1
|
Espiritu A, O’Sullivan KM. A Web of Challenges: The Therapeutic Struggle to Target NETs in Disease. Int J Mol Sci 2025; 26:4773. [PMID: 40429915 PMCID: PMC12111817 DOI: 10.3390/ijms26104773] [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: 04/08/2025] [Revised: 05/12/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Neutrophil extracellular traps (NETs) play a crucial role in the pathophysiology of many debilitating conditions, including autoimmune diseases, inflammatory diseases, and cancer. As a result, NET-targeted therapies have been investigated in search of effective treatment strategies. Despite promising preclinical findings, clinical translation of NET inhibitors has had limited success. These preclinical studies have faced limitations such as mouse models that inaccurately reflect human disease dynamics, as well as by the complexity of NETs-including their diverse morphology and convoluted pathways to formation relative to pathology. The NET inhibitors themselves have several limitations, including off-target effects and bioavailability issues. The challenges facing NET-targeted therapies reported here may explain what is required to go from bench to bedside successfully.
Collapse
Affiliation(s)
| | - Kim Maree O’Sullivan
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, VIC 3168, Australia;
| |
Collapse
|
2
|
Duong L, Wu Y, Kasallis SJ, Abbondante S, Hurst PJ, Marshall ME, McCarthy K, Reddy BJN, Bru JL, Perinbam K, Pearlman E, Patterson JP, Gross SP, Siryaporn A. Bactericidal activity of mammalian histones is caused by large membrane pore formation. Cell Rep 2025; 44:115658. [PMID: 40333180 DOI: 10.1016/j.celrep.2025.115658] [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: 12/09/2024] [Revised: 02/16/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
Histones have an important role in eukaryotic innate immunity, wherein histones co-localize with antimicrobial peptides (AMPs). The mechanism of histone cooperation with AMPs and the extent to which histones form pores both remain a mystery. Here, we show that histones form large pores in bacterial membranes that lack lipopolysaccharide (LPS) and that their antimicrobial effect is significantly stronger than that of the clinical AMP polymyxin B. We find that histones and AMPs together produce potent antimicrobial synergy through the formation of 26 nm pores, whereby the pore-forming activity of AMPs on LPS-containing membranes enables histones to enter the periplasmic space and subsequently attack unprotected membranes to create pores. We provide a mechanistic explanation for the long-standing observations of histone antimicrobial activity and demonstrate how antimicrobial synergy arises. The ubiquity of histones and AMPs in innate immunity has significant implications for organismal defense and can be leveraged for novel antibiotic strategies.
Collapse
Affiliation(s)
- Leora Duong
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Yonghan Wu
- Department of Physics and Astronomy, University of California, Irvine, Irvine, CA 92697, USA
| | - Summer J Kasallis
- Department of Physics and Astronomy, University of California, Irvine, Irvine, CA 92697, USA
| | - Serena Abbondante
- Department of Ophthalmology, University of California, Irvine, Irvine, CA 92697, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Paul J Hurst
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Michaela E Marshall
- Department of Ophthalmology, University of California, Irvine, Irvine, CA 92697, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Katherine McCarthy
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Babu J N Reddy
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Jean-Louis Bru
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Kumar Perinbam
- Department of Physics and Astronomy, University of California, Irvine, Irvine, CA 92697, USA; School of Basic and Applied Sciences, Dayananda Sagar University, Bengaluru, Karnataka 560078, India
| | - Eric Pearlman
- Department of Ophthalmology, University of California, Irvine, Irvine, CA 92697, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Joseph P Patterson
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Steven P Gross
- Department of Physics and Astronomy, University of California, Irvine, Irvine, CA 92697, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA.
| | - Albert Siryaporn
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Physics and Astronomy, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
3
|
Peng Y, Liu Y, Wu J, Zhang Y, Wang Q, Shao S. ArgR-dependent bacterial resistance to host lipid droplets in Edwardsiella piscicida. Commun Biol 2025; 8:333. [PMID: 40021749 PMCID: PMC11871307 DOI: 10.1038/s42003-025-07777-7] [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: 07/03/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Lipid droplets (LDs), as innate immune hubs, function in the front line of antimicrobial defense involved in the host-pathogen arms race. Particularly for intracellular bacterial pathogens, the endowed capacity to resist host LDs can effectively facilitate pathogen in vivo colonization and evasion from the host's innate immune response. Here, to investigate the genetic mechanisms of intracellular bacteria response to host LDs, we utilized transposon insertion sequencing to dissect critical fitness determinants of Edwardsiella piscicida under the treatment of LDs isolated from its native host, turbot. Targeted metabolomics indicated that LD challenge resulted in the accumulation of intracellular arginine. The core arginine metabolism regulatory factor, ArgR, was found to play a pivotal role in combating LDs, emphasizing the importance of orchestrating intracellular arginine levels for bacterial LD adaptation. Specifically, ArgR enhanced the expressions of genes involved in arginine catabolism (speA/B and arcC) and diminished gene transcripts associated with arginine import (artP) and synthesis (argD/E/H). Furthermore, ArgR contributed to the pathogenesis of E. piscicida, promoting the proliferation in host cells and virulence in turbot. Collectively, our results shed light on the underlying mechanism of intracellular pathogens resisting LDs during bacterial infections and highlighting the crucial role of arginine in the host-pathogen interactions.
Collapse
Affiliation(s)
- Yue Peng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yihan Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Junze Wu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Yuanxing Zhang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), 519000, Zhuhai, China
| | - Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, China
- Laboratory of Aquatic Animal Diseases of MOA, Shanghai, China
| | - Shuai Shao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.
- Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, China.
- Laboratory of Aquatic Animal Diseases of MOA, Shanghai, China.
| |
Collapse
|
4
|
Sweet MJ, Ramnath D, Singhal A, Kapetanovic R. Inducible antibacterial responses in macrophages. Nat Rev Immunol 2025; 25:92-107. [PMID: 39294278 DOI: 10.1038/s41577-024-01080-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/20/2024]
Abstract
Macrophages destroy bacteria and other microorganisms through phagocytosis-coupled antimicrobial responses, such as the generation of reactive oxygen species and the delivery of hydrolytic enzymes from lysosomes to the phagosome. However, many intracellular bacteria subvert these responses, escaping to other cellular compartments to survive and/or replicate. Such bacterial subversion strategies are countered by a range of additional direct antibacterial responses that are switched on by pattern-recognition receptors and/or host-derived cytokines and other factors, often through inducible gene expression and/or metabolic reprogramming. Our understanding of these inducible antibacterial defence strategies in macrophages is rapidly evolving. In this Review, we provide an overview of the broad repertoire of antibacterial responses that can be engaged in macrophages, including LC3-associated phagocytosis, metabolic reprogramming and antimicrobial metabolites, lipid droplets, guanylate-binding proteins, antimicrobial peptides, metal ion toxicity, nutrient depletion, autophagy and nitric oxide production. We also highlight key inducers, signalling pathways and transcription factors involved in driving these different antibacterial responses. Finally, we discuss how a detailed understanding of the molecular mechanisms of antibacterial responses in macrophages might be exploited for developing host-directed therapies to combat antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| | - Divya Ramnath
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Amit Singhal
- Infectious Diseases Labs (ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ronan Kapetanovic
- INRAE, Université de Tours, Infectiologie et Santé Publique (ISP), Nouzilly, France
| |
Collapse
|
5
|
Li M, Zhao P, Wang J, Zhang X, Li J. Functional antimicrobial peptide-loaded 3D scaffolds for infected bone defect treatment with AI and multidimensional printing. MATERIALS HORIZONS 2025; 12:20-36. [PMID: 39484845 DOI: 10.1039/d4mh01124d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infection is the most prevalent complication of fractures, particularly in open fractures, and often leads to severe consequences. The emergence of bacterial resistance has significantly exacerbated the burden of infection in clinical practice, making infection control a significant treatment challenge for infectious bone defects. The implantation of a structural stent is necessary to treat large bone defects despite the increased risk of infection. Therefore, there is a need for the development of novel antibacterial therapies. The advancement in antibacterial biomaterials and new antimicrobial drugs offers fresh perspectives on antibacterial treatment. Although antimicrobial 3D scaffolds are currently under intense research focus, relying solely on material properties or antibiotic action remains insufficient. Antimicrobial peptides (AMPs) are one of the most promising new antibacterial therapy approaches. This review discusses the underlying mechanisms behind infectious bone defects and presents research findings on antimicrobial peptides, specifically emphasizing their mechanisms and optimization strategies. We also explore the potential prospects of utilizing antimicrobial peptides in treating infectious bone defects. Furthermore, we propose that artificial intelligence (AI) algorithms can be utilized for predicting the pharmacokinetic properties of AMPs, including absorption, distribution, metabolism, and excretion, and by combining information from genomics, proteomics, metabolomics, and clinical studies with computational models driven by machine learning algorithms, scientists can gain a comprehensive understanding of AMPs' mechanisms of action, therapeutic potential, and optimizing treatment strategies tailored to individual patients, and through interdisciplinary collaborations between computer scientists, biologists, and clinicians, the full potential of AI in accelerating the discovery and development of novel AMPs will be realized. Besides, with the continuous advancements in 3D/4D/5D/6D technology and its integration into bone scaffold materials, we anticipate remarkable progress in the field of regenerative medicine. This review summarizes relevant research on the optimal future for the treatment of infectious bone defects, provides guidance for future novel treatment strategies combining multi-dimensional printing with new antimicrobial agents, and provides a novel and effective solution to the current challenges in the field of bone regeneration.
Collapse
Affiliation(s)
- Mengmeng Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Peizhang Zhao
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jingwen Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xincai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
6
|
Grinat J, Shriever NP, Christophorou MA. Fantastic proteins and where to find them - histones, in the nucleus and beyond. J Cell Sci 2024; 137:jcs262071. [PMID: 39704565 PMCID: PMC11827605 DOI: 10.1242/jcs.262071] [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: 12/21/2024] Open
Abstract
Animal genomes are packaged into chromatin, a highly dynamic macromolecular structure of DNA and histone proteins organised into nucleosomes. This accommodates packaging of lengthy genomic sequences within the physical confines of the nucleus while also enabling precise regulation of access to genetic information. However, histones existed before chromatin and have lesser-known functions beyond genome regulation. Most notably, histones are potent antimicrobial agents, and the release of chromatin to the extracellular space is a defence mechanism nearly as ancient and widespread as chromatin itself. Histone sequences have changed very little throughout evolution, suggesting the possibility that some of their 'non-canonical' functions are at play in parallel or in concert with their genome regulatory functions. In this Review, we take an evolutionary perspective of histone, nuclear chromatin and extracellular chromatin biology and describe the known extranuclear and extracellular functions of histones. We detail molecular mechanisms of chromatin release and extracellular chromatin sensing, and we discuss their roles in physiology and disease. Finally, we present evidence and give a perspective on the potential of extracellular histones to act as bioactive, cell modulatory factors.
Collapse
|
7
|
Driouich A, Gueye MLF, Vicré M, Moore JP. The root extracellular trap; a complex and dynamic biomatrix network essential for plant protection. CURRENT OPINION IN PLANT BIOLOGY 2024; 82:102656. [PMID: 39536647 DOI: 10.1016/j.pbi.2024.102656] [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: 07/18/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Plants have evolved a number of defense mechanisms to protect themselves against biotic stresses. Each cell, tissue, and organ is able to perceive and fight off attackers using a combination of chemical and physical defense mechanisms. Root cells employ similar defense response patterning. They develop immune responses upon pathogen attack and release a variety of compounds able to defend the root proper as well as the entire plant body. Currently, one of the most effective mechanisms of root defense involves the root extracellular trap (RET) that is produced at the tip of the root. The RET consists of root cap-derived cells embedded in mucilaginous secretions containing cell wall-derived polysaccharides, defense-related (glyco)proteins, phytoalexins, histones, and extracellular DNA (eDNA). The RET network plays a central role in root immunity and fulfills biological functions similar to those performed by neutrophil extracellular traps in mammals.
Collapse
Affiliation(s)
- Azeddine Driouich
- Normandie Université, Laboratoire Glycobiologie et Matrice Extracellulaire Végétale EA 4358, Fédération de Recherche -Normandie Végétal- FED 4277, Université de Rouen Normandie, 76000, Rouen, France.
| | - Marie-Laure Follet Gueye
- Normandie Université, Laboratoire Glycobiologie et Matrice Extracellulaire Végétale EA 4358, Fédération de Recherche -Normandie Végétal- FED 4277, Université de Rouen Normandie, 76000, Rouen, France
| | - Maïté Vicré
- Normandie Université, Laboratoire Glycobiologie et Matrice Extracellulaire Végétale EA 4358, Fédération de Recherche -Normandie Végétal- FED 4277, Université de Rouen Normandie, 76000, Rouen, France
| | - John P Moore
- South African Grape and Wine Research Institute, Department of Viticulture and Oenology, Faculty of AgriSciences, Stellenbosch University, Matieland, 7602, South Africa
| |
Collapse
|
8
|
Marsman G, Zheng X, Čerina D, Lacey KA, Liu M, Humme D, Goosmann C, Brinkmann V, Harbort CJ, Torres VJ, Zychlinsky A. Histone H1 kills MRSA. Cell Rep 2024; 43:114969. [PMID: 39546397 DOI: 10.1016/j.celrep.2024.114969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/06/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
The antimicrobial activity of histones was discovered in the 1940s, but their mechanism of action is not fully known. Here we show that methicillin-resistant Staphylococcus aureus (MRSA) is susceptible to histone H1 (H1), even in the presence of divalent cations and serum. Through selective evolution and a genome-wide screen of a transposon library, as well as physiological and pharmacological experiments, we elucidated how H1 kills MRSA. We show that H1 first binds to wall teichoic acids with high affinity. Once bound, H1 requires a potentiated membrane and a metabolically active bacterium to permeabilize the membrane and enter the cell. Upon entry, H1 accumulates intracellularly, in close association with the bacterial DNA. Of note, anti-H1 antibodies inhibit neutrophil extracellular trap killing of MRSA. Moreover, H1 colocalizes with bacterial DNA in abscess samples of MRSA-infected patients, suggesting a role for H1 in combating MRSA in vivo.
Collapse
Affiliation(s)
- Gerben Marsman
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Xuhui Zheng
- Department of Microbiology, New York University Grossman School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Dora Čerina
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Menghan Liu
- Department of Microbiology, New York University Grossman School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Daniel Humme
- Department of Dermatology, Venerology and Allergology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Christian Goosmann
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Volker Brinkmann
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - C J Harbort
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, 430 East 29th Street, New York, NY 10016, USA; Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| | - Arturo Zychlinsky
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
9
|
Arévalo-Jaimes BV, Salinas-Pena M, Ponte I, Jordan A, Roque A, Torrents E. Antimicrobial and antibiofilm activity of human recombinant H1 histones against bacterial infections. mSystems 2024; 9:e0070424. [PMID: 39470247 PMCID: PMC11575268 DOI: 10.1128/msystems.00704-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Histones possess significant antimicrobial potential, yet their activity against biofilms remains underexplored. Moreover, concerns regarding adverse effects limit their clinical implementation. We investigated the antibacterial efficacy of human recombinant histone H1 subtypes against Pseudomonas aeruginosa PAO1, both planktonic and in biofilms. After the in vitro tests, toxicity and efficacy were assessed in a P. aeruginosa PAO1 infection model using Galleria mellonella larvae. Histones were also evaluated in combination with ciprofloxacin (Cpx) and gentamicin (Gm). Our results demonstrate antimicrobial activity of all three histones against P. aeruginosa PAO1, with H1.0 and H1.4 showing efficacy at lower concentrations. The bactericidal effect was associated with a mechanism of membrane disruption. In vitro studies using static and dynamic models showed that H1.4 had antibiofilm potential by reducing cell biomass. Neither H1.0 nor H1.4 showed toxicity in G. mellonella larvae, and both increased larvae survival when infected with P. aeruginosa PAO1. Although in vitro synergism was observed between ciprofloxacin and H1.0, no improvement over the antibiotic alone was noted in vivo. Differences in antibacterial and antibiofilm activity were attributed to sequence and structural variations among histone subtypes. Moreover, the efficacy of H1.0 and H1.4 was influenced by the presence and strength of the extracellular matrix. These findings suggest histones hold promise for combating acute and chronic infections caused by pathogens such as P. aeruginosa.IMPORTANCEThe constant increase of multidrug-resistant bacteria is a critical global concern. The inefficacy of current therapies to treat bacterial infections is attributed to multiple mechanisms of resistance, including the capacity to form biofilms. Therefore, the identification of novel and safe therapeutic strategies is imperative. This study confirms the antimicrobial potential of three histone H1 subtypes against both Gram-negative and Gram-positive bacteria. Furthermore, histones H1.0 and H1.4 demonstrated in vivo efficacy without associated toxicity in an acute infection model of Pseudomonas aeruginosa PAO1 in Galleria mellonella larvae. The bactericidal effect of these proteins also resulted in biomass reduction of P. aeruginosa PAO1 biofilms. Given the clinical significance of this opportunistic pathogen, our research provides a comprehensive initial evaluation of the efficacy, toxicity, and mechanism of action of a potential new therapeutic approach against acute and chronic bacterial infections.
Collapse
Affiliation(s)
- Betsy Verónica Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | | | - Inmaculada Ponte
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Jordan
- Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Alicia Roque
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
10
|
Ma X, Wang Q, Ren K, Xu T, Zhang Z, Xu M, Rao Z, Zhang X. A Review of Antimicrobial Peptides: Structure, Mechanism of Action, and Molecular Optimization Strategies. FERMENTATION-BASEL 2024; 10:540. [DOI: 10.3390/fermentation10110540] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Antimicrobial peptides (AMPs) are bioactive macromolecules that exhibit antibacterial, antiviral, and immunomodulatory functions. They come from a wide range of sources and are found in all forms of life, from bacteria to plants, vertebrates, and invertebrates, and play an important role in controlling the spread of pathogens, promoting wound healing and treating tumors. Consequently, AMPs have emerged as promising alternatives to next-generation antibiotics. With advancements in systems biology and synthetic biology technologies, it has become possible to synthesize AMPs artificially. We can better understand their functional activities for further modification and development by investigating the mechanism of action underlying their antimicrobial properties. This review focuses on the structural aspects of AMPs while highlighting their significance for biological activity. Furthermore, it elucidates the membrane targeting mechanism and intracellular targets of these peptides while summarizing molecular modification approaches aimed at enhancing their antibacterial efficacy. Finally, this article outlines future challenges in the functional development of AMPs along with proposed strategies to overcome them.
Collapse
Affiliation(s)
- Xu Ma
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China
| | - Qiang Wang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China
| | - Kexin Ren
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China
| | - Tongtong Xu
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China
| | - Zigang Zhang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China
| | - Meijuan Xu
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China
| | - Zhiming Rao
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China
| | - Xian Zhang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food and Biotechnology Co., Ltd., Yixing 214200, China
| |
Collapse
|
11
|
Muñoz-Camargo C, Cruz JC. From inside to outside: exploring extracellular antimicrobial histone-derived peptides as multi-talented molecules. J Antibiot (Tokyo) 2024; 77:553-568. [PMID: 38871806 PMCID: PMC11347383 DOI: 10.1038/s41429-024-00744-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 06/15/2024]
Abstract
The emergence of bacterial resistance to antibiotics poses a global health threat, necessitating innovative solutions. The contemporary challenge lies in bacterial resistance, impacting morbidity, mortality, and global economies. Antimicrobial peptides (AMPs) offer a promising avenue for addressing antibiotic resistance. The Antimicrobial Peptide Database catalogs 3569 peptides from various organisms, representing a rich resource for drug development. Histones, traditionally recognized for their role in nucleosome structures, have gained attention for their extracellular functions, including antimicrobial and immunomodulatory properties. This review aims to thoroughly investigate antimicrobial peptides derived from histones in various organisms, elucidating their mechanisms. In addition, it gives us clues about how extracellular histones might be used in drug delivery systems to fight bacterial infections. This comprehensive analysis emphasizes the importance of histone-derived peptides in developing innovative therapeutic strategies for evolving bacterial challenges.
Collapse
Affiliation(s)
- Carolina Muñoz-Camargo
- Grupo de investigación en Nanobiomateriales, Ingeniería Celular y Bioimpresión (GINIB), Departamento de Ingeniería Biomédica, Universidad de los Andes, Bogotá, Colombia.
| | - Juan C Cruz
- Grupo de investigación en Nanobiomateriales, Ingeniería Celular y Bioimpresión (GINIB), Departamento de Ingeniería Biomédica, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
12
|
Ye Z, Fu L, Li S, Chen Z, Ouyang J, Shang X, Liu Y, Gao L, Wang Y. Synergistic collaboration between AMPs and non-direct antimicrobial cationic peptides. Nat Commun 2024; 15:7319. [PMID: 39183339 PMCID: PMC11345435 DOI: 10.1038/s41467-024-51730-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Non-direct antimicrobial cationic peptides (NDACPs) are components of the animal innate immune system. But their functions and association with antimicrobial peptides (AMPs) are incompletely understood. Here, we reveal a synergistic interaction between the AMP AW1 and the NDACP AW2, which are co-expressed in the frog Amolops wuyiensis. AW2 enhances the antibacterial activity of AW1 both in vitro and in vivo, while mitigating the development of bacterial resistance and eradicating biofilms. AW1 and AW2 synergistically damage bacterial membranes, facilitating cellular uptake and interaction of AW2 with the intracellular target bacterial genomic DNA. Simultaneously, they trigger the generation of ROS in bacteria, contributing to cell death upon reaching a threshold level. Moreover, we demonstrate that this synergistic antibacterial effect between AMPs and NDACPs is prevalent across diverse animal species. These findings unveil a robust and previously unknown correlation between AMPs and NDACPs as a widespread antibacterial immune defense strategy in animals.
Collapse
Affiliation(s)
- Zifan Ye
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Lei Fu
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Shuangyu Li
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Ziying Chen
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Jianhong Ouyang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xinci Shang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yanli Liu
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Lianghui Gao
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China.
| | - Yipeng Wang
- Department of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.
| |
Collapse
|
13
|
Parveen S, Bhat CV, Sagilkumar AC, Aziz S, Arya J, Dutta A, Dutta S, Show S, Sharma K, Rakshit S, Johnson JB, Nongthomba U, Banerjee A, Subramanian K. Bacterial pore-forming toxin pneumolysin drives pathogenicity through host extracellular vesicles released during infection. iScience 2024; 27:110589. [PMID: 39211544 PMCID: PMC11357855 DOI: 10.1016/j.isci.2024.110589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Streptococcus pneumoniae is a global priority respiratory pathogen that kills over a million people annually. The pore-forming cytotoxin, pneumolysin (PLY) is a major virulence factor. Here, we found that recombinant PLY as well as wild-type pneumococcal strains, but not the isogenic PLY mutant, upregulated the shedding of extracellular vesicles (EVs) harboring membrane-bound toxin from human THP-1 monocytes. PLY-EVs induced cytotoxicity and hemolysis dose-dependently upon internalization by recipient monocyte-derived dendritic cells. Proteomics analysis revealed that PLY-EVs are selectively enriched in key inflammatory host proteins such as IFI16, NLRC4, PTX3, and MMP9. EVs shed from PLY-challenged or infected cells induced dendritic cell maturation and primed them to infection. In vivo, zebrafish administered with PLY-EVs showed pericardial edema and mortality. Adoptive transfer of bronchoalveolar-lavage-derived EVs from infected mice to healthy recipients induced lung damage and inflammation in a PLY-dependent manner. Our findings identify that host EVs released during infection mediate pneumococcal pathogenesis.
Collapse
Affiliation(s)
- Saba Parveen
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Chinmayi V Bhat
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Aswathy C Sagilkumar
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
- Regional Centre for Biotechnology, Faridabad 121001, India
| | - Shaheena Aziz
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - J Arya
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Asmita Dutta
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Somit Dutta
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Sautan Show
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Kuldeep Sharma
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sumit Rakshit
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - John Bernet Johnson
- Virology Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Upendra Nongthomba
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Anirban Banerjee
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Karthik Subramanian
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
- Regional Centre for Biotechnology, Faridabad 121001, India
| |
Collapse
|
14
|
Habjan E, Schouten GK, Speer A, van Ulsen P, Bitter W. Diving into drug-screening: zebrafish embryos as an in vivo platform for antimicrobial drug discovery and assessment. FEMS Microbiol Rev 2024; 48:fuae011. [PMID: 38684467 PMCID: PMC11078164 DOI: 10.1093/femsre/fuae011] [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: 11/01/2023] [Revised: 02/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
The rise of multidrug-resistant bacteria underlines the need for innovative treatments, yet the introduction of new drugs has stagnated despite numerous antimicrobial discoveries. A major hurdle is a poor correlation between promising in vitro data and in vivo efficacy in animal models, which is essential for clinical development. Early in vivo testing is hindered by the expense and complexity of existing animal models. Therefore, there is a pressing need for cost-effective, rapid preclinical models with high translational value. To overcome these challenges, zebrafish embryos have emerged as an attractive model for infectious disease studies, offering advantages such as ethical alignment, rapid development, ease of maintenance, and genetic manipulability. The zebrafish embryo infection model, involving microinjection or immersion of pathogens and potential antibiotic hit compounds, provides a promising solution for early-stage drug screening. It offers a cost-effective and rapid means of assessing the efficacy, toxicity and mechanism of action of compounds in a whole-organism context. This review discusses the experimental design of this model, but also its benefits and challenges. Additionally, it highlights recently identified compounds in the zebrafish embryo infection model and discusses the relevance of the model in predicting the compound's clinical potential.
Collapse
Affiliation(s)
- Eva Habjan
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Gina K Schouten
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Alexander Speer
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Peter van Ulsen
- Section Molecular Microbiology of A-LIFE, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Section Molecular Microbiology of A-LIFE, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
15
|
Wang Y, Du C, Zhang Y, Zhu L. Composition and Function of Neutrophil Extracellular Traps. Biomolecules 2024; 14:416. [PMID: 38672433 PMCID: PMC11048602 DOI: 10.3390/biom14040416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are intricate fibrous structures released by neutrophils in response to specific stimuli. These structures are composed of depolymerized chromatin adorned with histones, granule proteins, and cytosolic proteins. NETs are formed via two distinct pathways known as suicidal NETosis, which involves NADPH oxidase (NOX), and vital NETosis, which is independent of NOX. Certain proteins found within NETs exhibit strong cytotoxic effects against both pathogens and nearby host cells. While NETs play a defensive role against pathogens, they can also contribute to tissue damage and worsen inflammation. Despite extensive research on the pathophysiological role of NETs, less attention has been paid to their components, which form a unique structure containing various proteins that have significant implications in a wide range of diseases. This review aims to elucidate the components of NETs and provide an overview of their impact on host defense against invasive pathogens, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Yijie Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Beijing Institute of Infectious Diseases, Beijing 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Chunjing Du
- Department of Critical Care Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yue Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Beijing Institute of Infectious Diseases, Beijing 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Liuluan Zhu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Beijing Institute of Infectious Diseases, Beijing 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| |
Collapse
|
16
|
Eskandarian HA, Chen YX, Toniolo C, Belardinelli JM, Palcekova Z, Hom L, Ashby PD, Fantner GE, Jackson M, McKinney JD, Javid B. Mechanical morphotype switching as an adaptive response in mycobacteria. SCIENCE ADVANCES 2024; 10:eadh7957. [PMID: 38170768 PMCID: PMC10776010 DOI: 10.1126/sciadv.adh7957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
Invading microbes face a myriad of cidal mechanisms of phagocytes that inflict physical damage to microbial structures. How intracellular bacterial pathogens adapt to these stresses is not fully understood. Here, we report the discovery of a virulence mechanism by which changes to the mechanical stiffness of the mycobacterial cell surface confer refraction to killing during infection. Long-term time-lapse atomic force microscopy was used to reveal a process of "mechanical morphotype switching" in mycobacteria exposed to host intracellular stress. A "soft" mechanical morphotype switch enhances tolerance to intracellular macrophage stress, including cathelicidin. Both pharmacologic treatment, with bedaquiline, and a genetic mutant lacking uvrA modified the basal mechanical state of mycobacteria into a soft mechanical morphotype, enhancing survival in macrophages. Our study proposes microbial cell mechanical adaptation as a critical axis for surviving host-mediated stressors.
Collapse
Affiliation(s)
- Haig Alexander Eskandarian
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94143, USA
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Yu-Xiang Chen
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Chiara Toniolo
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Juan M. Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523-1682, USA
| | - Zuzana Palcekova
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523-1682, USA
| | - Lesley Hom
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Paul D. Ashby
- Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Materials Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Georg E. Fantner
- School of Engineering, Swiss Federal Institute of Technology (EPFL), 1015 Lausanne, Switzerland
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523-1682, USA
| | - John D. McKinney
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Babak Javid
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
17
|
Masadeh MM, Alshogran H, Alsaggar M, Sabi SH, Al Momany EM, Masadeh MM, Alrabadi N, Alzoubi KH. Evaluation of Novel HLM Peptide Activity and Toxicity against Planktonic and Biofilm Bacteria: Comparison to Standard Antibiotics. Curr Protein Pept Sci 2024; 25:826-843. [PMID: 38910428 DOI: 10.2174/0113892037291252240528110516] [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: 12/15/2023] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Antibiotic resistance is one of the main concerns of public health, and the whole world is trying to overcome such a challenge by finding novel therapeutic modalities and approaches. This study has applied the sequence hybridization approach to the original sequence of two cathelicidin natural parent peptides (BMAP-28 and LL-37) to design a novel HLM peptide with broad antimicrobial activity. METHODS The physicochemical characteristics of the newly designed peptide were determined. As well, the new peptide's antimicrobial activity (Minimum Inhibitory Concentration (MIC), Minimum Bacterial Eradication Concentration (MBEC), and antibiofilm activity) was tested on two control (Staphylococcus aureus ATCC 29213, Escherichia coli ATCC 25922) and two resistant (Methicillin-resistant Staphylococcus aureus (MRSA) ATCC BAA41, New Delhi metallo-beta- lactamase-1 Escherichia coli ATCC BAA-2452) bacterial strains. Furthermore, synergistic studies have been applied to HLM-hybridized peptides with five conventional antibiotics by checkerboard assays. Also, the toxicity of HLM-hybridized peptide was studied on Vero cell lines to obtain the IC50 value. Besides the percentage of hemolysis action, the peptide was tested in freshly heparinized blood. RESULTS The MIC values for the HLM peptide were obtained as 20, 10, 20, and 20 μM, respectively. Also, the results showed no hemolysis action, with low to slightly moderate toxicity action against mammalian cells, with an IC50 value of 10.06. The Biomatik corporate labs, where HLM was manufactured, determined the stability results of the product by Mass Spectrophotometry (MS) and High-performance Liquid Chromatography (HPLC) methods. The HLM-hybridized peptide exhibited a range of synergistic to additive antimicrobial activities upon combination with five commercially available different antibiotics. It has demonstrated the biofilm-killing effects in the same concentration required to eradicate the control strains. CONCLUSION The results indicated that HLM-hybridized peptide displayed a broad-spectrum activity toward different bacterial strains in planktonic and biofilm forms. It showed synergistic or additive antimicrobial activity upon combining with commercially available different antibiotics.
Collapse
Affiliation(s)
- Majed M Masadeh
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Haneen Alshogran
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad Alsaggar
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Salsabeel H Sabi
- Department of Biology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Enaam M Al Momany
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, P.O. box 330127, Zarqa 13133, Jordan
| | - Majd M Masadeh
- Discipline of Clinical Pharmacy, School of Pharmaceutical Sciences, University Sains Malaysia, 11800, Penang, Malaysia
| | - Nasr Alrabadi
- Department of Pharmacology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Karem H Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, UAE
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
18
|
Liu F, Greenwood AI, Xiong Y, Miceli RT, Fu R, Anderson KW, McCallum SA, Mihailescu M, Gross R, Cotten ML. Host Defense Peptide Piscidin and Yeast-Derived Glycolipid Exhibit Synergistic Antimicrobial Action through Concerted Interactions with Membranes. JACS AU 2023; 3:3345-3365. [PMID: 38155643 PMCID: PMC10751773 DOI: 10.1021/jacsau.3c00506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 12/30/2023]
Abstract
Developing new antimicrobials as alternatives to conventional antibiotics has become an urgent race to eradicate drug-resistant bacteria and to save human lives. Conventionally, antimicrobial molecules are studied independently even though they can be cosecreted in vivo. In this research, we investigate two classes of naturally derived antimicrobials: sophorolipid (SL) esters as modified yeast-derived glycolipid biosurfactants that feature high biocompatibility and low production cost; piscidins, which are host defense peptides (HDPs) from fish. While HDPs such as piscidins target the membrane of pathogens, and thus result in low incidence of resistance, SLs are not well understood on a mechanistic level. Here, we demonstrate that combining SL-hexyl ester (SL-HE) with subinhibitory concentration of piscidins 1 (P1) and 3 (P3) stimulates strong antimicrobial synergy, potentiating a promising therapeutic window. Permeabilization assays and biophysical studies employing circular dichroism, NMR, mass spectrometry, and X-ray diffraction are performed to investigate the mechanism underlying this powerful synergy. We reveal four key mechanistic features underlying the synergistic action: (1) P1/3 binds to SL-HE aggregates, becoming α-helical; (2) piscidin-glycolipid assemblies synergistically accumulate on membranes; (3) SL-HE used alone or bound to P1/3 associates with phospholipid bilayers where it induces defects; (4) piscidin-glycolipid complexes disrupt the bilayer structure more dramatically and differently than either compound alone, with phase separation occurring when both agents are present. Overall, dramatic enhancement in antimicrobial activity is associated with the use of two membrane-active agents, with the glycolipid playing the roles of prefolding the peptide, coordinating the delivery of both agents to bacterial surfaces, recruiting the peptide to the pathogenic membranes, and supporting membrane disruption by the peptide. Given that SLs are ubiquitously and safely used in consumer products, the SL/peptide formulation engineered and mechanistically characterized in this study could represent fertile ground to develop novel synergistic agents against drug-resistant bacteria.
Collapse
Affiliation(s)
- Fei Liu
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Alexander I. Greenwood
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| | - Yawei Xiong
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| | - Rebecca T. Miceli
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Riqiang Fu
- Center
of Interdisciplinary Magnetic Resonance, National High Magnetic Field Laboratory, Tallahassee, Florida 32310, United States
| | - Kyle W. Anderson
- National
Institute of Standards and Technology, Rockville, Maryland 20850, United States
| | - Scott A. McCallum
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Mihaela Mihailescu
- Institute
for Bioscience and Biotechnology Research, University of Maryland, Rockville, Maryland 20850, United States
| | - Richard Gross
- Department
of Chemistry, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Center
for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Myriam L. Cotten
- Department
of Applied Science, William & Mary, Williamsburg, Virginia 23185, United States
| |
Collapse
|
19
|
Yang Z, He S, Wei Y, Li X, Shan A, Wang J. Antimicrobial peptides in combination with citronellal efficiently kills multidrug resistance bacteria. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155070. [PMID: 37729771 DOI: 10.1016/j.phymed.2023.155070] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Antimicrobial peptides (AMPs) are considered as the most potential alternatives to antibiotics, but they have several drawbacks, including high cost, medium antimicrobial efficacy, poor cell selectivity, which limit clinical application. To overcome the above problems, combination therapy of AMPs with adjuvants might maximize the effectiveness of AMPs. We found that citronellal can substantially potentiate the ZY4R peptide efficacy against Escherichia coli ATCC25922. However, it is unclear whether ZY4R/citronellal combination poses synergistic antimicrobial effects against most bacteria, and their synergy mechanism has not been elucidated. PURPOSE To investigate synergistic antimicrobial efficacies, biosafety, and synergy mechanism of ZY4R/citronellal combination. METHOD Checkerboard, time-kill curves, cytotoxicity assays, and in vivo animal models were conducted to assess synergistic antimicrobial effects and biosafety of the ZY4R/citronellal combination. To evaluate their synergy mechanism, a series of cell-based assays and transcriptome analysis were performed. RESULTS ZY4R/citronellal combination exhibited synergistic antimicrobial effects against 20 clinically significant pathogens, with the fractional inhibitory concentration index (FICI) ranging from 0.313 to 0.047. Meanwhile, ZY4R/citronellal combination enhanced antimicrobial efficacies without compromising cell selectivity, contributing to decreasing drug dosage and improving biosafety. Compared with ZY4R (4 mg/kg) and citronellal (25 mg/kg) alone, ZY4R (4 mg/kg)/citronellal (25 mg/kg) combination significantly decreased the bacterial load in peritoneal fluid, liver, and kidney (P < 0.05) and alleviated pathological damage of the organs of mice. Mechanistic studies showed that ZY4R allowed citronellal to pass through the outer membrane rapidly and acted on the inner membrane together with citronellal, causing more potent membrane damage. The membrane damage prompted the continuous accumulation of citronellal in cells, and citronellal further induced energy breakdown and inhibited exopolysaccharide (EPS) production, which aggravated ZY4R-induced outer membrane damage, thereby resulting in bacterial death. CONCLUSIONS ZY4R/citronellal combination exhibited broad-spectrum synergy with a low resistance development and high biosafety. Their synergy mechanism acted on two important cellular targets (energy metabolism and membrane integrity). Combination therapy of ZY4R with citronellal may be a promising mixture to combat bacterial infections facing an antibiotic-resistance crisis.
Collapse
Affiliation(s)
- Zhanyi Yang
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Shiqi He
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yingxin Wei
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xuefeng Li
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Anshan Shan
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Jiajun Wang
- College of animal science and technology, Northeast Agricultural University, Harbin 150030, P. R. China.
| |
Collapse
|
20
|
Safi R, Sánchez-Álvarez M, Bosch M, Demangel C, Parton RG, Pol A. Defensive-lipid droplets: Cellular organelles designed for antimicrobial immunity. Immunol Rev 2023; 317:113-136. [PMID: 36960679 DOI: 10.1111/imr.13199] [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: 03/25/2023]
Abstract
Microbes have developed many strategies to subvert host organisms, which, in turn, evolved several innate immune responses. As major lipid storage organelles of eukaryotes, lipid droplets (LDs) are an attractive source of nutrients for invaders. Intracellular viruses, bacteria, and protozoan parasites induce and physically interact with LDs, and the current view is that they "hijack" LDs to draw on substrates for host colonization. This dogma has been challenged by the recent demonstration that LDs are endowed with a protein-mediated antibiotic activity, which is upregulated in response to danger signals and sepsis. Dependence on host nutrients could be a generic "Achilles' heel" of intracellular pathogens and LDs a suitable chokepoint harnessed by innate immunity to organize a front-line defense. Here, we will provide a brief overview of the state of the conflict and discuss potential mechanisms driving the formation of the 'defensive-LDs' functioning as hubs of innate immunity.
Collapse
Affiliation(s)
- Rémi Safi
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Miguel Sánchez-Álvarez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (IIB), Madrid, Spain
| | - Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Caroline Demangel
- Immunobiology and Therapy Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Robert G Parton
- Institute for Molecular Bioscience (IMB), Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis (CMM), University of Queensland, Brisbane, Queensland, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
21
|
Hüsler D, Stauffer P, Hilbi H. Tapping lipid droplets: A rich fat diet of intracellular bacterial pathogens. Mol Microbiol 2023; 120:194-209. [PMID: 37429596 DOI: 10.1111/mmi.15120] [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: 05/03/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023]
Abstract
Lipid droplets (LDs) are dynamic and versatile organelles present in most eukaryotic cells. LDs consist of a hydrophobic core of neutral lipids, a phospholipid monolayer coat, and a variety of associated proteins. LDs are formed at the endoplasmic reticulum and have diverse roles in lipid storage, energy metabolism, membrane trafficking, and cellular signaling. In addition to their physiological cellular functions, LDs have been implicated in the pathogenesis of several diseases, including metabolic disorders, cancer, and infections. A number of intracellular bacterial pathogens modulate and/or interact with LDs during host cell infection. Members of the genera Mycobacterium, Legionella, Coxiella, Chlamydia, and Salmonella exploit LDs as a source of intracellular nutrients and membrane components to establish their distinct intracellular replicative niches. In this review, we focus on the biogenesis, interactions, and functions of LDs, as well as on their role in lipid metabolism of intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Dario Hüsler
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Pia Stauffer
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Ioannou P, Baliou S, Kofteridis DP. Antimicrobial Peptides in Infectious Diseases and Beyond-A Narrative Review. Life (Basel) 2023; 13:1651. [PMID: 37629508 PMCID: PMC10455936 DOI: 10.3390/life13081651] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Despite recent medical research and clinical practice developments, the development of antimicrobial resistance (AMR) significantly limits therapeutics for infectious diseases. Thus, novel treatments for infectious diseases, especially in this era of increasing AMR, are urgently needed. There is ongoing research on non-classical therapies for infectious diseases utilizing alternative antimicrobial mechanisms to fight pathogens, such as bacteriophages or antimicrobial peptides (AMPs). AMPs are evolutionarily conserved molecules naturally produced by several organisms, such as plants, insects, marine organisms, and mammals, aiming to protect the host by fighting pathogenic microorganisms. There is ongoing research regarding developing AMPs for clinical use in infectious diseases. Moreover, AMPs have several other non-medical applications in the food industry, such as preservatives, animal husbandry, plant protection, and aquaculture. This review focuses on AMPs, their origins, biology, structure, mechanisms of action, non-medical applications, and clinical applications in infectious diseases.
Collapse
Affiliation(s)
- Petros Ioannou
- School of Medicine, University of Crete, 71003 Heraklion, Greece
- Internal Medicine, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Stella Baliou
- Internal Medicine, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Diamantis P. Kofteridis
- School of Medicine, University of Crete, 71003 Heraklion, Greece
- Internal Medicine, University Hospital of Heraklion, 71110 Heraklion, Greece
| |
Collapse
|
23
|
Agidigbi TS, Kwon HK, Knight JR, Zhao D, Lee FY, Oh I. Transcriptomic identification of genes expressed in invasive S. aureus diabetic foot ulcer infection. Front Cell Infect Microbiol 2023; 13:1198115. [PMID: 37434783 PMCID: PMC10332306 DOI: 10.3389/fcimb.2023.1198115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction Infection in diabetic foot ulcers (DFUs) is one of the major complications associated with patients with diabetes. Staphylococcus aureus is the most common offending pathogen in patients with infected DFU. Previous studies have suggested the application of species-specific antibodies against S. aureus for diagnosis and monitoring treatment response. Early and accurate identification of the main pathogen is critical for management of DFU infection. Understanding the host immune response against species-specific infection may facilitate diagnosis and may suggest potential intervention options to promote healing infected DFUs. We sought to investigate evolving host transcriptome associated with surgical treatment of S. aureus- infected DFU. Methods This study compared the transcriptome profile of 21 patients with S. aureus- infected DFU who underwent initial foot salvage therapy with irrigation and debridement followed by intravenous antibiotic therapy. Blood samples were collected at the recruitment (0 weeks) and 8 weeks after therapy to isolate peripheral blood mononuclear cells (PBMCs). We analyzed the PBMC expression of transcriptomes at two different time points (0 versus 8 weeks). Subjects were further divided into two groups at 8 weeks: healed (n = 17, 80.95%) versus non-healed (n = 4, 19.05%) based on the wound healing status. DESeq2 differential gene analysis was performed. Results and discussion An increased expression of IGHG1, IGHG2, IGHG3, IGLV3-21, and IGLV6-57 was noted during active infection at 0 weeks compared with that at 8 weeks. Lysine- and arginine-rich histones (HIST1H2AJ, HIST1H2AL, HIST1H2BM, HIST1H3B, and HIST1H3G) were upregulated at the initial phase of active infection at 0 weeks. CD177 and RRM2 were also upregulated at the initial phase of active infection (0 weeks) compared with that at 8 weeks of follow-up. Genes of heat shock protein members (HSPA1A, HSPE1, and HSP90B1) were high in not healed patients compared with that in healed patients 8 weeks after therapy. The outcome of our study suggests that the identification of genes evolution based on a transcriptomic profiling could be a useful tool for diagnosing infection and assessing severity and host immune response to therapies.
Collapse
Affiliation(s)
- Taiwo Samuel Agidigbi
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| | - Hyuk-Kwon Kwon
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
- Division of Life Science, Gyeongsang National University, Jinju, Republic of Korea
| | - James R. Knight
- Yale Center for Genome Analysis, Department of Genetics, Yale School of Medicine, New Haven, CT, United States
| | - Dejian Zhao
- Yale Center for Genome Analysis, Department of Genetics, Yale School of Medicine, New Haven, CT, United States
| | - Francis Y. Lee
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| | - Irvin Oh
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
24
|
Martinez V, Dettleff P, Zamorano P, Galarce N, Borie C, Naish K. Host-pathogen interaction involving cytoskeleton changes as well as non-coding regulation as primary mechanisms for SRS resistance in Atlantic salmon. FISH & SHELLFISH IMMUNOLOGY 2023; 136:108711. [PMID: 37004895 DOI: 10.1016/j.fsi.2023.108711] [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: 11/26/2021] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 06/19/2023]
Abstract
The salmonid rickettsial syndrome (SRS) is a systemic bacterial infection caused by Piscirickettsia salmonis that generates significant economic losses in Atlantic salmon (Salmo salar) aquaculture. Despite this disease's relevance, the mechanisms involved in resistance against P. salmonis infection are not entirely understood. Thus, we aimed at studying the pathways explaining SRS resistance using different approaches. First, we determined the heritability using pedigree data from a challenge test. Secondly, a genome-wide association analysis was performed following a complete transcriptomic profile of fish from genetically susceptible and resistant families within the challenge infection with P. salmonis. We found differentially expressed transcripts related to immune response, pathogen recognition, and several new pathways related to extracellular matrix remodelling and intracellular invasion. The resistant background showed a constrained inflammatory response, mediated by the Arp2/3 complex actin cytoskeleton remodelling polymerization pathway, probably leading to bacterial clearance. A series of biomarkers of SRS resistance, such as the beta-enolase (ENO-β), Tubulin G1 (TUBG1), Plasmin (PLG) and ARP2/3 Complex Subunit 4 (ARPC4) genes showed consistent overexpression in resistant individuals, showing promise as biomarkers for SRS resistance. All these results together with the differential expression of several long non-coding RNAs show the complexity of the host-pathogen interaction of S. salar and P. salmonis. These results provide valuable information on new models describing host-pathogen interaction and its role in SRS resistance.
Collapse
Affiliation(s)
- Victor Martinez
- FAVET-INBIOGEN, Faculty of Veterinary Sciences, University of Chile, Avda. Santa Rosa, 11735, Santiago, Chile.
| | - Phillip Dettleff
- FAVET-INBIOGEN, Faculty of Veterinary Sciences, University of Chile, Avda. Santa Rosa, 11735, Santiago, Chile
| | - Pedro Zamorano
- Cell and Molecular Biology-Genetics Unit, University of Antofagasta, Antofagasta, Chile
| | - Nicolás Galarce
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, 8370146, Chile
| | - Consuelo Borie
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, 8370146, Chile
| | - Kerry Naish
- School of Aquatic and Fishery Sciences, University of Washington, Seattle, United States
| |
Collapse
|
25
|
Yang T, Yu J, Ahmed T, Nguyen K, Nie F, Zan R, Li Z, Han P, Shen H, Zhang X, Takayama S, Song Y. Synthetic neutrophil extracellular traps dissect bactericidal contribution of NETs under regulation of α-1-antitrypsin. SCIENCE ADVANCES 2023; 9:eadf2445. [PMID: 37115934 PMCID: PMC10146876 DOI: 10.1126/sciadv.adf2445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/30/2023] [Indexed: 05/03/2023]
Abstract
Deciphering the complex interplay of neutrophil extracellular traps (NETs) with the surrounding environment is a challenge with notable clinical implications. To bridge the gap in knowledge, we report our findings on the antibacterial activity against Pseudomonas aeruginosa of synthetic NET-mimetic materials composed of nanofibrillated DNA-protein complexes. Our synthetic system makes component-by-component bottom-up analysis of NET protein effects possible. When the antimicrobial enzyme neutrophil elastase (NE) is incorporated into the bactericidal DNA-histone complexes, the resulting synthetic NET-like structure exhibits an unexpected reduction in antimicrobial activity. This critical immune function is rescued upon treatment with alpha-1-antitrypsin (AAT), a physiological tissue-protective protease inhibitor. This suggests a direct causal link between AAT inhibition of NE and preservation of histone-mediated antimicrobial activity. These results help better understand the complex and, at times, contradictory observations of in vivo antimicrobial effects of NETs and AAT by excluding neutrophil, cytokine, and chemoattractant contributions.
Collapse
Affiliation(s)
- Ting Yang
- School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| | - Jinlong Yu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Tasdiq Ahmed
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| | - Katherine Nguyen
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| | - Fang Nie
- Renji Hospital affiliated to Shanghai Jiao Tong University, Shanghai 200127, China
| | - Rui Zan
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai 200032, China
| | - Zhiwei Li
- Renji Hospital affiliated to Shanghai Jiao Tong University, Shanghai 200127, China
| | - Pei Han
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Shen
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xiaonong Zhang
- School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai 200032, China
| | - Shuichi Takayama
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA 30332, USA
| | - Yang Song
- School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
26
|
He S, Yang Z, Li X, Wu H, Zhang L, Shan A, Wang J. Boosting stability and therapeutic potential of proteolysis-resistant antimicrobial peptides by end-tagging β-naphthylalanine. Acta Biomater 2023; 164:175-194. [PMID: 37100185 DOI: 10.1016/j.actbio.2023.04.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023]
Abstract
Recently, much emphasis has been placed on solving the intrinsic defects of antimicrobial peptides (AMPs), especially their susceptibility to protease digestion for the systemic application of antibacterial biomaterials. Although many strategies have increased the protease stability of AMPs, antimicrobial activity was severely compromised, thereby substantially weakening their therapeutic effect. To address this issue, we introduced hydrophobic group modifications at the N-terminus of proteolysis-resistant AMPs D1 (AArIIlrWrFR) through end-tagging with stretches of natural amino acids (W and I), unnatural amino acid (Nal) and fatty acids. Of these peptides, N1 tagged with a Nal at N-terminus showed the highest selectivity index (GMSI = 19.59), with a 6.73-fold improvement over D1. In addition to potent broad-spectrum antimicrobial activity, N1 also exhibited high antimicrobial stability toward salts, serum and proteases in vitro and ideal biocompatibility and therapeutic efficacy in vivo. Furthermore, N1 killed bacteria through multiple mechanisms, involving disruption of bacterial membranes and inhibition of bacterial energy metabolism. Indeed, appropriate terminal hydrophobicity modification opens up new avenues for developing and applying high-stability peptide-based antibacterial biomaterials. STATEMENT OF SIGNIFICANCE: To improve the potency and stability of proteolysis-resistant antimicrobial peptides (AMPs) without increasing toxicity, we constructed a convenient and tunable platform based on different compositions and lengths of hydrophobic end modifications. By tagging an Nal at the N-terminal, the obtained target compound N1 exhibited strong antimicrobial activity and desirable stability under multifarious environments in vitro (protease, salts and serum), and also showed favorable biocompatibility and therapeutic efficacy in vivo. Notably, N1exerted its bactericidal effect by damaging bacterial cell membranes and inhibiting bacterial energy metabolism in a dual mode. The findings provide a potential method for designing or optimizing proteolysis-resistant AMPs thus promoting the development and application of peptide-based antibacterial biomaterial.
Collapse
Affiliation(s)
- Shiqi He
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Zhanyi Yang
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xuefeng Li
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Hua Wu
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Licong Zhang
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Anshan Shan
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Jiajun Wang
- Animal Science and Technology College, Northeast Agricultural University, Harbin 150030, P. R. China.
| |
Collapse
|
27
|
Shen C, Guo Z, Liang H, Zhang M. Preliminary investigation of the immune activity of PmH2A-derived antimicrobial peptides from the pearl oyster Pinctada fucata martensii. FISH & SHELLFISH IMMUNOLOGY 2023; 135:108691. [PMID: 36924911 DOI: 10.1016/j.fsi.2023.108691] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 06/18/2023]
Abstract
Antimicrobial peptides (AMPs) play important roles in the immune defense against pathogenic microorganisms. For instance, histone 2A (H2A)-derived AMPs is an antimicrobial peptide involved in the host's innate immune defense and immunoregulation. AMPs have been isolated from the pearl oyster Pinctada fucata martensii but their role in host defense remains poorly understood. To elucidate the structural features of P. f. martensii H2A (PmH2A)-derived AMPs and their potential immune functions, we synthesized a series of laboratory-designed synthetic analogs of PmH2A and examined their antimicrobial properties, as well as their mechanisms of action. This analysis revealed inhibitory effects on the growth of Gram-positive and Gram-negative bacteria. Further assessment by transmission electron microscopy (TEM) of two of the three peptides, PmH2A-AMP and PmH2A-AMP(5-13)[KLLK]3, confirmed that it exerted an anti-bacterial activity through membrane lysis. Finally, we found that the hemocytes and gills of P. f. martensii released antimicrobial H2A histones in response to LPS exposure, mimicking tissue damage and infection. This immune response is reminiscent of the neutrophil extracellular traps (NETs) recently described in oysters. Thus, the LPS challenge is sufficient to induce histone-derived peptide accumulation in pearl oyster P.f. martensii.
Collapse
Affiliation(s)
- Chenghao Shen
- Fisheries College of Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Zhijie Guo
- Fisheries College of Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Haiying Liang
- Fisheries College of Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, Guangdong, 524088, China.
| | - Meizhen Zhang
- Fisheries College of Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| |
Collapse
|
28
|
Moreno J, Zoghebi K, Salehi D, Kim L, Shoushtari SK, Tiwari RK, Parang K. Amphiphilic Cell-Penetrating Peptides Containing Arginine and Hydrophobic Residues as Protein Delivery Agents. Pharmaceuticals (Basel) 2023; 16:469. [PMID: 36986567 PMCID: PMC10053436 DOI: 10.3390/ph16030469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
The entry of proteins through the cell membrane is challenging, thus limiting their use as potential therapeutics. Seven cell-penetrating peptides, designed in our laboratory, were evaluated for the delivery of proteins. Fmoc solid-phase peptide synthesis was utilized for the synthesis of seven cyclic or hybrid cyclic-linear amphiphilic peptides composed of hydrophobic (tryptophan (W) or 3,3-diphenylalanine (Dip) and positively-charged arginine (R) residues, such as [WR]4, [WR]9, [WWRR]4, [WWRR]5, [(RW)5K](RW)5, [R5K]W7, and [DipR]5. Confocal microscopy was used to screen the peptides as a protein delivery system of model cargo proteins, green and red fluorescein proteins (GFP and RFP). Based on the confocal microscopy results, [WR]9 and [DipR]5 were found to be more efficient among all the peptides and were selected for further studies. [WR]9 (1-10 µM) + protein (GFP and RFP) physical mixture did not show high cytotoxicity (>90% viability) in triple-negative breast cancer cells (MDA-MB-231) after 24 h, while [DipR]5 (1-10 µM) physical mixture with GFP exhibited more than 81% cell viability. Confocal microscopy images revealed internalization of GFP and RFP in MDA-MB-231 cells using [WR]9 (2-10 μM) and [DipR]5 (1-10 µM). Fluorescence-activated cell sorting (FACS) analysis indicated that the cellular uptake of GFP was concentration-dependent in the presence of [WR]9 in MDA-MB-231 cells after 3 h of incubation at 37 °C. The concentration-dependent uptake of GFP and RFP was also observed in the presence of [DipR5] in SK-OV-3 and MDA-MB-231 cells after 3 h of incubation at 37 °C. FACS analysis indicated that the cellular uptake of GFP in the presence of [WR]9 was partially decreased by methyl-β-cyclodextrin and nystatin as endocytosis inhibitors after 3 h of incubation in MDA-MB-231 cells, whereas nystatin and chlorpromazine as endocytosis inhibitors slightly reduced the uptake of GFP in the presence of [DipR]5 after 3 h of incubation in MDA-MB-231. [WR]9 was able to deliver therapeutically relevant proteins (Histone H2A) at different concentrations. These results provide insight into the use of amphiphilic cyclic peptides in the delivery of protein-related therapeutics.
Collapse
Affiliation(s)
- Jonathan Moreno
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Khalid Zoghebi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 82826, Saudi Arabia
| | - David Salehi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Lois Kim
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Sorour Khayyatnejad Shoushtari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Rakesh K. Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| |
Collapse
|
29
|
Yang Z, Wei Y, Wu W, Zhang L, Wang J, Shan A. Characterization of simplified nonapeptides with broad-spectrum antimicrobial activities as potential food preservatives, and their antibacterial mechanism. Food Funct 2023; 14:3139-3154. [PMID: 36892465 DOI: 10.1039/d2fo03861g] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Antimicrobial peptides (AMPs) have attracted attention in the field of food preservatives due to their favorable biosafety and potential antimicrobial activity. However, high synthetic cost, systemic toxicity, a narrow antimicrobial spectrum, and poor antimicrobial activity become the main bottlenecks for their practical applications. To address these questions, a set of derived nonapeptides were designed based on a previously discovered ultra-short peptide sequence template (RXRXRXRXL-NH2) and screened to identify an optimal peptide-based food preservative with excellent antimicrobial properties. Among these nonapeptides, the designed peptides 3IW (RIRIRIRWL-NH2) and W2IW (RWRIRIRWL-NH2) presented a membrane-disruptive and reactive oxygen species (ROS) accumulation mechanism to execute potent and rapid broad-spectrum antimicrobial activity without observed cytotoxicity. Moreover, they exhibited favorable antimicrobial stability regardless of high ionic strength, heat, and excessive acid-base conditions, retaining potent antimicrobial effects for chicken meat preservation. Collectively, their ultra-short sequence length and potent broad-spectrum antimicrobial capacity may be beneficial for the further development of green and safe peptide-based food preservatives.
Collapse
Affiliation(s)
- Zhanyi Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Yingxin Wei
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Wanpeng Wu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Jiajun Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
30
|
Si Z, Pethe K, Chan-Park MB. Chemical Basis of Combination Therapy to Combat Antibiotic Resistance. JACS AU 2023; 3:276-292. [PMID: 36873689 PMCID: PMC9975838 DOI: 10.1021/jacsau.2c00532] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 06/10/2023]
Abstract
The antimicrobial resistance crisis is a global health issue requiring discovery and development of novel therapeutics. However, conventional screening of natural products or synthetic chemical libraries is uncertain. Combination therapy using approved antibiotics with inhibitors targeting innate resistance mechanisms provides an alternative strategy to develop potent therapeutics. This review discusses the chemical structures of effective β-lactamase inhibitors, outer membrane permeabilizers, and efflux pump inhibitors that act as adjuvant molecules of classical antibiotics. Rational design of the chemical structures of adjuvants will provide methods to impart or restore efficacy to classical antibiotics for inherently antibiotic-resistant bacteria. As many bacteria have multiple resistance pathways, adjuvant molecules simultaneously targeting multiple pathways are promising approaches to combat multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Zhangyong Si
- School
of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459
| | - Kevin Pethe
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 636921
- Singapore
Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551
| | - Mary B. Chan-Park
- School
of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459
- Lee
Kong Chian School of Medicine, Nanyang Technological
University, Singapore 636921
| |
Collapse
|
31
|
Shen C, Liang H, Guo Z, Zhang M. Members of the histone-derived antimicrobial peptide family from the pearl oyster Pinctada fucata martensii: Inhibition of bacterial growth. FISH & SHELLFISH IMMUNOLOGY 2023; 132:108439. [PMID: 36423807 DOI: 10.1016/j.fsi.2022.11.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 06/16/2023]
Abstract
Because it is difficult to isolate standard antimicrobial peptides (AMPs) using traditional biochemical approaches, we designed, synthesized, and evaluated a series of structurally altered histone-derived AMPs (HDAPs) from the pearl oyster Pinctada fucata martensii using molecular cloning approaches. Four histone-homolog genes (PmH2A, PmH2B, PmH3, and PmH4-1) were identified, of which PmH2A and PmH2B had yet to be described. PmH2A and PmH2B were therefore cloned using Rapid Amplification of cDNA Ends (RACE) and characterized. Constitutive PmH2A and PmH2B mRNA expression was detected in all six pearl oyster tissues tested, with comparatively greater transcript abundance in the gonads. Because α-helical content, hydrophilicity index, and the presence of a proline hinge may be the three important factors influencing the antimicrobial efficacy of HDAPs, we synthesized a series of eight N- and C-terminally truncated or amino acid-substituted synthetic candidate HDAP analogs derived from PmH2A, PmH2B, PmH3, and PmH4-1. Only the PmH2A- and PmH4-derived AMPs inhibited bacterial growth. The PmH2A-derived AMPs were α-helical proteins, while the PmH4-derived AMPs were extended strand/random coil proteins. Our results suggested that having an α-helical structure was particularly important for the antibacterial efficacy of the PmH2A-derived peptides; amphipathic structures (hydrophilic index, 0.3 to -0.3) may enhance the antimicrobial function of both the PmH2A- and PmH4-derived peptides. The high antibacterial efficacy of one of the HDAP analogs studied, PmH2A-AMP (5-13) [KLLK]3, indicated that this protein may represent a promising candidate for the treatment of bacterial infections in aquaculture mollusk species. This first study of HDAPs from the pearl oyster P. f. martensii provides new insights into the design and function of highly effective antimicrobial peptides.
Collapse
Affiliation(s)
- Chenghao Shen
- Fisheries College of Guangdong Ocean University, Zhanjiang, Guangdong, 524088, PR China
| | - Haiying Liang
- Fisheries College of Guangdong Ocean University, Zhanjiang, Guangdong, 524088, PR China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, Guangdong, 524088, PR China.
| | - Zhijie Guo
- Fisheries College of Guangdong Ocean University, Zhanjiang, Guangdong, 524088, PR China
| | - Meizhen Zhang
- Fisheries College of Guangdong Ocean University, Zhanjiang, Guangdong, 524088, PR China
| |
Collapse
|
32
|
Li X, Ye Y, Peng K, Zeng Z, Chen L, Zeng Y. Histones: The critical players in innate immunity. Front Immunol 2022; 13:1030610. [PMID: 36479112 PMCID: PMC9720293 DOI: 10.3389/fimmu.2022.1030610] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
The highly conserved histones in different species seem to represent a very ancient and universal innate host defense system against microorganisms in the biological world. Histones are the essential part of nuclear matter and act as a control switch for DNA transcription. However, histones are also found in the cytoplasm, cell membranes, and extracellular fluid, where they function as host defenses and promote inflammatory responses. In some cases, extracellular histones can act as damage-associated molecular patterns (DAMPs) and bind to pattern recognition receptors (PRRs), thereby triggering innate immune responses and causing initial organ damage. Histones and their fragments serve as antimicrobial peptides (AMPs) to directly eliminate bacteria, viruses, fungi, and parasites in vitro and in vivo. Histones are also involved in phagocytes-related innate immune response as components of neutrophil extracellular traps (NETs), neutrophil activators, and plasminogen receptors. In addition, as a considerable part of epigenetic regulation, histone modifications play a vital role in regulating the innate immune response and expression of corresponding defense genes. Here, we review the regulatory role of histones in innate immune response, which provides a new strategy for the development of antibiotics and the use of histones as therapeutic targets for inflammatory diseases, sepsis, autoimmune diseases, and COVID-19.
Collapse
Affiliation(s)
- Xia Li
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Youyuan Ye
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Kailan Peng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhuo Zeng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Li Chen
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Yanhua Zeng
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China,Department of Dermatology and Venereology, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China,*Correspondence: Yanhua Zeng, ;
| |
Collapse
|
33
|
Ji X, Han L, Zhang W, Sun L, Xu S, Qiu X, Fan S, Li Z. Molecular, cellular and neurological consequences of infection by the neglected human pathogen Nocardia. BMC Biol 2022; 20:251. [PMID: 36352407 PMCID: PMC9647956 DOI: 10.1186/s12915-022-01452-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/29/2022] [Indexed: 11/10/2022] Open
Abstract
Background Nocardia is a facultative intracellular pathogen that infects the lungs and brains of immunocompromised patients with consequences that can be fatal. The incidence of such infections is rising, immunocompetent individuals are also being infected, and there is a need to learn more about this neglected bacterial pathogen and the interaction with its human host. Results We have applied dual RNA-seq to assess the global transcriptome changes that occur simultaneously in Nocardia farcinica (N. farcinica) and infected human epithelial alveolar host cells, and have tested a series of mutants in this in vitro system to identify candidate determinants of virulence. Using a mouse model, we revealed the profiles of inflammation-related factors in the lung after intranasal infection and confirmed that nbtB and nbtS are key virulence genes for Nocardia infection in vivo. Regarding the host response to infection, we found that the expression of many histones was dysregulated during the infection of lung cells, indicating that epigenetic modification might play a crucial role in the host during Nocardia infection. In our mouse model, Nocardia infection led to neurological symptoms and we found that 15 of 22 Nocardia clinical strains tested could cause obvious PD-like symptoms. Further experiments indicated that Nocardia infection could activate microglia and drive M1 microglial polarization, promote iNOS and CXCL-10 production, and cause neuroinflammation in the substantia nigra, all of which may be involved in causing PD-like symptoms. Importantly, the deletion of nbtS in N. farcinica completely attenuated the neurological symptoms. Conclusions Our data contribute to an in-depth understanding of the characteristics of both the host and Nocardia during infection and provide valuable clues for future studies of this neglected human pathogen, especially those addressing the underlying causes of infection-related neurological symptoms. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01452-7.
Collapse
|
34
|
Chen X, Han J, Cai X, Wang S. Antimicrobial peptides: Sustainable application informed by evolutionary constraints. Biotechnol Adv 2022; 60:108012. [PMID: 35752270 DOI: 10.1016/j.biotechadv.2022.108012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/02/2022] [Accepted: 06/19/2022] [Indexed: 01/10/2023]
Abstract
The proliferation and global expansion of multidrug-resistant (MDR) bacteria have deepened the need to develop novel antimicrobials. Antimicrobial peptides (AMPs) are regarded as promising antibacterial agents because of their broad-spectrum antibacterial activity and multifaceted mechanisms of action with non-specific targets. However, if AMPs are to be applied sustainably, knowledge of how they induce resistance in pathogenic bacteria must be mastered to avoid repeating the traditional antibiotic resistance mistakes currently faced. Furthermore, the evolutionary constraints on the acquisition of AMP resistance by microorganisms in the natural environment, such as functional compatibility and fitness trade-offs, inform the translational application of AMPs. Consequently, the shortcut to achieve sustainable utilization of AMPs is to uncover the evolutionary constraints of bacteria on AMP resistance in nature and find the tricks to exploit these constraints, such as applying AMP cocktails to minimize the efficacy of selection for resistance or combining nanomaterials to maximize the costs of AMP resistance. Altogether, this review dissects the benefits, challenges, and opportunities of utilizing AMPs against disease-causing bacteria, and highlights the use of AMP cocktails or nanomaterials to proactively address potential AMP resistance crises in the future.
Collapse
Affiliation(s)
- Xuan Chen
- College of Chemical Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Jinzhi Han
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Xixi Cai
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Shaoyun Wang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
35
|
Xia M, Xu F, Ni H, Wang Q, Zhang R, Lou Y, Zhou J. Neutrophil activation and NETosis are the predominant drivers of airway inflammation in an OVA/CFA/LPS induced murine model. Respir Res 2022; 23:289. [PMID: 36271366 PMCID: PMC9587569 DOI: 10.1186/s12931-022-02209-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/03/2022] [Indexed: 12/03/2022] Open
Abstract
Background Asthma is one of the most common chronic diseases that affects more than 300 million people worldwide. Though most asthma can be well controlled, individuals with severe asthma experience recurrent exacerbations and impose a substantial economic burden on healthcare system. Neutrophil inflammation often occurs in patients with severe asthma who have poor response to glucocorticoids, increasing the difficulty of clinical treatment. Methods We established several neutrophil-dominated allergic asthma mouse models, and analyzed the airway hyperresponsiveness, airway inflammation and lung pathological changes. Neutrophil extracellular traps (NETs) formation was analyzed using confocal microscopy and western blot. Results We found that the ovalbumin (OVA)/complete Freund’s adjuvant (CFA)/low-dose lipopolysaccharide (LPS)-induced mouse model best recapitulated the complex alterations in the airways of human severe asthmatic patients. We also observed OVA/CFA/LPS-exposed mice produced large quantities of neutrophil extracellular traps (NETs) in lung tissue and bone marrow neutrophils. Furthermore, we found that reducing the production of NETs or increasing the degradation of NETs can reduce airway inflammation and airway hyperresponsiveness. Conclusion Our findings identify a novel mouse model of neutrophilic asthma. We have also identified NETs play a significant role in neutrophilic asthma models and contribute to neutrophilic asthma pathogenesis. NETs may serve as a promising therapeutic target for neutrophilic asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02209-0.
Collapse
Affiliation(s)
- Mengling Xia
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qingchun Road, 310003, Hangzhou, China
| | - Fei Xu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qingchun Road, 310003, Hangzhou, China
| | - Hangqi Ni
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qingchun Road, 310003, Hangzhou, China
| | - Qing Wang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qingchun Road, 310003, Hangzhou, China
| | - Ruhui Zhang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qingchun Road, 310003, Hangzhou, China
| | - Yafang Lou
- Department of Respiratory Medicine, Hangzhou Hospital of Traditional Chinese Medicine, No. 453, Tiyuchang Road, 310013, Hangzhou, China.
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qingchun Road, 310003, Hangzhou, China.
| |
Collapse
|
36
|
He S, Yang Z, Li X, Wu H, Zhang L, Wang J, Shan A. Optimized proteolytic resistance motif (DabW)-based U1-2WD: A membrane-induced self-aggregating peptide to trigger bacterial agglutination and death. Acta Biomater 2022; 153:540-556. [PMID: 36162762 DOI: 10.1016/j.actbio.2022.09.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 12/13/2022]
Abstract
The biggest application bottleneck of antimicrobial peptides (AMPs) is the low oral bioavailability caused by the poor stability of digestive enzymes in the gastrointestinal tract. However, the research methods and evaluation criteria of available studies about anti-proteolytic strategies are not uniform and far from the actual environment in vivo. Here, we developed a research system and evaluation criteria for proteolytic resistance and systematically evaluated the effectiveness of different strategies for improving the protease stability of AMPs on the same platform for the first time. After a comprehensive analysis, Dab modification is identified as the most effective strategy to improve the trypsin stability of AMPs. By further modulating the proteolytic resistance optimization motif (DabW)n, U1-2WD is obtained with ideal stability and antimicrobial properties in vivo and in vitro. Notably, U1-2WD has a unique antibacterial mechanism, which forms amorphous aggregates in the bacteria environment to trigger the agglutination of bacterial cells to prevent bacterial escape. It then kills bacteria by disrupting bacterial membranes and inhibiting bacterial energy metabolism. Overall, our work has led to a new understanding of the effectiveness of proteolytic resistance strategies and accelerated the development of anti-proteolytic AMPs to combat multidrug-resistant bacterial infections. STATEMENT OF SIGNIFICANCE: We developed research system and evaluation criteria for proteolytic resistance and systematically evaluated the effectiveness of different strategies for improving protease stability of AMPs on the same platform for the first time. we found effective strategies to resist trypsin hydrolysis: modification with backbone (β-Arg), D-enantiomer (D-Arg) and L-2,4-diaminobutanoic acid (Dab). Further, the proteolytic resistance optimization motif (DabW)n was designed. When n=3, derivative U1-2WD was obtained with desirable stability and antimicrobial properties in vivo and in vitro. Notably, U1-2WD has a unique antibacterial mechanism, which can self-aggregate into amorphous aggregates in the bacteria environment to mediate the agglutination and sedimentation of bacterial cells to prevent bacterial escape, and then kill bacteria by destroying bacterial membranes and inhibiting bacterial energy metabolism.
Collapse
Affiliation(s)
- Shiqi He
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Zhanyi Yang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Xuefeng Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Hua Wu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Licong Zhang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Jiajun Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China.
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, P. R. China.
| |
Collapse
|
37
|
Johnstone KF, Herzberg MC. Antimicrobial peptides: Defending the mucosal epithelial barrier. FRONTIERS IN ORAL HEALTH 2022; 3:958480. [PMID: 35979535 PMCID: PMC9376388 DOI: 10.3389/froh.2022.958480] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
The recent epidemic caused by aerosolized SARS-CoV-2 virus illustrates the importance and vulnerability of the mucosal epithelial barrier against infection. Antimicrobial proteins and peptides (AMPs) are key to the epithelial barrier, providing immunity against microbes. In primitive life forms, AMPs protect the integument and the gut against pathogenic microbes. AMPs have also evolved in humans and other mammals to enhance newer, complex innate and adaptive immunity to favor the persistence of commensals over pathogenic microbes. The canonical AMPs are helictical peptides that form lethal pores in microbial membranes. In higher life forms, this type of AMP is exemplified by the defensin family of AMPs. In epithelial tissues, defensins, and calprotectin (complex of S100A8 and S100A9) have evolved to work cooperatively. The mechanisms of action differ. Unlike defensins, calprotectin sequesters essential trace metals from microbes, which inhibits growth. This review focuses on defensins and calprotectin as AMPs that appear to work cooperatively to fortify the epithelial barrier against infection. The antimicrobial spectrum is broad with overlap between the two AMPs. In mice, experimental models highlight the contribution of both AMPs to candidiasis as a fungal infection and periodontitis resulting from bacterial dysbiosis. These AMPs appear to contribute to innate immunity in humans, protecting the commensal microflora and restricting the emergence of pathobionts and pathogens. A striking example in human innate immunity is that elevated serum calprotectin protects against neonatal sepsis. Calprotectin is also remarkable because of functional differences when localized in epithelial and neutrophil cytoplasm or released into the extracellular environment. In the cytoplasm, calprotectin appears to protect against invasive pathogens. Extracellularly, calprotectin can engage pathogen-recognition receptors to activate innate immune and proinflammatory mechanisms. In inflamed epithelial and other tissue spaces, calprotectin, DNA, and histones are released from degranulated neutrophils to form insoluble antimicrobial barriers termed neutrophil extracellular traps. Hence, calprotectin and other AMPs use several strategies to provide microbial control and stimulate innate immunity.
Collapse
Affiliation(s)
| | - Mark C. Herzberg
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
38
|
Sánchez-Álvarez M, del Pozo MÁ, Bosch M, Pol A. Insights Into the Biogenesis and Emerging Functions of Lipid Droplets From Unbiased Molecular Profiling Approaches. Front Cell Dev Biol 2022; 10:901321. [PMID: 35756995 PMCID: PMC9213792 DOI: 10.3389/fcell.2022.901321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/17/2022] [Indexed: 11/30/2022] Open
Abstract
Lipid droplets (LDs) are spherical, single sheet phospholipid-bound organelles that store neutral lipids in all eukaryotes and some prokaryotes. Initially conceived as relatively inert depots for energy and lipid precursors, these highly dynamic structures play active roles in homeostatic functions beyond metabolism, such as proteostasis and protein turnover, innate immunity and defense. A major share of the knowledge behind this paradigm shift has been enabled by the use of systematic molecular profiling approaches, capable of revealing and describing these non-intuitive systems-level relationships. Here, we discuss these advances and some of the challenges they entail, and highlight standing questions in the field.
Collapse
Affiliation(s)
- Miguel Sánchez-Álvarez
- Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Ángel del Pozo
- Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
39
|
Sultana MF, Suzuki M, Yamasaki F, Kubota W, Takahashi K, Abo H, Kawashima H. Identification of Crucial Amino Acid Residues for Antimicrobial Activity of Angiogenin 4 and Its Modulation of Gut Microbiota in Mice. Front Microbiol 2022; 13:900948. [PMID: 35733962 PMCID: PMC9207454 DOI: 10.3389/fmicb.2022.900948] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/09/2022] [Indexed: 01/15/2023] Open
Abstract
Angiogenin 4 bearing ribonuclease activity is an endogenous antimicrobial protein expressed in small and large intestine. However, the crucial amino acid residues responsible for the antibacterial activity of Ang4 and its impact on gut microbiota remain unknown. Here, we report the contribution of critical amino acid residues in the functional regions of Ang4 to its activity against Salmonella typhimurium LT2 and the effect of Ang4 on gut microbiota in mice. We found that Ang4 binds S. typhimurium LT2 through two consecutive basic amino acid residues, K58 and K59, in the cell-binding segment and disrupts the bacterial membrane integrity at the N-terminal α-helix containing residues K7 and K30, as evidenced by the specific mutations of cationic residues of Ang4. We also found that the RNase activity of Ang4 was not involved in its bactericidal activity, as shown by the H12 mutant, which lacks RNase activity. In vivo administration of Ang4 through the mouse rectum and subsequent bacterial 16S rRNA gene sequencing analyses demonstrated that administration of Ang4 not only increased beneficial bacteria such as Lactobacillus, Akkermansia, Dubosiella, Coriobacteriaceae UCG-002, and Adlercreutzia, but also decreased certain pathogenic bacteria, including Alistipes and Enterohabdus, indicating that Ang4 regulates the shape of gut microbiota composition. We conclude that Ang4 kills bacteria by disrupting bacterial membrane integrity through critical basic amino acid residues with different functionalities rather than overall electrostatic interactions and potentially maintains gut microflora in vivo under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Mst. Farzana Sultana
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Maki Suzuki
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Fumiya Yamasaki
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Wataru Kubota
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Kohta Takahashi
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hirohito Abo
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroto Kawashima
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
40
|
Greve JM, Cowan JA. Tackling antimicrobial stewardship through synergy and antimicrobial peptides. RSC Med Chem 2022; 13:511-521. [PMID: 35694695 PMCID: PMC9132191 DOI: 10.1039/d2md00048b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/04/2022] [Indexed: 11/21/2022] Open
Abstract
The unrestricted use of antibiotics has led to rapid development of antibiotic resistance (AR) and renewed calls to address this serious problem. This review summarizes the most common mechanisms of antibiotic action, and in turn antibiotic resistance, as well as pathways to mitigate the harm. Focus is then turned to emerging antibiotic strategies, including antimicrobial peptides (AMPs), with a discussion of their modes of action, biochemical features, and potential challenges for their use as antibiotics. The role of synergy in antimicrobials is also examined, with a focus on the synergy of AMPs and other emerging interactions with synergistic potential.
Collapse
Affiliation(s)
- Jenna M Greve
- Department of Chemistry and Biochemistry, The Ohio State University 100 West 18th Avenue Columbus Ohio 43210 USA +1 614 292 2703
| | - James A Cowan
- Department of Chemistry and Biochemistry, The Ohio State University 100 West 18th Avenue Columbus Ohio 43210 USA +1 614 292 2703
| |
Collapse
|
41
|
Qu X, Wang M, Wang M, Tang H, Zhang S, Yang H, Yuan W, Wang Y, Yang J, Yue B. Multi-Mode Antibacterial Strategies Enabled by Gene-Transfection and Immunomodulatory Nanoparticles in 3D-Printed Scaffolds for Synergistic Exogenous and Endogenous Treatment of Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2200096. [PMID: 35267223 DOI: 10.1002/adma.202200096] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/24/2022] [Indexed: 06/14/2023]
Abstract
As research on refractory Staphylococcus aureus-related implant infection intensifies, certain challenges remain, including low antibiotic concentrations within infected areas, immune escape achieved by intracellular bacteria, myeloid-derived suppressor cells (MDSCs) inducing regional immunosuppression, and recurrence of residual pathogenic bacteria after drug suspension. Herein, a novel antimicrobial system to simultaneously address these issues is proposed. Specifically, an oxygen-species-responsive 3D-printed scaffold with shell-core nanoparticles is designed, which are loaded with an antimicrobial peptide plasmid (LL37 plasmid) and have LL37 grafted on their surface (LL37@ZIF8-LL37). The surface-grafted LL37 directly kills S. aureus and, following entry into cells, the nanoparticles kill intracellular bacteria. Moreover, in vitro and in vivo, following translation of the LL37 plasmid, cells function as factories of the antimicrobial peptide, thereby generating a continuous, prolonged antibacterial effect at the site of infection. This system significantly reduces the abnormal increase in MDSCs within the infected microenvironment, thus relieving the immunosuppressive state and restoring a protective antimicrobial immune response. Hence, this proposed antimicrobial system provides an antimicrobial immune response and a novel strategy for S. aureus-related infections by offering a combined active antimicrobial and immunotherapeutic strategy, thereby significantly reducing the recurrence rate following recovery from implant-associated infections.
Collapse
Affiliation(s)
- Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Miaochen Wang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, P. R. China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Hongtao Yang
- School of Medical Science and Engineering, Beihang University, Beijing, 100191, P. R. China
| | - Weien Yuan
- Pharm-X Center, Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - You Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Jianping Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| |
Collapse
|
42
|
Bosch M, Pol A. Eukaryotic lipid droplets: metabolic hubs, and immune first responders. Trends Endocrinol Metab 2022; 33:218-229. [PMID: 35065875 DOI: 10.1016/j.tem.2021.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022]
Abstract
As major eukaryotic lipid storage organelles, lipid droplets (LDs) are metabolic hubs coordinating energy flux and building block distribution. Infectious pathogens often promote accumulation and physically interact with LDs. The most accepted view is that host LDs are hijacked by invaders to draw on nutrients for host colonisation. However, unique traits such as biogenesis plasticity, dynamic proteome, signalling capacity, and ability to interact with other organelles endow LDs with competencies to face complex biological challenges. Here, we focus on published data suggesting that LDs are not usurped organelles but innate immunity first responders. By comparison with analogous mechanisms activated on LDs in nutrient-poor environments, our review supports the hypothesis that host LDs actively participate in immunometabolism, immune signalling, and microbial killing.
Collapse
Affiliation(s)
- Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain.
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona
| |
Collapse
|
43
|
Howell M, Wenc AK, Donaghy CM, Wasche DV, Abissi I, Naing MD, Pierce S, Angeles-Boza AM. Exploring synergy and its role in antimicrobial peptide biology. Methods Enzymol 2022; 663:99-130. [PMID: 35168799 DOI: 10.1016/bs.mie.2021.09.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Antimicrobial peptides will be an essential component in combating the escalating issue of antibiotic resistance. Identifying synergistic combinations of two or more substances will increase the value of these peptides further. Several potential pitfalls in conducting synergy testing with peptides are discussed in detail. As case studies, we describe observations of AMP synergy with peptides, antibiotics, and metal ions as well as some of the mechanistic details that have been uncovered. The Bliss and Loewe models for synergy are presented prior to recommending protocols for conducting checkerboard, minimal inhibitory concentration, and time-kill assays. Establishing mechanisms of action and exploring the potential for resistance will be crucial to translate these studies into the clinic.
Collapse
Affiliation(s)
- Matthew Howell
- Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Antonina K Wenc
- Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Caroline M Donaghy
- Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Devon V Wasche
- Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Izabela Abissi
- Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Marvin D Naing
- Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Scott Pierce
- Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, United States
| | - Alfredo M Angeles-Boza
- Department of Chemistry and Institute of Materials Science, University of Connecticut, Storrs, CT, United States.
| |
Collapse
|
44
|
Duong L, Gross SP, Siryaporn A. Developing Antimicrobial Synergy With AMPs. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 3:640981. [PMID: 35047912 PMCID: PMC8757689 DOI: 10.3389/fmedt.2021.640981] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/12/2021] [Indexed: 12/03/2022] Open
Abstract
Antimicrobial peptides (AMPs) have been extensively studied due to their vast natural abundance and ability to kill microbes. In an era critically lacking in new antibiotics, manipulating AMPs for therapeutic application is a promising option. However, bacterial pathogens resistant to AMPs remain problematic. To improve AMPs antimicrobial efficacy, their use in conjunction with other antimicrobials has been proposed. How might this work? AMPs kill bacteria by forming pores in bacterial membranes or by inhibiting bacterial macromolecular functions. What remains unknown is the duration for which AMPs keep bacterial pores open, and the extent to which bacteria can recover by repairing these pores. In this mini-review, we discuss various antimicrobial synergies with AMPs. Such synergies might arise if the antimicrobial agents helped to keep bacterial pores open for longer periods of time, prevented pore repair, perturbed bacterial intracellular functions at greater levels, or performed other independent bacterial killing mechanisms. We first discuss combinations of AMPs, and then focus on histones, which have antimicrobial activity and co-localize with AMPs on lipid droplets and in neutrophil extracellular traps (NETs). Recent work has demonstrated that histones can enhance AMP-induced membrane permeation. It is possible that histones, histone fragments, and histone-like peptides could amplify the antimicrobial effects of AMPs, giving rise to antimicrobial synergy. If so, clarifying these mechanisms will thus improve our overall understanding of the antimicrobial processes and potentially contribute to improved drug design.
Collapse
Affiliation(s)
- Leora Duong
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Steven P Gross
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States.,Department of Physics & Astronomy, University of California, Irvine, Irvine, CA, United States
| | - Albert Siryaporn
- Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA, United States.,Department of Physics & Astronomy, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
45
|
Zhu X, Sokol CL. Caspase 8 sounds the alarm for allergic inflammation. J Allergy Clin Immunol 2021; 149:1218-1220. [PMID: 34699886 DOI: 10.1016/j.jaci.2021.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022]
Affiliation(s)
- Xueping Zhu
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Charlestown, Mass; Harvard Medical School, Boston, Mass
| | - Caroline L Sokol
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Charlestown, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
46
|
Gera S, Kankuri E, Kogermann K. Antimicrobial peptides - Unleashing their therapeutic potential using nanotechnology. Pharmacol Ther 2021; 232:107990. [PMID: 34592202 DOI: 10.1016/j.pharmthera.2021.107990] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023]
Abstract
Antimicrobial peptides (AMPs) are potent, mostly cationic, and amphiphilic broad-spectrum host defense antimicrobials that are produced by all organisms ranging from prokaryotes to humans. In addition to their antimicrobial actions, they modulate inflammatory and immune responses and promote wound healing. Although they have clear benefits over traditional antibiotic drugs, their wide therapeutic utilization is compromised by concerns of toxicity, stability, and production costs. Recent advances in nanotechnology have attracted increasing interest to unleash the AMPs' immense potential as broad-spectrum antibiotics and anti-biofilm agents, against which the bacteria have less chances to develop resistance. Topical application of AMPs promotes migration of keratinocytes and fibroblasts, and contributes significantly to an accelerated wound healing process. Delivery of AMPs by employing nanotechnological approaches avoids the major disadvantages of AMPs, such as instability and toxicity, and provides a controlled delivery profile together with prolonged activity. In this review, we provide an overview of the key properties of AMPs and discuss the latest developments in topical AMP therapy using nanocarriers. We use chronic hard-to-heal wounds-complicated by infections, inflammation, and stagnated healing-as an example of an unmet medical need for which the AMPs' wide range of therapeutic actions could provide the most potential benefit. The use of innovative materials and sophisticated nanotechnological approaches offering various possibilities are discussed in more depth.
Collapse
Affiliation(s)
- Sonia Gera
- Institute of Pharmacy, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland.
| | - Karin Kogermann
- Institute of Pharmacy, University of Tartu, Nooruse 1, 50411 Tartu, Estonia.
| |
Collapse
|
47
|
Shi Y, Zhou K, Li D, Guyonnet V, Hincke MT, Mine Y. Avian Eggshell Membrane as a Novel Biomaterial: A Review. Foods 2021; 10:foods10092178. [PMID: 34574286 PMCID: PMC8466381 DOI: 10.3390/foods10092178] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 12/20/2022] Open
Abstract
The eggshell membrane (ESM), mainly composed of collagen-like proteins, is readily available as a waste product of the egg industry. As a novel biomaterial, ESM is attractive for its applications in the nutraceutical, cosmetic, and pharmaceutical fields. This review provides the main information about the structure and chemical composition of the ESM as well as some approaches for its isolation and solubilization. In addition, the review focuses on the role and performance of bioactive ESM-derived products in various applications, while a detailed literature survey is provided. The evaluation of the safety of ESM is also summarized. Finally, new perspectives regarding the potential of ESM as a novel biomaterial in various engineering fields are discussed. This review provides promising future directions for comprehensive application of ESM.
Collapse
Affiliation(s)
- Yaning Shi
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (K.Z.); (D.L.)
- Correspondence: (Y.S.); (Y.M.)
| | - Kai Zhou
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (K.Z.); (D.L.)
| | - Dandan Li
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (K.Z.); (D.L.)
| | - Vincent Guyonnet
- FFI Consulting Ltd., 2488 Lyn Road, Brockville, ON K6V 5T3, Canada;
| | - Maxwell T. Hincke
- Department of Cellular and Molecular Medicine, University of Ottawa, 75 Laurier Ave. E, Ottawa, ON K1N 6N5, Canada;
| | - Yoshinori Mine
- Department of Food Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
- Correspondence: (Y.S.); (Y.M.)
| |
Collapse
|
48
|
Yang T, Yang S, Ahmed T, Nguyen K, Yu J, Cao X, Zan R, Zhang X, Shen H, Fay ME, Williams EK, Lam WA, VanEpps JS, Takayama S, Song Y. Dosage-dependent antimicrobial activity of DNA-histone microwebs against Staphylococcus aureus. ADVANCED MATERIALS INTERFACES 2021; 8:2100717. [PMID: 34540532 PMCID: PMC8447838 DOI: 10.1002/admi.202100717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Indexed: 05/15/2023]
Abstract
Neutrophil extracellular traps (NETs) is an antimicrobial cobweb-structured material produced by immune cells for clearance of pathogens in the body, but paradoxically associated with biofilm formation and exacerbated lung infections. To provide a better materials perspective on the pleiotropic roles played by NETs at diverse compositions/concentrations, a NETs-like material (called 'microwebs', abbreviated as μwebs) is synthesized for decoding the antimicrobial activity of NETs against Staphylococcus aureus in infection-relevant conditions. We show that μwebs composed of low-to-intermediate concentrations of DNA-histone complexes successfully trap and inhibit S. aureus growth and biofilm formation. However, with growing concentrations and histone proportions, the resulting microwebs appear gel-like structures accompanied by reduced antimicrobial activity that can even promote formation of S. aureus biofilms. Our simplified model of NETs provides a materials-based evidence on NETs-relevant pathology in the development of biofilms.
Collapse
Affiliation(s)
- Ting Yang
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Shi Yang
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tasdiq Ahmed
- Wallace H Coulter Department of Biomedical Engineering & Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology & Emory School of Medicine, Atlanta, GA 30332 USA
| | - Katherine Nguyen
- Wallace H Coulter Department of Biomedical Engineering & Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology & Emory School of Medicine, Atlanta, GA 30332 USA
| | - Jinlong Yu
- Department of Orthopedics, Shanghai No.6 People's hospital, Shanghai 200233, China
| | - Xuejun Cao
- State Key Laboratory of Bioreactor Engineering, Department of Bioengineering, East China University of Science and Technology, Shanghai 200237, China
| | - Rui Zan
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaonong Zhang
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Shen
- Department of Orthopedics, Shanghai No.6 People's hospital, Shanghai 200233, China
| | - Meredith E Fay
- Wallace H Coulter Department of Biomedical Engineering & Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology & Emory School of Medicine, Atlanta, GA 30332 USA
| | - Evelyn Kendall Williams
- Wallace H Coulter Department of Biomedical Engineering & Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology & Emory School of Medicine, Atlanta, GA 30332 USA
| | - Wilbur A Lam
- Wallace H Coulter Department of Biomedical Engineering & Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology & Emory School of Medicine, Atlanta, GA 30332 USA
| | - J Scott VanEpps
- Department of Emergency Medicine, Michigan Center for Integrative Research in Critical Care, Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Shuichi Takayama
- Wallace H Coulter Department of Biomedical Engineering & Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology & Emory School of Medicine, Atlanta, GA 30332 USA
| | - Yang Song
- State Key Laboratory of Metal Matrix Composites, School of Material Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
49
|
Bosch M, Sweet MJ, Parton RG, Pol A. Lipid droplets and the host-pathogen dynamic: FATal attraction? J Cell Biol 2021; 220:e202104005. [PMID: 34165498 PMCID: PMC8240858 DOI: 10.1083/jcb.202104005] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
In the ongoing conflict between eukaryotic cells and pathogens, lipid droplets (LDs) emerge as a choke point in the battle for nutrients. While many pathogens seek the lipids stored in LDs to fuel an expensive lifestyle, innate immunity rewires lipid metabolism and weaponizes LDs to defend cells and animals. Viruses, bacteria, and parasites directly and remotely manipulate LDs to obtain substrates for metabolic energy, replication compartments, assembly platforms, membrane blocks, and tools for host colonization and/or evasion such as anti-inflammatory mediators, lipoviroparticles, and even exosomes. Host LDs counterattack such advances by synthesizing bioactive lipids and toxic nucleotides, organizing immune signaling platforms, and recruiting a plethora of antimicrobial proteins to provide a front-line defense against the invader. Here, we review the current state of this conflict. We will discuss why, when, and how LDs efficiently coordinate and precisely execute a plethora of immune defenses. In the age of antimicrobial resistance and viral pandemics, understanding innate immune strategies developed by eukaryotic cells to fight and defeat dangerous microorganisms may inform future anti-infective strategies.
Collapse
Affiliation(s)
- Marta Bosch
- Lipid Trafficking and Disease Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Matthew J. Sweet
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| |
Collapse
|
50
|
Zada S, Lu H, Yang F, Zhang Y, Cheng Y, Tang S, Wei W, Qiao Y, Fu P, Dong H, Zhang X. V 2C Nanosheets as Dual-Functional Antibacterial Agents. ACS APPLIED BIO MATERIALS 2021; 4:4215-4223. [PMID: 35006834 DOI: 10.1021/acsabm.1c00008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Antibiotic-resistant bacterial strains have been continuously increasing and becoming a supreme threat to public health globally. The nanoparticle-based photothermal treatment has emerged as a powerful tool to combat toxic bacteria. Photothermal agents (PTAs) with cost-effective and high photothermal conversion efficiency are highly desirable. Herein, we unite the green process for delamination of V2AlC to produce a high yield mass of two-dimensional (2D) V2C nanosheets (NSs) by using algae extracts and demonstrate their high antibacterial efficiency. The resultant V2C NSs present decent structural reliability and intrinsic antibacterial ability. Powerful near-infrared (NIR) absorption and extraordinary photothermal conversion proficiency make it a good PTA for the photothermal treatment of bacteria. The antibacterial efficiency evaluation indicated that V2C NSs could effectively kill both Gram-positive S. aureus and Gram-negative E. coli. About 99.5% of both types of bacteria could be killed with low-dose of V2C NSs suspension (40 μg/mL) with 5 min NIR irradiation due to the intrinsic antibacterial ability and photothermal effect of V2C NSs, which is much higher than previous reports on Ta4C3, Ti3C2, MoSe2, and Nb2C. This work expands the application of MXene V2C NSs for rapid bacteria-killing and would gain promising attention for applications in the sterilization industry.
Collapse
Affiliation(s)
- Shah Zada
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Huiting Lu
- School of Chemistry and Biological Engineering, University of Science & Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Fan Yang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Yiyi Zhang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Yaru Cheng
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Songsong Tang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Wei Wei
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Yuchun Qiao
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
| | - Pengcheng Fu
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University 58 Renmin Avenue, Meilan District Haikou, Hainan 570228, China
| | - Haifeng Dong
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Centre, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Xueji Zhang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Research Centre for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing 100083, China
- Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Health Science Centre, Shenzhen University, Shenzhen, Guangdong 518060, China
| |
Collapse
|