- Tripti Kumari
- Biochemistry and Structural Biology Division, CDRI, Lucknow 226031, India
| | | | - Jitendra Kuldeep
- Biochemistry and Structural Biology Division, CDRI, Lucknow 226031, India
| | | | - Neeraj Kumar Verma
- Biochemistry and Structural Biology Division, CDRI, Lucknow 226031, India
| | - Rohit Sahai
- Electron Microscopy Unit, CDRI, Lucknow 226031, India
| | | | - Jyotshana Saroj
- Biochemistry and Structural Biology Division, CDRI, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Mehmood Ali
- Biochemistry and Structural Biology Division, CDRI, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Kalyan Mitra
- Electron Microscopy Unit, CDRI, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Mohammad Imran Siddiqi
- Biochemistry and Structural Biology Division, CDRI, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore
| | - Jimut Kanti Ghosh
- Biochemistry and Structural Biology Division, CDRI, Lucknow 226031, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
7
|
Ferguson PM, Clarke M, Manzo G, Hind CK, Clifford M, Sutton JM, Lorenz CD, Phoenix DA, Mason AJ. Temporin B Forms Hetero-Oligomers with Temporin L, Modifies Its Membrane Activity, and Increases the Cooperativity of Its Antibacterial Pharmacodynamic Profile. Biochemistry 2022; 61:1029-1040. [PMID: 35609188 PMCID: PMC9178791 DOI: 10.1021/acs.biochem.1c00762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
The pharmacodynamic
profile of antimicrobial peptides (AMPs) and
their in vivo synergy are two factors that are thought
to restrict resistance evolution and ensure their conservation. The
frog Rana temporaria secretes a family of closely
related AMPs, temporins A–L, as an effective chemical dermal
defense. The antibacterial potency of temporin L has been shown to
increase synergistically in combination with both temporins B and
A, but this is modest. Here we show that the less potent temporin
B enhances the cooperativity of the in vitro antibacterial
activity of the more potent temporin L against EMRSA-15 and that this
may be associated with an altered interaction with the bacterial plasma
membrane, a feature critical for the antibacterial activity of most
AMPs. Addition of buforin II, a histone H2A fragment, can further
increase the cooperativity. Molecular dynamics simulations indicate
temporins B and L readily form hetero-oligomers in models of Gram-positive
bacterial plasma membranes. Patch-clamp studies show transmembrane
ion conductance is triggered with lower amounts of both peptides and
more quickly when used in combination, but conductance is of a lower
amplitude and pores are smaller. Temporin B may therefore act by forming
temporin L/B hetero-oligomers that are more effective than temporin
L homo-oligomers at bacterial killing and/or by reducing the probability
of the latter forming until a threshold concentration is reached.
Exploration of the mechanism of synergy between AMPs isolated from
the same organism may therefore yield antibiotic combinations with
advantageous pharmacodynamic properties.
Collapse
Affiliation(s)
- Philip M Ferguson
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Maria Clarke
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Giorgia Manzo
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Charlotte K Hind
- Technology Development Group, UKHSA, Salisbury SP4 0JG, United Kingdom
| | - Melanie Clifford
- Technology Development Group, UKHSA, Salisbury SP4 0JG, United Kingdom
| | - J Mark Sutton
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom.,Technology Development Group, UKHSA, Salisbury SP4 0JG, United Kingdom
| | - Christian D Lorenz
- Department of Physics, King's College London, London WC2R 2LS, United Kingdom
| | - David A Phoenix
- School of Applied Science, London South Bank University, 103 Borough Road, London SE1 0AA, United Kingdom
| | - A James Mason
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| |
Collapse
|
8
|
Bellavita R, Casciaro B, Di Maro S, Brancaccio D, Carotenuto A, Falanga A, Cappiello F, Buommino E, Galdiero S, Novellino E, Grossmann TN, Mangoni ML, Merlino F, Grieco P. First-in-Class Cyclic Temporin L Analogue: Design, Synthesis, and Antimicrobial Assessment. J Med Chem 2021; 64:11675-11694. [PMID: 34296619 PMCID: PMC8389922 DOI: 10.1021/acs.jmedchem.1c01033] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The pharmacodynamic and pharmacokinetic properties of bioactive peptides can be modulated by introducing conformational constraints such as intramolecular macrocyclizations, which can involve either the backbone and/or side chains. Herein, we aimed at increasing the α-helicity content of temporin L, an isoform of an intriguing class of linear antimicrobial peptides (AMPs), endowed with a wide antimicrobial spectrum, by the employment of diverse side-chain tethering strategies, including lactam, 1,4-substituted [1,2,3]-triazole, hydrocarbon, and disulfide linkers. Our approach resulted in a library of cyclic temporin L analogues that were biologically assessed for their antimicrobial, cytotoxic, and antibiofilm activities, leading to the development of the first-in-class cyclic peptide related to this AMP family. Our results allowed us to expand the knowledge regarding the relationship between the α-helical character of temporin derivatives and their biological activity, paving the way for the development of improved antibiotic cyclic AMP analogues.
Collapse
Affiliation(s)
- Rosa Bellavita
- Department
of Pharmacy, University of Naples “Federico
II”, Naples 80131, Italy
| | - Bruno Casciaro
- Center
for Life Nano- & Neuro-Science, Fondazione
Istituto Italiano di Tecnologia (IIT), Rome 00161, Italy
| | - Salvatore Di Maro
- DiSTABiF, University of Campania “Luigi
Vanvitelli”, Caserta 81100, Italy
| | - Diego Brancaccio
- Department
of Pharmacy, University of Naples “Federico
II”, Naples 80131, Italy
| | - Alfonso Carotenuto
- Department
of Pharmacy, University of Naples “Federico
II”, Naples 80131, Italy
| | - Annarita Falanga
- Department
of Agricultural Sciences, University of
Naples “Federico II”, Portici 80055, Italy
| | - Floriana Cappiello
- Department
of Biochemical Sciences, Laboratory affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, Rome 00185, Italy
| | - Elisabetta Buommino
- Department
of Pharmacy, University of Naples “Federico
II”, Naples 80131, Italy
| | - Stefania Galdiero
- Department
of Pharmacy, University of Naples “Federico
II”, Naples 80131, Italy
| | - Ettore Novellino
- Department
of Pharmacy, University of Naples “Federico
II”, Naples 80131, Italy
| | - Tom N. Grossmann
- Department
of Chemistry & Pharmaceutical Sciences, VU University Amsterdam, Amsterdam 1081 HZ, The Netherlands
| | - Maria Luisa Mangoni
- Department
of Biochemical Sciences, Laboratory affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, Rome 00185, Italy
| | - Francesco Merlino
- Department
of Pharmacy, University of Naples “Federico
II”, Naples 80131, Italy
| | - Paolo Grieco
- Department
of Pharmacy, University of Naples “Federico
II”, Naples 80131, Italy
| |
Collapse
|
9
|
Zai Y, Xi X, Ye Z, Ma C, Zhou M, Chen X, Siu SWI, Chen T, Wang L, Kwok HF. Aggregation and Its Influence on the Bioactivities of a Novel Antimicrobial Peptide, Temporin-PF, and Its Analogues. Int J Mol Sci 2021; 22:4509. [PMID: 33925935 PMCID: PMC8123395 DOI: 10.3390/ijms22094509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Temporin is an antimicrobial peptide (AMP) family discovered in the skin secretion of ranid frog that has become a promising alternative for conventional antibiotic therapy. Herein, a novel temporin peptide, Temporin-PF (TPF), was successfully identified from Pelophylax fukienensis. It exhibited potent activity against Gram-positive bacteria, but no effect on Gram-negative bacteria. Additionally, TPF exhibited aggregation effects in different solutions. Three analogs were further designed to study the relationship between the aggregation patterns and bioactivities, and the MD simulation was performed for revealing the pattern of the peptide assembly. As the results showed, all peptides were able to aggregate in the standard culture media and salt solutions, especially CaCl2 and MgCl2 buffers, where the aggregation was affected by the concentration of the salts. MD simulation reported that all peptides were able to form oligomers. The parent peptide assembly depended on the hydrophobic interaction via the residues in the middle domain of the sequence. However, the substitution of Trp/D-Trp resulted in an enhanced inter-peptide interaction in the zipper-like domain and eliminated overall biological activities. Our study suggested that introducing aromaticity at the zipper-like domain for temporin may not improve the bioactivities, which might be related to the formation of aggregates via the inter-peptide contacts at the zipper-like motif domain, and it could reduce the binding affinity to the lipid membrane of microorganisms.
Collapse
Affiliation(s)
- Yu Zai
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida da Univesidade, Taipa, Macau, China;
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Z.Y.); (C.M.); (M.Z.); (X.C.); (T.C.); (L.W.)
- Jiangsu Key Laboratory of Biofunctional Molecule, College of Life Sciences and Chemistry, Jiangsu Second Normal University, Nanjing 210013, China
| | - Xinping Xi
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Z.Y.); (C.M.); (M.Z.); (X.C.); (T.C.); (L.W.)
| | - Zhuming Ye
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Z.Y.); (C.M.); (M.Z.); (X.C.); (T.C.); (L.W.)
| | - Chengbang Ma
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Z.Y.); (C.M.); (M.Z.); (X.C.); (T.C.); (L.W.)
| | - Mei Zhou
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Z.Y.); (C.M.); (M.Z.); (X.C.); (T.C.); (L.W.)
| | - Xiaoling Chen
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Z.Y.); (C.M.); (M.Z.); (X.C.); (T.C.); (L.W.)
| | - Shirley W. I. Siu
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Avenida da Universidade, Taipa, Macau, China;
| | - Tianbao Chen
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Z.Y.); (C.M.); (M.Z.); (X.C.); (T.C.); (L.W.)
| | - Lei Wang
- School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (Z.Y.); (C.M.); (M.Z.); (X.C.); (T.C.); (L.W.)
| | - Hang Fai Kwok
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida da Univesidade, Taipa, Macau, China;
| |
Collapse
|
10
|
Petruk G, Puthia M, Petrlova J, Samsudin F, Strömdahl AC, Cerps S, Uller L, Kjellström S, Bond PJ, Schmidtchen AA. SARS-CoV-2 spike protein binds to bacterial lipopolysaccharide and boosts proinflammatory activity. J Mol Cell Biol 2021; 12:916-932. [PMID: 33295606 PMCID: PMC7799037 DOI: 10.1093/jmcb/mjaa067] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/12/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
There is a link between high lipopolysaccharide (LPS) levels in the blood and the metabolic syndrome, and metabolic syndrome predisposes patients to severe COVID-19. Here, we define an interaction between SARS-CoV-2 spike (S) protein and LPS, leading to aggravated inflammation in vitro and in vivo. Native gel electrophoresis demonstrated that SARS-CoV-2 S protein binds to LPS. Microscale thermophoresis yielded a KD of ∼47 nM for the interaction. Computational modeling and all-atom molecular dynamics simulations further substantiated the experimental results, identifying a main LPS-binding site in SARS-CoV-2 S protein. S protein, when combined with low levels of LPS, boosted nuclear factor-kappa B (NF-κB) activation in monocytic THP-1 cells and cytokine responses in human blood and peripheral blood mononuclear cells, respectively. The in vitro inflammatory response was further validated by employing NF-κB reporter mice and in vivo bioimaging. Dynamic light scattering, transmission electron microscopy, and LPS-FITC analyses demonstrated that S protein modulated the aggregation state of LPS, providing a molecular explanation for the observed boosting effect. Taken together, our results provide an interesting molecular link between excessive inflammation during infection with SARS-CoV-2 and comorbidities involving increased levels of bacterial endotoxins.
Collapse
Affiliation(s)
- Ganna Petruk
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Manoj Puthia
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Jitka Petrlova
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Firdaus Samsudin
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore
| | - Ann-Charlotte Strömdahl
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Samuel Cerps
- Unit of Respiratory Immunopharmacology, Department of Experimental Medicine, Lund University, SE-22184 Lund, Sweden
| | - Lena Uller
- Unit of Respiratory Immunopharmacology, Department of Experimental Medicine, Lund University, SE-22184 Lund, Sweden
| | - Sven Kjellström
- Division of Mass Spectrometry, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Peter J Bond
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - And Artur Schmidtchen
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden.,Copenhagen Wound Healing Center, Bispebjerg Hospital, Department of Biomedical Sciences, University of Copenhagen, DK-2400 Copenhagen, Denmark.,Dermatology, Skåne University Hospital, SE-22185 Lund, Sweden
| |
Collapse
|
11
|
Antimicrobial Peptides with Enhanced Salt Resistance and Antiendotoxin Properties. Int J Mol Sci 2020; 21:ijms21186810. [PMID: 32948086 PMCID: PMC7554977 DOI: 10.3390/ijms21186810] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/08/2020] [Accepted: 09/12/2020] [Indexed: 12/14/2022] Open
Abstract
A strategy was described to design antimicrobial peptides (AMPs) with enhanced salt resistance and antiendotoxin activities by linking two helical AMPs with the Ala-Gly-Pro (AGP) hinge. Among the designed peptides, KR12AGPWR6 demonstrated the best antimicrobial activities even in high salt conditions (NaCl ~300 mM) and possessed the strongest antiendotoxin activities. These activities may be related to hydrophobicity, membrane-permeability, and α-helical content of the peptide. Amino acids of the C-terminal helices were found to affect the peptide-induced permeabilization of LUVs, the α-helicity of the designed peptides under various LUVs, and the LPS aggregation and size alternation. A possible model was proposed to explain the mechanism of LPS neutralization by the designed peptides. These findings could provide a new approach for designing AMPs with enhanced salt resistance and antiendotoxin activities for potential therapeutic applications.
Collapse
|
12
|
Kumari T, Verma DP, Afshan T, Verma NK, Pant G, Ali M, Shukla PK, Mitra K, Ghosh JK. A Noncytotoxic Temporin L Analogue with In Vivo Antibacterial and Antiendotoxin Activities and a Nonmembrane-Lytic Mode of Action. ACS Infect Dis 2020; 6:2369-2385. [PMID: 32786286 DOI: 10.1021/acsinfecdis.0c00022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cytotoxic frog antimicrobial peptide Temporin L (TempL) is an attractive molecule for the design of lead antimicrobial agents due to its short size and versatile biological activities. However, noncytotoxic TempL variants with desirable biological activities have rarely been reported. TempL analogue Q3K,TempL is water-soluble and possesses a significant antiendotoxin property along with comparable cytotoxicity to TempL. A phenylalanine residue, located at the hydrophobic face of Q3K,TempL and the "d" position of its phenylalanine zipper sequence, was replaced with a cationic lysine residue. This analogue, Q3K,F8K,TempL, showed reduced hydrophobic moment and was noncytotoxic with lower antimicrobial activity. Interestingly, swapping between tryptophan at the fourth and serine at the sixth positions turned Q3K,F8K,TempL totally amphipathic as reflected by its helical wheel projection with clusters of hydrophobic and hydrophilic residues and the highest hydrophobic moment among these peptides. Surprisingly, this analogue, SW,Q3K,F8K,TempL, was as noncytotoxic as Q3K,F8K,TempL but showed augmented antimicrobial and antiendotoxin properties, comparable to that of TempL and Q3K,TempL. SW,Q3K,F8K,TempL exhibited appreciable survival of mice against P. aeruginosa infection and a lipopolysaccharide (LPS) challenge. Unlike TempL and Q3K,TempL, SW,Q3K,F8K,TempL adopted an unordered secondary structure in bacterial membrane mimetic lipid vesicles and did not permeabilize them or depolarize the bacterial membrane. Overall, the results demonstrate the design of a nontoxic TempL analogue that possesses clusters of hydrophobic and hydrophilic residues with impaired secondary structure and shows a nonmembrane-lytic mechanism and in vivo antiendotoxin and antimicrobial activities. This paradigm of design of antimicrobial peptide with clusters of hydrophobic and hydrophilic residues and high hydrophobic moment but low secondary structure could be attempted further.
Collapse
Affiliation(s)
- Tripti Kumari
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Devesh Pratap Verma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Tayyaba Afshan
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Neeraj Kumar Verma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Garima Pant
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Mehmood Ali
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - P. K. Shukla
- Microbiology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Kalyan Mitra
- Electron Microscopy Unit, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| | - Jimut Kanti Ghosh
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh 226 031, India
| |
Collapse
|
13
|
Bhattacharjya S, Straus SK. Design, Engineering and Discovery of Novel α-Helical and β-Boomerang Antimicrobial Peptides against Drug Resistant Bacteria. Int J Mol Sci 2020; 21:ijms21165773. [PMID: 32796755 PMCID: PMC7460851 DOI: 10.3390/ijms21165773] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022] Open
Abstract
In an era where the pipeline of new antibiotic development is drying up, the continuous rise of multi-drug resistant (MDR) and extensively drug resistant (XDR) bacteria are genuine threats to human health. Although antimicrobial peptides (AMPs) may serve as promising leads against drug resistant bacteria, only a few AMPs are in advanced clinical trials. The limitations of AMPs, namely their low in vivo activity, toxicity, and poor bioavailability, need to be addressed. Here, we review engineering of frog derived short α-helical AMPs (aurein, temporins) and lipopolysaccharide (LPS) binding designed β-boomerang AMPs for further development. The discovery of novel cell selective AMPs from the human proprotein convertase furin is also discussed.
Collapse
Affiliation(s)
- Surajit Bhattacharjya
- School of Biological Sciences, 60 Nanyang Drive, Nanyang Technological University, Singapore 637551, Singapore
- Correspondence: (S.B.); (S.K.S.)
| | - Suzana K. Straus
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
- Correspondence: (S.B.); (S.K.S.)
| |
Collapse
|
14
|
Mercer DK, Torres MDT, Duay SS, Lovie E, Simpson L, von Köckritz-Blickwede M, de la Fuente-Nunez C, O'Neil DA, Angeles-Boza AM. Antimicrobial Susceptibility Testing of Antimicrobial Peptides to Better Predict Efficacy. Front Cell Infect Microbiol 2020; 10:326. [PMID: 32733816 PMCID: PMC7358464 DOI: 10.3389/fcimb.2020.00326] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
During the development of antimicrobial peptides (AMP) as potential therapeutics, antimicrobial susceptibility testing (AST) stands as an essential part of the process in identification and optimisation of candidate AMP. Standard methods for AST, developed almost 60 years ago for testing conventional antibiotics, are not necessarily fit for purpose when it comes to determining the susceptibility of microorganisms to AMP. Without careful consideration of the parameters comprising AST there is a risk of failing to identify novel antimicrobials at a time when antimicrobial resistance (AMR) is leading the planet toward a post-antibiotic era. More physiologically/clinically relevant AST will allow better determination of the preclinical activity of drug candidates and allow the identification of lead compounds. An important consideration is the efficacy of AMP in biological matrices replicating sites of infection, e.g., blood/plasma/serum, lung bronchiolar lavage fluid/sputum, urine, biofilms, etc., as this will likely be more predictive of clinical efficacy. Additionally, specific AST for different target microorganisms may help to better predict efficacy of AMP in specific infections. In this manuscript, we describe what we believe are the key considerations for AST of AMP and hope that this information can better guide the preclinical development of AMP toward becoming a new generation of urgently needed antimicrobials.
Collapse
Affiliation(s)
| | - Marcelo D. T. Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Penn Institute for Computational Science, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Searle S. Duay
- Department of Chemistry, Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Emma Lovie
- NovaBiotics Ltd, Aberdeen, United Kingdom
| | | | | | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, Penn Institute for Computational Science, and Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Alfredo M. Angeles-Boza
- Department of Chemistry, Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
15
|
Wu CL, Hsueh JY, Yip BS, Chih YH, Peng KL, Cheng JW. Antimicrobial Peptides Display Strong Synergy with Vancomycin Against Vancomycin-Resistant E. faecium, S. aureus, and Wild-Type E. coli. Int J Mol Sci 2020; 21:ijms21134578. [PMID: 32605123 PMCID: PMC7369893 DOI: 10.3390/ijms21134578] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 01/25/2023] Open
Abstract
There is an urgent and imminent need to develop new antimicrobials to fight against antibiotic-resistant bacterial and fungal strains. In this study, a checkerboard method was used to evaluate the synergistic effects of the antimicrobial peptide P-113 and its bulky non-nature amino acid substituted derivatives with vancomycin against vancomycin-resistant Enterococcus faecium, Staphylococcus aureus, and wild-type Escherichia coli. Boron-dipyrro-methene (BODIPY) labeled vancomycin was used to characterize the interactions between the peptides, vancomycin, and bacterial strains. Moreover, neutralization of antibiotic-induced releasing of lipopolysaccharide (LPS) from E. coli by the peptides was obtained. Among these peptides, Bip-P-113 demonstrated the best minimal inhibitory concentrations (MICs), antibiotics synergism, bacterial membrane permeabilization, and supernatant LPS neutralizing activities against the bacteria studied. These results could help in developing antimicrobial peptides that have synergistic activity with large size glycopeptides such as vancomycin in therapeutic applications.
Collapse
Affiliation(s)
- Chih-Lung Wu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; (C.-L.W.); (J.-Y.H.); (B.-S.Y.); (Y.-H.C.); (K.-L.P.)
| | - Ju-Yun Hsueh
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; (C.-L.W.); (J.-Y.H.); (B.-S.Y.); (Y.-H.C.); (K.-L.P.)
| | - Bak-Sau Yip
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; (C.-L.W.); (J.-Y.H.); (B.-S.Y.); (Y.-H.C.); (K.-L.P.)
- Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu 300, Taiwan
| | - Ya-Han Chih
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; (C.-L.W.); (J.-Y.H.); (B.-S.Y.); (Y.-H.C.); (K.-L.P.)
| | - Kuang-Li Peng
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; (C.-L.W.); (J.-Y.H.); (B.-S.Y.); (Y.-H.C.); (K.-L.P.)
| | - Jya-Wei Cheng
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; (C.-L.W.); (J.-Y.H.); (B.-S.Y.); (Y.-H.C.); (K.-L.P.)
- Correspondence: ; Tel.: +886-3-574-2763; Fax: +886-3-571-5934
| |
Collapse
|
16
|
Immunomodulatory Functions of the Human Cathelicidin LL-37 (aa 13-31)-Derived Peptides are Associated with Predicted α-Helical Propensity and Hydrophobic Index. Biomolecules 2019; 9:biom9090501. [PMID: 31540479 PMCID: PMC6769993 DOI: 10.3390/biom9090501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/03/2019] [Accepted: 09/15/2019] [Indexed: 12/18/2022] Open
Abstract
The anti-endotoxin activity of the cationic peptide LL-37 and its derivative IG-19 is attributed to electrostatic interaction of the peptides’ positive charge with negatively charged bacterial lipopolysaccharides (LPS), and in part to the alteration of intracellular mechanisms independent of peptide binding to LPS. We examined the immunomodulatory responses induced by IG-19 and four IG-19-derived scrambled peptides (IG-19a–d), in the presence and absence of LPS, in macrophages and peripheral blood-derived mononuclear cells. All peptides had identical net charge (+5) and amino acid composition, but different hydrophobicity and α-helical propensity. Peptide IG-19 suppressed LPS-induced cytokine/chemokine production by >90%, IG-19a and IG-19b suppressed it by 40–50%, and IG-19c and IG-19d did not suppress cytokine/chemokine production at all. In silico prediction algorithms and the peptide retention time (RT) on a C18 RP HPLC column indicated a linear association between α-helical propensity and hydrophobicity with the ability of the peptides to inhibit LPS-induced responses. Peptide RT exhibited a significant correlation (>70%) between the suppression of LPS-induced cytokine/chemokine production and peptide-induced production of the anti-inflammatory cytokine IL-1RA. These results indicate that RT on a C18 column can be used as a predictor for the immunomodulatory functions of cationic peptides. Overall, we demonstrated that the immunomodulatory functions of LL-37-derived peptides with identical positive charge and amino acid composition are directly associated with the predicted α-helical propensity and hydrophobicity of the peptides.
Collapse
|
17
|
Temporin L and aurein 2.5 have identical conformations but subtly distinct membrane and antibacterial activities. Sci Rep 2019; 9:10934. [PMID: 31358802 PMCID: PMC6662694 DOI: 10.1038/s41598-019-47327-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 07/16/2019] [Indexed: 11/22/2022] Open
Abstract
Frogs such as Rana temporaria and Litoria aurea secrete numerous closely related antimicrobial peptides (AMPs) as an effective chemical dermal defence. Damage or penetration of the bacterial plasma membrane is considered essential for AMP activity and such properties are commonly ascribed to their ability to form secondary amphipathic, α-helix conformations in membrane mimicking milieu. Nevertheless, despite the high similarity in physical properties and preference for adopting such conformations, the spectrum of activity and potency of AMPs often varies considerably. Hence distinguishing apparently similar AMPs according to their behaviour in, and effects on, model membranes will inform understanding of primary-sequence-specific antimicrobial mechanisms. Here we use a combination of molecular dynamics simulations, circular dichroism and patch-clamp to investigate the basis for differing anti-bacterial activities in representative AMPs from each species; temporin L and aurein 2.5. Despite adopting near identical, α-helix conformations in the steady-state in a variety of membrane models, these two AMPs can be distinguished both in vitro and in silico based on their dynamic interactions with model membranes, notably their differing conformational flexibility at the N-terminus, ability to form higher order aggregates and the characteristics of induced ion conductance. Taken together, these differences provide an explanation of the greater potency and broader antibacterial spectrum of activity of temporin L over aurein 2.5. Consequently, while the secondary amphipathic, α-helix conformation is a key determinant of the ability of a cationic AMP to penetrate and disrupt the bacterial plasma membrane, the exact mechanism, potency and spectrum of activity is determined by precise structural and dynamic contributions from specific residues in each AMP sequence.
Collapse
|
18
|
Malekkhaiat Häffner S, Malmsten M. Influence of self-assembly on the performance of antimicrobial peptides. Curr Opin Colloid Interface Sci 2018. [DOI: 10.1016/j.cocis.2018.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
19
|
Liang X, Wang R, Dou W, Zhao L, Zhou L, Zhu J, Wang K, Yan J. Arminin 1a-C, a novel antimicrobial peptide from ancient metazoan Hydra, shows potent antileukemia activity against drug-sensitive and drug-resistant leukemia cells. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:3691-3703. [PMID: 30464401 PMCID: PMC6217004 DOI: 10.2147/dddt.s181188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Purpose Due to the emergence of multidrug resistance (MDR), traditional antileukemia drugs no longer meet the treatment needs. Therefore, new antileukemia drugs with different action mechanisms are urgently needed to cope with this situation. Materials and methods Arminin 1a-C is an antimicrobial peptide (AMP) developed from the ancient metazoan marine Hydra. In this study, we first explored its antileukemia activity. Results Our results showed that Arminin 1a-C formed an α-helical structure and efficaciously suppressed the viability of leukemia cell lines whether or not they were multidrug resistant or sensitive, and there were no obvious differences between these cell lines. Arminin 1a-C exhibited distinct selectivity between noncancerous and cancerous cell lines. Arminin 1a-C interfered with K562/adriamycin (ADM) cell (a kind of multidrug-resistant leukemia cell line) proliferation in a very rapid manner and formed pores in its cell membrane, making it difficult to develop resistance against Arminin 1a-C. Conclusion Our data show that Arminin 1a-C possesses great potential as a therapeutic candidate for the treatment of multidrug-resistant leukemia.
Collapse
Affiliation(s)
- Xiaolei Liang
- The Reproductive Medicine Special Hospital of the First Hospital of Lanzhou University, Key Laboratory for Reproductive Medicine and Embryo, Lanzhou, China
| | - Ruirui Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wenshan Dou
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Li Zhao
- The Key Laboratory, The First Hospital of Lanzhou University, Lanzhou, China,
| | - Lanxia Zhou
- The Key Laboratory, The First Hospital of Lanzhou University, Lanzhou, China,
| | - Junfang Zhu
- The Key Laboratory, The First Hospital of Lanzhou University, Lanzhou, China,
| | - Kairong Wang
- School of Basic Medical Sciences, Institute of Biochemistry and Molecular Biology, Lanzhou University, Lanzhou, China
| | - Jiexi Yan
- The Key Laboratory, The First Hospital of Lanzhou University, Lanzhou, China,
| |
Collapse
|
20
|
Chih YH, Wang SY, Yip BS, Cheng KT, Hsu SY, Wu CL, Yu HY, Cheng JW. Dependence on size and shape of non-nature amino acids in the enhancement of lipopolysaccharide (LPS) neutralizing activities of antimicrobial peptides. J Colloid Interface Sci 2018; 533:492-502. [PMID: 30176540 DOI: 10.1016/j.jcis.2018.08.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/11/2022]
Abstract
HYPOTHESIS Release of lipopolysaccharides (LPS) from bacteria into bloodstream may cause serious unwanted stimulation of the host immune system. P-113 is a clinically active histidine-rich antimicrobial peptide. Nal-P-113, a β-naphthylalanine-substituted P-113, is salt-resistant but has limited LPS neutralizing activity. We suspected the size and shape of the non-natural bulky amino acid may affect its LPS neutralizing activity. Herein, antimicrobial, LPS neutralizing, and antiproteolytic effects of phenylalanine- (Phe-P-113), β-naphthylalanine- (Nal-P-113), β-diphenylalanine- (Dip-P-113), and β-(4,4'-biphenyl)alanine- (Bip-P-113) substituted P-113 were studied. EXPERIMENTS Structure-activity relationships of P-113, Phe-P-113, Nal-P-113, Dip-P-113, and Bip-P-113 were evaluated using antimicrobial activity assays, serum proteolytic assays, peptide-induced permeabilization of large unilamellar vesicles, zeta potential measurements, dynamic light scattering measurement of LPS aggregation, and Limulus amebocyte lysate assays for measuring LPS neutralization. In vitro and in vivo LPS neutralizing activities were further confirmed by LPS-induced inflammation inhibition in an endotoxemia mouse model. FINDINGS Bip-P-113 and Dip-P-113 had the longest and widest non-nature amino acids, respectively. Bip-P-113 enhanced salt resistance, serum proteolytic stability, peptide-induced permeabilization, zeta potential measurements, LPS aggregation, and in vitro and in vivo LPS neutralizing activities. These results could help design novel antimicrobial peptides that have enhanced stability in vivo and that can have potential therapeutic applications.
Collapse
Affiliation(s)
- Ya-Han Chih
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Siou-Ying Wang
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Bak-Sau Yip
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu 300, Taiwan
| | - Kuang-Ting Cheng
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Su-Ya Hsu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chih-Lung Wu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hui-Yuan Yu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jya-Wei Cheng
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
21
|
Sayeed M, Gautam S, Verma DP, Afshan T, Kumari T, Srivastava AK, Ghosh JK. A collagen domain-derived short adiponectin peptide activates APPL1 and AMPK signaling pathways and improves glucose and fatty acid metabolisms. J Biol Chem 2018; 293:13509-13523. [PMID: 29991592 DOI: 10.1074/jbc.ra118.001801] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 07/02/2018] [Indexed: 12/13/2022] Open
Abstract
Adiponectin is a fat tissue-derived adipokine with beneficial effects against diabetes, cardiovascular diseases, and cancer. Accordingly, adiponectin-mimetic molecules possess significant pharmacological potential. Oligomeric states of adiponectin appear to determine its biological activity. We identified a highly conserved, 13-residue segment (ADP-1) from adiponectin's collagen domain, which comprises GXXG motifs and has one asparagine and two histidine residues that assist in oligomeric protein assembly. We therefore hypothesized that ADP-1 promotes oligomeric assembly and thereby mediates potential metabolic effects. We observed here that ADP-1 is stable in human serum and oligomerizes in aqueous environments. We also found that ADP-1 activates AMP-activated protein kinase (AMPK) in an adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1)-dependent pathway and stimulates glucose uptake in rat skeletal muscle cells (L6 myotubes). ADP-1-induced glucose transport coincided with ADP-1-induced biosynthesis of glucose transporter 4 and its translocation to the plasma membrane. ADP-1 induced an interaction between APPL1 and the small GTPase Rab5, resulting in AMPK phosphorylation, in turn leading to phosphorylation of p38 mitogen-activated protein kinase (MAPK), acetyl-CoA carboxylase, and peroxisome proliferator-activated receptor α. Similar to adiponectin, ADP-1 increased the expression of the adiponectin receptor 1 (AdipoR1) gene. Of note, ADP-1 decreased blood glucose levels and enhanced insulin production in pancreatic β cells in db/db mice. Further, ADP-1 beneficially affected lipid metabolism by enhancing lipid globule formation in mouse 3T3-L1 adipocytes. To our knowledge, this is the first report on identification of a short peptide from adiponectin with positive effects on glucose or fatty acid metabolism.
Collapse
Affiliation(s)
- Mohd Sayeed
- From the Molecular and Structural Biology Division and
| | - Sudeep Gautam
- the Biochemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow-226 031, India
| | | | | | - Tripti Kumari
- From the Molecular and Structural Biology Division and
| | - Arvind Kumar Srivastava
- the Biochemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow-226 031, India
| | | |
Collapse
|
22
|
Almaaytah A, Qaoud MT, Abualhaijaa A, Al-Balas Q, Alzoubi KH. Hybridization and antibiotic synergism as a tool for reducing the cytotoxicity of antimicrobial peptides. Infect Drug Resist 2018; 11:835-847. [PMID: 29910626 PMCID: PMC5987794 DOI: 10.2147/idr.s166236] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Introduction As the development of new antimicrobial agents faces a historical decline, the issue of bacterial drug resistance has become a serious dilemma that threatens the human population worldwide. Antimicrobial peptides (AMPs) represent an attractive and a promising class of antimicrobial agents. Aim The hybridization of AMPs aimed at merging two individual active fragments of native peptides to generate a new AMP with altered physicochemical properties that translate into an enhanced safety profile. Materials and methods In this study, we have rationally designed a new hybrid peptide via combining two individual α-helical fragments of both BMAP-27 and OP-145. The resultant peptide, was evaluated for its antimicrobial and antibiofilm activity against a range of microbial strains. The resultant peptide was also evaluated for its toxicity against mammalian cells using hemolytic and anti proliferative assays. Results The antimicrobial activity of H4 revealed that the peptide is displaying a broad spectrum of activity against both Gram-positive and Gram-negative bacteria including standard and multidrug-resistant bacterial strains in the range of 2.5-25 μM. The new hybrid peptide displayed potent activity in eradicating biofilm-forming cells, and the reported minimum biofilm eradication concentrations were equal to the minimum inhibitory concentration values reported for planktonic cells. Additionally, H4 exhibited reduced toxicity profiles against eukaryotic cells. Combining H4 peptide with conventional antibiotics has led to a dramatic enhancement of the antimicrobial activity of both agents with synergistic or additive outcomes. Conclusion Overall, this study indicates the success of both the hybridization and synergism strategy in developing AMPs as potential antimicrobial therapeutics with reduced toxicity profiles that could be efficiently employed to eradicate resistant bacterial strains and enhance the selectivity and toxicity profiles of native AMPs.
Collapse
Affiliation(s)
- Ammar Almaaytah
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammed T Qaoud
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Ahmad Abualhaijaa
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| | - Qosay Al-Balas
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H Alzoubi
- Department Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
23
|
Recent Advances in Antibacterial and Antiendotoxic Peptides or Proteins from Marine Resources. Mar Drugs 2018; 16:md16020057. [PMID: 29439417 PMCID: PMC5852485 DOI: 10.3390/md16020057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/23/2018] [Accepted: 02/02/2018] [Indexed: 12/17/2022] Open
Abstract
Infectious diseases caused by Gram-negative bacteria and sepsis induced by lipopolysaccharide (LPS) pose a major threat to humans and animals and cause millions of deaths each year. Marine organisms are a valuable resource library of bioactive products with huge medicinal potential. Among them, antibacterial and antiendotoxic peptides or proteins, which are composed of metabolically tolerable residues, are present in many marine species, including marine vertebrates, invertebrates and microorganisms. A lot of studies have reported that these marine peptides and proteins or their derivatives exhibit potent antibacterial activity and antiendotoxic activity in vitro and in vivo. However, their categories, heterologous expression in microorganisms, physicochemical factors affecting peptide or protein interactions with bacterial LPS and LPS-neutralizing mechanism are not well known. In this review, we highlight the characteristics and anti-infective activity of bifunctional peptides or proteins from marine resources as well as the challenges and strategies for further study.
Collapse
|
24
|
Mohamed MF, Brezden A, Mohammad H, Chmielewski J, Seleem MN. A short D-enantiomeric antimicrobial peptide with potent immunomodulatory and antibiofilm activity against multidrug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii. Sci Rep 2017; 7:6953. [PMID: 28761101 PMCID: PMC5537347 DOI: 10.1038/s41598-017-07440-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 06/28/2017] [Indexed: 11/26/2022] Open
Abstract
Antimicrobial peptides (AMPs) represent a promising therapeutic alternative for the treatment of antibiotic-resistant bacterial infections. The present study investigates the antimicrobial activity of new, rationally-designed derivatives of a short α-helical peptide, RR. From the peptides designed, RR4 and its D-enantiomer, D-RR4, emerged as the most potent analogues with a more than 32-fold improvement in antimicrobial activity observed against multidrug-resistant strains of Pseudomonas aeruginosa and Acinetobacter baumannii. Remarkably, D-RR4 demonstrated potent activity against colistin-resistant strains of P. aeruginosa (isolated from cystic fibrosis patients) indicating a potential therapeutic advantage of this peptide over several AMPs. In contrast to many natural AMPs, D-RR4 retained its activity under challenging physiological conditions (high salts, serum, and acidic pH). Furthermore, D-RR4 was more capable of disrupting P. aeruginosa and A. baumannii biofilms when compared to conventional antibiotics. Of note, D-RR4 was able to bind to lipopolysaccharide to reduce the endotoxin-induced proinflammatory cytokine response in macrophages. Finally, D-RR4 protected Caenorhabditis elegans from lethal infections of P. aeruginosa and A. baumannii and enhanced the activity of colistin in vivo against colistin-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Mohamed F Mohamed
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, 47907, USA
| | - Anna Brezden
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Haroon Mohammad
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, 47907, USA
| | - Jean Chmielewski
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA.,Purdue Institute for Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, 47907, USA
| | - Mohamed N Seleem
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, 47907, USA. .,Purdue Institute for Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
25
|
Farrotti A, Conflitti P, Srivastava S, Ghosh JK, Palleschi A, Stella L, Bocchinfuso G. Molecular Dynamics Simulations of the Host Defense Peptide Temporin L and Its Q3K Derivative: An Atomic Level View from Aggregation in Water to Bilayer Perturbation. Molecules 2017; 22:molecules22071235. [PMID: 28737669 PMCID: PMC6152314 DOI: 10.3390/molecules22071235] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/20/2017] [Accepted: 07/20/2017] [Indexed: 12/03/2022] Open
Abstract
Temporin L (TempL) is a 13 residue Host Defense Peptide (HDP) isolated from the skin of frogs. It has a strong affinity for lipopolysaccharides (LPS), which is related to its high activity against Gram-negative bacteria and also to its strong tendency to neutralize the pro-inflammatory response caused by LPS release from inactivated bacteria. A designed analog with the Q3K substitution shows an enhancement in both these activities. In the present paper, Molecular Dynamics (MD) simulations have been used to investigate the origin of these improved properties. To this end, we have studied the behavior of the peptides both in water solution and in the presence of LPS lipid-A bilayers, demonstrating that the main effect through which the Q3K substitution improves the peptide activities is the destabilization of peptide aggregates in water.
Collapse
Affiliation(s)
- Andrea Farrotti
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma "Tor Vergata", Rome 00133, Italy.
| | - Paolo Conflitti
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma "Tor Vergata", Rome 00133, Italy.
| | - Saurabh Srivastava
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India.
| | - Jimut Kanti Ghosh
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India.
| | - Antonio Palleschi
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma "Tor Vergata", Rome 00133, Italy.
| | - Lorenzo Stella
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma "Tor Vergata", Rome 00133, Italy.
| | - Gianfranco Bocchinfuso
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma "Tor Vergata", Rome 00133, Italy.
| |
Collapse
|
26
|
Grassi L, Maisetta G, Maccari G, Esin S, Batoni G. Analogs of the Frog-skin Antimicrobial Peptide Temporin 1Tb Exhibit a Wider Spectrum of Activity and a Stronger Antibiofilm Potential as Compared to the Parental Peptide. Front Chem 2017; 5:24. [PMID: 28443279 PMCID: PMC5387044 DOI: 10.3389/fchem.2017.00024] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/23/2017] [Indexed: 11/13/2022] Open
Abstract
The frog skin-derived peptide Temporin 1Tb (TB) has gained increasing attention as novel antimicrobial agent for the treatment of antibiotic-resistant and/or biofilm-mediated infections. Nevertheless, such a peptide possesses a preferential spectrum of action against Gram-positive bacteria. In order to improve the therapeutic potential of TB, the present study evaluated the antibacterial and antibiofilm activities of two TB analogs against medically relevant bacterial species. Of the two analogs, TB_KKG6A has been previously described in the literature, while TB_L1FK is a new analog designed by us through statistical-based computational strategies. Both TB analogs displayed a faster and stronger bactericidal activity than the parental peptide, especially against Gram-negative bacteria in planktonic form. Differently from the parental peptide, TB_KKG6A and TB_L1FK were able to inhibit the formation of Staphylococcus aureus biofilms by more than 50% at 12 μM, while only TB_KKG6A prevented the formation of Pseudomonas aeruginosa biofilms at 24 μM. A marked antibiofilm activity against preformed biofilms of both bacterial species was observed for the two TB analogs when used in combination with EDTA. Analysis of synergism at the cellular level suggested that the antibiofilm activity exerted by the peptide-EDTA combinations against mature biofilms might be due mainly to a disaggregating effect on the extracellular matrix in the case of S. aureus, and to a direct activity on biofilm-embedded cells in the case of P. aeruginosa. Both analogs displayed a low hemolytic effect at the active concentrations and, overall, TB_L1FK resulted less cytotoxic toward mammalian cells. Collectively, the results obtained demonstrated that subtle changes in the primary sequence of TB may provide TB analogs that, used alone or in combination with adjuvant molecules such as EDTA, exhibit promising features against both planktonic and biofilm cells of medically relevant bacteria.
Collapse
Affiliation(s)
- Lucia Grassi
- Department of Translational Research and new Technologies in Medicine and Surgery, University of PisaPisa, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and new Technologies in Medicine and Surgery, University of PisaPisa, Italy
| | - Giuseppe Maccari
- Center for Nanotechnology Innovation @NEST, Italian Institute of TechnologyPisa, Italy
| | - Semih Esin
- Department of Translational Research and new Technologies in Medicine and Surgery, University of PisaPisa, Italy
| | - Giovanna Batoni
- Department of Translational Research and new Technologies in Medicine and Surgery, University of PisaPisa, Italy
| |
Collapse
|
27
|
Mechanistic and structural basis of bioengineered bovine Cathelicidin-5 with optimized therapeutic activity. Sci Rep 2017; 7:44781. [PMID: 28322271 PMCID: PMC5359555 DOI: 10.1038/srep44781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/13/2017] [Indexed: 11/08/2022] Open
Abstract
Peptide-drug discovery using host-defense peptides becomes promising against antibiotic-resistant pathogens and cancer cells. Here, we customized the therapeutic activity of bovine cathelicidin-5 targeting to bacteria, protozoa, and tumor cells. The membrane dependent conformational adaptability and plasticity of cathelicidin-5 is revealed by biophysical analysis and atomistic simulations over 200 μs in thymocytes, leukemia, and E. coli cell-membranes. Our understanding of energy-dependent cathelicidin-5 intrusion in heterogeneous membranes aided in designing novel loss/gain-of-function analogues. In vitro findings identified leucine-zipper to phenylalanine substitution in cathelicidin-5 (1-18) significantly enhance the antimicrobial and anticancer activity with trivial hemolytic activity. Targeted mutants of cathelicidin-5 at kink region and N-terminal truncation revealed loss-of-function. We ensured the existence of a bimodal mechanism of peptide action (membranolytic and non-membranolytic) in vitro. The melanoma mouse model in vivo study further supports the in vitro findings. This is the first structural report on cathelicidin-5 and our findings revealed potent therapeutic application of designed cathelicidin-5 analogues.
Collapse
|
28
|
Qiu S, Zhu R, Zhao Y, An X, Jia F, Peng J, Ma Z, Zhu Y, Wang J, Su J, Wang Q, Wang H, Li Y, Wang K, Yan W, Wang R. Antimicrobial activity and stability of protonectin withD-amino acid substitutions. J Pept Sci 2017; 23:392-402. [DOI: 10.1002/psc.2989] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Shuai Qiu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Ranran Zhu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Yanyan Zhao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Xiaoping An
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Fengjing Jia
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Jinxiu Peng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Zelin Ma
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Yuanyuan Zhu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Jiayi Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Jinhuan Su
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Qingjun Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Hailin Wang
- The People's Hospital in Gansu Province; 204 West Donggang Road Lanzhou 730000 China
| | - Yuan Li
- The People's Hospital in Gansu Province; 204 West Donggang Road Lanzhou 730000 China
| | - Kairong Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Wenjin Yan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences; Institute of Biochemistry and Molecular Biology, School of Life Sciences, Lanzhou University; 222 Tian Shui South Road Lanzhou 730000 China
| |
Collapse
|
29
|
Yu HY, Chen YA, Yip BS, Wang SY, Wei HJ, Chih YH, Chen KH, Cheng JW. Role of β-naphthylalanine end-tags in the enhancement of antiendotoxin activities: Solution structure of the antimicrobial peptide S1-Nal-Nal in complex with lipopolysaccharide. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1114-1123. [PMID: 28288781 DOI: 10.1016/j.bbamem.2017.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/08/2017] [Accepted: 03/09/2017] [Indexed: 12/18/2022]
Abstract
Lipopolysaccharide (LPS, endotoxin) is the major component of Gram-negative bacterial outer surface membrane. LPS released from bacteria into bloodstream during infection may cause serious unwanted stimulation of host's immune system and lead to septic shock of the patient. Recently, we have developed a strategy to increase salt resistance and LPS neutralization of short antimicrobial peptides by adding β-naphthylalanine end-tags to their termini. Herein, correlations between membrane immersion depth, orientation, and antiendotoxin activities of the antimicrobial peptides S1 and S1-Nal-Nal have been investigated via solution structure, paramagnetic resonance enhancement, and saturation transfer difference NMR studies. Unlike the parent peptide S1, S1-Nal-Nal rotated its two terminal β-naphthylalanine residues into the hydrophobic lipid A motif of LPS micelles. The LPS-induced inflammation may then be prohibited by the blocked lipid A motif.
Collapse
Affiliation(s)
- Hui-Yuan Yu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-An Chen
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Bak-Sau Yip
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu 300, Taiwan
| | - Siou-Ying Wang
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hsiu-Ju Wei
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ya-Han Chih
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Kuan-Hao Chen
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jya-Wei Cheng
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
30
|
Targeting biofilms and persisters of ESKAPE pathogens with P14KanS, a kanamycin peptide conjugate. Biochim Biophys Acta Gen Subj 2017; 1861:848-859. [PMID: 28132897 DOI: 10.1016/j.bbagen.2017.01.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/19/2017] [Accepted: 01/26/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND The worldwide emergence of antibiotic resistance represents a serious medical threat. The ability of these resistant pathogens to form biofilms that are highly tolerant to antibiotics further aggravates the situation and leads to recurring infections. Thus, new therapeutic approaches that adopt novel mechanisms of action are urgently needed. To address this significant problem, we conjugated the antibiotic kanamycin with a novel antimicrobial peptide (P14LRR) to develop a kanamycin peptide conjugate (P14KanS). METHODS Antibacterial activities were evaluated in vitro and in vivo using a Caenorhabditis elegans model. Additionally, the mechanism of action, antibiofilm activity and anti-inflammatory effect of P14KanS were investigated. RESULTS P14KanS exhibited potent antimicrobial activity against ESKAPE pathogens. P14KanS demonstrated a ≥128-fold improvement in MIC relative to kanamycin against kanamycin-resistant strains. Mechanistic studies confirmed that P14KanS exerts its antibacterial effect by selectively disrupting the bacterial cell membrane. Unlike many antibiotics, P14KanS demonstrated rapid bactericidal activity against stationary phases of both Gram-positive and Gram-negative pathogens. Moreover, P14KanS was superior in disrupting adherent bacterial biofilms and in killing intracellular pathogens as compared to conventional antibiotics. Furthermore, P14KanS demonstrated potent anti-inflammatory activity via the suppression of LPS-induced proinflammatory cytokines. Finally, P14KanS protected C. elegans from lethal infections of both Gram-positive and Gram-negative pathogens. CONCLUSIONS The potent in vitro and in vivo activity of P14KanS warrants further investigation as a potential therapeutic agent for bacterial infections. GENERAL SIGNIFICANCE This study demonstrates that equipping kanamycin with an antimicrobial peptide is a promising method to tackle bacterial biofilms and address bacterial resistance to aminoglycosides.
Collapse
|
31
|
Piscidin-1-analogs with double L- and D-lysine residues exhibited different conformations in lipopolysaccharide but comparable anti-endotoxin activities. Sci Rep 2017; 7:39925. [PMID: 28051162 PMCID: PMC5209718 DOI: 10.1038/srep39925] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/10/2016] [Indexed: 01/11/2023] Open
Abstract
To become clinically effective, antimicrobial peptides (AMPs) should be non-cytotoxic to host cells. Piscidins are a group of fish-derived AMPs with potent antimicrobial and antiendotoxin activities but limited by extreme cytotoxicity. We conjectured that introduction of cationic residue(s) at the interface of polar and non-polar faces of piscidins may control their insertion into hydrophobic mammalian cell membrane and thereby reducing cytotoxicity. We have designed several novel analogs of piscidin-1 by substituting threonine residue(s) with L and D-lysine residue(s). L/D-lysine-substituted analogs showed significantly reduced cytotoxicity but exhibited either higher or comparable antibacterial activity akin to piscidin-1. Piscidin-1-analogs demonstrated higher efficacy than piscidin-1 in inhibiting lipopolysaccharide (LPS)-induced pro-inflammatory responses in THP-1 cells. T15,21K-piscidin-1 (0.5 mg/Kg) and T15,21dK-piscidin-1 (1.0 mg/Kg) demonstrated 100% survival of LPS (12.0 mg/Kg)-administered mice. High resolution NMR studies revealed that both piscidin-1 and T15,21K-piscidin-1 adopted helical structures, with latter showing a shorter helix, higher amphipathicity and cationic residues placed at optimal distances to form ionic/hydrogen bond with lipid A of LPS. Remarkably, T15,21dK-piscidin-1 showed a helix-loop-helix structure in LPS and its interactions with LPS could be sustained by the distance of separation of side chains of R7 and D-Lys-15 which is close to the inter-phosphate distance of lipid A.
Collapse
|
32
|
Modulation of anti-endotoxin property of Temporin L by minor amino acid substitution in identified phenylalanine zipper sequence. Biochem J 2016; 473:4045-4062. [PMID: 27609815 DOI: 10.1042/bcj20160713] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 09/07/2016] [Indexed: 11/17/2022]
Abstract
A 13-residue frog antimicrobial peptide Temporin L (TempL) possesses versatile antimicrobial activities and is considered a lead molecule for the development of new antimicrobial agents. To find out the amino acid sequences that influence the anti-microbial property of TempL, a phenylalanine zipper-like sequence was identified in it which was not reported earlier. Several alanine-substituted analogs and a scrambled peptide having the same composition of TempL were designed for evaluating the role of this motif. To investigate whether leucine residues instead of phenylalanine residues at 'a' and/or 'd' position(s) of the heptad repeat sequence could alter its antimicrobial property, several TempL analogs were synthesized after replacing these phenylalanine residues with leucine residues. Replacing phenylalanine residues with alanine residues in the phenylalanine zipper sequence significantly compromised the anti-endotoxin property of TempL. This is evident from the higher production of tumor necrosis factor-α and interleukin-6 in lipopolysaccharide (LPS)-stimulated rat bone-marrow-derived macrophage cells in the presence of its alanine-substituted analogs than TempL itself. However, replacement of these phenylalanine residues with leucine residues significantly augmented anti-endotoxin property of TempL. A single alanine-substituted TempL analog (F8A-TempL) showed significantly reduced cytotoxicity but retained the antibacterial activity of TempL, while the two single leucine-substituted analogs (F5L-TempL and F8L-TempL), although exhibiting lower cytotoxicity, were able to retain the antibacterial activity of the parent peptide. The results demonstrate how minor amino acid substitutions in the identified phenylalanine zipper sequence in TempL could yield analogs with better antibacterial and/or anti-endotoxin properties with their plausible mechanism of action.
Collapse
|
33
|
Antibiotic combinations for controlling colistin-resistant Enterobacter cloacae. J Antibiot (Tokyo) 2016; 70:122-129. [PMID: 27381521 DOI: 10.1038/ja.2016.77] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 04/16/2016] [Accepted: 05/26/2016] [Indexed: 02/02/2023]
Abstract
Enterobacter cloacae is a Gram-negative bacterium associated with high morbidity and mortality in intensive care patients due to its resistance to multiple antibiotics. Currently, therapy against multi-resistant bacteria consists of using colistin, in spite of its toxic effects at higher concentrations. In this context, colistin-resistant E. cloacae strains were challenged with lower levels of colistin combined with other antibiotics to reduce colistin-associated side effects. Colistin-resistant E. cloacae (ATCC 49141) strains were generated by serial propagation in subinhibitory colistin concentrations. After this, three colistin-resistant and three nonresistant replicates were isolated. The identity of all the strains was confirmed by MALDI-TOF MS, VITEK 2 and MicroScan analysis. Furthermore, cross-resistance to other antibiotics was checked by disk diffusion and automated systems. The synergistic effects of the combined use of colistin and chloramphenicol were observed via the broth microdilution checkerboard method. First, data here reported showed that all strains presented intrinsic resistance to penicillin, cephalosporin (except fourth generation), monobactam, and some associations of penicillin and β-lactamase inhibitors. Moreover, a chloramphenicol and colistin combination was capable of inhibiting the induced colistin-resistant strains as well as two colistin-resistant clinical strains. Furthermore, no cytotoxic effect was observed by using such concentrations. In summary, the data reported here showed for the first time the possible therapeutic use of colistin-chloramphenicol for infections caused by colistin-resistant E. cloacae.
Collapse
|
34
|
Single Amino Acid Substitutions at Specific Positions of the Heptad Repeat Sequence of Piscidin-1 Yielded Novel Analogs That Show Low Cytotoxicity and In Vitro and In Vivo Antiendotoxin Activity. Antimicrob Agents Chemother 2016; 60:3687-99. [PMID: 27067326 DOI: 10.1128/aac.02341-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 03/27/2016] [Indexed: 12/16/2022] Open
Abstract
Piscidin-1 possesses significant antimicrobial and cytotoxic activities. To recognize the primary amino acid sequence(s) in piscidin-1 that could be important for its biological activity, a long heptad repeat sequence located in the region from amino acids 2 to 19 was identified. To comprehend the possible role of this motif, six analogs of piscidin-1 were designed by selectively replacing a single isoleucine residue at a d (5th) position or at an a (9th or 16th) position with either an alanine or a valine residue. Two more analogs, namely, I5F,F6A-piscidin-1 and V12I-piscidin-1, were designed for investigating the effect of interchanging an alanine residue at a d position with an adjacent phenylalanine residue and replacing a valine residue with an isoleucine residue at another d position of the heptad repeat of piscidin-1, respectively. Single alanine-substituted analogs exhibited significantly reduced cytotoxicity against mammalian cells compared with that of piscidin-1 but appreciably retained the antibacterial and antiendotoxin activities of piscidin-1. All the single valine-substituted piscidin-1 analogs and I5F,F6A-piscidin-1 showed cytotoxicity greater than that of the corresponding alanine-substituted analogs, antibacterial activity marginally greater than or similar to that of the corresponding alanine-substituted analogs, and also antiendotoxin activity superior to that of the corresponding alanine-substituted analogs. Interestingly, among these peptides, V12I-piscidin-1 showed the highest cytotoxicity and antibacterial and antiendotoxin activities. Lipopolysaccharide (12 mg/kg of body weight)-treated mice, further treated with I16A-piscidin-1, the piscidin-1 analog with the highest therapeutic index, at a single dose of 1 or 2 mg/kg of body weight, showed 80 and 100% survival, respectively. Structural and functional characterization of these peptides revealed the basis of their biological activity and demonstrated that nontoxic piscidin-1 analogs with significant antimicrobial and antiendotoxin activities can be designed by incorporating single alanine substitutions in the piscidin-1 heptad repeat.
Collapse
|
35
|
Mohanram H, Bhattacharjya S. 'Lollipop'-shaped helical structure of a hybrid antimicrobial peptide of temporin B-lipopolysaccharide binding motif and mapping cationic residues in antibacterial activity. Biochim Biophys Acta Gen Subj 2016; 1860:1362-72. [PMID: 27015761 DOI: 10.1016/j.bbagen.2016.03.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/08/2016] [Accepted: 03/20/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Temporins are attractive templates for the development of antibiotics. However, many temporins are inactive against Gram-negative bacteria. Previously, we demonstrated conjugation of a lipopolysaccharide binding motif peptide to temporins yielded hybrid non-haemolytic AMPs that killed several Gram-negative bacteria. METHODS We carried out a systematic Ala replacement of individual cationic and polar amino acid residues of LG21, a hybrid AMP consisted of temporin B (TB) and LPS binding motif. These Ala containing analogs of LG21 were examined for antibacterial activity, cell membrane permeabilization and liposome leakage assays using optical spectroscopic methods. Atomic resolution structure of LG21 was determined in zwitterionic dodecyl phosphocholine (DPC) micelles by NMR spectroscopy. RESULTS Cationic residues in the LPS binding motif of LG21 were critical for bactericidal and membrane permeabilization. Detergent bound structure of LG21 revealed helical conformation containing extensive sidechain/sidechain packing including cation/π interactions in the LPS binding motif. The helical structure of LG21 resembled a 'lollipop' like shape that was sustained by a compacted bulky aromatic/cationic head with a comparatively thinner 'stick' at the N-terminal region. The 'head' of the structure could be localized into micelle-water interfacial region whereas the 'stick' region may be inserted into the hydrophobic core of micelle. CONCLUSIONS The LPS binding motif of LG21 played dominant roles in broad spectrum activity and the 3-D structure provided plausible mechanistic insights for permeabilization of bacterial membrane. GENERAL SIGNIFICANCE Hybrid AMPs containing LPS binding motif could be useful for the structure based development of broad spectrum antibiotics.
Collapse
Affiliation(s)
- Harini Mohanram
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
36
|
Shang D, Zhang Q, Dong W, Liang H, Bi X. The effects of LPS on the activity of Trp-containing antimicrobial peptides against Gram-negative bacteria and endotoxin neutralization. Acta Biomater 2016; 33:153-65. [PMID: 26804205 DOI: 10.1016/j.actbio.2016.01.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/11/2016] [Accepted: 01/18/2016] [Indexed: 02/06/2023]
Abstract
A series of synthesized Trp-containing antimicrobial peptides showed significantly different antimicrobial activity against Gram-negative bacteria despite having similar components and amino acid sequences and the same net positive charge and hydrophobicity. Lipopolysaccharide (LPS) in the outer membrane is a permeability barrier to prevent antimicrobial peptides from crossing into Gram-negative bacteria. We investigated the interaction of five Trp-containing peptides, I1W, I4W, L5W, L11W and L12W, with LPS using circular dichroism (CD), IR spectroscopy, isothermal titration calorimetry (ITC), dynamic light scattering (DLS), zeta-potential measurements and confocal laser scanning microscopy, to address whether bacterial LPS is responsible for the different susceptibilities of Gram-negative bacteria to Trp-containing peptides. Our data indicate that I1W and I4W penetrated the LPS layer and killed Gram-negative bacteria by a "self-promoted uptake" pathway in which the peptides first approach LPS by electrostatic forces and then dissociate LPS micelle. This process results in disorganization of the LPS leaflet and promotes the ability of the peptide to cross the outer membrane into the inner membrane and disrupt the cytoplasmic membrane. Although L5W, L11W and L12W strongly bind to LPS bilayers and depolarize bacterial cytoplasmic membranes, similar to I1W and I4W, they are unable to destabilize LPS aggregates and traverse through the tightly packed LPS molecules. This study increases our understanding of the mechanism of action of these peptides in the LPS outer membrane and will help in the development of a potent broad-spectrum antibiotic for future therapeutic purposes. STATEMENT OF SIGNIFICANCE Tryptophan (Trp) residues show a strong preference for the interfacial region of biological membranes, and this property endows Trp-containing peptides with the unique ability to interact with the surface of bacterial cell membranes. In this manuscript, we report the membrane interaction of Trp-containing peptide to address whether bacterial LPS is responsible for the different susceptibilities of Gram-negative bacteria to Trp-containing peptides. Based on the data collected, we propose a molecular mechanism for the peptide-LPS interactions that allows the peptides to traverse or prevents them from transversing the LPS layer and the target inner membrane. The data should help in the development of a potent broad-spectrum antibiotic for future therapeutic purposes.
Collapse
|
37
|
Rajasekaran G, Kamalakannan R, Shin SY. Enhancement of the anti-inflammatory activity of temporin-1Tl-derived antimicrobial peptides by tryptophan, arginine and lysine substitutions. J Pept Sci 2015; 21:779-785. [DOI: doi 10.1002/psc.2807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2023]
Affiliation(s)
- Ganesan Rajasekaran
- Department of Medical Science, Graduate School; Chosun University; Gwangju 501-759 Korea
| | | | - Song Yub Shin
- Department of Medical Science, Graduate School; Chosun University; Gwangju 501-759 Korea
- Department of Cellular and Molecular Medicine, School of Medicine; Chosun University; Gwangju 501-759 Korea
| |
Collapse
|
38
|
Rajasekaran G, Kamalakannan R, Shin SY. Enhancement of the anti-inflammatory activity of temporin-1Tl-derived antimicrobial peptides by tryptophan, arginine and lysine substitutions. J Pept Sci 2015; 21:779-85. [DOI: 10.1002/psc.2807] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/10/2015] [Accepted: 07/10/2015] [Indexed: 12/23/2022]
Affiliation(s)
- Ganesan Rajasekaran
- Department of Medical Science, Graduate School; Chosun University; Gwangju 501-759 Korea
| | | | - Song Yub Shin
- Department of Medical Science, Graduate School; Chosun University; Gwangju 501-759 Korea
- Department of Cellular and Molecular Medicine, School of Medicine; Chosun University; Gwangju 501-759 Korea
| |
Collapse
|
39
|
Ultrashort Antimicrobial Peptides with Antiendotoxin Properties. Antimicrob Agents Chemother 2015; 59:5052-6. [PMID: 26033727 DOI: 10.1128/aac.00519-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/25/2015] [Indexed: 11/20/2022] Open
Abstract
Release of lipopolysaccharide (LPS) (endotoxin) from bacteria into the bloodstream may cause serious unwanted stimulation of the host immune system. Some but not all antimicrobial peptides can neutralize LPS-stimulated proinflammatory responses. Salt resistance and serum stability of short antimicrobial peptides can be boosted by adding β-naphthylalanine to their termini. Herein, significant antiendotoxin effects were observed in vitro and in vivo with the β-naphthylalanine end-tagged variants of the short antimicrobial peptides S1 and KWWK.
Collapse
|
40
|
An unprecedented alteration in mode of action of IsCT resulting its translocation into bacterial cytoplasm and inhibition of macromolecular syntheses. Sci Rep 2015; 5:9127. [PMID: 25773522 PMCID: PMC4360471 DOI: 10.1038/srep09127] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/30/2015] [Indexed: 01/12/2023] Open
Abstract
IsCT, a 13-residue, non-cell-selective antimicrobial peptide is comprised of mostly hydrophobic residues and lesser cationic residues. Assuming that placement of an additional positive charge in the non-polar face of IsCT could reduce its hydrophobic interaction, resulting in its reduction of cytotoxicity, an analog, I9K-IsCT was designed. Two more analogs, namely, E7K-IsCT and E7K,I9K-IsCT, were designed to investigate the impact of positive charges in the polar face as well as polar and non-polar faces at a time. These amino acid substitutions resulted in a significant enhancement of therapeutic potential of IsCT. IsCT and E7K-IsCT seem to target bacterial membrane for their anti-bacterial activity. However, I9K-IsCT and E7K,I9K-IsCT inhibited nucleic acid and protein syntheses in tested E. coli without perturbing its membrane. This was further supported by the observation that NBD-IsCT localized onto bacterial membrane while NBD-labeled I9K-IsCT and E7K,I9K-IsCT translocated into bacterial cytoplasm. Interestingly, IsCT and E7K-IsCT were significantly helical while I9K-IsCT and E7K,I9K-IsCT were mostly unstructured with no helix content in presence of mammalian and bacterial membrane-mimetic lipid vesicles. Altogether, the results identify two novel cell-selective analogs of IsCT with new prototype amino acid sequences that can translocate into bacterial cytoplasm without any helical structure and inhibit macromolecular syntheses.
Collapse
|
41
|
Sun Y, Dong W, Sun L, Ma L, Shang D. Insights into the membrane interaction mechanism and antibacterial properties of chensinin-1b. Biomaterials 2014; 37:299-311. [PMID: 25453959 DOI: 10.1016/j.biomaterials.2014.10.041] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 10/02/2014] [Indexed: 11/29/2022]
Abstract
Antimicrobial peptides (AMPs) with non-specific membrane disrupting activities are thought to exert their antimicrobial activity as a result of their cationicity, hydrophobicity and α-helical or β-sheet structures. Chensinin-1, a native peptide from skin secretions of Rana chensinensis, fails to manifest its desired biological properties because its low hydrophobic nature and an adopted random coil structure in a membrane-mimetic environment. In this study, chensinin-1b was designed by rearranging the amino acid sequence of its hydrophilic/polar residues on one face and its hydrophobic/nonpolar residues on the opposite face according to its helical diagram, and by replacing three Gly residues with three Trp residues. Introduction of Trp residues significantly promoted the binding of the peptide to the bacterial outer membrane and exerted bactericidal activity through cytoplasmic membrane damage. Chensinin-1b demonstrates higher antimicrobial activity and greater cell selectivity than its parent peptide, chensinin-1. The electrostatic interactions between chensinin-1b and lipopolysaccharide (LPS) may have facilitated the uptake of the peptide into Gram-negative cells and be also helpful to disrupt the bacterial cytoplasmic membrane, as evidenced by depolarisation of the membrane potential and leakage of calceins from the liposomes of Escherichia coli and Staphylococcus aureus. Chensinin-1b was also found to penetrate mouse skin and was also effective in vivo, as measured by hydroxyproline levels in a wound infection mouse model, and could therefore act as an anti-infective agent for wound healing.
Collapse
Affiliation(s)
- Yue Sun
- School of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Weibing Dong
- School of Life Science, Liaoning Normal University, Dalian 116081, China; Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian 116081, China
| | - Li Sun
- School of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Lijie Ma
- School of Life Science, Liaoning Normal University, Dalian 116081, China
| | - Dejing Shang
- School of Life Science, Liaoning Normal University, Dalian 116081, China; Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian 116081, China.
| |
Collapse
|
42
|
Lorenzón EN, Piccoli JP, Cilli EM. Interaction between the antimicrobial peptide Aurein 1.2 dimer and mannans. Amino Acids 2014; 46:2627-31. [PMID: 25209238 DOI: 10.1007/s00726-014-1832-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/27/2014] [Indexed: 02/04/2023]
Abstract
We have previously described the structure and the ability of a dimeric analog of the antimicrobial peptide Aurein 1.2 to aggregate Candida albicans. In this study, circular dichroism and fluorescence spectroscopy data showed that this aggregation is related to the interaction between the peptide and mannans, the main component of yeast cell wall. In this context, we propose a model in which dimers interact with the polysaccharide leading to cells aggregation.
Collapse
Affiliation(s)
- Esteban N Lorenzón
- Institute of Chemistry, UNESP - Univ Estadual Paulista, Rua Prof. Francisco Degni, 55, Bairro, Quitandinha, CEP 14800-060, Araraquara, São Paulo, Brazil
| | | | | |
Collapse
|
43
|
|