1
|
Sahu A, Ruhal R. Immune system dynamics in response to Pseudomonas aeruginosa biofilms. NPJ Biofilms Microbiomes 2025; 11:104. [PMID: 40506442 PMCID: PMC12162861 DOI: 10.1038/s41522-025-00738-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 05/24/2025] [Indexed: 06/16/2025] Open
Abstract
Pseudomonas aeruginosa biofilms contribute to chronic infections by resisting immune attacks and antibiotics. This review explores how innate immunity, including neutrophils, macrophages, and dendritic cells, responds to biofilms and how adaptive mechanisms involving T cells, B cells, and immunoglobulins contribute to infection persistence. Additionally, it highlights immune evasion strategies and discusses emerging therapies such as immunotherapy, monoclonal antibodies, and vaccines, offering insights into enhancing biofilm clearance and improving treatment outcomes.
Collapse
Affiliation(s)
- Abhijeet Sahu
- School of Bio Science and Technology, VIT Vellore, Vellore, Tamil Nadu, India
| | - Rohit Ruhal
- School of Bio Science and Technology, VIT Vellore, Vellore, Tamil Nadu, India.
| |
Collapse
|
2
|
Sarkar T, Rajalakshmi VS, K R R, Thummer RP, Chatterjee S. Serum-Stable, Cationic, α-Helical AMPs to Combat Infections of ESKAPE Pathogens and C. albicans. ACS APPLIED BIO MATERIALS 2025; 8:3941-3957. [PMID: 40305093 DOI: 10.1021/acsabm.5c00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Expedition in the rate of development of antimicrobial resistance accompanied by the slowdown in the development of new antimicrobials has led to a dire necessity to develop an alternate class of antimicrobial agents. Antimicrobial peptides (AMPs), available in nature, are effective molecules that can combat microbial infections. However, due to several inherent shortcomings such as salt sensitivity of their potency, short systemic half-lives owing to protease and serum degradation, and cytotoxicity, their commercial success is limited. Inspired by α helical AMPs present in nature, here in this work, we have developed two short, cationic, helical AMPs RR-12 and FL-13. Both peptides exhibited high broad-spectrum antimicrobial activity, salt tolerance, prompt bactericidal activity, considerable serum stability, remaining non-cytotoxic and non-hemolytic at relevant microbicidal concentrations. The designed AMPs were membranolytic toward the microbial strains, though there were subtle differences in the mechanism owing to the variation in the composition of the cell membranes in different microbes. Rigorous experimental techniques and molecular dynamics (MD) simulations were performed to understand the structure, activity, and their mechanisms in detail. Positive charge, balanced hydrophobicity-hydrophilicity, and helical conformation were the different attributes that led to the development of the superior performance of the AMPs, making them valuable additions to the repertoire of therapeutically promising antimicrobials.
Collapse
Affiliation(s)
- Tanumoy Sarkar
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, Assam-781039, India
| | | | - Ronima K R
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam-781039, India
| | - Rajkumar P Thummer
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam-781039, India
| | - Sunanda Chatterjee
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, Assam-781039, India
| |
Collapse
|
3
|
Benmamoun Z, Kinard T, Chandar P, Jankolovits J, Ducker WA. Effect of Salt on Synthetic Cationic Antimicrobial Polymer-Cell Interactions. Biomacromolecules 2025. [PMID: 40387209 DOI: 10.1021/acs.biomac.4c01706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Cationic antiseptics are deployed in a variety of settings, where salinity ranges from almost pure water to hypertonic salt. Here, we examine how dissolved NaCl affects the antimicrobial action of a model antimicrobial, polydiallyldimethylammonium chloride (PDADMAC) to the bacterium Escherichia coli (E. coli). Fluorescence microscopy is used to measure the time course of both the adsorption of PDADMAC to E. coli and the cell viability. NaCl decreases the density of adsorbed PDADMAC and diminishes its efficacy. At NaCl concentrations at or above 0.15 M, PDADMAC no longer kills bacteria but still prevents reproduction by halting the growth in cell length. Reproduction can be restarted if PDADMAC is removed. Fluorescence depolarization measurements show that PDADMAC rigidifies model membranes, but salt reduces the rigidity. We therefore attribute the halt in cell growth to reversible bridging by the polymer on the cell surface that prevents expansion of the cell membrane.
Collapse
Affiliation(s)
- Zachary Benmamoun
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Thomas Kinard
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24060, United States
| | - Prem Chandar
- Unilever Research & Development, Trumbull, Connecticut 06611, United States
| | - Joe Jankolovits
- Unilever Research & Development, Trumbull, Connecticut 06611, United States
| | - William A Ducker
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24060, United States
| |
Collapse
|
4
|
Schöpf C, Knapp M, Scheler J, Coraça-Huber DC, Romanelli A, Ladurner P, Seybold AC, Binder U, Würzner R, Marx F. The antibacterial activity and therapeutic potential of the amphibian-derived peptide TB_KKG6K. mSphere 2025:e0101624. [PMID: 40387366 DOI: 10.1128/msphere.01016-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/13/2025] [Indexed: 05/20/2025] Open
Abstract
Antimicrobial peptides (AMPs) have great potential to be developed as topical treatments for microbial infections of the skin, including those caused by the gram-positive human pathogen Staphylococcus aureus. Among the AMPs, temporin B (TB) is of particular interest. This 13-amino-acid-long cationic peptide is secreted by the granular glands of the European frog Rana temporaria and represents a primary line of defense against invading pathogens. The objective of this study was to investigate the antibacterial efficacy and the mode of action of the synthetic TB analog, TB_KKG6K, in a drug-resistant clinical isolate of S. aureus and assess the peptide's tolerance and curative potential in an in vitro infection model using three-dimensional human epidermis equivalents (HEEs). The results revealed a high bactericidal efficacy of TB_KKG6K at low micromolar concentrations. The peptide perturbed the bacterial cell membrane integrity by permeabilization and depolarization. TB_KKG6K showed no toxicity in the invertebrate mini-host model Galleria mellonella and a high level of tolerance when topically applied in HEEs. Importantly, the therapeutic potential of TB_KKG6K was confirmed in HEEs infected with S. aureus. The topical application of TB_KKG6K significantly reduced the bacterial load and lowered the pro-inflammatory response in the infected HEEs. These findings reinforce the antibacterial potential and therapeutic efficacy of TB_KKG6K against S. aureus infection, particularly in the context of a cutaneous infection.IMPORTANCEThe emergence of multidrug-resistant bacteria has rendered the exploration of novel therapeutic treatment strategies a pivotal area of research. Among the most promising candidates are amphibian-derived antimicrobial peptides (AMPs), which are ideal for the development of novel drugs due to their multifaceted mode of action. Extensive studies have been conducted on these peptides over the last decade, resulting in the development of temporin B (TB) peptide analogs that have undergone modifications to their primary sequence. These modified analogs have demonstrated enhanced antibacterial and antifungal efficacy, while exhibiting reduced hemolytic activity. TB_KKG6K has the potential to be a promising candidate for topical treatments due to its small size and high antimicrobial activity against pathogens of the human skin. In particular, it demonstrated efficacy against Staphylococcus aureus, a skin commensal that can become an opportunistic pathogen, causing a range of infections from minor skin infections to life-threatening diseases such as bacteremia and sepsis.
Collapse
Affiliation(s)
- Cristina Schöpf
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Knapp
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Jakob Scheler
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Débora C Coraça-Huber
- Research Laboratory for Implant Associated Infections (BIOFILM LAB), Experimental Orthopaedics, University Hospital for Orthopaedics and Traumatology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Peter Ladurner
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Anna C Seybold
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Ulrike Binder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Reinhard Würzner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
5
|
Ma A, Deng X, Wei L, Dong Y, Zhang P, Xuan S, Zhang Z. High Antibacterial Activity and Selectivity of Cationic Disubstituted Polypeptoids with Stable Helices and Enzymatic Resistance. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27950-27963. [PMID: 40311149 DOI: 10.1021/acsami.5c02994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
High antibacterial activity, low mammalian cell toxicity, and serum stability are crucial parameters for designing efficient antibacterial materials under physiological conditions. This relies on a deep understanding of the structure-property relationship of antibacterial materials. In this study, a series of cationic amphiphilic disubstituted polypeptoids were synthesized by using ring-opening polymerization (ROP) followed by thiol-ene click reactions. This new class of peptidomimetic materials, with chiral centers at backbones and ammonium alkyl N-substituents, exhibited remarkably stable helical structures independent of pH, temperature, salt, and denaturing agents. The helical analogs were found to show higher antibacterial activity against both Gram-negative and Gram-positive strains than the racemic, nonhelical counterparts. The helical structure and the balance of cationic charges and hydrophobicity were key parameters to achieve high selectivity for bacteria over mammalian cells. Moreover, unlike poly(l-lysine), the disubstituted polypeptoids, with stable helices and enzymatic resistance, retained high antibacterial activity even in the presence of salts, human serum albumin (HSA), and protease trypsin at physiological concentrations. This study deepens our understanding of how structural elements correlate with antibacterial activity and selectivity. In addition, the helical and enzymatically stable disubstituted polypeptoids have shown promise as an attractive platform for the design of new antibacterial materials with high efficiency and low toxicity.
Collapse
Affiliation(s)
- Anyao Ma
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Xuehua Deng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Luxin Wei
- The Fourth Affiliated Hospital of Soochow University, Suzhou 215124, China
| | - Yutong Dong
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | | | - Sunting Xuan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| |
Collapse
|
6
|
Damica FZ, Lucas DR, Toledo EB, de Carvalho Ribeiro M, Façanha ALO, Zeraik AE, Seabra SH, da Silva JA, Gomes VM, de Oliveira Carvalho A. A role in intracellular K + in protecting pathogenic dimorphic fungi against induced cell death by bioinspired antimicrobial peptides. Biochim Biophys Acta Gen Subj 2025; 1869:130795. [PMID: 40118348 DOI: 10.1016/j.bbagen.2025.130795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/23/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Antimicrobial peptides (AMPs) are promising drugs, though their fungal combat mechanisms remain partly unclear. We designed three AMPs (dAMPs) based on the γ-core of the Vu-Def1 seed defensin from Vigna unguiculata L. Walp. named RR, D-RR, and WR, and assessed their actions on Candida tropicalis and Candida albicans. Amidst their actions are cell shrinkage caused by K+ efflux from fungal cells. K+ involvement in fungal death by these peptides was explored. We assessed cell shrinkage, oxidative stress, mitochondria hyperpolarization, membrane permeabilization, medium acidification, antimicrobial activity under hypoosmotic conditions, and cellular degradation. Viability assays were performed with channel blockers and K+ addition at various times. The interactions of dAMPs with salts and fungal cells were analyzed using circular dichroism and microscopy. K+ and Cl- channels were not directly involved in dAMPs-induced death. Supplementation with K+ protected fungal cells from death. In tests, cations often deactivated them through charge neutralization. Peptides maintained their conformation with K+ and were found in cell cytoplasm indicating K+ did not neutralize charges. K+ did not prevent oxidative stress, but protected from cell shrinkage and mitochondria hyperpolarization. dAMPs rapidly stimulated medium acidification, followed by inhibition after 1 min, and K+ prevented acidification. Membrane permeabilization occurred after 20 min, faster with WR, explaining lack of protection from blockers. Fungal death was accelerated under hypoosmotic conditions. Electrophoresis revealed protein degradation, while ultrastructural analysis of the cells showed vacuolization, indicative of cytoplasmic degradation. Thus, K+ prevented cell death by maintaining internal levels, averting activation of cell degradation process.
Collapse
Affiliation(s)
- Filipe Zaniratti Damica
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Douglas Ribeiro Lucas
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Estefany Bras Toledo
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Marilúcia de Carvalho Ribeiro
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Anna Lvovna Okorokova Façanha
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Ana Eliza Zeraik
- Laboratório de Química e Função de Proteínas e Peptídeos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Sérgio Henrique Seabra
- Laboratório de Biologia Celular e Tecidual, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Juliana Azevedo da Silva
- Laboratório de Biologia do Reconhecer, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Valdirene Moreira Gomes
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - André de Oliveira Carvalho
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil.
| |
Collapse
|
7
|
Lee J, Mohammad N, Han K, Flagg-Dowie T, Magallon M, Brantly ML, Serban KA. Alpha-defensins increase NTHi binding but not engulfment by the macrophages enhancing airway inflammation in Alpha-1 antitrypsin deficiency. Front Immunol 2025; 16:1543729. [PMID: 40013145 PMCID: PMC11861504 DOI: 10.3389/fimmu.2025.1543729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
Neutrophilic inflammation and a high level of free α-defensins are main features of chronic airway inflammation in alpha-1 antitrypsin-deficient (AATD) individuals. Despite the antimicrobial activities of α-defensins by direct bacterial killing and by modulation of immune responses, AATD individuals are paradoxically burdened by recurrent exacerbation triggered by bacterial infections, frequently with nontypeable Haemophilus influenzae (NTHi). Previous studies demonstrated that high, rather than low α-defensin level could modulate the local pro-inflammatory milieu of bronchial epithelial cells and macrophages promoting chronic inflammation and lower pathogen phagocytosis. IgG-mediated phagocytosis and NTHi adherence, engulfment and phagocytosis were measured in human alveolar macrophages and monocyte-derived macrophages (MDM) isolated from patients with AATD and from healthy individuals. A high concentration of free α-defensins induced NTHi adherence to MDMs but decreased IgG-mediated phagocytosis by MDMs. The decreased phagocytosis was associated with TLR4 activation, downstream signaling via NF-κB p65 and marked increased secretion of inflammatory cytokines, CXCL8, IL-1b, and TNFα by the α-defensin-treated and NTHi-infected MDMs. Exogenous AAT treatment and TLR4 inhibitor decreased TNFα expression in α-defensin-treated cells. Dampening the downstream effects of a high concentration of α-defensins may render AAT and TLR4 inhibitors as potential therapies to decrease NTHi colonization and increase its clearance by phagocytosis in AATD individuals.
Collapse
Affiliation(s)
- Jungnam Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Naweed Mohammad
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Kyudong Han
- Department of Microbiology, College of Bio-convergence, Dankook University, Cheonan, Republic of Korea
- Center for Bio-Medical Engineering Core Facility, Dankook University, Cheonan, Republic of Korea
| | - Tammy Flagg-Dowie
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Maria Magallon
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Mark L. Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Karina A. Serban
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO, United States
| |
Collapse
|
8
|
Horvath M, Yang R, Castaneda DC, Callender M, Aiken ES, Voigt AY, Caldwell R, Fachi J, Di Luccia B, Scholar Z, Yu P, Salner A, Colonna M, Palucka K, Oh J. Species- and strain-specific microbial modulation of interferon, innate immunity, and epithelial barrier in 2D air-liquid interface respiratory epithelial cultures. BMC Biol 2025; 23:28. [PMID: 39875977 PMCID: PMC11776145 DOI: 10.1186/s12915-025-02129-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The microbiome regulates the respiratory epithelium's immunomodulatory functions. To explore how the microbiome's biodiversity affects microbe-epithelial interactions, we screened 58 phylogenetically diverse microbes for their transcriptomic effect on human primary bronchial air-liquid interface (ALI) cell cultures. RESULTS We found distinct species- and strain-level differences in host innate immunity and epithelial barrier response. Strikingly, we found that host interferon, an antiviral response, was one of the most variable host processes. This variability was not driven by microbial phylogenetic diversity, bioburden, nor by the microbe's ability to stimulate other innate immunity pathways. CONCLUSIONS Microbial colonization differentially stimulates host gene expression with variations observed across phylogenetically diverse microbes and across different strains of the same species. Our study provides a foundation for understanding how the respiratory microbiome's biodiversity affects epithelial, and particularly antiviral, innate immunity.
Collapse
Affiliation(s)
- Mian Horvath
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- UCONN Health (University of Connecticut), Farmington, CT, 06030, USA
| | - Ruoyu Yang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- UCONN Health (University of Connecticut), Farmington, CT, 06030, USA
- Duke University School of Medicine, Durham, NC, 27708, USA
| | | | - Megan Callender
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Elizabeth S Aiken
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Anita Y Voigt
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- Duke University School of Medicine, Durham, NC, 27708, USA
| | - Ryan Caldwell
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - José Fachi
- Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Blanda Di Luccia
- Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Zoe Scholar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Peter Yu
- Hartford Healthcare Cancer Institute, Hartford, CT, 06102, USA
| | - Andrew Salner
- Hartford Healthcare Cancer Institute, Hartford, CT, 06102, USA
| | - Marco Colonna
- Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Julia Oh
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA.
- Duke University School of Medicine, Durham, NC, 27708, USA.
| |
Collapse
|
9
|
Sarkar T, Vignesh SR, Kumar Sundaravadivelu P, Thummer RP, Satpati P, Chatterjee S. De Novo Design of Tryptophan Containing Broad-Spectrum Cationic Antimicrobial Octapeptides. ChemMedChem 2025; 20:e202400566. [PMID: 39402809 DOI: 10.1002/cmdc.202400566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/07/2024] [Indexed: 11/14/2024]
Abstract
With the advent of antibiotic resistant organisms, development of alternate classes of molecules other than antibiotics to combat microbial infections, have become extremely important. In this context, antimicrobial peptides have taken center stage of antimicrobial therapeutic research. In this work, we have reported two cationic antimicrobial octapeptides WRL and LWRF, with broad spectrum antimicrobial activities against several strains of ESKAPE pathogens. Both the peptides were membrane associative and induced microbial cell death through membranolysis, being selective towards microbial membranes over mammalian membranes. The AMPs were unstructured in water, adopting partial helical conformation in the presence of microbial membrane mimics. Electrostatic interaction formed the primary basis of peptide-membrane interactions. WRL was more potent, salt tolerant and faster acting of the two AMPs, owing to the presence of two tryptophan residues against that of one in LWRF. Increased tryptophan number in WRL enhanced its membrane association ability, resulting in higher antimicrobial potency but lower selectivity. This experimental and computational work, established that an optimum number of tryptophan residues and their position was critical for obtaining high antimicrobial potency and selectivity simultaneously in the designed cationic AMPs. Understanding the peptide membrane interactions in atomistic details can lead to development of better antimicrobial therapeutics in future.
Collapse
Affiliation(s)
- Tanumoy Sarkar
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - S R Vignesh
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Pradeep Kumar Sundaravadivelu
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Rajkumar P Thummer
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Priyadarshi Satpati
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Sunanda Chatterjee
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| |
Collapse
|
10
|
Atif M, Babuççu G, Riool M, Zaat S, Jonas U. Antimicrobial Peptide SAAP-148-Functionalized Hydrogels from Photocrosslinkable Polymers with Broad Antibacterial Activity. Macromol Rapid Commun 2024; 45:e2400785. [PMID: 39530205 DOI: 10.1002/marc.202400785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Antimicrobial peptides (AMPs) are promising alternatives to traditional antibiotics for treating skin wound infections. Nonetheless, their short half-life in biological environments restricts clinical applicability. Covalent immobilization of AMPs onto suitable substrates offers a comprehensive solution, creating contact-killing surfaces with long-term functionality. Here, a copolymer of poly[(hydroxy ethyl acrylamide)-co-(4-benzophenone acrylamide)-co-(pentafluorophenyl acrylate)-co-(ECOSURF EH-3 acrylate)], in short poly(HEAAm-co-BPAAm-co-PFPA-co-EH3A), is synthesized by free radical polymerization. Subsequent modification of active ester groups with the amine groups of SAAP-148, results in a copolymer, that is non-cytotoxic to human lung fibroblasts. UV photocrosslinking of the benzophenone units yields a polymer network that forms a hydrogel after swelling with aqueous medium. Both the SAAP-148-modified polymer in solution and the photocrosslinked hydrogels show good antimicrobial activity against strains of Escherichia coli, Staphylococcus aureus, Pseudomonas aeruginosa, and Acinetobacter baumannii, including multidrug-resistant strains, frequently found in wound infections. The covalent attachment of SAAP-148 prevents leaching, ensuring sustained antimicrobial activity for at least 48 h in diluted human blood plasma and 14 days in PBS. This prolonged retention of antimicrobial activity in human blood plasma significantly enhances its clinical potential. Overall, this study shows the potential of the AMP-functionalized photocrosslinkable polymer as antimicrobial wound dressings, providing an effective alternative to antibiotics.
Collapse
Affiliation(s)
- Muhammad Atif
- Macromolecular Chemistry, Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Strasse 2, 57076, Siegen, Germany
| | - Gizem Babuççu
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Immunology and Infectious Diseases, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Martijn Riool
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Immunology and Infectious Diseases, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Am Biopark 9, 93053, Regensburg, Germany
| | - Sebastian Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Immunology and Infectious Diseases, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Ulrich Jonas
- Macromolecular Chemistry, Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Strasse 2, 57076, Siegen, Germany
| |
Collapse
|
11
|
Hampton TH, Barnaby R, Roche C, Nymon A, Fukutani KF, MacKenzie TA, Charpentier LA, Stanton BA. Gene expression responses of CF airway epithelial cells exposed to elexacaftor/tezacaftor/ivacaftor suggest benefits beyond improved CFTR channel function. Am J Physiol Lung Cell Mol Physiol 2024; 327:L905-L916. [PMID: 39437760 PMCID: PMC11684945 DOI: 10.1152/ajplung.00272.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
The combination of elexacaftor/tezacaftor/ivacaftor (ETI, Trikafta) reverses the primary defect in cystic fibrosis (CF) by improving CFTR-mediated Cl- and HCO3- secretion by airway epithelial cells (AECs), leading to improved lung function and less frequent exacerbations and hospitalizations. However, studies have shown that CFTR modulators like ivacaftor, a component of ETI, have numerous effects on CF cells beyond improved CFTR channel function. Because little is known about the effect of ETI on CF AEC gene expression, we exposed primary human AEC to ETI for 48 h and interrogated the transcriptome by RNA-seq and qPCR. ETI increased CFTR Cl- secretion, and defensin gene expression (DEFB1), an observation consistent with reports of decreased bacterial burden in the lungs of people with CF (pwCF). ETI decreased MMP10 and MMP12 gene expression, suggesting that ETI may reduce proteolytic-induced lung destruction in CF. ETI also reduced the expression of the stress response gene heme oxygenase (HMOX1). qPCR analysis confirmed DEFB1, HMOX1, MMP10, and MMP12 gene expression results observed by RNA-seq. Gene pathway analysis revealed that ETI decreased inflammatory signaling, cellular proliferation, and MHC class II antigen presentation. Collectively, these findings suggest that the clinical observation that ETI reduces lung infections in pwCF is related in part to drug-induced increases in DEFB1 and that ETI may reduce lung damage by reducing MMP10 and MMP12 gene expression. Moreover, pathway analysis also identified several other genes responsible for the ETI-induced reduction in inflammation observed in pwCF.NEW & NOTEWORTHY Gene expression responses by CF AECs exposed to ETI suggest that in addition to improving CFTR channel function, ETI is likely to enhance resistance to bacterial infection by increasing levels of beta-defensin 1 (hBD-1). ETI may also reduce lung damage by suppressing MMP10 and MMP12 and reduce airway inflammation by repressing proinflammatory cytokine secretion by CF AECs.
Collapse
Affiliation(s)
- Thomas H Hampton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Roxanna Barnaby
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Carolyn Roche
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Amanda Nymon
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Kiyoshi Ferreira Fukutani
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Todd A MacKenzie
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States
| | - Lily A Charpentier
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| | - Bruce A Stanton
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States
| |
Collapse
|
12
|
Du J, Yang C, Deng Y, Guo H, Gu M, Chen D, Liu X, Huang J, Yan W, Liu J. Discovery of AMPs from random peptides via deep learning-based model and biological activity validation. Eur J Med Chem 2024; 277:116797. [PMID: 39197254 DOI: 10.1016/j.ejmech.2024.116797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/31/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
The ample peptide field is the best source for discovering clinically available novel antimicrobial peptides (AMPs) to address emerging drug resistance. However, discovering novel AMPs is complex and expensive, representing a major challenge. Recent advances in artificial intelligence (AI) have significantly improved the efficiency of identifying antimicrobial peptides from large libraries, whereas using random peptides as negative data increases the difficulty of discovering antimicrobial peptides from random peptides using discriminative models. In this study, we constructed three multi-discriminator models using deep learning and successfully screened twelve AMPs from a library of 30,000 random peptides. three candidate peptides (P2, P11, and P12) were screened by antimicrobial experiments, and further experiments showed that they not only possessed excellent antimicrobial activity but also had extremely low hemolytic activity. Mechanistic studies showed that these peptides exerted their bactericidal effects through membrane disruption, thus reducing the possibility of bacterial resistance. Notably, peptide 12 (P12) showed significant efficacy in a mouse model of Staphylococcus aureus wound infection with low toxicity to major organs at the highest tested dose (400 mg/kg). These results suggest deep learning-based multi-discriminator models can identify AMPs from random peptides with potential clinical applications.
Collapse
Affiliation(s)
- Jun Du
- School of Basic Medical Sciences, Lanzhou University, Donggang West Road, Lanzhou, 730000, China; Gansu Provincial Maternity and Child Care Hospital, North Road 143, Qilihe District, Lanzhou, 730000, China
| | - Changyan Yang
- School of Basic Medical Sciences, Lanzhou University, Donggang West Road, Lanzhou, 730000, China; Gansu Provincial Maternity and Child Care Hospital, North Road 143, Qilihe District, Lanzhou, 730000, China
| | - Yabo Deng
- School of Basic Medical Sciences, Lanzhou University, Donggang West Road, Lanzhou, 730000, China
| | - Hai Guo
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Mengyun Gu
- School of Basic Medical Sciences, Lanzhou University, Donggang West Road, Lanzhou, 730000, China
| | - Danna Chen
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Xia Liu
- School of Basic Medical Sciences, Lanzhou University, Donggang West Road, Lanzhou, 730000, China.
| | - Jinqi Huang
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| | - Wenjin Yan
- School of Basic Medical Sciences, Lanzhou University, Donggang West Road, Lanzhou, 730000, China.
| | - Jian Liu
- Gansu Provincial Maternity and Child Care Hospital, North Road 143, Qilihe District, Lanzhou, 730000, China.
| |
Collapse
|
13
|
Shepperson OA, Harris PWR, Brimble MA, Cameron AJ. Thanatin and vinyl sulfide analogues as narrow spectrum antimicrobial peptides that synergise with polymyxin B. Front Pharmacol 2024; 15:1487338. [PMID: 39564120 PMCID: PMC11573584 DOI: 10.3389/fphar.2024.1487338] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
Thanatin is a β-hairpin antimicrobial peptide cyclised by a single disulfide bond that has shown potent broad-spectrum activity towards bacterial and fungal pathogens. Towards Gram-negative species, thanatin acts both by forming trans-membranal pores and inhibiting outer membrane biogenesis by binding to LptA and blocking lipopolysaccharide (LPS) transport. Inspired by previous modifications of thanatin, an analogue was prepared which demonstrated potent but selective activity towards E. coli. Furthermore, this compound was shown to act in synergy with the highly potent FDA-approved lipopeptide antibiotic polymyxin B, which engages LPS at the cytoplasmic membrane. Four analogues of thanatin in which the disulfide was substituted for vinyl sulfide bridge mimetics were prepared, all of which retained similar secondary structures. Two of these retained substantial potency and selectivity towards E. coli. Importantly, synergy with polymyxin B was also maintained for the lead analogue. The vinyl sulfide potentially offers a facile replacement strategy for labile disulfide bonds and the selective activity and drug synergy of the reported thanatin analogues is promising for the development of narrow spectrum antimicrobials with reduced likelihood of resistance emerging in clinical settings.
Collapse
Affiliation(s)
- Oscar A Shepperson
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Bio-Discovery, The University of Auckland, Auckland, New Zealand
| | - Paul W R Harris
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Bio-Discovery, The University of Auckland, Auckland, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Bio-Discovery, The University of Auckland, Auckland, New Zealand
| | - Alan J Cameron
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Bio-Discovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Jandl B, Dighe S, Gasche C, Makristathis A, Muttenthaler M. Intestinal biofilms: pathophysiological relevance, host defense, and therapeutic opportunities. Clin Microbiol Rev 2024; 37:e0013323. [PMID: 38995034 PMCID: PMC11391705 DOI: 10.1128/cmr.00133-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
SUMMARYThe human intestinal tract harbors a profound variety of microorganisms that live in symbiosis with the host and each other. It is a complex and highly dynamic environment whose homeostasis directly relates to human health. Dysbiosis of the gut microbiota and polymicrobial biofilms have been associated with gastrointestinal diseases, including irritable bowel syndrome, inflammatory bowel diseases, and colorectal cancers. This review covers the molecular composition and organization of intestinal biofilms, mechanistic aspects of biofilm signaling networks for bacterial communication and behavior, and synergistic effects in polymicrobial biofilms. It further describes the clinical relevance and diseases associated with gut biofilms, the role of biofilms in antimicrobial resistance, and the intestinal host defense system and therapeutic strategies counteracting biofilms. Taken together, this review summarizes the latest knowledge and research on intestinal biofilms and their role in gut disorders and provides directions toward the development of biofilm-specific treatments.
Collapse
Affiliation(s)
- Bernhard Jandl
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Satish Dighe
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Christoph Gasche
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
- Loha for Life, Center for Gastroenterology and Iron Deficiency, Vienna, Austria
| | - Athanasios Makristathis
- Department of Laboratory Medicine, Division of Clinical Microbiology, Medical University of Vienna, Vienna, Austria
| | - Markus Muttenthaler
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
15
|
Romano Ibarra GS, Lei L, Yu W, Thurman AL, Gansemer ND, Meyerholz DK, Pezzulo AA, McCray PB, Thornell IM, Stoltz DA. IL-13 induces loss of CFTR in ionocytes and reduces airway epithelial fluid absorption. J Clin Invest 2024; 134:e181995. [PMID: 39255033 PMCID: PMC11527443 DOI: 10.1172/jci181995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024] Open
Abstract
The airway surface liquid (ASL) plays a crucial role in lung defense mechanisms, and its composition and volume are regulated by the airway epithelium. The cystic fibrosis transmembrane conductance regulator (CFTR) is abundantly expressed in a rare airway epithelial cell type called an ionocyte. Recently, we demonstrated that ionocytes can increase liquid absorption through apical CFTR and basolateral barttin/chloride channels, while airway secretory cells mediate liquid secretion through apical CFTR channels and basolateral NKCC1 transporters. Th2-driven (IL-4/IL-13) airway diseases, such as asthma, cause goblet cell metaplasia, accompanied by increased mucus production and airway secretions. In this study, we investigate the effect of IL-13 on chloride and liquid transport performed by ionocytes. IL-13 treatment of human airway epithelia was associated with reduced epithelial liquid absorption rates and increased ASL volume. Additionally, IL-13 treatment reduced the abundance of CFTR-positive ionocytes and increased the abundance of CFTR-positive secretory cells. Increasing ionocyte abundance attenuated liquid secretion caused by IL-13. Finally, CFTR-positive ionocytes were less common in asthma and chronic obstructive pulmonary disease and were associated with airflow obstruction. Our findings suggest that loss of CFTR in ionocytes contributes to the liquid secretion observed in IL-13-mediated airway diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paul B. McCray
- Department of Internal Medicine
- Department of Pediatrics
- Pappajohn Biomedical Institute, and
| | - Ian M. Thornell
- Department of Internal Medicine
- Pappajohn Biomedical Institute, and
| | - David A. Stoltz
- Department of Internal Medicine
- Pappajohn Biomedical Institute, and
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
16
|
Lamba S, Heruka De Zoysa G, Wang K, Lu J, Swift S, Sarojni V. Homo and Hetero-Branched Lipopeptide Dendrimers: Synthesis and Antimicrobial Activity. Bioorg Chem 2024; 150:107567. [PMID: 38936047 DOI: 10.1016/j.bioorg.2024.107567] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/30/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024]
Abstract
Di-branched and tetra-branched versions of a previously reported analogue of the lipopeptide battacin were successfully synthesised using thiol-maleimide click and 1, 2, 3-triazole click chemistry. Antimicrobial studies against drug resistant clinical isolates of Escherichia coli (ESBL E. coli Ctx-M14), Pseudomonas aeruginosa (P. aeruginosa Q502), and Methicillin resistant Staphylococcus aureus (MRSA ATCC 33593), as well as clinically isolated Acinetobacter baumannii (A. baumannii ATCC 19606), and P. aeruginosa (ATCC 27853), revealed that the dendrimeric peptides have antimicrobial activity in the low micromolar range (0.5 -- 4 μM) which was 10 times more potent than the monomer peptides. Under high salt concentrations (150 mM NaCl, 2 mM MgCl2, and 2.5 mM CaCl2) the di-branched lipopeptides retained their antimicrobial activity while the monomer peptides were not active (>100 μM). The di-branched triazole click lipopeptide, Peptide 12, was membrane lytic, showed faster killing kinetics, and exhibited antibiofilm activity against A. baumannii and MRSA and eradicated > 85 % preformed biofilms at low micromolar concentrations. The di-branched analogues were > 30-fold potent than the monomers against Candida albicans. Peptide 12 was not haemolytic (HC10 = 932.12 μM) and showed up to 40-fold higher selectivity against bacteria and fungi than the monomer peptide. Peptide 12 exhibited strong proteolytic stability (>80 % not degraded) in rat serum over 24 h whereas > 95 % of the thiol-maleimide analogue (Peptide 10) was degraded. The tetra-branched peptides showed comparable antibacterial potency to the di-branched analogues. These findings indicate that dual branching using triazole click chemistry is a promising strategy to improve the antimicrobial activity and proteolytic stability of battacin based lipopeptides. The information gathered can be used to build effective antimicrobial dendrimeric peptides as new peptide antibiotics.
Collapse
Affiliation(s)
- Saurabh Lamba
- School of Chemical Sciences and The Centre for Green Chemical Science, University of Auckland, Auckland 1142, New Zealand; The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6012, New Zealand
| | - Gayan Heruka De Zoysa
- School of Chemical Sciences and The Centre for Green Chemical Science, University of Auckland, Auckland 1142, New Zealand
| | - Kelvin Wang
- Auckland Bioengineering Institute, University of Auckland, Auckland 1142, New Zealand
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland 1142, New Zealand
| | - Simon Swift
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Vijayalekshmi Sarojni
- School of Chemical Sciences and The Centre for Green Chemical Science, University of Auckland, Auckland 1142, New Zealand; The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6012, New Zealand.
| |
Collapse
|
17
|
Zhao H, Zhao S, Wang S, Liu Y. Human β-defensins: The multi-functional natural peptide. Biochem Pharmacol 2024; 227:116451. [PMID: 39059771 DOI: 10.1016/j.bcp.2024.116451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
The increasing threat of antibiotic resistance among pathogenic microorganisms and the urgent demand for new antibiotics require immediate attention. Antimicrobial peptides exhibit effectiveness against microorganisms, fungi, viruses, and protozoa. The discovery of human β-defensins represents a major milestone in biomedical research, opening new avenues for scientific investigation into the innate immune system and its resistance mechanisms against pathogenic microorganisms. Multiple defensins present a promising alternative in the context of antibiotic abuse. However, obstacles to the practical application of defensins as anti-infective therapies persist due to the unique properties of human β-defensins themselves and serious pharmacological and technical challenges. To overcome these challenges, diverse delivery vehicles have been developed and progressively improved for the conjugation or encapsulation of human β-defensins. This review briefly introduces the biology of human β-defensins, focusing on their multistage structure and diverse functions. It also discusses several heterologous systems for producing human β-defensins, various delivery systems created for these peptides, and patent applications related to their utilization, concluding with a summary of current challenges and potential solutions.
Collapse
Affiliation(s)
- Haile Zhao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock Jointly Constructed by Ministry and Province, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Hohhot, Inner Mongolia 010020, China
| | - Shuli Zhao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock Jointly Constructed by Ministry and Province, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Hohhot, Inner Mongolia 010020, China
| | - Simeng Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock Jointly Constructed by Ministry and Province, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Hohhot, Inner Mongolia 010020, China
| | - Ying Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock Jointly Constructed by Ministry and Province, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Hohhot, Inner Mongolia 010020, China.
| |
Collapse
|
18
|
Hampton TH, Barnaby R, Roche C, Nymon A, Fukutani KF, MacKenzie TA, Stanton BA. Gene expression responses of CF airway epithelial cells exposed to elexacaftor/tezacaftor/ivacaftor (ETI) suggest benefits beyond improved CFTR channel function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610162. [PMID: 39257747 PMCID: PMC11383677 DOI: 10.1101/2024.08.28.610162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The combination of elexacaftor/tezacaftor/ivacaftor (ETI, Trikafta) reverses the primary defect in Cystic Fibrosis (CF) by improving CFTR mediated Cl - and HCO 3 - secretion by airway epithelial cells (AEC), leading to improved lung function and less frequent exacerbations and hospitalizations. However, studies have shown that CFTR modulators like ivacaftor, a component of ETI, has numerous effects on CF cells beyond improved CFTR channel function. Because little is known about the effect of ETI on CF AEC gene expression we exposed primary human AEC to ETI for 48 hours and interrogated the transcriptome by RNA-seq and qPCR. ETI increased defensin gene expression ( DEFB1 ) an observation consistent with reports of decreased bacterial burden in the lungs of people with CF (pwCF). ETI also decreased MMP10 and MMP12 gene expression, suggesting that ETI may reduce proteolytic induced lung destruction in CF. ETI also reduced the expression of the stress response gene heme oxygenase ( HMOX1 ). qPCR analysis confirmed DEFB1, HMOX1, MMP10 and MMP12 gene expression results observed by RNA-seq. Gene pathway analysis revealed that ETI decreased inflammatory signaling, cellular proliferation and MHC Class II antigen presentation. Collectively, these findings suggest that the clinical observation that ETI reduces lung infections in pwCF is related in part to drug induced increases in DEFB1 , and that ETI may reduce lung damage by reducing MMP10 and MMP12 gene expression, which is predicted to reduce matrix metalloprotease activity. Moreover, pathway analysis also identified several genes responsible for the ETI induced reduction in inflammation observed in people with CF. New and Noteworthy Gene expression responses by CF AEC exposed to ETI suggest that in addition to improving CFTR channel function, ETI is likely to increase resistance to bacterial infection by increasing levels of beta defensin 1 (hBD-1). ETI may also reduce lung damage by suppressing MMP10, and reduce airway inflammation by repressing proinflammatory cytokine secretion by AEC cells.
Collapse
|
19
|
Walsh D, Bevan J, Harrison F. How Does Airway Surface Liquid Composition Vary in Different Pulmonary Diseases, and How Can We Use This Knowledge to Model Microbial Infections? Microorganisms 2024; 12:732. [PMID: 38674677 PMCID: PMC11052052 DOI: 10.3390/microorganisms12040732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Growth environment greatly alters many facets of pathogen physiology, including pathogenesis and antimicrobial tolerance. The importance of host-mimicking environments for attaining an accurate picture of pathogen behaviour is widely recognised. Whilst this recognition has translated into the extensive development of artificial cystic fibrosis (CF) sputum medium, attempts to mimic the growth environment in other respiratory disease states have been completely neglected. The composition of the airway surface liquid (ASL) in different pulmonary diseases is far less well characterised than CF sputum, making it very difficult for researchers to model these infection environments. In this review, we discuss the components of human ASL, how different lung pathologies affect ASL composition, and how different pathogens interact with these components. This will provide researchers interested in mimicking different respiratory environments with the information necessary to design a host-mimicking medium, allowing for better understanding of how to treat pathogens causing infection in these environments.
Collapse
Affiliation(s)
- Dean Walsh
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK (F.H.)
| | | | | |
Collapse
|
20
|
Chen Y, Han Z, Zhang S, Liu H, Wang K, Liu J, Liu F, Yu S, Sai N, Mai H, Zhou X, Zhou C, Wen Q, Ma L. ERK1/2-CEBPB Axis-Regulated hBD1 Enhances Anti-Tuberculosis Capacity in Alveolar Type II Epithelial Cells. Int J Mol Sci 2024; 25:2408. [PMID: 38397085 PMCID: PMC10889425 DOI: 10.3390/ijms25042408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), remains a global health crisis with substantial morbidity and mortality rates. Type II alveolar epithelial cells (AEC-II) play a critical role in the pulmonary immune response against Mtb infection by secreting effector molecules such as antimicrobial peptides (AMPs). Here, human β-defensin 1 (hBD1), an important AMP produced by AEC-II, has been demonstrated to exert potent anti-tuberculosis activity. HBD1 overexpression effectively inhibited Mtb proliferation in AEC-II, while mice lacking hBD1 exhibited susceptibility to Mtb and increased lung tissue inflammation. Mechanistically, in A549 cells infected with Mtb, STAT1 negatively regulated hBD1 transcription, while CEBPB was the primary transcription factor upregulating hBD1 expression. Furthermore, we revealed that the ERK1/2 signaling pathway activated by Mtb infection led to CEBPB phosphorylation and nuclear translocation, which subsequently promoted hBD1 expression. Our findings suggest that the ERK1/2-CEBPB-hBD1 regulatory axis can be a potential therapeutic target for anti-tuberculosis therapy aimed at enhancing the immune response of AEC-II cells.
Collapse
Affiliation(s)
- Yaoxin Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Zhenyu Han
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Sian Zhang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Honglin Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Ke Wang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Jieyu Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Feichang Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Shiyun Yu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Na Sai
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Haiyan Mai
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Xinying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Chaoying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China; (Y.C.); (Z.H.); (S.Z.); (H.L.); (K.W.); (J.L.); (F.L.); (S.Y.); (N.S.); (H.M.); (X.Z.); (C.Z.)
- Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Guangzhou 510515, China
| |
Collapse
|
21
|
Casanova M, Maresca M, Poncin I, Point V, Olleik H, Boidin-Wichlacz C, Tasiemski A, Mabrouk K, Cavalier JF, Canaan S. Promising antibacterial efficacy of arenicin peptides against the emerging opportunistic pathogen Mycobacterium abscessus. J Biomed Sci 2024; 31:18. [PMID: 38287360 PMCID: PMC10823733 DOI: 10.1186/s12929-024-01007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Mycobacterium abscessus, a fast-growing non-tuberculous mycobacterium, is an emerging opportunistic pathogen responsible for chronic bronchopulmonary infections in people with respiratory diseases such as cystic fibrosis (CF). Due to its intrinsic polyresistance to a wide range of antibiotics, most treatments for M. abscessus pulmonary infections are poorly effective. In this context, antimicrobial peptides (AMPs) active against bacterial strains and less prompt to cause resistance, represent a good alternative to conventional antibiotics. Herein, we evaluated the effect of three arenicin isoforms, possessing two or four Cysteines involved in one (Ar-1, Ar-2) or two disulfide bonds (Ar-3), on the in vitro growth of M. abscessus. METHODS The respective disulfide-free AMPs, were built by replacing the Cysteines with alpha-amino-n-butyric acid (Abu) residue. We evaluated the efficiency of the eight arenicin derivatives through their antimicrobial activity against M. abscessus strains, their cytotoxicity towards human cell lines, and their hemolytic activity on human erythrocytes. The mechanism of action of the Ar-1 peptide was further investigated through membrane permeabilization assay, electron microscopy, lipid insertion assay via surface pressure measurement, and the induction of resistance assay. RESULTS Our results demonstrated that Ar-1 was the safest peptide with no toxicity towards human cells and no hemolytic activity, and the most active against M. abscessus growth. Ar-1 acts by insertion into mycobacterial lipids, resulting in a rapid membranolytic effect that kills M. abscessus without induction of resistance. CONCLUSION Overall, the present study emphasized Ar-1 as a potential new alternative to conventional antibiotics in the treatment of CF-associated bacterial infection related to M. abscessus.
Collapse
Affiliation(s)
- Magali Casanova
- CNRS, Aix-Marseille Univ, LISM UMR7255, IMM FR3479, Marseille, France.
| | - Marc Maresca
- Aix Marseille Univ, CNRS, Centrale Marseille, iSm2 (UMR7313), Marseille, France
| | - Isabelle Poncin
- CNRS, Aix-Marseille Univ, LISM UMR7255, IMM FR3479, Marseille, France
| | - Vanessa Point
- CNRS, Aix-Marseille Univ, LISM UMR7255, IMM FR3479, Marseille, France
| | - Hamza Olleik
- Aix Marseille Univ, CNRS, Centrale Marseille, iSm2 (UMR7313), Marseille, France
| | - Céline Boidin-Wichlacz
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL - Center for Infection and Immunity of Lille, 59000, Lille, France
| | - Aurélie Tasiemski
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL - Center for Infection and Immunity of Lille, 59000, Lille, France
| | - Kamel Mabrouk
- Aix-Marseille Univ, CNRS, UMR7273, ICR, 13013, Marseille, France
| | | | - Stéphane Canaan
- CNRS, Aix-Marseille Univ, LISM UMR7255, IMM FR3479, Marseille, France
| |
Collapse
|
22
|
Thakur A, Ganesan R, Ray Dutta J. Antimicrobial Peptide-Based Nanomaterials in Combating Multidrug-Resistant Bacteria. NANOTECHNOLOGY BASED STRATEGIES FOR COMBATING ANTIMICROBIAL RESISTANCE 2024:177-201. [DOI: 10.1007/978-981-97-2023-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
23
|
Bonvin E, Personne H, Paschoud T, Reusser J, Gan BH, Luscher A, Köhler T, van Delden C, Reymond JL. Antimicrobial Peptide-Peptoid Hybrids with and without Membrane Disruption. ACS Infect Dis 2023; 9:2593-2606. [PMID: 38062792 PMCID: PMC10714400 DOI: 10.1021/acsinfecdis.3c00421] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023]
Abstract
Among synthetic analogues of antimicrobial peptides (AMPs) under investigation to address antimicrobial resistance, peptoids (N-alkylated oligoglycines) have been reported to act both by membrane disruption and on intracellular targets. Here we gradually introduced peptoid units into the membrane-disruptive undecapeptide KKLLKLLKLLL to test a possible transition toward intracellular targeting. We found that selected hybrids containing up to five peptoid units retained the parent AMP's α-helical folding, membrane disruption, and antimicrobial effects against Gram-negative bacteria including multidrug-resistant (MDR) strains of Pseudomonas aeruginosa and Klebsiella pneumoniae while showing reduced hemolysis and cell toxicities. Furthermore, some hybrids containing as few as three peptoid units as well as the full peptoid lost folding, membrane disruption, hemolysis, and cytotoxicity but displayed strong antibacterial activity under dilute medium conditions typical for proline-rich antimicrobial peptides (PrAMPs), pointing to intracellular targeting. These findings parallel previous reports that partially helical amphiphilic peptoids are privileged oligomers for antibiotic development.
Collapse
Affiliation(s)
- Etienne Bonvin
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Hippolyte Personne
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Thierry Paschoud
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Jérémie Reusser
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Bee-Ha Gan
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| | - Alexandre Luscher
- Department
of Microbiology and Molecular Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Service of
Infectious Diseases, University Hospital
of Geneva, CH-1211 Geneva, Switzerland
| | - Thilo Köhler
- Department
of Microbiology and Molecular Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Service of
Infectious Diseases, University Hospital
of Geneva, CH-1211 Geneva, Switzerland
| | - Christian van Delden
- Department
of Microbiology and Molecular Medicine, University of Geneva, CH-1211 Geneva, Switzerland
- Service of
Infectious Diseases, University Hospital
of Geneva, CH-1211 Geneva, Switzerland
| | - Jean-Louis Reymond
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, CH-3012 Bern, Switzerland
| |
Collapse
|
24
|
R PA, Anbarasu A. Antimicrobial Peptides as Immunomodulators and Antimycobacterial Agents to Combat Mycobacterium tuberculosis: a Critical Review. Probiotics Antimicrob Proteins 2023; 15:1539-1566. [PMID: 36576687 DOI: 10.1007/s12602-022-10018-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 12/29/2022]
Abstract
Tuberculosis (TB) is a devastating disease foisting a significantly high morbidity, prepotent in low- and middle-income developing countries. Evolution of drug resistance among Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, has made the TB treatment more complicated. The protracted nature of present TB treatment, persistent and tolerant Mtb populations, interaction with antiretroviral therapy and existing toxicity concerned with conventional anti-TB drugs are the four major challenges inflicted with emergence of drug-resistant mycobacterial strains, and the standard medications are unable to combat these strains. These factors emphasize an exigency to develop new drugs to overcome these barriers in current TB therapy. With this regard, antimycobacterial peptides derived from various sources such as human cells, bacterial sources, mycobacteriophages, fungal, plant and animal sources could be considered as antituberculosis leads as most of these peptides are associated with dual advantages of having both bactericidal activity towards Mtb as well as immuno-regulatory property. Some of the peptides possess the additional advantage of interacting synergistically with antituberculosis medications too, thereby increasing their efficiency, underscoring the vigour of antimicrobial peptides (AMPs) as best possible alternative therapeutic candidates or adjuvants in TB treatment. Albeit the beneficiary features of these peptides, few obstacles allied with them like cytotoxicity and proteolytic degradation are matter of concerns too. In this review, we have focused on structural hallmarks, targeting mechanisms and specific structural aspects contributing to antimycobacterial activity and discovered natural and synthetic antimycobacterial peptides along with their sources, anti-TB, immuno-regulatory properties, merits and demerits and possible delivery methods of AMPs.
Collapse
Affiliation(s)
- Preethi A R
- Medical & Biological Computing Laboratory, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore-632014, India
- Department of Biotechnology, SBST, VIT, Vellore-632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical & Biological Computing Laboratory, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore-632014, India.
- Department of Biotechnology, SBST, VIT, Vellore-632014, Tamil Nadu, India.
| |
Collapse
|
25
|
Skłodowski K, Suprewicz Ł, Chmielewska-Deptuła SJ, Kaliniak S, Okła S, Zakrzewska M, Minarowski Ł, Mróz R, Daniluk T, Savage PB, Fiedoruk K, Bucki R. Ceragenins exhibit bactericidal properties that are independent of the ionic strength in the environment mimicking cystic fibrosis sputum. Front Microbiol 2023; 14:1290952. [PMID: 38045035 PMCID: PMC10693459 DOI: 10.3389/fmicb.2023.1290952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
The purpose of the work was to investigate the impact of sodium chloride (NaCl) on the antimicrobial efficacy of ceragenins (CSAs) and antimicrobial peptides (AMPs) against bacterial and fungal pathogens associated with cystic fibrosis (CF) lung infections. CF-associated bacterial (Pseudomonas aeruginosa, Ochrobactrum spp., and Staphylococcus aureus), and fungal pathogens (Candida albicans, and Candida tropicalis) were used as target organisms for ceragenins (CSA-13 and CSA-131) and AMPs (LL-37 and omiganan). Susceptibility to the tested compounds was assessed using minimal inhibitory concentrations (MICs) and bactericidal concentrations (MBCs), as well as by colony counting assays in CF sputum samples supplemented with various concentrations of NaCl. Our results demonstrated that ceragenins exhibit potent antimicrobial activity in CF sputum regardless of the NaCl concentration when compared to LL-37 and omiganan. Given the broad-spectrum antimicrobial activity of ceragenins in the microenvironments mimicking the airways of CF patients, ceragenins might be promising agents in managing CF disease.
Collapse
Affiliation(s)
- Karol Skłodowski
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | | | | | - Sławomir Okła
- Holy Cross Cancer Center, Kielce, Poland
- Institute of Health Science, Collegium Medicum, Jan Kochanowski University of Kielce, Kielce, Poland
| | - Magdalena Zakrzewska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Łukasz Minarowski
- 2nd Department of Lung Diseases and Tuberculosis, Medical University of Bialystok, Bialystok, Poland
| | - Robert Mróz
- 2nd Department of Lung Diseases and Tuberculosis, Medical University of Bialystok, Bialystok, Poland
| | - Tamara Daniluk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Paul B. Savage
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, United States
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
26
|
Routsias JG, Marinou D, Mavrouli M, Tsakris A, Pitiriga V. Serum β-Defensin 2, A Novel Biomarker for the Diagnosis of Acute Infections. Diagnostics (Basel) 2023; 13:diagnostics13111885. [PMID: 37296737 DOI: 10.3390/diagnostics13111885] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Defensins are natural antimicrobial peptides that the human body secretes to protect itself from an infection. Thus, they are ideal molecules to serve as biomarkers for infection. This study was conducted to evaluate the levels of human β-defensins in patients with inflammation. METHODS CRP, hBD2 and procalcitonin were measured in 423 sera of 114 patients with inflammation and healthy individuals using nephelometry and commercial ELISA assays. RESULTS Levels of hBD2 in the serum of patients with an infection were markedly elevated compared to those of hBD2 in patients with inflammation of non-infectious etiology (p < 0.0001, t = 10.17) and healthy individuals. ROC analysis demonstrated that hBD2 showed the highest detection performance for infection (AUC 0.897; p < 0.001) followed by PCT (AUC 0.576; p = ns) and CRP (AUC 0.517; p = ns). In addition, analysis of hBD2 and CRP in patients' sera collected at different time points showed that hBD2 levels could help differentiate inflammation of infectious and non-infectious etiology during the first 5 days of hospitalization, while CRP levels could not. CONCLUSIONS hBD2 has the potential to serve as a diagnostic biomarker for infection. In addition, the levels of hBD2 may reflect the efficacy of antibiotic treatment.
Collapse
Affiliation(s)
- John G Routsias
- Department of Microbiology, School of Health Sciences, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Attica, Greece
| | - Dionysia Marinou
- Department of Microbiology, School of Health Sciences, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Attica, Greece
| | - Maria Mavrouli
- Department of Microbiology, School of Health Sciences, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Attica, Greece
| | - Athanasios Tsakris
- Department of Microbiology, School of Health Sciences, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Attica, Greece
| | - Vassiliki Pitiriga
- Department of Microbiology, School of Health Sciences, National and Kapodistrian University of Athens, Mikras Asias 75, 11527 Athens, Attica, Greece
| |
Collapse
|
27
|
van Dijk A, Guabiraba R, Bailleul G, Schouler C, Haagsman HP, Lalmanach AC. Evolutionary diversification of defensins and cathelicidins in birds and primates. Mol Immunol 2023; 157:53-69. [PMID: 36996595 DOI: 10.1016/j.molimm.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
Divergent evolution for more than 310 million years has resulted in an avian immune system that is complex and more compact than that of primates, sharing much of its structure and functions. Not surprisingly, well conserved ancient host defense molecules, such as defensins and cathelicidins, have diversified over time. In this review, we describe how evolution influenced the host defense peptides repertoire, its distribution, and the relationship between structure and biological functions. Marked features of primate and avian HDPs are linked to species-specific characteristics, biological requirements, and environmental challenge.
Collapse
|
28
|
Feng J, Jia Z, Yuan G, Zhu X, Liu Q, Wu K, Wang J, Zou J. Expression and functional characterization of three β-defensins in grass carp (Ctenopharyngodon idella). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 140:104616. [PMID: 36565823 DOI: 10.1016/j.dci.2022.104616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/07/2022] [Accepted: 12/11/2022] [Indexed: 06/17/2023]
Abstract
β-defensins (BDs) are a group of cysteine-rich cationic antimicrobial peptides and play important roles in the first line of defense against infection. In this study, the expression and antibacterial activities of three grass carp (Ctenopharyngodon idella) (Ci) β-defensin (BD) peptides were comparatively investigated. Expression analysis reveals that CiBD1-3 were constitutively expressed in tissues, with the highest expression detected in the skin. The CiBD-1 transcripts were more abundant than CiBD-2 and CiBD-3. In the primary head kidney leukocytes, CiBDs were induced by PHA, LPS, poly(I:C) and cytokines such as IL-1β and IFN-γ. In vivo challenge of fish with Aeromonas hydrophila resulted in the up-regulation of CiBDs in the head kidney and hindgut. To determine the biological activities, recombinant CiBD proteins were produced in the HEK293-F cells and purified for the minimum inhibitory concentration assay. It was found that all three recombinant CiBD proteins were effective to inhibit the growth of Gram-negative fish bacterial pathogens including Aeromonas hydrophila, Edwardsiella tarda, Flavobacterium columnare and Klebsiella pneumoniae and Gram-positive Staphylococcus aureus. CiBD-2 and CiBD-3 were more effective than CiBD-1. Our results demonstrate that all the three CiBDs have broad antibacterial activity against fish bacterial pathogens.
Collapse
Affiliation(s)
- Jianhua Feng
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Zhao Jia
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Gaoliang Yuan
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiaozhen Zhu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Qin Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Kaizheng Wu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Junya Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jun Zou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China.
| |
Collapse
|
29
|
Botryllin, a Novel Antimicrobial Peptide from the Colonial Ascidian Botryllus schlosseri. Mar Drugs 2023; 21:md21020074. [PMID: 36827115 PMCID: PMC9966394 DOI: 10.3390/md21020074] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
By mining the transcriptome of the colonial ascidian Botryllus schlosseri, we identified a transcript for a novel styelin-like antimicrobial peptide, which we named botryllin. The gene is constitutively transcribed by circulating cytotoxic morula cells (MCs) as a pre-propeptide that is then cleaved to mature peptide. The synthetic peptide, obtained from in silico translation of the transcript, shows robust killing activity of bacterial and unicellular yeast cells, causing breakages of both the plasma membrane and the cell wall. Specific monoclonal antibodies were raised against the epitopes of the putative amino acid sequence of the propeptide and the mature peptide; in both cases, they label the MC granular content. Upon MC degranulation induced by the presence of nonself, the antibodies recognise the extracellular nets with entrapped bacteria nearby MC remains. The obtained results suggest that the botryllin gene carries the information for the synthesis of an AMP involved in the protection of B. schlosseri from invading foreign cells.
Collapse
|
30
|
Jiang M, Chen R, Zhang J, Chen F, Wang KJ. A Novel Antimicrobial Peptide Spampcin 56-86 from Scylla paramamosain Exerting Rapid Bactericidal and Anti-Biofilm Activity In Vitro and Anti-Infection In Vivo. Int J Mol Sci 2022; 23:ijms232113316. [PMID: 36362111 PMCID: PMC9653689 DOI: 10.3390/ijms232113316] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
The abuse of antibiotics leads to the increase of bacterial resistance, which seriously threatens human health. Therefore, there is an urgent need to find effective alternatives to antibiotics, and antimicrobial peptides (AMPs) are the most promising antibacterial agents and have received extensive attention. In this study, a novel potential AMP was identified from the marine invertebrate Scylla paramamosain and named Spampcin. After bioinformatics analysis and AMP database prediction, four truncated peptides (Spa31, Spa22, Spa20 and Spa14) derived from Spampcin were screened, all of which showed potent antimicrobial activity with different antibacterial spectrum. Among them, Spampcin56-86 (Spa31 for short) exhibited strong bactericidal activity against a variety of clinical pathogens and could rapidly kill the tested bacteria within minutes. Further analysis of the antibacterial mechanism revealed that Spa31 disrupted the integrity of the bacterial membrane (as confirmed by scanning electron microscopy observation, NPN, and PI staining assays), leading to bacterial rupture, leakage of cellular contents (such as elevated extracellular ATP), increased ROS production, and ultimately cell death. Furthermore, Spa31 was found to interact with LPS and effectively inhibit bacterial biofilms. The antibacterial activity of Spa31 had good thermal stability, certain ion tolerance, and no obvious cytotoxicity. It is worth noting that Spa31 could significantly improve the survival rate of zebrafish Danio rerio infected with Pseudomonas aeruginosa, indicating that Spa31 played an important role in anti-infection in vivo. This study will enrich the database of marine animal AMPs and provide theoretical reference and scientific basis for the application of marine AMPs in medical fields.
Collapse
Affiliation(s)
- Manyu Jiang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Roushi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Jingrong Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (F.C.); (K.-J.W.)
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (F.C.); (K.-J.W.)
| |
Collapse
|
31
|
Ghosh S, Chatterjee S, Satpati P. Effect of a monovalent salt on the energetics of an antimicrobial-peptide: micelle dissociation. Phys Chem Chem Phys 2022; 24:23669-23678. [PMID: 36148810 DOI: 10.1039/d2cp02735f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Antimicrobial peptides (AMPs) are promising antimicrobial and therapeutic agents. Recently, we synthesized a cationic 14 residue AMP (LL-14: LKWLKKLLKWLKKL), which showed high broad-spectrum antimicrobial activity. However, the antimicrobial activity of LL-14 was compromised in the presence of NaCl. Salt sensitivity of antimicrobial potency is one of the fundamental limitations of AMP therapeutics. Thus, understanding the thermodynamics of AMP binding to simple membrane-mimetic systems and the effect of NaCl that contributes to their stability is crucial for designing promising AMPs against microbial infection. In this work, we reported computational analysis of LL-14 binding to SDS micelles (the simplest bacterial membrane mimic) at various NaCl concentrations (0.0%, 0.5%, 1.0% w/v). The thermodynamics of LL-14 dissociation from the SDS micelles was estimated by employing steered molecular dynamics (SMD) simulation followed by umbrella sampling. The results indicated that the increase in NaCl concentration systematically disfavoured the LL-14:SDS binding, primarily by stabilizing the dissociative state (i.e., free LL-14 and free micelles in water). We proposed a kinetic scheme in which the salt-induced selective stabilization of the dissociative state increased the activation barrier for the peptide:micelle binding event resulting in reduced affinity. Center-of-mass pulling indicated that the interactions involving the N-terminal of the LL-14 (residues 1-6) and SDS micelle were crucial for the stability of the LL-14:SDS complex, and LL-14 underwent a conformational change (helix → unstructured) before dissociating from the SDS micelle. The observed structural features from the peptide:micelle dissociation pathway corroborate our previous simulations as well as circular dichroism (CD), and fluorescence experiments.
Collapse
Affiliation(s)
- Suvankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Sunanda Chatterjee
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Priyadarshi Satpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
32
|
Hill DB, Button B, Rubinstein M, Boucher RC. Physiology and pathophysiology of human airway mucus. Physiol Rev 2022; 102:1757-1836. [PMID: 35001665 PMCID: PMC9665957 DOI: 10.1152/physrev.00004.2021] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 12/13/2021] [Accepted: 12/19/2021] [Indexed: 01/27/2023] Open
Abstract
The mucus clearance system is the dominant mechanical host defense system of the human lung. Mucus is cleared from the lung by cilia and airflow, including both two-phase gas-liquid pumping and cough-dependent mechanisms, and mucus transport rates are heavily dependent on mucus concentration. Importantly, mucus transport rates are accurately predicted by the gel-on-brush model of the mucociliary apparatus from the relative osmotic moduli of the mucus and periciliary-glycocalyceal (PCL-G) layers. The fluid available to hydrate mucus is generated by transepithelial fluid transport. Feedback interactions between mucus concentrations and cilia beating, via purinergic signaling, coordinate Na+ absorptive vs Cl- secretory rates to maintain mucus hydration in health. In disease, mucus becomes hyperconcentrated (dehydrated). Multiple mechanisms derange the ion transport pathways that normally hydrate mucus in muco-obstructive lung diseases, e.g., cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), non-CF bronchiectasis (NCFB), and primary ciliary dyskinesia (PCD). A key step in muco-obstructive disease pathogenesis is the osmotic compression of the mucus layer onto the airway surface with the formation of adherent mucus plaques and plugs, particularly in distal airways. Mucus plaques create locally hypoxic conditions and produce airflow obstruction, inflammation, infection, and, ultimately, airway wall damage. Therapies to clear adherent mucus with hydrating and mucolytic agents are rational, and strategies to develop these agents are reviewed.
Collapse
Affiliation(s)
- David B Hill
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina
| | - Brian Button
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael Rubinstein
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Mechanical Engineering and Materials Science, Biomedical Engineering, Physics, and Chemistry, Duke University, Durham, North Carolina
| | - Richard C Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
33
|
Wu Z, Chan B, Low J, Chu JJH, Hey HWD, Tay A. Microbial resistance to nanotechnologies: An important but understudied consideration using antimicrobial nanotechnologies in orthopaedic implants. Bioact Mater 2022; 16:249-270. [PMID: 35415290 PMCID: PMC8965851 DOI: 10.1016/j.bioactmat.2022.02.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
Microbial resistance to current antibiotics therapies is a major cause of implant failure and adverse clinical outcomes in orthopaedic surgery. Recent developments in advanced antimicrobial nanotechnologies provide numerous opportunities to effective remove resistant bacteria and prevent resistance from occurring through unique mechanisms. With tunable physicochemical properties, nanomaterials can be designed to be bactericidal, antifouling, immunomodulating, and capable of delivering antibacterial compounds to the infection region with spatiotemporal accuracy. Despite its substantial advancement, an important, but under-explored area, is potential microbial resistance to nanomaterials and how this can impact the clinical use of antimicrobial nanotechnologies. This review aims to provide a better understanding of nanomaterial-associated microbial resistance to accelerate bench-to-bedside translations of emerging nanotechnologies for effective control of implant associated infections.
Collapse
Affiliation(s)
- Zhuoran Wu
- Institute of Health Innovation & Technology, National University of Singapore, 117599, Singapore
| | - Brian Chan
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Jessalyn Low
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
| | - Justin Jang Hann Chu
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
- Infectious Disease Programme, Yong Loo Lin School of Medicine, National University of Singapore, 117547, Singapore
- Institute of Molecular and Cell Biology, 35 Agency for Science, Technology and Research, 138673, Singapore
| | - Hwee Weng Dennis Hey
- National University Health System, National University of Singapore, 119228, Singapore
| | - Andy Tay
- Institute of Health Innovation & Technology, National University of Singapore, 117599, Singapore
- Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
- Tissue Engineering Programme, National University of Singapore, 117510, Singapore
| |
Collapse
|
34
|
Ruhluel D, O'Brien S, Fothergill JL, Neill DR. Development of liquid culture media mimicking the conditions of sinuses and lungs in cystic fibrosis and health. F1000Res 2022; 11:1007. [PMID: 36519007 PMCID: PMC9718992 DOI: 10.12688/f1000research.125074.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 11/25/2023] Open
Abstract
The respiratory tract is a compartmentalised and heterogenous environment. The nasopharynx and sinuses of the upper airways have distinct properties from the lungs and these differences may shape bacterial adaptation and evolution. Upper airway niches act as early colonisation sites for respiratory bacterial pathogens, including those, such as Pseudomonas aeruginosa, that can go on to establish chronic infection of the lungs in people with cystic fibrosis (CF). Despite the importance of upper airway environments in facilitating early adaptation to host environments, currently available in vitro models for study of respiratory infection in CF focus exclusively on the lungs. Furthermore, animal models, widely used to bridge the gap between in vitro systems and the clinical scenario, do not allow the upper and lower airways to be studied in isolation. We have developed a suite of culture media reproducing key features of the upper and lower airways, for the study of bacterial adaptation and evolution in different respiratory environments. For both upper and lower airway-mimicking media, we have developed formulations that reflect airway conditions in health and those that reflect the altered environment of the CF respiratory tract. Here, we describe the development and validation of these media and their use for study of genetic and phenotypic adaptations in P. aeruginosa during growth under upper or lower airway conditions in health and in CF.
Collapse
Affiliation(s)
- Dilem Ruhluel
- University of Liverpool, Institute of Infection, Veterinary and Ecological Sciences,, Liverpool, L69 7BE, UK
| | - Siobhan O'Brien
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, 2, Ireland
| | - Joanne L Fothergill
- University of Liverpool, Institute of Infection, Veterinary and Ecological Sciences,, Liverpool, L69 7BE, UK
| | - Daniel R Neill
- University of Liverpool, Institute of Infection, Veterinary and Ecological Sciences,, Liverpool, L69 7BE, UK
| |
Collapse
|
35
|
Ruhluel D, O'Brien S, Fothergill JL, Neill DR. Development of liquid culture media mimicking the conditions of sinuses and lungs in cystic fibrosis and health. F1000Res 2022; 11:1007. [PMID: 36519007 PMCID: PMC9718992 DOI: 10.12688/f1000research.125074.2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/18/2022] [Indexed: 12/05/2022] Open
Abstract
The respiratory tract is a compartmentalised and heterogenous environment. The nasopharynx and sinuses of the upper airways have distinct properties from the lungs and these differences may shape bacterial adaptation and evolution. Upper airway niches act as early colonisation sites for respiratory bacterial pathogens, including those, such as Pseudomonas aeruginosa, that can go on to establish chronic infection of the lungs in people with cystic fibrosis (CF). Despite the importance of upper airway environments in facilitating early adaptation to host environments, currently available in vitro models for study of respiratory infection in CF focus exclusively on the lungs. Furthermore, animal models, widely used to bridge the gap between in vitro systems and the clinical scenario, do not allow the upper and lower airways to be studied in isolation. We have developed a suite of culture media reproducing key features of the upper and lower airways, for the study of bacterial adaptation and evolution in different respiratory environments. For both upper and lower airway-mimicking media, we have developed formulations that reflect airway conditions in health and those that reflect the altered environment of the CF respiratory tract. Here, we describe the development and validation of these media and their use for study of genetic and phenotypic adaptations in P. aeruginosa during growth under upper or lower airway conditions in health and in CF.
Collapse
Affiliation(s)
- Dilem Ruhluel
- University of Liverpool, Institute of Infection, Veterinary and Ecological Sciences,, Liverpool, L69 7BE, UK
| | - Siobhan O'Brien
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, 2, Ireland
| | - Joanne L Fothergill
- University of Liverpool, Institute of Infection, Veterinary and Ecological Sciences,, Liverpool, L69 7BE, UK
| | - Daniel R Neill
- University of Liverpool, Institute of Infection, Veterinary and Ecological Sciences,, Liverpool, L69 7BE, UK
| |
Collapse
|
36
|
Bhat RAH, Thakuria D, Tandel RS, Khangembam VC, Dash P, Tripathi G, Sarma D. Tools and techniques for rational designing of antimicrobial peptides for aquaculture. FISH & SHELLFISH IMMUNOLOGY 2022; 127:1033-1050. [PMID: 35872334 DOI: 10.1016/j.fsi.2022.07.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Fisheries and aquaculture industries remain essential sources of food and nutrition for millions of people worldwide. Indiscriminate use of antibiotics has led to the emergence of antimicrobial-resistant bacteria and posed a severe threat to public health. Researchers have opined that antimicrobial peptides (AMPs) can be the best possible alternative to curb the rising tide of antimicrobial resistance in aquaculture. AMPs may also help to achieve the objectives of one health approach. The natural AMPs are associated with several shortcomings, like less in vivo stability, toxicity to host cell, high cost of production and low potency in a biological system. In this review, we have provided a comprehensive outline about the strategies for designing synthetic mimics of natural AMPs with high potency. Moreover, the freely available AMP databases and the information about the molecular docking tools are enlisted. We also provided in silico template for rationally designing the AMPs from fish piscidins or other peptides. The rationally designed piscidin (rP1 and rp2) may be used to tackle microbial infections in aquaculture. Further, the protocol can be used to develop the truncated mimics of natural AMPs having more potency and protease stability.
Collapse
Affiliation(s)
| | - Dimpal Thakuria
- ICAR-Directorate of Coldwater Fisheries Research, Bhimtal, 263136, Uttarakhand, India
| | | | - Victoria C Khangembam
- ICAR-Directorate of Coldwater Fisheries Research, Bhimtal, 263136, Uttarakhand, India
| | - Pragyan Dash
- ICAR-Directorate of Coldwater Fisheries Research, Bhimtal, 263136, Uttarakhand, India
| | - Gayatri Tripathi
- ICAR-Central Institute of Fisheries Education, Mumbai, 400061, Maharashtra, India
| | - Debajit Sarma
- ICAR-Directorate of Coldwater Fisheries Research, Bhimtal, 263136, Uttarakhand, India
| |
Collapse
|
37
|
Figueira MF, Ribeiro CMP, Button B. Mucus-targeting therapies of defective mucus clearance for cystic fibrosis: A short review. Curr Opin Pharmacol 2022; 65:102248. [PMID: 35689870 PMCID: PMC9891491 DOI: 10.1016/j.coph.2022.102248] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/19/2022] [Accepted: 05/05/2022] [Indexed: 02/03/2023]
Abstract
In the lungs, defective CFTR associated with cystic fibrosis (CF) represents the nidus for abnormal mucus clearance in the airways and consequently a progressive lung disease. Defective CFTR-mediated Cl- secretion results in altered mucus properties, including concentration, viscoelasticity, and the ratio of the two mucins, MUC5B and MUC5AC. In the past decades, therapies targeting the CF mucus defect, directly or indirectly, have been developed; nevertheless, better treatments to prevent the disease progression are still needed. This review summarizes the existing knowledge on the defective mucus in CF disease and highlights it as a barrier to the development of future inhaled genetic therapies. The use of new mucus-targeting treatments is also discussed, focusing on their potential role to halt the progress of CF lung disease.
Collapse
Affiliation(s)
- Miriam Frankenthal Figueira
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Carla M. P. Ribeiro
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina, Chapel Hill, NC 27599-7248, USA.,Department of Medicine, University of North Carolina, Chapel Hill, NC 27599-7248, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Brian Button
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina, Chapel Hill, NC 27599-7248, USA.,Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| |
Collapse
|
38
|
Antimicrobial peptides for tackling cystic fibrosis related bacterial infections: a review. Microbiol Res 2022; 263:127152. [DOI: 10.1016/j.micres.2022.127152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/18/2022]
|
39
|
Hanson MA, Kondo S, Lemaitre B. Drosophila immunity: the Drosocin gene encodes two host defence peptides with pathogen-specific roles. Proc Biol Sci 2022; 289:20220773. [PMID: 35730150 PMCID: PMC9233930 DOI: 10.1098/rspb.2022.0773] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Antimicrobial peptides (AMPs) are key to defence against infection in plants and animals. Use of AMP mutations in Drosophila has now revealed that AMPs can additively or synergistically contribute to defence in vivo. However, these studies also revealed high specificity, wherein just one AMP contributes an outsized role in combatting a specific pathogen. Here, we show the Drosocin locus (CG10816) is more complex than previously described. In addition to its namesake peptide 'Drosocin', it encodes a second mature peptide from a precursor via furin cleavage. This peptide corresponds to the previously uncharacterized 'Immune-induced Molecule 7'. A polymorphism (Thr52Ala) in the Drosocin precursor protein previously masked the identification of this peptide, which we name 'Buletin'. Using mutations differently affecting Drosocin and Buletin, we show that only Drosocin contributes to Drosocin gene-mediated defence against Enterobacter cloacae. Strikingly, we observed that Buletin, but not Drosocin, contributes to the Drosocin gene-mediated defence against Providencia burhodogranariea, including an importance of the Thr52Ala polymorphism for survival. Our study reveals that the Drosocin gene encodes two prominent host defence peptides with different specificity against distinct pathogens. This finding emphasizes the complexity of the Drosophila humoral response and demonstrates how natural polymorphisms can affect host susceptibility.
Collapse
Affiliation(s)
- M. A. Hanson
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - S. Kondo
- Invertebrate Genetics Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima, Japan
| | - B. Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
40
|
Czechtizky W, Su W, Ripa L, Schiesser S, Höijer A, Cox RJ. Advances in the design of new types of inhaled medicines. PROGRESS IN MEDICINAL CHEMISTRY 2022; 61:93-162. [PMID: 35753716 DOI: 10.1016/bs.pmch.2022.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Inhalation of small molecule drugs has proven very efficacious for the treatment of respiratory diseases due to enhanced efficacy and a favourable therapeutic index compared with other dosing routes. It enables targeted delivery to the lung with rapid onset of therapeutic action, low systemic drug exposure, and thereby reduced systemic side effects. An increasing number of pharmaceutical companies and biotechs are investing in new modalities-for this review defined as therapeutic molecules with a molecular weight >800Da and therefore beyond usual inhaled small molecule drug-like space. However, our experience with inhaled administration of PROTACs, peptides, oligonucleotides (antisense oligonucleotides, siRNAs, miRs and antagomirs), diverse protein scaffolds, antibodies and antibody fragments is still limited. Investigating the retention and metabolism of these types of molecules in lung tissue and fluid will contribute to understanding which are best suited for inhalation. Nonetheless, the first such therapeutic molecules have already reached the clinic. This review will provide information on the physiology of healthy and diseased lungs and their capacity for drug metabolism. It will outline the stability, aggregation and immunogenicity aspects of new modalities, as well as recap on formulation and delivery aspects. It concludes by summarising clinical trial outcomes with inhaled new modalities based on information available at the end of 2021.
Collapse
Affiliation(s)
- Werngard Czechtizky
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden.
| | - Wu Su
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Lena Ripa
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Stefan Schiesser
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Andreas Höijer
- Cardiovascular, Renal & Metabolism CMC Projects, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Rhona J Cox
- Department of Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal & Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| |
Collapse
|
41
|
Lee YCJ, Shirkey JD, Park J, Bisht K, Cowan AJ. An Overview of Antiviral Peptides and Rational Biodesign Considerations. BIODESIGN RESEARCH 2022; 2022:9898241. [PMID: 37850133 PMCID: PMC10521750 DOI: 10.34133/2022/9898241] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 10/19/2023] Open
Abstract
Viral diseases have contributed significantly to worldwide morbidity and mortality throughout history. Despite the existence of therapeutic treatments for many viral infections, antiviral resistance and the threat posed by novel viruses highlight the need for an increased number of effective therapeutics. In addition to small molecule drugs and biologics, antimicrobial peptides (AMPs) represent an emerging class of potential antiviral therapeutics. While AMPs have traditionally been regarded in the context of their antibacterial activities, many AMPs are now known to be antiviral. These antiviral peptides (AVPs) have been shown to target and perturb viral membrane envelopes and inhibit various stages of the viral life cycle, from preattachment inhibition through viral release from infected host cells. Rational design of AMPs has also proven effective in identifying highly active and specific peptides and can aid in the discovery of lead peptides with high therapeutic selectivity. In this review, we highlight AVPs with strong antiviral activity largely curated from a publicly available AMP database. We then compile the sequences present in our AVP database to generate structural predictions of generic AVP motifs. Finally, we cover the rational design approaches available for AVPs taking into account approaches currently used for the rational design of AMPs.
Collapse
Affiliation(s)
- Ying-Chiang J. Lee
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Jaden D. Shirkey
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Jongbeom Park
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Karishma Bisht
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Alexis J. Cowan
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| |
Collapse
|
42
|
Pandit G, Sarkar T, S. R. V, Debnath S, Satpati P, Chatterjee S. Delineating the Mechanism of Action of a Protease Resistant and Salt Tolerant Synthetic Antimicrobial Peptide against Pseudomonas aeruginosa. ACS OMEGA 2022; 7:15951-15968. [PMID: 35571791 PMCID: PMC9097201 DOI: 10.1021/acsomega.2c01089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/08/2022] [Indexed: 06/15/2023]
Abstract
Rapidly growing antimicrobial resistance (AMR) against antibiotics has propelled the development of synthetic antimicrobial peptides (AMPs) as potential antimicrobial agents. An antimicrobial peptide Nle-Dab-Trp-Nle-Dab-Dab-Nle-CONH2 (P36; Nle = norleucine, Dab = diaminobutyric acid, Trp = tryptophan) potent against Pseudomonas aeruginosa (P. aeruginosa) has been developed in the present study. Rational design strategy adopted in this study led to the improvisation of the therapeutic qualities such as activity, salt tolerance, cytotoxicity, and protease resistance of the template peptide P4, which was earlier reported from our group. P36 exhibited salt tolerant antimicrobial potency against P. aeruginosa, along with very low cytotoxicity against mammalian cell lines. P36 was found to be nonhemolytic and resistant toward protease degradation which qualified it as a potent antimicrobial agent. We have investigated the mechanism of action of this molecule in detail using several experimental techniques (spectroscopic, biophysical, and microscopic) and molecular dynamics simulations. P36 was a membrane active AMP with membrane destabilization and deformation abilities, leading to leakage of the intracellular materials and causing eventual cell death. The interaction between P36 and the microbial membrane/membrane mimics was primarily driven by electrostatics. P36 was unstructured in water and upon binding to the microbial membrane mimic SDS, suggesting no influence of secondary structure on its antimicrobial potency. Positive charge, optimum hydrophobic-hydrophilic balance, and chain length remained the most important concerns to be addressed while designing small cationic antimicrobial peptides.
Collapse
Affiliation(s)
- Gopal Pandit
- Department
of Chemistry, Indian Institute of Technology.
Guwahati (IITG), Guwahati, Assam 781039, India
| | - Tanumoy Sarkar
- Department
of Chemistry, Indian Institute of Technology.
Guwahati (IITG), Guwahati, Assam 781039, India
| | - Vignesh S. R.
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology. Guwahati (IITG), Guwahati, Assam 781039, India
| | - Swapna Debnath
- Department
of Chemistry, Indian Institute of Technology.
Guwahati (IITG), Guwahati, Assam 781039, India
| | - Priyadarshi Satpati
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology. Guwahati (IITG), Guwahati, Assam 781039, India
| | - Sunanda Chatterjee
- Department
of Chemistry, Indian Institute of Technology.
Guwahati (IITG), Guwahati, Assam 781039, India
| |
Collapse
|
43
|
Alzahrani NM, Booq RY, Aldossary AM, Bakr AA, Almughem FA, Alfahad AJ, Alsharif WK, Jarallah SJ, Alharbi WS, Alsudir SA, Alyamani EJ, Tawfik EA, Alshehri AA. Liposome-Encapsulated Tobramycin and IDR-1018 Peptide Mediated Biofilm Disruption and Enhanced Antimicrobial Activity against Pseudomonas aeruginosa. Pharmaceutics 2022; 14:960. [PMID: 35631547 PMCID: PMC9144307 DOI: 10.3390/pharmaceutics14050960] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
The inadequate eradication of pulmonary infections and chronic inflammation are significant complications in cystic fibrosis (CF) patients, who usually suffer from persistent and frequent lung infections caused by several pathogens, particularly Pseudomonas aeruginosa (P. aeruginosa). The ability of pathogenic microbes to protect themselves from biofilms leads to the development of an innate immune response and antibiotic resistance. In the present work, a reference bacterial strain of P. aeruginosa (PA01) and a multidrug-resistant isolate (MDR 7067) were used to explore the microbial susceptibility to three antibiotics (ceftazidime, imipenem, and tobramycin) and an anti-biofilm peptide (IDR-1018 peptide) using the minimum inhibition concentration (MIC). The most effective antibiotic was then encapsulated into liposomal nanoparticles and the IDR-1018 peptide with antibacterial activity, and the ability to disrupt the produced biofilm against PA01 and MDR 7067 was assessed. The MIC evaluation of the tobramycin antibacterial activity showed an insignificant effect on the liposomes loaded with tobramycin and liposomes encapsulating tobramycin and IDR-1018 against both P. aeruginosa strains to free tobramycin. Nevertheless, the biofilm formation was significantly reduced (p < 0.05) at concentrations of ≥4 μg/mL and ≤32 μg/mL for PA01 and ≤32 μg/mL for MDR 7067 when loading tobramycin into liposomes, with or without the anti-biofilm peptide compared to the free antibiotic, empty liposomes, and IDR-1018-loaded liposomes. A tobramycin concentration of ≤256 µg/mL was safe when exposed to a lung carcinoma cell line upon its encapsulation into the liposomal formulation. Tobramycin-loaded liposomes could be a potential candidate for treating lung-infected animal models owing to the high therapeutic efficacy and safety profile of this system compared to the free administration of the antibiotic.
Collapse
Affiliation(s)
- Nouf M. Alzahrani
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Rayan Y. Booq
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Ahmad M. Aldossary
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Abrar A. Bakr
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Fahad A. Almughem
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Ahmed J. Alfahad
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Wijdan K. Alsharif
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Somayah J. Jarallah
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Waleed S. Alharbi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Samar A. Alsudir
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Essam J. Alyamani
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Essam A. Tawfik
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| | - Abdullah A. Alshehri
- National Center of Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (N.M.A.); (R.Y.B.); (A.M.A.); (A.A.B.); (F.A.A.); (A.J.A.); (W.K.A.); (S.J.J.); (S.A.A.); (E.J.A.)
| |
Collapse
|
44
|
Recombinant human β-defensin130 inhibited the growth of foodborne bacteria through membrane disruption and exerted anti-inflammatory activity. Food Sci Biotechnol 2022; 31:893-904. [PMID: 35720462 PMCID: PMC9203618 DOI: 10.1007/s10068-022-01087-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 11/04/2022] Open
Abstract
Foodborne pathogens causing food poisoning and infections are detrimental to human health, and the abuse of antibiotics induced severe antibiotic resistance in past decades. Thus, it is urgent to develop new antimicrobial agents. In the current study, human β-defensin 130 (hBD130), which is an antimicrobial peptide identified in human macrophages in 2017, was initially produced in Pichia pastoris. The purified hBD130 demonstrated broad bactericidal spectrum against foodborne pathogens through a membrane disruption, with concentrations ranging from 10 to 45 μg/mL. Moreover, hBD130 showed a low hemolytic effect and nearly no cytotoxicity to mammalian cells with a dosage of 400 μg/mL. In addition, the secretion amounts and mRNA levels of NO, IL-6, IL-1β, and TNF-α in LPS-induced mouse macrophage were significantly decreased with 1 mg/mL of hBD130. Taken together, these results showed that hBD130 is a promising antimicrobial agent to treat foodborne bacterial infections and inflammation. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-022-01087-y.
Collapse
|
45
|
Effects of dietary antimicrobial peptides on intestinal morphology, antioxidant status, immune responses, microbiota and pathogen disease resistance in grass carp Ctenopharyngodon idellus. Microb Pathog 2022; 165:105386. [PMID: 35031411 DOI: 10.1016/j.micpath.2021.105386] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 12/29/2022]
Abstract
This study aims to highlight the effects (8 weeks) of dietary antimicrobial peptides (AMPs, a compound of 6 kDa and 5 kDa from intestine) on intestinal morphological functions and health status in grass carp (Ctenopharyngodon idellus). Fish were supplemented with various gradient concentrations of AMPs, including M0 (0 mg/kg), M1 (100 mg/kg), M2 (200 mg/kg), M3 (400 mg/kg), M4 (800 mg/kg) and M5 (1600 mg/kg). Our results showed that amylase, lipase, chymotrypsin enzymatic levels, and total antioxidant capacity (T-AOC) were significantly increased (p < 0.05), while malondialdehyde (MDA) content was significantly decreased in the intestines of the AMP treated groups compared to the M0. Histological analysis revealed villus height and crypt depth of foregut and midgut in the M4 group were significantly different (p < 0.05) compared to the M0. In the M3 group, the gene expression levels of IL-1β were significantly up-regulated, while levels of IL10 and TGF-β were significantly down-regulated than other treated and control groups. The abundance of Firmicutes was significantly increased (p < 0.05), while the Planctomycetes abundance was decreased at phylum level in M1-M5 groups. Subsequent to the AMP treatment, fish were injected with Aeromonas. hydrophila to assess disease resistant potential. In A. hydrophila injected M3-group, the gene expressions of IL-1β, IL8, and TNF-α were significantly down-regulated while that of TGF-β was significantly up-regulated, and IL10 showed no significant difference compared to the control. Further, AMPs also increased the abundance of the Acidobacteria, Proteobacteria, and Patescibacteria, and decreased the abundance of the Fusobacteria and Firmicutes. Therefore, dietary AMPs (400-800 mg/kg) boosted intestinal health by promoting intestinal morphology, digestive and antioxidant capacities, immunity, and intestinal microbiota in C. idellus.
Collapse
|
46
|
Ali W, Elsahn A, Ting DSJ, Dua HS, Mohammed I. Host Defence Peptides: A Potent Alternative to Combat Antimicrobial Resistance in the Era of the COVID-19 Pandemic. Antibiotics (Basel) 2022; 11:475. [PMID: 35453226 PMCID: PMC9032040 DOI: 10.3390/antibiotics11040475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 12/07/2022] Open
Abstract
One of the greatest challenges facing the medical community today is the ever-increasing trajectory of antimicrobial resistance (AMR), which is being compounded by the decrease in our antimicrobial armamentarium. From their initial discovery to the current day, antibiotics have seen an exponential increase in their usage, from medical to agricultural use. Benefits aside, this has led to an exponential increase in AMR, with the fear that over 10 million lives are predicted to be lost by 2050, according to the World Health Organisation (WHO). As such, medical researchers are turning their focus to discovering novel alternatives to antimicrobials, one being Host Defence Peptides (HDPs). These small cationic peptides have shown great efficacy in being used as an antimicrobial therapy for currently resistant microbial variants. With the sudden emergence of the SARS-CoV-2 variant and the subsequent global pandemic, the great versatility and potential use of HDPs as an alternative to conventional antibiotics in treating as well as preventing the spread of COVID-19 has been reviewed. Thus, to allow the reader to have a full understanding of the multifaceted therapeutic use of HDPs, this literature review shall cover the association between COVID-19 and AMR whilst discussing and evaluating the use of HDPs as an answer to antimicrobial resistance (AMR).
Collapse
Affiliation(s)
| | | | | | | | - Imran Mohammed
- Section of Ophthalmology, Larry A. Donoso Laboratory for Eye Research, Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Queens Medical Centre, Eye and ENT Building, Nottingham NG7 2UH, UK; (W.A.); (A.E.); (D.S.J.T.); (H.S.D.)
| |
Collapse
|
47
|
Nuti N, Rottmann P, Stucki A, Koch P, Panke S, Dittrich PS. A Multiplexed Cell-Free Assay to Screen for Antimicrobial Peptides in Double Emulsion Droplets. Angew Chem Int Ed Engl 2022; 61:e202114632. [PMID: 34989471 PMCID: PMC9303939 DOI: 10.1002/anie.202114632] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Indexed: 12/17/2022]
Abstract
The global surge in bacterial resistance against traditional antibiotics triggered intensive research for novel compounds, with antimicrobial peptides (AMPs) identified as a promising candidate. Automated methods to systematically generate and screen AMPs according to their membrane preference, however, are still lacking. We introduce a novel microfluidic system for the simultaneous cell-free production and screening of AMPs for their membrane specificity. On our device, AMPs are cell-free produced within water-in-oil-in-water double emulsion droplets, generated at high frequency. Within each droplet, the peptides can interact with different classes of co-encapsulated liposomes, generating a membrane-specific fluorescent signal. The double emulsions can be incubated and observed in a hydrodynamic trapping array or analyzed via flow cytometry. Our approach provides a valuable tool for the discovery and development of membrane-active antimicrobials.
Collapse
Affiliation(s)
- Nicola Nuti
- Department of Biosystems Science and EngineeringBioanalytics GroupETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Philipp Rottmann
- Department of Biosystems Science and EngineeringBioprocess LaboratoryETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Ariane Stucki
- Department of Biosystems Science and EngineeringBioanalytics GroupETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Philipp Koch
- Department of Biosystems Science and EngineeringBioprocess LaboratoryETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Sven Panke
- Department of Biosystems Science and EngineeringBioprocess LaboratoryETH ZürichMattenstrasse 264058BaselSwitzerland
| | - Petra S. Dittrich
- Department of Biosystems Science and EngineeringBioanalytics GroupETH ZürichMattenstrasse 264058BaselSwitzerland
| |
Collapse
|
48
|
Pérez-Cobas AE, Baquero F, de Pablo R, Soriano MC, Coque TM. Altered Ecology of the Respiratory Tract Microbiome and Nosocomial Pneumonia. Front Microbiol 2022; 12:709421. [PMID: 35222291 PMCID: PMC8866767 DOI: 10.3389/fmicb.2021.709421] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 12/21/2021] [Indexed: 12/23/2022] Open
Abstract
Nosocomial pneumonia is one of the most frequent infections in critical patients. It is primarily associated with mechanical ventilation leading to severe illness, high mortality, and prolonged hospitalization. The risk of mortality has increased over time due to the rise in multidrug-resistant (MDR) bacterial infections, which represent a global public health threat. Respiratory tract microbiome (RTM) research is growing, and recent studies suggest that a healthy RTM positively stimulates the immune system and, like the gut microbiome, can protect against pathogen infection through colonization resistance (CR). Physiological conditions of critical patients and interventions as antibiotics administration and mechanical ventilation dramatically alter the RTM, leading to dysbiosis. The dysbiosis of the RTM of ICU patients favors the colonization by opportunistic and resistant pathogens that can be part of the microbiota or acquired from the hospital environments (biotic or built ones). Despite recent evidence demonstrating the significance of RTM in nosocomial infections, most of the host-RTM interactions remain unknown. In this context, we present our perspective regarding research in RTM altered ecology in the clinical environment, particularly as a risk for acquisition of nosocomial pneumonia. We also reflect on the gaps in the field and suggest future research directions. Moreover, expected microbiome-based interventions together with the tools to study the RTM highlighting the "omics" approaches are discussed.
Collapse
Affiliation(s)
- Ana Elena Pérez-Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain
| | - Fernando Baquero
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain.,CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Raúl de Pablo
- Intensive Care Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - María Cruz Soriano
- Intensive Care Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - Teresa M Coque
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS), Ramón y Cajal University Hospital, Madrid, Spain.,CIBER in Infectious Diseases (CIBERINFEC), Madrid, Spain
| |
Collapse
|
49
|
|
50
|
Deepthi V, Mohanakumar KP, Rajamma U. Efficacy of defensins as neutralizing agents against the deadly SARS-CoV-2. J Biomol Struct Dyn 2022; 41:2911-2925. [PMID: 35189779 DOI: 10.1080/07391102.2022.2041487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
SARS-CoV-2 infection causes asymptomatic to severe human respiratory diseases. Vaccinations are effective only to a certain extent, and the disease recurs with milder symptoms even after booster doses. Hence, we hypothesize that antiviral therapy in conjunction with vaccination is the need of the hour for containing the disease. SARS-CoV-2 enters the host cell through interaction between viral spike (S) protein and human Angiotensin II converting enzyme2 (ACE2). So, any S-protein neutralizing molecule could be a potential antiviral moiety. The interaction-interface architecture indicates that cationic peptides effectively bind to anionic interface residues of S protein-receptor binding domain (S-RBD). Subsequently, we adopted molecular docking and simulation approaches to examine the binding affinity of cationic human α and β defensins, HNP1 and HBD2 with S-RBD. We observed strong hydrogen bonds, electrostatic, salt bridge, and hydrophobic interactions between these defensins and S-RBD with binding energy (BE) of -10.7 kcal/mol. Interestingly, defensins from Zea mays (ZmD32), Solanum lycopersicum (TPP3), and Sorghum bicolor (DEF1_SORBI) exhibited approximately similar BE of -11.1 kcal/mol, -11.9 kcal/mol, and -12.6 kcal/mol respectively, comparable to ACE2 (BE= -11.9 kcal/mol). Molecular dynamics simulation of S-RBD complexes formed with HBD2, ZmD32 and TPP3, showed stable associations for 100 ns. Results of in-silico studies demonstrated higher binding affinity of more positively-charged peptides with S-RBD, suggesting the potential of plant defensins to block ACE2 binding of S-RBD. These results warrant experimental validation. However these findings indicate the usefulness of plant defensin homologues as neutralizing antiviral agents for use as ideal prophylactic and therapeutic drugs for COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Varughese Deepthi
- Centre for Development and Aging Research, Inter University Centre for Biomedical Research & Super Speciality Hospital, Mahatma Gandhi University Campus at Thalappady, Kerala, India
| | - Kochupurackal P Mohanakumar
- Centre for Development and Aging Research, Inter University Centre for Biomedical Research & Super Speciality Hospital, Mahatma Gandhi University Campus at Thalappady, Kerala, India
- Virus Research and Diagnostic Centre, Inter University Centre for Biomedical Research & Super Speciality Hospital, Mahatma Gandhi University Campus at Thalappady, Kerala, India
| | - Usha Rajamma
- Centre for Development and Aging Research, Inter University Centre for Biomedical Research & Super Speciality Hospital, Mahatma Gandhi University Campus at Thalappady, Kerala, India
| |
Collapse
|