1
|
Kim J, Hasan M, Liao X, Ding T, Ahn J. Combined antimicrobial activity of short peptide and phage-derived endolysin against antibiotic-resistant Salmonella Typhimurium. Food Microbiol 2025; 125:104642. [PMID: 39448152 DOI: 10.1016/j.fm.2024.104642] [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: 03/01/2024] [Revised: 07/05/2024] [Accepted: 09/08/2024] [Indexed: 10/26/2024]
Abstract
This study was designed to evaluate the combination effects of antimicrobial peptides (FK13 and FK16) and phage-encoded endolysin (LysPB32) on the inhibition of growth of polymyxin B-resistant Salmonella Typhimurium ATCC 19585 (STPMB). The inhibitory effects of FK13, FK16, and LysPB32 against STPMB were evaluated by using antimicrobial susceptibility, membrane permeability, biofilm reduction, cross-resistance, and mutant frequency assay. The minimum inhibitory concentrations (MICs) of FK13 and FK16 treated with LysPB32 (FK13+LysPB32 and FK16+LysPB32) against STPMB were decreased from more than 512 to 128 μg/ml and from 64 to 32 μg/ml, respectively. Compared to the control, the number of STPMB in the growing culture was reduced by 4.2 and 5.2 log CFU/ml, respectively, for FK13+LysPB32 and FK16+LysPB32 after 12-h incubation at 37 °C. All treatments (FK13, FK16, FK13+LysPB32, FK16+LysPB32) significantly increased the permeability of the outer membrane of STPMB. Biofilms were significantly decreased from OD600 of 0.6 to 0.16 for FK13+LysPB32 and from 0.6 to 0.13 for FK16+LysPB32. The ratios of MICs of erythromycin, ceftriaxone, polymyxin B, and ciprofloxacin to MIC of the control against STPMB were decreased to 0.50 for FK13+LysPB32 and FK16+LysPB32. The bactericidal activities of amikacin and gentamicin were enhanced for FK13+LysPB32 and FK16+LysPB32 (2-fold < MBC/MIC ratio). The mutant frequencies of STPMB to antibiotics were decreased when treated with FK13+LysPB32 and FK16+LysPB32. The results suggest that the combination of antimicrobial peptides and endolysins can be a promising strategy to control polymyxin B-resistant S. Typhimurium.
Collapse
Affiliation(s)
- Junhwan Kim
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Mahadi Hasan
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Xinyu Liao
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang, 314100, China
| | - Tian Ding
- Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang, 314100, China; Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Juhee Ahn
- Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea; Future Food Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, Zhejiang, 314100, China.
| |
Collapse
|
2
|
Singh AN, Wu M, Ye TT, Brown AC, Wittenberg NJ. Engineering Planar Gram-Negative Outer Membrane Mimics Using Bacterial Outer Membrane Vesicles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024. [PMID: 39453730 DOI: 10.1021/acs.langmuir.4c02632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Antibiotic resistance is a major challenge in modern medicine. The unique double membrane structure of Gram-negative bacteria limits the efficacy of many existing antibiotics and adds complexity to antibiotic development by limiting transport of antibiotics to the bacterial cytosol. New methods to mimic this barrier would enable high-throughput studies for antibiotic development. In this study, we introduce an innovative approach to modify outer membrane vesicles (OMVs) from Aggregatibacter actinomycetemcomitans, to generate planar supported lipid bilayer membranes. Our method first involves the incorporation of synthetic lipids into OMVs using a rapid freeze-thaw technique to form outer membrane hybrid vesicles (OM-Hybrids). Subsequently, these OM-Hybrids can spontaneously rupture when in contact with SiO2 surfaces to form a planar outer membrane supported bilayer (OM-SB). We assessed the formation of OM-Hybrids using dynamic light scattering and a fluorescence quenching assay. To analyze the formation of OM-SBs from OM-Hybrids we used quartz crystal microbalance with dissipation monitoring (QCM-D) and fluorescence recovery after photobleaching (FRAP). Additionally, we conducted assays to detect surface-associated DNA and proteins on OM-SBs. The interaction of an antimicrobial peptide, polymyxin B, with the OM-SBs was also assessed. These findings emphasize the capability of our platform to produce planar surfaces of bacterial outer membranes, which in turn, could function as a valuable tool for streamlining the development of antibiotics.
Collapse
Affiliation(s)
- Aarshi N Singh
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Meishan Wu
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Tiffany T Ye
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Angela C Brown
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Nathan J Wittenberg
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
3
|
Heffernan LM, Lawrence ALE, Marcotte HA, Sharma A, Jenkins AX, Iguwe D, Rood J, Herke SW, O'Riordan MX, Abuaita BH. Heterogeneity of Salmonella enterica lipopolysaccharide counteracts macrophage and antimicrobial peptide defenses. Infect Immun 2024; 92:e0025124. [PMID: 39225472 PMCID: PMC11475854 DOI: 10.1128/iai.00251-24] [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: 06/11/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Salmonella enterica is comprised of over 2,500 serovars, in which non-typhoidal serovars (NTS), Enteritidis (SE), and Typhimurium (STM) are the most clinically associated with human infections. Although NTS have similar genetic elements to cause disease, phenotypic variation including differences in lipopolysaccharide (LPS) composition may control immune evasion. Here, we demonstrate that macrophage host defenses and LL-37 antimicrobial efficacy against SE and STM are substantially altered by LPS heterogeneity. We found that SE evades macrophage killing by inhibiting phagocytosis while STM survives better intracellularly post-phagocytosis. SE-infected macrophages failed to activate the inflammasomes and subsequently produced less interleukin-1β (IL-1β), IL-18, and interferon λ. Inactivation of LPS biosynthesis genes altered LPS composition, and the SE LPS-altered mutants could no longer inhibit phagocytosis, inflammasome activation, and type II interferon signaling. In addition, SE and STM showed differential susceptibility to the antimicrobials LL-37 and colistin, and alteration of LPS structure substantially increased susceptibility to these molecules. Collectively, our findings highlight that modification of LPS composition by Salmonella increases resistance to host defenses and antibiotics.
Collapse
Affiliation(s)
- Linda M. Heffernan
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Anna-Lisa E. Lawrence
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Haley A. Marcotte
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Amit Sharma
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Aria X. Jenkins
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Damilola Iguwe
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Jennifer Rood
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Scott W. Herke
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Mary X. O'Riordan
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Basel H. Abuaita
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| |
Collapse
|
4
|
Zhao M, Zhang Y, Wang M, Ma LZ. dsAMP and dsAMPGAN: Deep Learning Networks for Antimicrobial Peptides Recognition and Generation. Antibiotics (Basel) 2024; 13:948. [PMID: 39452213 PMCID: PMC11504993 DOI: 10.3390/antibiotics13100948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
Antibiotic resistance is a growing public health challenge. Antimicrobial peptides (AMPs) effectively target microorganisms through non-specific mechanisms, limiting their ability to develop resistance. Therefore, the prediction and design of new AMPs is crucial. Recently, deep learning has spurred interest in computational approaches to peptide drug discovery. This study presents a novel deep learning framework for AMP classification, function prediction, and generation. We developed discoverAMP (dsAMP), a robust AMP predictor using CNN Attention BiLSTM and transfer learning, which outperforms existing classifiers. In addition, dsAMPGAN, a Generative Adversarial Network (GAN)-based model, generates new AMP candidates. Our results demonstrate the superior performance of dsAMP in terms of sensitivity, specificity, Matthew correlation coefficient, accuracy, precision, F1 score, and area under the ROC curve, achieving >95% classification accuracy with transfer learning on a small dataset. Furthermore, dsAMPGAN successfully synthesizes AMPs similar to natural ones, as confirmed by comparisons of physical and chemical properties. This model serves as a reliable tool for the identification of novel AMPs in clinical settings and supports the development of AMPs to effectively combat antibiotic resistance.
Collapse
Affiliation(s)
- Min Zhao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; (M.Z.); (Y.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; (M.Z.); (Y.Z.)
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Maolin Wang
- CAAC Key Laboratory of General Aviation Operation, Civil Aviation Management Institute of China, Beijing 100102, China
| | - Luyan Z. Ma
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; (M.Z.); (Y.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Min JH, Sarlus H, Harris RA. MAD-microbial (origin of) Alzheimer's disease hypothesis: from infection and the antimicrobial response to disruption of key copper-based systems. Front Neurosci 2024; 18:1467333. [PMID: 39416952 PMCID: PMC11480022 DOI: 10.3389/fnins.2024.1467333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Microbes have been suspected to cause Alzheimer's disease since at least 1908, but this has generally remained unpopular in comparison to the amyloid hypothesis and the dominance of Aβ and Tau. However, evidence has been accumulating to suggest that these earlier theories are but a manifestation of a common cause that can trigger and interact with all the major molecular players recognized in AD. Aβ, Tau and ApoE, in particular appear to be molecules with normal homeostatic functions but also with alternative antimicrobial functions. Their alternative functions confer the non-immune specialized neuron with some innate intracellular defenses that appear to be re-appropriated from their normal functions in times of need. Indeed, signs of infection of the neurons by biofilm-forming microbial colonies, in synergy with herpes viruses, are evident from the clinical and preclinical studies we discuss. Furthermore, we attempt to provide a mechanistic understanding of the AD landscape by discussing the antimicrobial effect of Aβ, Tau and ApoE and Lactoferrin in AD, and a possible mechanistic link with deficiency of vital copper-based systems. In particular, we focus on mitochondrial oxidative respiration via complex 4 and ceruloplasmin for iron homeostasis, and how this is similar and possibly central to neurodegenerative diseases in general. In the case of AD, we provide evidence for the microbial Alzheimer's disease (MAD) theory, namely that AD could in fact be caused by a long-term microbial exposure or even long-term infection of the neurons themselves that results in a costly prolonged antimicrobial response that disrupts copper-based systems that govern neurotransmission, iron homeostasis and respiration. Finally, we discuss potential treatment modalities based on this holistic understanding of AD that incorporates the many separate and seemingly conflicting theories. If the MAD theory is correct, then the reduction of microbial exposure through use of broad antimicrobial and anti-inflammatory treatments could potentially alleviate AD although this requires further clinical investigation.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital at Solna, Stockholm, Sweden
| | | | | |
Collapse
|
6
|
Ziu T, Sambur E, Ruzsics Z, Hengel H, Grabherr R, Höfinger S, Harant H. In Vitro Profiling of the Antiviral Peptide TAT-I24. Int J Mol Sci 2024; 25:10463. [PMID: 39408791 PMCID: PMC11477294 DOI: 10.3390/ijms251910463] [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: 09/10/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The synthetic peptide TAT-I24 (GRKKRRQRRRPPQCLAFYACFC) exerts antiviral activity against several double-stranded (ds) DNA viruses, including herpes simplex viruses, cytomegalovirus, some adenoviruses, vaccinia virus and SV40 polyomavirus. In the present study, in vitro profiling of this peptide was performed with the aim of characterizing and improving its properties for further development. As TAT-I24 contains three free cysteine residues, a potential disadvantageous feature, peptide variants with replacements or deletions of specific residues were generated and tested in various cell systems and by biochemical analyses. Some cysteine replacements had no impact on the antiviral activity, such as the deletion of cysteine 14, which also showed improved biochemical properties, while the cyclization of cysteines 14 and 20 had the most detrimental effect on antiviral activity. At concentrations below 20 µM, TAT-I24 and selected variants did not induce hemolysis in red blood cells (RBCs) nor modulated lipopolysaccharide (LPS)-induced release of cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), in human peripheral blood mononuclear cells (PBMCs). These data indicate that TAT-I24 or its peptide variants are not expected to cause unwanted effects on blood cells.
Collapse
Affiliation(s)
- Theodhora Ziu
- Pivaris BioScience GmbH, Media Quarter Marx 3.4, Maria-Jacobi-Gasse 1, 1030 Vienna, Austria;
| | - Ezgi Sambur
- VSC Research Center, Technical University of Vienna, Operngasse 11/E057-09, 1040 Vienna, Austria; (E.S.); or (S.H.)
| | - Zsolt Ruzsics
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Hermann-Herder-Str.11, 79104 Freiburg, Germany; (Z.R.); (H.H.)
| | - Hartmut Hengel
- Institute of Virology, Medical Center and Faculty of Medicine, University of Freiburg, Hermann-Herder-Str.11, 79104 Freiburg, Germany; (Z.R.); (H.H.)
| | - Reingard Grabherr
- Institute of Molecular Biotechnology, Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria;
| | - Siegfried Höfinger
- VSC Research Center, Technical University of Vienna, Operngasse 11/E057-09, 1040 Vienna, Austria; (E.S.); or (S.H.)
- Department of Physics, Michigan Technological University, Houghton, MI 49931, USA
| | - Hanna Harant
- Pivaris BioScience GmbH, Media Quarter Marx 3.4, Maria-Jacobi-Gasse 1, 1030 Vienna, Austria;
| |
Collapse
|
7
|
Kim E, Park Y, Yun M, Kim B. Functions of Hemp-Induced Exosomes against Periodontal Deterioration Caused by Fine Dust. Int J Mol Sci 2024; 25:10331. [PMID: 39408660 PMCID: PMC11477052 DOI: 10.3390/ijms251910331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Although fine dust is linked to numerous health issues, including cardiovascular, neurological, respiratory, and cancerous diseases, research on its effects on oral health remains limited. In this study, we investigated the protective effects of mature hemp stem extract-induced exosomes (MSEIEs) on periodontal cells exposed to fine dust. Using various methods, including microRNA profiling, PCR, flow cytometry, immunocytochemistry, ELISA, and Alizarin O staining, we found that MSE treatment upregulated key microRNAs, such as hsa-miR-122-5p, hsa-miR-1301-3p, and hsa-let-7e-5p, associated with vital biological functions. MSEIEs exhibited three primary protective functions: suppressing inflammatory genes while activating anti-inflammatory ones, promoting the differentiation of periodontal ligament stem cells (PDLSCs) into osteoblasts and other cells, and regulating LL-37 and MCP-1 expression. These findings suggest that MSEIEs have potential as functional biomaterials for applications in pharmaceuticals, cosmetics, and food industries.
Collapse
Affiliation(s)
- Eunhee Kim
- Department of Food Science and Biotechnology, Andong National University, Andong 36729, Republic of Korea;
| | - Yoonjin Park
- Department of Bio-Hemp Technology, Andong Science College, Andong 36616, Republic of Korea;
| | - Mihae Yun
- Department of Dental Hygiene, Andong Science College, Andong 36616, Republic of Korea
| | - Boyong Kim
- EVERBIO, 131, Jukhyeon-gil, Gwanghyewon-myeon, Jincheon-gun 27809, Republic of Korea
| |
Collapse
|
8
|
Pascal W, Gotowiec M, Smoliński A, Suchecki M, Kopka M, Pascal AM, Włodarski PK. Biologic Brachytherapy: Genetically Modified Surgical Flap as a Therapeutic Tool-A Systematic Review of Animal Studies. Int J Mol Sci 2024; 25:10330. [PMID: 39408659 PMCID: PMC11476562 DOI: 10.3390/ijms251910330] [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: 09/02/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Surgical flaps are rudimentary tools in reconstructive surgery, especially following extensive solid tumour resections. They cover skin and soft tissue defects but are prone to ischaemia and necrosis. Since their primary aim is reconstruction, they rarely exhibit a therapeutic activity against the treated disease. Attempts have been made to develop a new therapeutic strategy-biologic brachytherapy, which uses genetically engineered surgical flaps as a drug delivery vehicle, allowing the flap tissue to act as a "biologic pump". This systematic review summarizes the preclinical evidence on using genetically modified surgical flaps. A literature search was conducted in PubMed, EMBASE, Scopus and Web of Science. The initial literature search yielded 714 papers, and, eventually, seventy-seven studies were included in qualitative analysis. The results show that genetic enhancement of flaps has been used as a local or systemic therapy for numerous disease models. Frequently, it has been used to increase flap survival and limit ischaemia or promote flap survival in a non-ischemic context, with some studies focusing on optimizing the technique of such gene therapy. The results show that genetically modified flaps can be successfully used in a variety of contexts, but we need more studies to implement this research into specific clinical scenarios.
Collapse
Affiliation(s)
- Wiktor Pascal
- Department of Methodology, Medical University of Warsaw, 1b Banacha Street, 02-091 Warsaw, Poland; (M.G.); (A.S.); (M.S.); (M.K.); (A.M.P.); (P.K.W.)
| | - Mateusz Gotowiec
- Department of Methodology, Medical University of Warsaw, 1b Banacha Street, 02-091 Warsaw, Poland; (M.G.); (A.S.); (M.S.); (M.K.); (A.M.P.); (P.K.W.)
| | - Antoni Smoliński
- Department of Methodology, Medical University of Warsaw, 1b Banacha Street, 02-091 Warsaw, Poland; (M.G.); (A.S.); (M.S.); (M.K.); (A.M.P.); (P.K.W.)
| | - Michał Suchecki
- Department of Methodology, Medical University of Warsaw, 1b Banacha Street, 02-091 Warsaw, Poland; (M.G.); (A.S.); (M.S.); (M.K.); (A.M.P.); (P.K.W.)
| | - Michał Kopka
- Department of Methodology, Medical University of Warsaw, 1b Banacha Street, 02-091 Warsaw, Poland; (M.G.); (A.S.); (M.S.); (M.K.); (A.M.P.); (P.K.W.)
- Doctoral School, Medical University of Warsaw, 81 Żwirki i Wigury Street, 02-091 Warsaw, Poland
| | - Adriana M. Pascal
- Department of Methodology, Medical University of Warsaw, 1b Banacha Street, 02-091 Warsaw, Poland; (M.G.); (A.S.); (M.S.); (M.K.); (A.M.P.); (P.K.W.)
| | - Paweł K. Włodarski
- Department of Methodology, Medical University of Warsaw, 1b Banacha Street, 02-091 Warsaw, Poland; (M.G.); (A.S.); (M.S.); (M.K.); (A.M.P.); (P.K.W.)
| |
Collapse
|
9
|
Subbarayudu S, Namasivayam SKR, Arockiaraj J. Immunomodulation in Non-traditional Therapies for Methicillin-resistant Staphylococcus aureus (MRSA) Management. Curr Microbiol 2024; 81:346. [PMID: 39240286 DOI: 10.1007/s00284-024-03875-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
The rise of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge in clinical settings due to its ability to evade conventional antibiotic treatments. This overview explores the potential of immunomodulatory strategies as alternative therapeutic approaches to combat MRSA infections. Traditional antibiotics are becoming less effective, necessitating innovative solutions that harness the body's immune system to enhance pathogen clearance. Recent advancements in immunotherapy, including the use of antimicrobial peptides, phage therapy, and mechanisms of immune cells, demonstrate promise in enhancing the body's ability to clear MRSA infections. However, the exact interactions between these therapies and immunomodulation are not fully understood, underscoring the need for further research. Hence, this review aims to provide a broad overview of the current understanding of non-traditional therapeutics and their impact on immune responses, which could lead to more effective MRSA treatment strategies. Additionally, combining immunomodulatory agents with existing antibiotics may improve outcomes, particularly for immunocompromised patients or those with chronic infections. As the landscape of antibiotic resistance evolves, the development of effective immunotherapeutic strategies could play a vital role in managing MRSA infections and reducing reliance on traditional antibiotics. Future research must focus on optimizing these approaches and validating their efficacy in diverse clinical populations to address the urgent need for effective MRSA management strategies.
Collapse
Affiliation(s)
- Suthi Subbarayudu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, 602105, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
10
|
Zuo Y, Dang R, Peng H, Hu P, Yang Y. LL37-mtDNA regulates viability, apoptosis, inflammation, and autophagy in lipopolysaccharide-treated RLE-6TN cells by targeting Hsp90aa1. Open Life Sci 2024; 19:20220943. [PMID: 39220589 PMCID: PMC11365468 DOI: 10.1515/biol-2022-0943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis-induced acute lung injury is associated with lung epithelial cell injury. This study analyzed the role of the antimicrobial peptide LL37 with mitochondrial DNA (LL37-mtDNA) and its potential mechanism of action in lipopolysaccharide (LPS)-treated rat type II alveolar epithelial cells (RLE-6TN cells). RLE-6TN cells were treated with LPS alone or with LL37-mtDNA, followed by transcriptome sequencing. Differentially expressed and pivotal genes were screened using bioinformatics tools. The effects of LL37-mtDNA on cell viability, inflammation, apoptosis, reactive oxygen species (ROS) production, and autophagy-related hallmark expression were evaluated in LPS-treated RLE-6TN cells. Additionally, the effects of Hsp90aa1 silencing following LL37-mtDNA treatment were investigated in vitro. LL37-mtDNA further suppressed cell viability, augmented apoptosis, promoted the release of inflammatory cytokines, increased ROS production, and elevated LC3B expression in LPS-treated RLE-6TN cells. Using transcriptome sequencing and bioinformatics, ten candidate genes were identified, of which three core genes were verified to be upregulated in the LPS + LL37-mtDNA group. Additionally, Hsp90aa1 downregulation attenuated the effects of LL37-mtDNA on LPS-treated RLE-6TN cells. Hsp90aa1 silencing possibly acted as a crucial target to counteract the effects of LL37-mtDNA on viability, apoptosis, inflammation, and autophagy activation in LPS-treated RLE-6TN cells.
Collapse
Affiliation(s)
- Yunlong Zuo
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Run Dang
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Hongyan Peng
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Peidan Hu
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Yiyu Yang
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, No. 318, Renmin Middle Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
11
|
Singh AN, Wu M, Ye TT, Brown AC, Wittenberg NJ. Engineering Planar Gram-Negative Outer Membrane Mimics Using Bacterial Outer Membrane Vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.11.570829. [PMID: 39229024 PMCID: PMC11370475 DOI: 10.1101/2023.12.11.570829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Antibiotic resistance is a major challenge in modern medicine. The unique double membrane structure of gram-negative bacteria limits the efficacy of many existing antibiotics and adds complexity to antibiotic development by limiting transport of antibiotics to the bacterial cytosol. New methods to mimic this barrier would enable high-throughput studies for antibiotic development. In this study, we introduce an innovative approach to modify outer membrane vesicles (OMVs) from Aggregatibacter actinomycetemcomitans, to generate planar supported lipid bilayer membranes. Our method first involves the incorporation of synthetic lipids into OMVs using a rapid freeze-thaw technique to form outer membrane hybrid vesicles (OM-Hybrids). Subsequently, these OM-Hybrids can spontaneously rupture when in contact with SiO2 surfaces to form a planar outer membrane supported bilayer (OM-SB). We assessed the formation of OM-Hybrids using dynamic light scattering and a fluorescence quenching assay. To analyze the formation of OM-SBs from OM-Hybrids we used quartz crystal microbalance with dissipation monitoring (QCM-D) and fluorescence recovery after photobleaching (FRAP). Additionally, we conducted assays to detect surface-associated DNA and proteins on OM-SBs. The interaction of an antimicrobial peptide, polymyxin B, with the OM-SBs was also assessed. These findings emphasize the capability of our platform to produce planar surfaces of bacterial outer membranes, which in turn, could function as a valuable tool for streamlining the development of antibiotics.
Collapse
Affiliation(s)
- Aarshi N. Singh
- Department of Chemistry, Lehigh University, Bethlehem, PA, USA
| | - Meishan Wu
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA, USA
| | - Tiffany T. Ye
- Department of Chemistry, Lehigh University, Bethlehem, PA, USA
| | - Angela C. Brown
- Department of Chemical and Biomolecular Engineering, Lehigh University, Bethlehem, PA, USA
| | | |
Collapse
|
12
|
Bostanghadiri N, Kouhzad M, Taki E, Elahi Z, Khoshbayan A, Navidifar T, Darban-Sarokhalil D. Oral microbiota and metabolites: key players in oral health and disorder, and microbiota-based therapies. Front Microbiol 2024; 15:1431785. [PMID: 39228377 PMCID: PMC11368800 DOI: 10.3389/fmicb.2024.1431785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024] Open
Abstract
The review aimed to investigate the diversity of oral microbiota and its influencing factors, as well as the association of oral microbiota with oral health and the possible effects of dysbiosis and oral disorder. The oral cavity harbors a substantial microbial burden, which is particularly notable compared to other organs within the human body. In usual situations, the microbiota exists in a state of equilibrium; however, when this balance is disturbed, a multitude of complications arise. Dental caries, a prevalent issue in the oral cavity, is primarily caused by the colonization and activity of bacteria, particularly streptococci. Furthermore, this environment also houses other pathogenic bacteria that are associated with the onset of gingival, periapical, and periodontal diseases, as well as oral cancer. Various strategies have been employed to prevent, control, and treat these disorders. Recently, techniques utilizing microbiota, like probiotics, microbiota transplantation, and the replacement of oral pathogens, have caught the eye. This extensive examination seeks to offer a general view of the oral microbiota and their metabolites concerning oral health and disease, and also the resilience of the microbiota, and the techniques used for the prevention, control, and treatment of disorders in this specific area.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mobina Kouhzad
- Department of Genetics, Faculty of Science, Islamic Azad University North Tehran Branch, Tehran, Iran
| | - Elahe Taki
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amin Khoshbayan
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tahereh Navidifar
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Pennone V, Angelini E, Sarlah D, Lovati AB. Antimicrobial Properties and Cytotoxicity of LL-37-Derived Synthetic Peptides to Treat Orthopedic Infections. Antibiotics (Basel) 2024; 13:764. [PMID: 39200064 PMCID: PMC11350787 DOI: 10.3390/antibiotics13080764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Open fractures and prosthetic joints are prone to bacterial infections, especially those involving biofilms, and are worsened by antibiotic inefficacy and resistance. This highlights the need for targeted treatments against orthopedic infections. LL-37, a human cathelicidin, is known for its antimicrobial properties. This study aimed to synthesize and evaluate LL-37-derived antimicrobial peptides (AMPs) for antibacterial efficacy and toxicity. Several truncated LL-37 analogues were created and tested against 18 bacterial strains, both ATCC and orthopedic clinical isolates, using MIC and MBC assays. Synergy with antibiotics and resistance development were also analyzed, alongside cytotoxicity on NIH-3T3 fibroblasts and hemolytic activity assessments. Six AMPs were synthesized, with FK-16 and GF-17 emerging as the most effective. The MIC values ranged from 4.69 to 18.75 µg/mL and 2.34 to 18.75 µg/mL, respectively, against S. epidermidis and S. aureus, with the MBC values matching the MIC values. Cytotoxicity tests showed no toxicity at concentrations below 75 µg/mL for GF-17 and 150 µg/mL for FK-16. Hemolytic activity was below 1% at 18.75 µg/mL for GF-17 and 75 µg/mL for FK-16. These AMPs showed no synergistic effects with antibiotics and no resistance development. FK-16 and GF-17 effectively removed biofilms, particularly against S. epidermidis. Incorporating these AMPs into surgical materials (hydrogels, cements, etc.) could enhance infection control in orthopedic procedures, warranting further in vivo studies.
Collapse
Affiliation(s)
- Vincenzo Pennone
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy;
| | - Elisa Angelini
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy; (E.A.); (D.S.)
| | - David Sarlah
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy; (E.A.); (D.S.)
- Department of Chemistry, Carl R. Woese Institute for Genomic Biology and Cancer Center at Illinois, University of Illinois, Urbana, IL 61801, USA
| | - Arianna B. Lovati
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, 20157 Milan, Italy;
| |
Collapse
|
14
|
Yang S, Stern A, Duncan G. Synthetic mucus biomaterials synergize with antibiofilm agents to combat Pseudomonas aeruginosa biofilms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607383. [PMID: 39149383 PMCID: PMC11326292 DOI: 10.1101/2024.08.09.607383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Bacterial biofilms are often highly resistant to antimicrobials causing persistent infections which when not effectively managed can significantly worsen clinical outcomes. As such, alternatives to standard antibiotic therapies have been highly sought after to address difficult-to-treat biofilm-associated infections. We hypothesized a biomaterial-based approach using the innate functions of mucins to modulate bacterial surface attachment and virulence could provide a new therapeutic strategy against biofilms. Based on our testing in Pseudomonas aeruginosa biofilms, we found synthetic mucus biomaterials can inhibit biofilm formation and significantly reduce the thickness of mature biofilms. In addition, we evaluated if synthetic mucus biomaterials could work synergistically with DNase and/or α-amylase for enhanced biofilm dispersal. Combination treatment with these antibiofilm agents and synthetic mucus biomaterials resulted in up to 3 log reductions in viability of mature P. aeruginosa biofilms. Overall, this work provides a new bio-inspired, combinatorial approach to address biofilms and antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Sydney Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Alexa Stern
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Gregg Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
15
|
Elafify M, Liao X, Feng J, Ahn J, Ding T. Biofilm formation in food industries: Challenges and control strategies for food safety. Food Res Int 2024; 190:114650. [PMID: 38945629 DOI: 10.1016/j.foodres.2024.114650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/02/2024]
Abstract
Various pathogens have the ability to grow on food matrices and instruments. This grow may reach to form biofilms. Bacterial biofilms are community of microorganisms embedded in extracellular polymeric substances (EPSs) containing lipids, DNA, proteins, and polysaccharides. These EPSs provide a tolerance and favorable living condition for microorganisms. Biofilm formations could not only contribute a risk for food safety but also have negative impacts on healthcare sector. Once biofilms form, they reveal resistances to traditional detergents and disinfectants, leading to cross-contamination. Inhibition of biofilms formation and abolition of mature biofilms is the main target for controlling of biofilm hazards in the food industry. Some novel eco-friendly technologies such as ultrasound, ultraviolet, cold plasma, magnetic nanoparticles, different chemicals additives as vitamins, D-amino acids, enzymes, antimicrobial peptides, and many other inhibitors provide a significant value on biofilm inhibition. These anti-biofilm agents represent promising tools for food industries and researchers to interfere with different phases of biofilms including adherence, quorum sensing molecules, and cell-to-cell communication. This perspective review highlights the biofilm formation mechanisms, issues associated with biofilms, environmental factors influencing bacterial biofilm development, and recent strategies employed to control biofilm-forming bacteria in the food industry. Further studies are still needed to explore the effects of biofilm regulation in food industries and exploit more regulation strategies for improving the quality and decreasing economic losses.
Collapse
Affiliation(s)
- Mahmoud Elafify
- Future Food Laboratory, Innovative Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Xinyu Liao
- Future Food Laboratory, Innovative Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
| | - Jinsong Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Juhee Ahn
- Future Food Laboratory, Innovative Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea.
| | - Tian Ding
- Future Food Laboratory, Innovative Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
16
|
Elbediwi M, Rolff J. Metabolic pathways and antimicrobial peptide resistance in bacteria. J Antimicrob Chemother 2024; 79:1473-1483. [PMID: 38742645 DOI: 10.1093/jac/dkae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
Antimicrobial resistance is a pressing concern that poses a significant threat to global public health, necessitating the exploration of alternative strategies to combat drug-resistant microbial infections. Recently, antimicrobial peptides (AMPs) have gained substantial attention as possible replacements for conventional antibiotics. Because of their pharmacodynamics and killing mechanisms, AMPs display a lower risk of bacterial resistance evolution compared with most conventional antibiotics. However, bacteria display different mechanisms to resist AMPs, and the role of metabolic pathways in the resistance mechanism is not fully understood. This review examines the intricate relationship between metabolic genes and AMP resistance, focusing on the impact of metabolic pathways on various aspects of resistance. Metabolic pathways related to guanosine pentaphosphate (pppGpp) and guanosine tetraphosphate (ppGpp) [collectively (p)ppGpp], the tricarboxylic acid (TCA) cycle, haem biosynthesis, purine and pyrimidine biosynthesis, and amino acid and lipid metabolism influence in different ways metabolic adjustments, biofilm formation and energy production that could be involved in AMP resistance. By targeting metabolic pathways and their associated genes, it could be possible to enhance the efficacy of existing antimicrobial therapies and overcome the challenges exhibited by phenotypic (recalcitrance) and genetic resistance toward AMPs. Further research in this area is needed to provide valuable insights into specific mechanisms, uncover novel therapeutic targets, and aid in the fight against antimicrobial resistance.
Collapse
Affiliation(s)
- Mohammed Elbediwi
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
- Animal Health Research Institute, Agriculture Research Centre, 12618 Cairo, Egypt
| | - Jens Rolff
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB), Berlin, Germany
| |
Collapse
|
17
|
Yeh KT, Wu WT, Wang CC, Lee RP. Adipose-derived stem cells and antibiotics: A novel synergistic approach for treating implant-related osteomyelitis. Tzu Chi Med J 2024; 36:271-274. [PMID: 38993834 PMCID: PMC11236081 DOI: 10.4103/tcmj.tcmj_48_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/01/2024] [Accepted: 04/16/2024] [Indexed: 07/13/2024] Open
Abstract
Implant-related osteomyelitis poses a significant challenge in orthopedic practice, particularly due to the increasing prevalence of antibiotic-resistant infections and biofilm-associated complications. This article focused on exploring the potential of combination therapy with adipose-derived stem cells (ADSCs) and antibiotics to overcome these challenges, thereby enhancing treatment efficacy. A systematic synthesis of the results of recent in vivo studies, predominantly those using rat models, was performed. Studies that evaluated the effectiveness of ADSCs combined with antibiotics against common pathogens in implant-related osteomyelitis, particularly Staphylococcus aureus and methicillin-resistant Staphylococcus epidermidis, were selected. A significant reduction in symptoms such as swelling, abscess formation, and bacterial burden in the ADSCs + antibiotic-treated group was observed in all studies. In addition, microcomputed tomography revealed reduced osteolysis, indicating enhanced bone preservation. Furthermore, histological examination revealed improved tissue structure and altered immune response, signifying the dual role of ADSCs in enhancing antibiotic action and modulating the immune system. This review highlights the promising role of the concurrent use of ADSCs and antibiotics in the treatment of implant-related osteomyelitis. This novel therapeutic strategy has the potential to revolutionize the management of complex orthopedic infections, especially those resistant to conventional treatments. However, further research is required to translate the results of animal studies into clinical applications and to develop optimized treatment protocols for human use.
Collapse
Affiliation(s)
- Kuang-Ting Yeh
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Clinical Pharmacy, Tzu Chi University, Hualien, Taiwan
| | - Wen-Tien Wu
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Chen-Chie Wang
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Orthopedic Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Ru-Ping Lee
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
18
|
Marissen J, Reichert L, Härtel C, Fortmann MI, Faust K, Msanga D, Harder J, Zemlin M, Gomez de Agüero M, Masjosthusmann K, Humberg A. Antimicrobial Peptides (AMPs) and the Microbiome in Preterm Infants: Consequences and Opportunities for Future Therapeutics. Int J Mol Sci 2024; 25:6684. [PMID: 38928389 PMCID: PMC11203687 DOI: 10.3390/ijms25126684] [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: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in various organisms, including humans. Beyond their direct antimicrobial effects, AMPs play essential roles in various physiological processes. They induce angiogenesis, promote wound healing, modulate immune responses, and serve as chemoattractants for immune cells. AMPs regulate the microbiome and combat microbial infections on the skin, lungs, and gastrointestinal tract. Produced in response to microbial signals, AMPs help maintain a balanced microbial community and provide a first line of defense against infection. In preterm infants, alterations in microbiome composition have been linked to various health outcomes, including sepsis, necrotizing enterocolitis, atopic dermatitis, and respiratory infections. Dysbiosis, or an imbalance in the microbiome, can alter AMP profiles and potentially lead to inflammation-mediated diseases such as chronic lung disease and obesity. In the following review, we summarize what is known about the vital role of AMPs as multifunctional peptides in protecting newborn infants against infections and modulating the microbiome and immune response. Understanding their roles in preterm infants and high-risk populations offers the potential for innovative approaches to disease prevention and treatment.
Collapse
Affiliation(s)
- Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Lilith Reichert
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Kirstin Faust
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Delfina Msanga
- Department of Pediatrics, Bugando Hospital, Catholic University of Health and Allied Sciences, Mwanza 33109, Tanzania;
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, 24105 Kiel, Germany;
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| | - Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| |
Collapse
|
19
|
Ferrucci V, Miceli M, Pagliuca C, Bianco O, Castaldo L, Izzo L, Cozzolino M, Zannella C, Oglio F, Polcaro A, Randazzo A, Colicchio R, Galdiero M, Berni Canani R, Salvatore P, Zollo M. Modulation of innate immunity related genes resulting in prophylactic antimicrobial and antiviral properties. J Transl Med 2024; 22:574. [PMID: 38886736 PMCID: PMC11184722 DOI: 10.1186/s12967-024-05378-2] [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: 03/14/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The innate immunity acts during the early phases of infection and its failure in response to a multilayer network of co-infections is cause of immune system dysregulation. Epidemiological SARS-CoV-2 infections data, show that Influenza Virus (FLU-A-B-C) and Respiratory Syncytial Virus (RSV) are co-habiting those respiratory traits. These viruses, especially in children (mostly affected by 'multi-system inflammatory syndrome in children' [MIS-C] and the winter pandemic FLU), in the aged population, and in 'fragile' patients are causing alteration in immune response. Then, bacterial and fungal pathogens are also co-habiting the upper respiratory traits (e.g., Staphylococcus aureus and Candida albicans), thus contributing to morbidity in those COVID-19 affected patients. METHODS Liquid chromatography coupled with high-resolution mass spectrometry using the quadrupole orbital ion trap analyser (i.e., UHPLC-Q-Orbitrap HRMS) was adopted to measure the polyphenols content of a new nutraceutical formula (Solution-3). Viral infections with SARS-CoV-2 (EG.5), FLU-A and RSV-A viruses (as performed in BLS3 authorised laboratory) and real time RT-PCR (qPCR) assay were used to test the antiviral action of the nutraceutical formula. Dilution susceptibility tests have been used to estimate the minimum inhibitory and bactericidal concentration (MIC and MBC, respectively) of Solution-3 on a variety of microorganisms belonging to Gram positive/ negative bacteria and fungi. Transcriptomic data analyses and functional genomics (i.e., RNAseq and data mining), coupled to qPCR and ELISA assays have been used to investigate the mechanisms of action of the nutraceutical formula on those processes involved in innate immune response. RESULTS Here, we have tested the combination of natural products containing higher amounts of polyphenols (i.e., propolis, Verbascum thapsus L., and Thymus vulgaris L.), together with the inorganic long chain polyphosphates 'polyPs' with antiviral, antibacterial, and antifungal behaviours, against SARS-CoV-2, FLU-A, RSV-A, Gram positive/ negative bacteria and fungi (i.e., Candida albicans). These components synergistically exert an immunomodulatory action by enhancing those processes involved in innate immune response (e.g., cytokines: IFNγ, TNFα, IL-10, IL-6/12; chemokines: CXCL1; antimicrobial peptides: HBD-2, LL-37; complement system: C3). CONCLUSION The prophylactic antimicrobial success of this nutraceutical formula against SARS-CoV-2, FLU-A and RSV-A viruses, together with the common bacteria and fungi co-infections as present in human oral cavity, is expected to be valuable.
Collapse
Affiliation(s)
- Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy.
- Elysium Cell Bio Ita, Via Gaetano Salvatore 486, 80145, Naples, Italy.
| | - Marco Miceli
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
| | - Orazio Bianco
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Luigi Castaldo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Luana Izzo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Marica Cozzolino
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Franca Oglio
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Antonio Polcaro
- Polcaro Fitopreparazioni S.R.L, Via Sant Agnello, 9 D; 80030, Roccarainola, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
- UOC of Virology and Microbiology, University Hospital of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Roberto Berni Canani
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy.
- Elysium Cell Bio Ita, Via Gaetano Salvatore 486, 80145, Naples, Italy.
- DAI Medicina di Laboratorio e Trasfusionale, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
20
|
Abdulrehman T, Qadri S, Haik Y, Sultan A, Skariah S, Kumar S, Mendoza Z, Yadav KK, Titus A, Khader S. Advances in the targeted theragnostics of osteomyelitis caused by Staphylococcus aureus. Arch Microbiol 2024; 206:288. [PMID: 38834761 DOI: 10.1007/s00203-024-04015-2] [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: 03/31/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
Bone infections caused by Staphylococcus aureus may lead to an inflammatory condition called osteomyelitis, which results in progressive bone loss. Biofilm formation, intracellular survival, and the ability of S. aureus to evade the immune response result in recurrent and persistent infections that present significant challenges in treating osteomyelitis. Moreover, people with diabetes are prone to osteomyelitis due to their compromised immune system, and in life-threatening cases, this may lead to amputation of the affected limbs. In most cases, bone infections are localized; thus, early detection and targeted therapy may prove fruitful in treating S. aureus-related bone infections and preventing the spread of the infection. Specific S. aureus components or overexpressed tissue biomarkers in bone infections could be targeted to deliver active therapeutics, thereby reducing drug dosage and systemic toxicity. Compounds like peptides and antibodies can specifically bind to S. aureus or overexpressed disease markers and combining these with therapeutics or imaging agents can facilitate targeted delivery to the site of infection. The effectiveness of photodynamic therapy and hyperthermia therapy can be increased by the addition of targeting molecules to these therapies enabling site-specific therapy delivery. Strategies like host-directed therapy focus on modulating the host immune mechanisms or signaling pathways utilized by S. aureus for therapeutic efficacy. Targeted therapeutic strategies in conjunction with standard surgical care could be potential treatment strategies for S. aureus-associated osteomyelitis to overcome antibiotic resistance and disease recurrence. This review paper presents information about the targeting strategies and agents for the therapy and diagnostic imaging of S. aureus bone infections.
Collapse
Affiliation(s)
- Tahir Abdulrehman
- eHealth Program, DeGroote School of Business, McMaster University, Hamilton, ON, Canada
- Health Policy, Management and Informatics, Allied Health, Credit Valley Hospital, Mississauga, ON, Canada
| | - Shahnaz Qadri
- School of Pharmacy, Texas A&M University, Kingsville, USA.
| | - Yousef Haik
- Department of Mechanical & Nuclear Engineering, University of Sharjah, Sharjah, UAE.
| | - Ali Sultan
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Sini Skariah
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Shourya Kumar
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Zachary Mendoza
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Kamlesh K Yadav
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Anoop Titus
- Department of Preventive Cardiology, Houston Methodist, Houston, TX, USA
| | - Shameer Khader
- School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
21
|
Farzi N, Oloomi M, Bahramali G, Siadat SD, Bouzari S. Antibacterial Properties and Efficacy of LL-37 Fragment GF-17D3 and Scolopendin A2 Peptides Against Resistant Clinical Strains of Staphylococcus aureus, Pseudomonas aeruginosa, and Acinetobacter baumannii In Vitro and In Vivo Model Studies. Probiotics Antimicrob Proteins 2024; 16:796-814. [PMID: 37148452 DOI: 10.1007/s12602-023-10070-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 05/08/2023]
Abstract
Pseudomonas aeruginosa, Staphylococcus aureus, and Acinetobacter baumannii have emerged as major clinical threats owing to the increasing prevalence of ventilator-associated pneumonia caused by multidrug-resistant or extensively drug-resistant strains. The present study aimed to assess the antibacterial effects and efficacy of LL-37 fragment GF-17D3 and synthetic Scolopendin A2 peptides against resistant clinical strains in vitro and in vivo models. P. aeruginosa, S. aureus, and A. baumannii were isolated from clinical infections. Their antibiotic resistance and minimum inhibitory concentration were assessed. LL-37 fragment GF-17D3 peptide was selected from available databases. Scolopendin A2 peptide's 6th amino acid (proline) was substituted with lysine and peptides and MICs were determined. The biofilm inhibitory activity was quantified at sub MIC concentrations. Synergetic effects of Scolopendin A2 and imipenem were assessed by checkerboard. After mice nasal infection with P. aeruginosa, peptides LD50 was determined. Isolates harbored complete resistance toward the majority of antibiotics and MIC values ranged between 1 and > 512 µg/ml. The majority of isolates exhibited strong biofilm activity. Synthetic peptides showed lower MIC values than antibiotic agents and the lowest MIC values were obtained for synthetic peptides in combination with antibiotics. The Synergisms effect of Scolopendin A2 with imipenem was also determined. Scolopendin A2 was found to have antibacterial efficacy against P. aeruginosa, S. aureus, and A. baumannii with MIC 64 µg/ml, 8 µg/ml, and 16 µg/ml, respectively, and LL37 showed antibacterial efficacy against P. aeruginosa, S. aureus, and A. baumannii with MIC 128 µg/ml, 32 µg/ml, and 32 µg/ml, respectively. Both AMPs decreased biofilms by ≥ 96% at 1 × MIC. The biofilm inhibitory activity was measured at sub MIC concentrations of the peptides and the results demonstrated that Scolopendin A2 exhibited anti-biofilm activity at 1/4 × MIC and 1/2 × MIC concentrations was 47.9 to 63.8%, although LL37 among 1/4 × MIC and 1/2 × MIC concentrations was 21.3 to 49.6% against three pathogens. The combination of Scolopendin A2 and antibiotics demonstrated synergistic activity-resistant strains with FIC values ≤ 0.5 for three pathogens, while LL37 and antibiotics showed synergistic activity FIC values ≤ 0.5 for only P. aeruginosa. Infection model Scolopendin A2 with Imipenem (2 × MIC) was efficacious in vivo, with a 100% survival rate following treatment at 2 × MIC after 120 h. The mRNA expression of biofilm-related genes was decreased for both peptides. Synthesis Scolopendin A2 decreased the expression of biofilm formation genes compared to the control group. Synthetic Scolopendin A2 exhibits antimicrobial activity without causing toxicity on the human epithelial cell line. Based on our findings, it seems that synthetic Scolopendin A2 is an appropriate antimicrobial source. That could be a promising option in combination with antibiotics for a topical medication and in the prevention of acute and chronic infections caused by multidrug-resistant bacteria. Nevertheless, additional experiments are required to assess another potential of this novel AMP.
Collapse
Affiliation(s)
- Nastaran Farzi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mana Oloomi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Golnaz Bahramali
- Department of Hepatitis and AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran.
- Department of Hepatitis and AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran.
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
22
|
Lim BY, Azmi F, Ng SF. LL37 Microspheres Loaded on Activated Carbon-chitosan Hydrogel: Anti-bacterial and Anti-toxin Wound Dressing for Chronic Wound Infections. AAPS PharmSciTech 2024; 25:110. [PMID: 38740721 DOI: 10.1208/s12249-024-02826-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Antimicrobial peptide LL37 is a promising antibacterial candidate due to its potent antimicrobial activity with no known bacterial resistance. However, intrinsically LL37 is susceptible to degradation in wound fluids limits its effectiveness. Bacterial toxins which are released after cell lysis are found to hinder wound healing. To address these challenges, encapsulating LL37 in microspheres (MS) and loading the MS onto activated carbon (AC)-chitosan (CS) hydrogel. This advanced wound dressing not only protects LL37 from degradation but also targets bacterial toxins, aiding in the healing of chronic wound infections. First, LL37 MS and LL37-AC-CS hydrogel were prepared and characterised in terms of physicochemical properties, drug release, and peptide-polymer compatibility. Antibacterial and antibiofilm activity, bacterial toxin elimination, cell migration, and cell cytotoxicity activities were investigated. LL37-AC-CS hydrogel was effective against Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus. LL37-AC-CS hydrogel bound more endotoxin than AC with CS hydrogel alone. The hydrogel also induced cell migration after 72 h and showed no cytotoxicity towards NHDF after 72 h of treatment. In conclusion, the LL37-AC-CS hydrogel was shown to be a stable, non-toxic advanced wound dressing method with enhanced antimicrobial and antitoxin activity, and it can potentially be applied to chronic wound infections to accelerate wound healing.
Collapse
Affiliation(s)
- Bee-Yee Lim
- National Pharmaceutical Regulatory Agency, 36, Jalan Profesor Diraja Ungku Aziz, PJS 13, Petaling Jaya, Selangor, 46200, Malaysia
- Centre for Drug Delivery Technology and Vaccine, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| | - Fazren Azmi
- Centre for Drug Delivery Technology and Vaccine, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| | - Shiow-Fern Ng
- Centre for Drug Delivery Technology and Vaccine, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia.
| |
Collapse
|
23
|
Pastuszak K, Jurak M, Kowalczyk B, Tarasiuk J, Wiącek AE, Palusińska-Szysz M. Susceptibility of Legionella gormanii Membrane-Derived Phospholipids to the Peptide Action of Antimicrobial LL-37-Langmuir Monolayer Studies. Molecules 2024; 29:1522. [PMID: 38611802 PMCID: PMC11013288 DOI: 10.3390/molecules29071522] [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: 02/16/2024] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
LL-37 is the only member of the cathelicidin-type host defense peptide family in humans. It exhibits broad-spectrum bactericidal activity, which represents a distinctive advantage for future therapeutic targets. The presence of choline in the growth medium for bacteria changes the composition and physicochemical properties of their membranes, which affects LL-37's activity as an antimicrobial agent. In this study, the effect of the LL-37 peptide on the phospholipid monolayers at the liquid-air interface imitating the membranes of Legionella gormanii bacteria was determined. The Langmuir monolayer technique was employed to prepare model membranes composed of individual classes of phospholipids-phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylglycerol (PG), cardiolipin (CL)-isolated from L. gormanii bacteria supplemented or non-supplemented with exogenous choline. Compression isotherms were obtained for the monolayers with or without the addition of the peptide to the subphase. Then, penetration tests were carried out for the phospholipid monolayers compressed to a surface pressure of 30 mN/m, followed by the insertion of the peptide into the subphase. Changes in the mean molecular area were observed over time. Our findings demonstrate the diversified effect of LL-37 on the phospholipid monolayers, depending on the bacteria growth conditions. The substantial changes in membrane properties due to its interactions with LL-37 enable us to propose a feasible mechanism of peptide action at a molecular level. This can be associated with the stable incorporation of the peptide inside the monolayer or with the disruption of the membrane leading to the removal (desorption) of molecules into the subphase. Understanding the role of antimicrobial peptides is crucial for the design and development of new strategies and routes for combating resistance to conventional antibiotics.
Collapse
Affiliation(s)
- Katarzyna Pastuszak
- Department of Interfacial Phenomena, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Skłodowska University, Maria Curie-Skłodowska Sq. 3, 20-031 Lublin, Poland; (K.P.); (A.E.W.)
| | - Małgorzata Jurak
- Department of Interfacial Phenomena, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Skłodowska University, Maria Curie-Skłodowska Sq. 3, 20-031 Lublin, Poland; (K.P.); (A.E.W.)
| | - Bożena Kowalczyk
- Department of Genetics and Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (B.K.); (J.T.); (M.P.-S.)
| | - Jacek Tarasiuk
- Department of Genetics and Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (B.K.); (J.T.); (M.P.-S.)
| | - Agnieszka Ewa Wiącek
- Department of Interfacial Phenomena, Institute of Chemical Sciences, Faculty of Chemistry, Maria Curie-Skłodowska University, Maria Curie-Skłodowska Sq. 3, 20-031 Lublin, Poland; (K.P.); (A.E.W.)
| | - Marta Palusińska-Szysz
- Department of Genetics and Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (B.K.); (J.T.); (M.P.-S.)
| |
Collapse
|
24
|
Vieira APGC, de Souza AN, Lima WG, Brito JCM, Simião DC, Gonçalves LVR, Cordeiro LPB, de Oliveira Scoaris D, Fernandes SOA, Resende JM, Bechinger B, Verly RM, de Lima ME. The Synthetic Peptide LyeTx I mn∆K, Derived from Lycosa erythrognatha Spider Toxin, Is Active against Methicillin-Resistant Staphylococcus aureus (MRSA) In Vitro and In Vivo. Antibiotics (Basel) 2024; 13:248. [PMID: 38534683 DOI: 10.3390/antibiotics13030248] [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: 01/27/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
The urgent global health challenge posed by methicillin-resistant Staphylococcus aureus (MRSA) infections demands effective solutions. Antimicrobial peptides (AMPs) represent promising tools of research of new antibacterial agents and LyeTx I mn∆K, a short synthetic peptide based on the Lycosa erythrognatha spider venom, is a good representative. This study focused on analyzing the antimicrobial activities of LyeTx I mn∆K, including minimum inhibitory and bactericidal concentrations, synergy and resensitization assays, lysis activity, the effect on biofilm, and the bacterial death curve in MRSA. Additionally, its characterization was conducted through isothermal titration calorimetry, dynamic light scattering, calcein release, and finally, efficacy in a mice wound model. The peptide demonstrates remarkable efficacy against planktonic cells (MIC 8-16 µM) and biofilms (>30% of inhibition) of MRSA, and outperforms vancomycin in terms of rapid bactericidal action and anti-biofilm effects. The mechanism involves significant membrane damage. Interactions with bacterial model membranes, including those with lysylphosphatidylglycerol (LysylPOPG) modifications, highlight the versatility and selectivity of this compound. Also, the peptide has the ability to sensitize resistant bacteria to conventional antibiotics, showing potential for combinatory therapy. Furthermore, using an in vivo model, this study showed that a formulated gel containing the peptide proved superior to vancomycin in treating MRSA-induced wounds in mice. Together, the results highlight LyeTx I mnΔK as a promising prototype for the development of effective therapeutic strategies against superficial MRSA infections.
Collapse
Affiliation(s)
- Ana Paula Gonçalves Coelho Vieira
- Faculdade de Saúde Santa Casa de Belo Horizonte, Programa de Pós-Graduação Stricto Sensu em Medicina e Biomedicina, Belo Horizonte 30150-240, Brazil
| | - Amanda Neves de Souza
- Departamento de Química, FACET, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM)-Campus JK, Diamantina 39100-000, Brazil
- Institut de Chimie, Centre National de la Recherche Scientifique, UMR7177, Université de Strasbourg, 67070 Strasbourg, France
| | - William Gustavo Lima
- Faculdade de Saúde Santa Casa de Belo Horizonte, Programa de Pós-Graduação Stricto Sensu em Medicina e Biomedicina, Belo Horizonte 30150-240, Brazil
| | | | - Daniela Carolina Simião
- Laboratório de Radioisótopos, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia-Campus Pampulha, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Lucas Vinícius Ribeiro Gonçalves
- Faculdade de Saúde Santa Casa de Belo Horizonte, Programa de Pós-Graduação Stricto Sensu em Medicina e Biomedicina, Belo Horizonte 30150-240, Brazil
| | - Lídia Pereira Barbosa Cordeiro
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | | | - Simone Odília Antunes Fernandes
- Laboratório de Radioisótopos, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia-Campus Pampulha, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Jarbas Magalhães Resende
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil
| | - Burkhard Bechinger
- Institut de Chimie, Centre National de la Recherche Scientifique, UMR7177, Université de Strasbourg, 67070 Strasbourg, France
- Institut Universitaire de France (IUF), 75005 Paris, France
| | - Rodrigo Moreira Verly
- Departamento de Química, FACET, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM)-Campus JK, Diamantina 39100-000, Brazil
| | - Maria Elena de Lima
- Faculdade de Saúde Santa Casa de Belo Horizonte, Programa de Pós-Graduação Stricto Sensu em Medicina e Biomedicina, Belo Horizonte 30150-240, Brazil
| |
Collapse
|
25
|
Giedraitiene A, Tatarunas V, Kaminskaite K, Meskauskaite U, Boieva S, Ajima Y, Ciapiene I, Veikutiene A, Zvikas V, Kupstyte-Kristapone N, Jakstas V, Luksiene D, Tamosiunas A, Lesauskaite V. Enterobacterales Biofilm-Specific Genes and Antimicrobial and Anti-Inflammatory Biomarkers in the Blood of Patients with Ischemic Heart Disease. Diagnostics (Basel) 2024; 14:546. [PMID: 38473018 DOI: 10.3390/diagnostics14050546] [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: 02/03/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Ischemic heart disease (IHD) is the most prevalent type of cardiovascular disease. The main cause of IHD is atherosclerosis, which is a multifactorial inflammatory disease of blood vessels. Studies show that bacteria might have a significant impact on the pathogenesis of atherosclerosis and plaque rupture. This study aimed to evaluate the complexity of interactions between bacteria and the human body concerning metabolites and bacterial genes in patients with ischemic heart disease. METHODS Bacterial 16S rDNA and wcaF, papC, and sdhC genes were detected in whole blood using a real-time PCR methodology. An enzyme-linked immunosorbent assay was used to measure the concentration of the LL-37 protein. An analysis of ARA in blood plasma was performed. RESULTS Bacterial 16S rDNA was detected in 31% of the study patients, and the genes wcaF and sdhC in 20%. Enterobacterales genes were detected more frequently in patients younger than 65 years than in patients aged 65 years and older (p = 0.018) and in patients with type 2 diabetes (p = 0.048). Concentrations of the human antimicrobial peptide LL-37 and 12S-HETE concentrations were determined to be higher if patients had 16S rDNA and biofilm-specific genes. CONCLUSIONS The results of this study enhance the understanding that Enterobacterales bacteria may participate in the pathogenesis of atherosclerosis and IHD. Bacterial DNA and host metabolites in higher concentrations appear to be detected.
Collapse
Affiliation(s)
- Agne Giedraitiene
- Institute of Microbiology and Virology, Lithuanian University of Health Sciences, Eiveniu 4, LT 50161 Kaunas, Lithuania
| | - Vacis Tatarunas
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania
| | - Kornelija Kaminskaite
- Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| | - Ugne Meskauskaite
- Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| | - Svitlana Boieva
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania
| | - Yu Ajima
- Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
- School of Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Ieva Ciapiene
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania
| | - Audrone Veikutiene
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania
| | - Vaidotas Zvikas
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Sukileliu 13, LT 50161 Kaunas, Lithuania
| | - Nora Kupstyte-Kristapone
- Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| | - Valdas Jakstas
- Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Sukileliu 13, LT 50161 Kaunas, Lithuania
| | - Dalia Luksiene
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania
| | - Abdonas Tamosiunas
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania
| | - Vaiva Lesauskaite
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT 50103 Kaunas, Lithuania
| |
Collapse
|
26
|
Gao C, Qin S, Wang M, Li R, Ampomah-Wireko M, Chen S, Qu Y, Zhang E. Effective ciprofloxacin cationic antibacterial agent against persister bacteria with low hemolytic toxicity. Eur J Med Chem 2024; 267:116215. [PMID: 38354522 DOI: 10.1016/j.ejmech.2024.116215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/16/2024]
Abstract
With the widespread use of antibiotics, bacterial resistance has developed rapidly. To make matters worse, infections caused by persistent bacteria and biofilms often cannot be completely eliminated, which brings great difficulties to clinical medication. In this work, three series of quinolone pyridinium quaternary ammonium small molecules were designed and synthesized. Most of the compounds showed good antibacterial activity against Gram-positive bacteria (S. aureus and E. faecalis) and Gram-negative bacteria (E. coli and S. maltophilia). The activity of the para-pyridine quaternary ammonium salt was better than that of the meta-pyridine. 3f was the optimal compound with good stability in body fluids and was unlikely to induce bacterial resistance. The hemolysis rate of erythrocytes at 1280 μg/mL for 3f was only 5.1%. Encouragingly, 3f rapidly killed bacteria within 4 h at 4 × MIC concentration and was effective in killing persistent bacteria in biofilms. The antibacterial mechanism experiments showed that 3f could cause disorder of bacterial membrane potential, increase bacterial membrane permeability, dissolve and destroy the membrane. Incomplete bacterial membranes lead to leakage of bacterial genetic material, concomitant production of ROS, and bacterial death due to these multiple effects.
Collapse
Affiliation(s)
- Chen Gao
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Meng Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Ruirui Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Maxwell Ampomah-Wireko
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Shengcong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - Ye Qu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China
| | - En Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China Zhengzhou University, Zhengzhou 450001, PR China; Pingyuan Laboratory (Zhengzhou University), PR China.
| |
Collapse
|
27
|
Surekha S, Lamiyan AK, Gupta V. Antibiotic Resistant Biofilms and the Quest for Novel Therapeutic Strategies. Indian J Microbiol 2024; 64:20-35. [PMID: 38468748 PMCID: PMC10924852 DOI: 10.1007/s12088-023-01138-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/03/2023] [Indexed: 03/13/2024] Open
Abstract
Antimicrobial resistance (AMR) is one of the major leading causes of death around the globe. Present treatment pipelines are insufficient to overcome the critical situation. Prominent biofilm forming human pathogens which can thrive in infection sites using adaptive features results in biofilm persistence. Considering the present scenario, prudential investigations into the mechanisms of resistance target them to improve antibiotic efficacy is required. Regarding this, developing newer and effective treatment options using edge cutting technologies in medical research is the need of time. The reasons underlying the adaptive features in biofilm persistence have been centred on different metabolic and physiological aspects. The high tolerance levels against antibiotics direct researchers to search for novel bioactive molecules that can help combat the problem. In view of this, the present review outlines the focuses on an opportunity of different strategies which are in testing pipeline can thus be developed into products ready to use.
Collapse
Affiliation(s)
- Saumya Surekha
- Department of Biochemistry, Panjab University, Chandigarh, India
| | | | - Varsha Gupta
- GMCH: Government Medical College and Hospital, Chandigarh, India
| |
Collapse
|
28
|
Tang S, Peel E, Belov K, Hogg CJ, Farquharson KA. Multi-omics resources for the Australian southern stuttering frog (Mixophyes australis) reveal assorted antimicrobial peptides. Sci Rep 2024; 14:3991. [PMID: 38368484 PMCID: PMC10874372 DOI: 10.1038/s41598-024-54522-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/13/2024] [Indexed: 02/19/2024] Open
Abstract
The number of genome-level resources for non-model species continues to rapidly expand. However, frog species remain underrepresented, with up to 90% of frog genera having no genomic or transcriptomic data. Here, we assemble the first genomic and transcriptomic resources for the recently described southern stuttering frog (Mixophyes australis). The southern stuttering frog is ground-dwelling, inhabiting naturally vegetated riverbanks in south-eastern Australia. Using PacBio HiFi long-read sequencing and Hi-C scaffolding, we generated a high-quality genome assembly, with a scaffold N50 of 369.3 Mb and 95.1% of the genome contained in twelve scaffolds. Using this assembly, we identified the mitochondrial genome, and assembled six tissue-specific transcriptomes. We also bioinformatically characterised novel sequences of two families of antimicrobial peptides (AMPs) in the southern stuttering frog, the cathelicidins and β-defensins. While traditional peptidomic approaches to peptide discovery have typically identified one or two AMPs in a frog species from skin secretions, our bioinformatic approach discovered 12 cathelicidins and two β-defensins that were expressed in a range of tissues. We investigated the novelty of the peptides and found diverse predicted activities. Our bioinformatic approach highlights the benefits of multi-omics resources in peptide discovery and contributes valuable genomic resources in an under-represented taxon.
Collapse
Affiliation(s)
- Simon Tang
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Emma Peel
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Katherine Belov
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Carolyn J Hogg
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia.
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Katherine A Farquharson
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
29
|
Czarnowski M, Słowińska M, Sawieljew M, Wnorowska U, Daniluk T, Król G, Karasiński M, Okła S, Savage PB, Piktel E, Bucki R. Efficacy of Ceragenins in Controlling the Growth of Oral Microorganisms: Implications for Oral Hygiene Management. Pharmaceuticals (Basel) 2024; 17:204. [PMID: 38399419 PMCID: PMC10893225 DOI: 10.3390/ph17020204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Ensuring proper dental hygiene is of paramount importance for individuals' general well-being, particularly for patients receiving medical care. There is a prevailing utilization of conventional oral hygiene items, including toothbrushes and mouthwashes, which have gained widespread acceptance; nevertheless, their limitations encourage investigating novel options in this domain. Our study indicates that ceragenins (CSAs) being lipid analogs of host defense peptides, well-recognized for their wide-ranging antimicrobial properties, may be a potentially efficacious means to augment oral hygiene in hospitalized individuals. We demonstrate that ceragenins CSA-13, CSA-44, and CSA-131 as well as undescribed to date CSA-255 display potent antimicrobial activities against isolates of fungi, aerobic, and anaerobic bacteria from Candida, Streptococcus, Enterococcus, and Bacteroides species, which are well-recognized representatives of microbes found in the oral cavity. These effects were further confirmed against mono- and dual-species fungal and bacterial biofilms. While the ceragenins showed similar or slightly diminished efficacy compared to commercially available mouthwashes, they demonstrated a highly favorable toxicity profile toward host cells, that may translate into better maintenance of host mucosal membrane stability. This suggests that incorporating ceragenins into oral hygiene products could be a valuable strategy for reducing the risk of both oral cavity-localized and secondary systemic infections and for improving the overall health outcomes of individuals receiving medical treatment.
Collapse
Affiliation(s)
- Michał Czarnowski
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.C.); (M.S.); (M.S.); (U.W.); (T.D.); (M.K.)
| | - Monika Słowińska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.C.); (M.S.); (M.S.); (U.W.); (T.D.); (M.K.)
| | - Mariusz Sawieljew
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.C.); (M.S.); (M.S.); (U.W.); (T.D.); (M.K.)
| | - Urszula Wnorowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.C.); (M.S.); (M.S.); (U.W.); (T.D.); (M.K.)
| | - Tamara Daniluk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.C.); (M.S.); (M.S.); (U.W.); (T.D.); (M.K.)
| | - Grzegorz Król
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, 25-317 Kielce, Poland; (G.K.); (S.O.)
| | - Maciej Karasiński
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.C.); (M.S.); (M.S.); (U.W.); (T.D.); (M.K.)
| | - Sławomir Okła
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, 25-317 Kielce, Poland; (G.K.); (S.O.)
- Holy Cross Cancer Center, 25-734 Kielce, Poland
| | - Paul B. Savage
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA;
| | - Ewelina Piktel
- Independent Laboratory of Nanomedicine, Medical University of Białystok, 15-222 Białystok, Poland;
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland; (M.C.); (M.S.); (M.S.); (U.W.); (T.D.); (M.K.)
| |
Collapse
|
30
|
Zhang Y, Zheng M, Wang Z, Liu Z, Chen S, Li X, Shi Y, Hu H. Discovery of novel antibacterial agent for the infected wound treatment: all-hydrocarbon stapling optimization of LL-37. Theranostics 2024; 14:1181-1194. [PMID: 38323312 PMCID: PMC10845205 DOI: 10.7150/thno.87916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024] Open
Abstract
Rationale: Antimicrobial peptide LL-37 has been recognized as a favorable alternative to antibiotics due to its broad antibacterial spectrum, low resistance development and diverse biological activities. However, its high manufactory cost, poor proteolytic stability, and unpredictable cytotoxicity seriously hindered its medical translation. Methods: To push the frontiers of its clinical application, all-hydrocarbon stapling strategy was exploited here for the structural modification of KR-12, the core and minimal fragment of LL-37. Results: Based on a library of KR-12 derivatives that designed and synthesized to be stapled at positions of either i, i+4 or i, i+7, structure to activity relationship was investigated. Among them, KR-12(Q5, D9) with the glutamine and aspartic acid residues stapled displayed increased helical content and positive charge. The reinforced α-helical conformation not only protected it from proteolytic hydrolysis but also improved its antibacterial efficacy via effective membrane perturbation and anti-inflammatory efficacy via compact LPS binding. Besides, the increased positive charge endowed it with an enhanced therapeutic index. On infected wound mouse model, it was demonstrated to eliminate bacteria and promote wound closure and regeneration effectively. Conclusion: Overall, the all-hydrocarbon stapling was proven to lay the foundation for the future development of antibacterial agents. KR-12(Q5, D9) could serve as a lead compound for the clinical treatment of bacterial infections.
Collapse
Affiliation(s)
- Yanan Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Mengjun Zheng
- School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Zhe Wang
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhinan Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Sumeng Chen
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Xiang Li
- School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Yejiao Shi
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Honggang Hu
- School of Medicine, Shanghai University, Shanghai 200444, China
| |
Collapse
|
31
|
Lei R, Yang C, Sun Y, Li D, Hao L, Li Y, Wu S, Li H, Lan C, Fang X. Turning cationic antimicrobial peptide KR-12 into self-assembled nanobiotics with potent bacterial killing and LPS neutralizing activities. NANOSCALE 2024; 16:887-902. [PMID: 38105768 DOI: 10.1039/d3nr05174a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Gram-negative sepsis has become a substantial and escalating global healthcare challenge due to the growing antibiotic resistance crisis and the sluggish development of new antibiotics. LL-37, a unique Cathelicidin species found in humans, exhibits a wide range of bioactive properties, including direct bactericidal effects, inflammation regulation, and LPS neutralization. KR-12, the smallest yet potent peptide fragment of LL-37, has been modified to create more effective antimicrobials. In this study, we designed two myristoylated derivatives of KR-12, referred to as Myr-KR-12N and Myr-KR-12C. These derivatives displayed remarkable ability to spontaneously assemble into nanoparticles when mixed with deionized water. Myristoylated KR-12 derivatives exhibited broad-spectrum and intensified bactericidal activity by disrupting bacterial cell membranes. In particular, Myr-KR-12N showed superior capability to rescue mice from lethal E. coli-induced sepsis in comparison with the conventional antibiotic meropenem. We also confirmed that the myristoylated KR-12 nanobiotic possesses significant LPS binding capacity and effectively reduces inflammation in vitro. In an in vivo context, Myr-KR-12N outperformed polymyxin B in rescuing mice from LPS-induced sepsis. Crucially, toxicological assessments revealed that neither Myr-KR-12N nor Myr-KR-12C nanobiotics induced meaningful hemolysis or caused damage to the liver and kidneys. Collectively, our study has yielded an innovative nanobiotic with dual capabilities of bactericidal action and LPS-neutralization, offering substantial promise for advancing the clinical translation of antimicrobial peptides and the development of novel antibiotics. This addresses the critical need for effective solutions to combat Gram-negative sepsis, a pressing global medical challenge.
Collapse
Affiliation(s)
- Ruyi Lei
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Chujun Yang
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Yaqi Sun
- China National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Dejian Li
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Liman Hao
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Yang Li
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Shuijing Wu
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Hui Li
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Chao Lan
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Xiangming Fang
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
32
|
Kulawik P, Jamróz E, Tkaczewska J, Vlčko T, Zając M, Guzik P, Janik M, Tadele W, Golian J, Milosavljević V. Application of antimicrobial chitosan-Furcellaran-hydrolysate gelatin edible coatings enriched with bioactive peptides in shelf-life extension of pork loin stored at 4 and -20 °C. Int J Biol Macromol 2024; 254:127865. [PMID: 37939757 DOI: 10.1016/j.ijbiomac.2023.127865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/03/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
In this study, the authors investigate the preservative capabilities of edible coatings comprising a blend of chitosan, furcellaran and gelatin hydrolysate enhanced with the bioactive peptides RW4 and LL37. The preservative effects on pork samples stored for 21 days at 4 °C and 6 months at -20 °C were studied, while examining changes in microbiological contamination, pH levels, water activity and sensory attributes. Microbiological analyses reveal the coatings' antimicrobial efficacy against aerobic bacteria, microscopic fungi and yeasts, particularly during the initial storage period, when coated samples exhibit microbial reductions of 0.5-2 log CFU/g compared to the controls. The coatings have no discernible impact on water activity during storage in refrigerated or freezer conditions. Notably, differences in pH development can be observed between the coated and control samples, potentially attributable to the antimicrobial action of the coatings. Sensory analysis allows to highlight the inhibition of deterioration related to sensory attributes through the use of edible coatings. In conclusion, employing bioactive peptide-enriched edible coatings holds promise for extending the shelf-life of perishable foods.
Collapse
Affiliation(s)
- Piotr Kulawik
- Department of Animal Products Processing, University of Agriculture, Balicka 122, PL-30-149, Kraków, Poland.
| | - Ewelina Jamróz
- Department of Chemistry, University of Agriculture, Balicka 122, PL-30-149, Kraków, Poland
| | - Joanna Tkaczewska
- Department of Animal Products Processing, University of Agriculture, Balicka 122, PL-30-149, Kraków, Poland
| | - Tomáš Vlčko
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949-76, Nitra, Slovakia
| | - Marzena Zając
- Department of Animal Products Processing, University of Agriculture, Balicka 122, PL-30-149, Kraków, Poland
| | - Paulina Guzik
- Department of Animal Products Processing, University of Agriculture, Balicka 122, PL-30-149, Kraków, Poland
| | - Magdalena Janik
- Department of Chemistry, University of Agriculture, Balicka 122, PL-30-149, Kraków, Poland
| | - Wondyfraw Tadele
- Department of Animal Products Processing, University of Agriculture, Balicka 122, PL-30-149, Kraków, Poland; Department of Food Engineering, Faculty of Chemical and Food Engineering, Bahir Dar Institute of Technology, Bahir Dar University, P.O. Box 26, Bahir Dar, Ethiopia
| | - Jozef Golian
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949-76, Nitra, Slovakia
| | - Vedran Milosavljević
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
| |
Collapse
|
33
|
Demarbaix T, Daele UV, Meirte J, Anthonissen M, Maertens K, Moortgat P. Possible benefits of food supplementation or diet in scar management: A scoping review. Scars Burn Heal 2024; 10:20595131241282105. [PMID: 39280762 PMCID: PMC11402062 DOI: 10.1177/20595131241282105] [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] [Indexed: 09/18/2024] Open
Abstract
Aim The evidence regarding a potential role of food supplementation as an adjunct therapy in scar aftercare is limited. In this scoping review we aim to provide an overview of the possible beneficial role of supplementations in aftercare settings. Method After formulating the research question and accompanying key words, a comprehensive search for relevant publications was performed using PubMed and Web of Science. Two authors independently identified and checked each study against the inclusion criteria. All data was collected and summarized for further discussion. Results After screening, 11 studies were included in the qualitative synthesis. Four studies including human subjects showed a promising connection between scar improvement and supplementation of vitamin D, omega-3 fatty-acids or a Solanaceae-free diet and lower omega-6 fatty-acid intake. Most of the studies were performed on in-vitro models. Preliminary evidence confirmed the beneficial role of vitamin D. Curcumin- and quercetin-supplementation were linked to decreased fibroblast proliferation. Vitamin C enhanced collagen production in healthy as well as keloidal dermal fibroblasts. Chitin stimulated cell-proliferation in human fibroblasts and keratinocytes. Conclusion The findings suggest early potential benefits of additional food supplementation in scar management for scars but provide no clear evidence. To establish guidelines or gather more evidence on food supplementation, studies involving human subjects (in vivo) are essential. The intricacies associated with nutritional studies in vivo present multifaceted challenges. It should be emphasized that substantial additional evidence is required before aspects such as timing and dosage of supplementation could be addressed for clinical application. Lay Summary Aim: This scoping review looks at whether taking food supplements might help with scar care alongside standard scar management following burn injury. Little information is thought to be available on this subject. An up-to-date review of the literature was undertaken to assimilate the body of evidence and determine if a consensus could be drawn.Method: A specific research question was designed and search conducted in scientific databases like PubMed and Web of Science. Two of our team members carefully selected and reviewed each study to determine which studies met the inclusion or exclusion criteria. All studies that met the inclusion criteria were then reviewed and the information collated to enable conclusions to be drawn.Results: Eleven studies met the inclusion criteria and were used to formulate the conclusions drawn. Four studies showed that taking vitamin D, omega-3 fatty acids, a diet without certain vegetables (Solanaceae), and eating less omega-6 fatty acids might help improve scars. It is important to note that most studies (seven out of 11) were carried out in a laboratory and not with real people. These lab studies showed that vitamin D might be helpful. Supplements like curcumin and quercetin seemed to slow down the growth of skin cells like fibroblasts and keratinocytes. Vitamin C aided collagen synthesis, which is important for healthy skin, in both normal and keloid scar cells. Another substance, chitin, was also found to help skin cells and keratinocytes grow better.Conclusion: Our findings point to some early possible benefits of taking extra nutrient supplements for managing scars but do not provide clear evidence. More research is required to enable the development of supplement recommendation and guidelines to be produced. Future research should focus on human trials but do keep in mind that carrying out supplement studies with people is more complicated. The evidence provided by this scoping review is insufficient to recommend the intake of any supplements or the imposition of dietary restrictions for the purpose of managing scars.
Collapse
Affiliation(s)
- Thibau Demarbaix
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
| | - Ulrike Van Daele
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
- Department of Rehabilitation Sciences & Physiotherapy, Research group MOVANT, University of Antwerp, Antwerp, Belgium
| | - Jill Meirte
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
- Department of Rehabilitation Sciences & Physiotherapy, Research group MOVANT, University of Antwerp, Antwerp, Belgium
| | - Mieke Anthonissen
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
- Department of Rehabilitation Sciences & Physiotherapy, Research group MOVANT, University of Antwerp, Antwerp, Belgium
- Department of Rehabilitation Sciences, KU Leuven, Leuven, Belgium
| | - Koen Maertens
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
- Vrije Universiteit Brussel, Clinical and Lifespan Psychology, Brussels, Belgium
| | - Peter Moortgat
- OSCARE, Organisation for Burns, Scar Aftercare and Research, Antwerp, Belgium
| |
Collapse
|
34
|
McCain J, Martínez SR, Fungo F, Sakaya A, Cosa G. Two-Pronged Dormant Photosensitizer-Antibiotic Bacterial Inactivation: Mechanism, Dosage, and Cellular Evolution Visualized at the Single-Cell Level. J Am Chem Soc 2023; 145:28124-28136. [PMID: 38095965 DOI: 10.1021/jacs.3c10034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Innovative therapeutic approaches are required to battle the rise of antibiotic-resistant bacterial strains. Tapping on reactive oxygen species (ROS) generation in bacteria induced by bactericidal antibiotics, here we report a two-pronged strategy for bacterial inactivation relying on the synergistic combination of a bactericidal antibiotic and newly designed dormant photosensitizers (DoPSs) that activate in the presence of ROS. Intramolecular quenching renders DoPS inert in the presence of light. ROS trapping by DoPS aborts the quenching mechanism unmasking, in equal proportions, singlet oxygen (1O2) sensitization and fluorescence emission. Juxtaposed antioxidant-prooxidant activity built within our DoPS enables (i) initial activation of a few molecules by ROS and (ii) subsequent rapid activation of all DoPS in a bacterium via a domino effect mediated by photogenerated 1O2. Bulk colony forming unit studies employing the minimum inhibitory concentration of the antibiotic illustrate rapid and selective inactivation of Escherichia coli and Pseudomonas aeruginosa only in the presence of light, antibiotic, and DoPS. Single-cell, real-time imaging studies on E. coli reveal an autocatalytic progression of DoPS activation from focal points, providing a unique amplification system for sensing. Single-cell analysis further illustrates the impact of DoPS cellular loading on the rate of DoPS activation and cell death times and on the 1O2 dosing necessary for cell death to occur. Our two-pronged therapy discriminates based on cell metabolites and has the potential to result in lower toxicity, pave the way to reduced drug resistance, and provide insightful mechanistic information about bacterial membrane response to 1O2.
Collapse
Affiliation(s)
- Julia McCain
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, Montreal, QC H3A 0B8, Canada
| | - Sol R Martínez
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, Montreal, QC H3A 0B8, Canada
| | - Florencia Fungo
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, Montreal, QC H3A 0B8, Canada
| | - Aya Sakaya
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, Montreal, QC H3A 0B8, Canada
| | - Gonzalo Cosa
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, Montreal, QC H3A 0B8, Canada
| |
Collapse
|
35
|
Ozola L, Pilmane M. Local Defense Factors in Cleft-Affected Palate in Children before and during Milk Dentition Age: A Pilot Study. J Pers Med 2023; 14:27. [PMID: 38248728 PMCID: PMC10817640 DOI: 10.3390/jpm14010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
One of the most frequent congenital orofacial defects is the cleft lip and palate. Local tissue defense factors are known to be important in immune response and inflammatory and healing processes in the cleft tissue; however, they have only been researched in older children during mixed dentition. Thus, the aim of this study is to assess the distribution of LL-37, CD-163, IL-10, HBD-2, HBD-3, and HBD-4 in children before and during milk dentition. The unique and rare material of palate tissue was obtained from 13 patients during veloplastic surgeries during the time span of 20 years. Immunohistochemistry, light microscopy, semi-quantitative evaluation, and non-parametric statistical analysis were used. A significant decrease in HBD-3 and HBD-4 in the connective tissue was found, as well as several mutual statistically significant and strong correlations between HBD-2, HBD-3, HBD-4, and LL-37. Deficiency of HBD-3 and HBD-4 suggests promotion of chronic inflammation. The scarcity of HBD-4 could be connected to the different signaling pathways of dental pulp cells. Mutual correlations imply changes in the epithelial barrier, amplified healing efficiency, and increased antibacterial line of defense. Deprivation of changes in IL-10 quantity points to possible suppression of the factor. The presence of similar CD-163 immunoreactive substances produced by M2 macrophages was also observed.
Collapse
Affiliation(s)
- Laura Ozola
- Institute of Anatomy and Anthropology, Riga Stradins University, Kronvalda Boulevard 9, LV-1010 Riga, Latvia
| | - Mara Pilmane
- Institute of Anatomy and Anthropology, Riga Stradins University, Kronvalda Boulevard 9, LV-1010 Riga, Latvia
| |
Collapse
|
36
|
Salinas-Restrepo C, Naranjo-Duran AM, Quintana J, Bueno J, Guzman F, Hoyos Palacio LM, Segura C. Short Antimicrobial Peptide Derived from the Venom Gland Transcriptome of Pamphobeteus verdolaga Increases Gentamicin Susceptibility of Multidrug-Resistant Klebsiella pneumoniae. Antibiotics (Basel) 2023; 13:6. [PMID: 38275316 PMCID: PMC10812672 DOI: 10.3390/antibiotics13010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 01/27/2024] Open
Abstract
Infectious diseases account for nine percent of annual human deaths, and the widespread emergence of antimicrobial resistances threatens to significantly increase this number in the coming decades. The prospect of antimicrobial peptides (AMPs) derived from venomous animals presents an interesting alternative for developing novel active pharmaceutical ingredients (APIs). Small, cationic and amphiphilic peptides were predicted from the venom gland transcriptome of Pamphobeteus verdolaga using a custom database of the arthropod's AMPs. Ninety-four candidates were chemically synthesized and screened against ATCC® strains of Escherichia coli and Staphylococcus aureus. Among them, one AMP, named PvAMP66, showed broad-spectrum antimicrobial properties with selectivity towards Gram-negative bacteria. It also exhibited activity against Pseudomonas aeruginosa, as well as both an ATCC® and a clinically isolated multidrug-resistant (MDR) strain of K. pneumoniae. The scanning electron microscopy analysis revealed that PvAMP66 induced morphological changes of the MDR K. pneumoniae strain suggesting a potential "carpet model" mechanism of action. The isobologram analysis showed an additive interaction between PvAMP66 and gentamicin in inhibiting the growth of MDR K. pneumoniae, leading to a ten-fold reduction in gentamicin's effective concentration. A cytotoxicity against erythrocytes or peripheral blood mononuclear cells was observed at concentrations three to thirteen-fold higher than those exhibited against the evaluated bacterial strains. This evidence suggests that PvAMP66 can serve as a template for the development of AMPs with enhanced activity and deserves further pre-clinical studies as an API in combination therapy.
Collapse
Affiliation(s)
- Cristian Salinas-Restrepo
- Grupo Toxinología, Alternativas Terapéuticas y Alimentarias, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín 050012, Colombia; (C.S.-R.); (A.M.N.-D.)
| | - Ana María Naranjo-Duran
- Grupo Toxinología, Alternativas Terapéuticas y Alimentarias, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín 050012, Colombia; (C.S.-R.); (A.M.N.-D.)
| | - Juan Quintana
- Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín 050012, Colombia;
| | - Julio Bueno
- Grupo Reproducción, Facultad de Medicina, Universidad de Antioquia, Medellín 050012, Colombia;
| | - Fanny Guzman
- Núcleo Biotecnología Curauma (NBC), Pontificia Universidad Católica de Valparaíso, Valparaíso 3100000, Chile;
| | - Lina M. Hoyos Palacio
- Escuela de Ciencias de la Salud, Grupo de Investigación Biología de Sistemas, Universidad Pontificia Bolivariana, Medellín 050031, Colombia;
| | - Cesar Segura
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Medellín 050012, Colombia
| |
Collapse
|
37
|
Bouhrour N, van der Reijden TJK, Voet MM, Schonkeren-Ravensbergen B, Cordfunke RA, Drijfhout JW, Bendali F, Nibbering PH. Novel Antibacterial Agents SAAP-148 and Halicin Combat Gram-Negative Bacteria Colonizing Catheters. Antibiotics (Basel) 2023; 12:1743. [PMID: 38136778 PMCID: PMC10741160 DOI: 10.3390/antibiotics12121743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
The antibiotic management of catheter-related infections (CRIs) often fails owing to the emergence of antimicrobial-resistant strains and/or biofilm/persister apparitions. Thus, we investigated the efficacy of two novel antimicrobial agents, i.e., the synthetic peptide SAAP-148 and the novel antibiotic halicin, against Gram-negative bacteria (GNB) colonizing catheters. The antibacterial, anti-biofilm, and anti-persister activities of both agents were evaluated against Acinetobacter baumannii, Escherichia coli, and Klebsiella pneumoniae strains. The enrolled strains were isolated from catheters and selected based on their resistance to at least three antibiotic classes and biofilm formation potential. Furthermore, the hemolysis and endotoxin neutralization abilities of these agents were explored. The bactericidal activity of both agents was reduced in urine and plasma as compared to buffered saline. In a dose-dependent manner, SAAP-148 and halicin reduced bacterial counts in 24 h preformed biofilms on silicone elastomer discs and eliminated persisters originating from antibiotic-exposed mature 7-day biofilms, with halicin being less effective than SAAP-148. Importantly, SAAP-148 and halicin acted synergistically on E. coli and K. pneumoniae biofilms but not on A. baumannii biofilms. The peptide, but not halicin, decreased the production of IL-12p40 upon exposure to UV-killed bacteria. This preliminary study showed that SAAP-148 and halicin alone/in combination are promising candidates to fight GNB colonizing catheters.
Collapse
Affiliation(s)
- Nesrine Bouhrour
- Laboratoire de Microbiologie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (T.J.K.v.d.R.); (M.M.V.); (B.S.-R.); (P.H.N.)
| | - Tanny J. K. van der Reijden
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (T.J.K.v.d.R.); (M.M.V.); (B.S.-R.); (P.H.N.)
| | - Michella M. Voet
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (T.J.K.v.d.R.); (M.M.V.); (B.S.-R.); (P.H.N.)
| | - Bep Schonkeren-Ravensbergen
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (T.J.K.v.d.R.); (M.M.V.); (B.S.-R.); (P.H.N.)
| | - Robert A. Cordfunke
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (R.A.C.); (J.W.D.)
| | - Jan Wouter Drijfhout
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (R.A.C.); (J.W.D.)
| | - Farida Bendali
- Laboratoire de Microbiologie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia 06000, Algeria
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (T.J.K.v.d.R.); (M.M.V.); (B.S.-R.); (P.H.N.)
| |
Collapse
|
38
|
Subaer S, Hartati H, Ramadhan I, Ismayanti H, Setiawan A. A Simple Synthesis and Microstructure Analysis of Human Peptide LL-37@Gold Nanoparticles (Known as LL-37@AuNPs) Conjugates as Antimicrobials and Substances for Wound Healing. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7675. [PMID: 38138816 PMCID: PMC10744923 DOI: 10.3390/ma16247675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
The basis of the present study is a straightforward method involving fewer chemical species for conjugating gold nanoparticles (AuNPs) with the antimicrobial peptide LL-37 designated as LL-37@AuNPs. Investigating the microstructure characteristics of the resulting materials and their potential as antibacterial and wound-healing substances are the main objectives of this study. Zeta (ζ) potential, Fourier transform infrared (FTIR), X-ray diffraction (XRD), field effect scanning electron microscopy (FE-SEM), energy dispersive X-ray diffraction (EDS), transmission electron microscopy (TEM), and UV-Vis spectrophotometry were used to analyze the physico-chemical properties of LL-37@AuNPs. The magnitude of LL-37's zeta potential and the LL-37@AuNPs show that the specimens are electrically stable and resistant to flocculation and coagulation. The surface plasmon resonance (RPS) of AuNPs, which is positioned at a wavelength of about 531 nm, was found to be unaffected by the presence of the LL-37 antimicrobial peptide. The FTIR data show the functional group characteristics of the LL-37@AuNPs vibration bands, and the XRD diffractogram confirms the formation of the LL-37@AuNPs conjugate nanocomposite. Based on FE-SEM and TEM data, the bulk of AuNPs were found to have a circular shape, with an average size of about 22.88 ± 8.21 nm. It was discovered that the LL-37@AuNPs had a good ability to inhibit S. aureus from growing. The wound-healing percentage reached 85% on day 12 of the trial, significantly greater than the results of the negative controls. LL-37@AuNPs(4) is the sample that had the highest percentage of wound healing between days 3 and 12. Moreover, sample LL-37@AuNPs(4) contains 0.45 µL of LL-37, whereas sample LL-37@AuNPs(2) contains 0.22 µL of LL-37. The faster wound-healing rate in LL-37@AuNPs(4) was believed to be due to a higher concentration of LL-37, which was able to stop S. aureus from developing while suppressing the inflammation surrounding the wound. The study's findings reveal that LL-37@AuNPs might be made using a straightforward process, making them a powerful antibacterial and therapeutic substance. However, before this discovery is applied in the field of medicine, a more thorough investigation is necessary.
Collapse
Affiliation(s)
- Subaer Subaer
- Physics Department, Faculty of Mathematics and Natural Science, Universitas Negeri Makassar, Makassar 90222, Indonesia; (I.R.); (H.I.)
- Green of Excellence of Green Materials & Technology (CeoGM-Tech) FMIPA, Universitas Negeri Makassar, Makassar 90222, Indonesia;
| | - Hartati Hartati
- Green of Excellence of Green Materials & Technology (CeoGM-Tech) FMIPA, Universitas Negeri Makassar, Makassar 90222, Indonesia;
- Biology Department, Faculty of Mathematics and Natural Science, Universitas Negeri Makassar, Makassar 90222, Indonesia
| | - Imam Ramadhan
- Physics Department, Faculty of Mathematics and Natural Science, Universitas Negeri Makassar, Makassar 90222, Indonesia; (I.R.); (H.I.)
| | - Harlyenda Ismayanti
- Physics Department, Faculty of Mathematics and Natural Science, Universitas Negeri Makassar, Makassar 90222, Indonesia; (I.R.); (H.I.)
| | - Agung Setiawan
- Research Center for Mining Technology, National Research and Innovation Agency (BRIN), Building 820, KST B.J. Habibie, Banten 15314, Indonesia;
| |
Collapse
|
39
|
Chen SY, Chang CK, Lan CY. Antimicrobial peptide LL-37 disrupts plasma membrane and calcium homeostasis in Candida albicans via the Rim101 pathway. Microbiol Spectr 2023; 11:e0255123. [PMID: 37888991 PMCID: PMC10715129 DOI: 10.1128/spectrum.02551-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023] Open
Abstract
IMPORTANCE Candida albicans is a major human fungal pathogen, and antimicrobial peptides are key components of innate immunity. Studying the interplay between C. albicans and human antimicrobial peptides would enhance a better understanding of pathogen-host interactions. Moreover, potential applications of antimicrobial peptides in antifungal therapy have aroused great interest. This work explores new mechanisms of LL-37 against C. albicans and reveals the complex connection among calcium homeostasis, oxidative stress, signaling, and possibly organelle interaction. Notably, these findings support the possible use of antimicrobial peptides to prevent and treat fungal infections.
Collapse
Affiliation(s)
- Sheng-Yuan Chen
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Che-Kang Chang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chung-Yu Lan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
40
|
Koskela SA, Figueiredo CR. From antimicrobial to anticancer: the pioneering works of Prof. Luiz Rodolpho Travassos on bioactive peptides. Braz J Microbiol 2023; 54:2561-2570. [PMID: 37725261 PMCID: PMC10689714 DOI: 10.1007/s42770-023-01118-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
Prof. Luiz Rodolpho Travassos, a distinguished Brazilian scientist, was instrumental in fostering an interdisciplinary research approach that seamlessly combined microbiology and oncology. This work has opened new pathways into the understanding of tumorigenesis and aided in the development of innovative therapeutic tools. One significant area of his work has been the exploration of bioactive peptides, many of which were first identified for their antimicrobial properties. These peptides demonstrate promise as potential cancer therapeutics due to their selectivity, cost-effectiveness, ease of synthesis, low antigenicity, and excellent tissue penetration. Prof. Travassos' pioneering work uncovered on the potential of peptides derived from microbiological sources, such as those obtained using phage display techniques. More importantly, in international cooperation, peptides derived from complementarity-determining regions (CDRs) that showed antimicrobial activity against Candida albicans further showed to be promising tools with cytotoxic properties against cancer cells. Similarly, peptides derived from natural sources, such as the gomesin peptide, not only had shown antimicrobial properties but could treat cutaneous melanoma in experimental models. These therapeutic tools allowed Prof. Travassos and his group to navigate the intricate landscape of factors and pathways that drive cancer development, including persistent proliferative signaling, evasion of tumor suppressor genes, inhibition of programmed cell death, and cellular immortality. This review examines the mechanisms of action of these peptides, aligning them with the universally recognized hallmarks of cancer, and evaluates their potential as drug candidates. It highlights the crucial need for more selective, microbiology-inspired anti-cancer strategies that spare healthy cells, a challenge that current therapies often struggle to address. By offering a comprehensive assessment of Prof. Travassos' innovative contributions and a detailed discussion on the increasing importance of microbiology-derived peptides, this review presents an informed and robust perspective on the possible future direction of cancer therapy.
Collapse
Affiliation(s)
- Saara A Koskela
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Carlos R Figueiredo
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
| |
Collapse
|
41
|
Bhatt T, Dam B, Khedkar SU, Lall S, Pandey S, Kataria S, Ajnabi J, Gulzar SEJ, Dias PM, Waskar M, Raut J, Sundaramurthy V, Vemula PK, Ghatlia N, Majumdar A, Jamora C. Niacinamide enhances cathelicidin mediated SARS-CoV-2 membrane disruption. Front Immunol 2023; 14:1255478. [PMID: 38022563 PMCID: PMC10663372 DOI: 10.3389/fimmu.2023.1255478] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
The continual emergence of SARS-CoV-2 variants threatens to compromise the effectiveness of worldwide vaccination programs, and highlights the need for complementary strategies for a sustainable containment plan. An effective approach is to mobilize the body's own antimicrobial peptides (AMPs), to combat SARS-CoV-2 infection and propagation. We have found that human cathelicidin (LL37), an AMP found at epithelial barriers as well as in various bodily fluids, has the capacity to neutralise multiple strains of SARS-CoV-2. Biophysical and computational studies indicate that LL37's mechanism of action is through the disruption of the viral membrane. This antiviral activity of LL37 is enhanced by the hydrotropic action of niacinamide, which may increase the bioavailability of the AMP. Interestingly, we observed an inverse correlation between LL37 levels and disease severity of COVID-19 positive patients, suggesting enhancement of AMP response as a potential therapeutic avenue to mitigate disease severity. The combination of niacinamide and LL37 is a potent antiviral formulation that targets viral membranes of various variants and can be an effective strategy to overcome vaccine escape.
Collapse
Affiliation(s)
- Tanay Bhatt
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Binita Dam
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Department of Biological Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Sneha Uday Khedkar
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Sahil Lall
- National Centre for Biological Sciences (TIFR), Bangalore, Karnataka, India
| | - Subhashini Pandey
- Integrative Chemical Biology, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Sunny Kataria
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Johan Ajnabi
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Department of Biological Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | | | | | | | | | | | - Praveen Kumar Vemula
- Integrative Chemical Biology, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | | | | | - Colin Jamora
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| |
Collapse
|
42
|
He J, Wang C, Lin G, Xia X, Chen H, Feng Z, Huang Z, Pan X, Zhang X, Wu C, Huang Y. Guard against internal and external: An antibacterial, anti-inflammation and healing-promoting spray gel based on lyotropic liquid crystals for the treatment of diabetic wound. Int J Pharm 2023; 646:123442. [PMID: 37774758 DOI: 10.1016/j.ijpharm.2023.123442] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/05/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023]
Abstract
The diabetic wound is a prevalent and serious complication of diabetes, which easily deteriorates due to susceptibility to infection and difficulty in healing, causing a high risk of amputation and economic burden to patients. Bacterial infection, persistent excessive inflammation, and cellular and angiogenesis disorders are the main reasons for the difficulty of diabetic wound healing. In this study, glycerol monooleate (GMO) was used to prepare lyotropic liquid crystal hydrogel (LLC) containing the natural antimicrobial peptide LL37 and carbenoxolone (CBX) to achieve antibacterial, anti-inflammation, and healing promotion for the treatment of diabetic wounds. The shear-thinning properties of the LLC precursor solution allowed it to be administered in the form of a spray, which perfectly fitted the shape of the wound and transformed into a gel after absorbing wound exudate to act as a wound protective barrier. The faster release of LL37 realized rapid sterilization of wounds, controlled the source of inflammation, and accelerated wound healing. The inflammatory signaling pathway was blocked by the subsequently released CBX, and the spread of the inflammatory response was inhibited and then further weakened. In addition, CBX down-regulated connexin (Cx43) to assist LL37 to promote cell migration and proliferation better. Combined with the pro-angiogenic effect of LL37, the healing of diabetic wounds was significantly accelerated. All these advantages made LL37-CBX-LLC a promising approach for the treatment of chronic diabetic wounds.
Collapse
Affiliation(s)
- Jingyu He
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China
| | - Chen Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China
| | - Gan Lin
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, Guangdong, PR China
| | - Xiao Xia
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China
| | - Hangping Chen
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China
| | - Zhiyuan Feng
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China.
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China
| | - Xuejuan Zhang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China.
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China
| | - Ying Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China.
| |
Collapse
|
43
|
Ng WNI, Kalimuthu S, Law COK, Lee AHC, Lau TCK, Leung YY, Cheung GSP, Neelakantan P. Intracellular bacterial eradication using a novel peptide in vitro. Int Endod J 2023; 56:1360-1372. [PMID: 37615967 DOI: 10.1111/iej.13965] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/09/2023] [Indexed: 08/25/2023]
Abstract
AIM To determine the effect of a novel antimicrobial peptide (AMP; OP145) and cell-penetrating peptide (Octa-arginine/R8) conjugate on the killing of intracellular Enterococcus faecalis, compared to OP145 and an antibiotic combination recommended for regenerative endodontic procedures. METHODOLOGY The biocompatible concentrations of OP145 and OP145-R8 were determined by assessing their cytotoxicity against human macrophages and red blood cells. Spatiotemporal internalization of the peptides into macrophages was investigated qualitatively and quantitatively by confocal laser scanning microscopy and flow cytometry respectively. Killing of extracellular and intracellular E. faecalis OG1RF by the peptides was determined by counting the colony-forming units (CFU). Intracellular antibacterial activity of the peptides was compared to a double antibiotic combination. Confocal microscopy was used to confirm the intracellular bacterial eradication. Significant differences between the different test groups were analysed using one-way analysis of variance. p < .05 was considered to be statistically significant. RESULTS Peptides at a concentration of 7.5 μmol/L were chosen for subsequent experiments based on the results of the alamarBlue™ cell viability assay and haemolytic assay. OP145-R8 selectively internalized into lysosomal compartments and the cytosol of macrophages. Conjugation with R8 improved the internalization of OP145 into macrophages in a temporal manner (70.53% at 1 h to 77.13% at 2 h), while no temporal increase was observed for OP145 alone (60.53% at 1 h with no increase at 2 h). OP145-R8 demonstrated significantly greater extracellular and intracellular antibacterial activity compared to OP145 at all investigated time-points and concentrations (p < .05). OP145-R8 at 7.5 μmol/L eradicated intracellular E. faecalis after 2 h (3.5 log reduction compared to the control; p < .05), while the antibiotics could not reduce more than 0.5 log CFU compared to the control (p > .05). Confocal microscopy showed complete absence of E. faecalis within the OP145-R8 treated macrophages. CONCLUSIONS The results of this study demonstrated that the conjugation of an AMP OP145 to a cell-penetrating peptide R8 eradicated extracellular and intracellular E. faecalis OG1RF without toxic effects on the host cells.
Collapse
Affiliation(s)
- Wing Nok Isaac Ng
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, Hong Kong
| | | | - Carmen Oi Kwan Law
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, Hong Kong
| | | | - Terrence Chi Kong Lau
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, Hong Kong
| | - Yiu Yan Leung
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, Hong Kong
| | - Gary Shun Pan Cheung
- Department of Dental Surgery, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Prasanna Neelakantan
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, Hong Kong
- Department of Endodontics, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, California, USA
| |
Collapse
|
44
|
Yang X, Hua C, Lin L, Ganting Z. Antimicrobial peptides as potential therapy for gastrointestinal cancers. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2831-2841. [PMID: 37249612 DOI: 10.1007/s00210-023-02536-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023]
Abstract
Since conventional therapy faces limitations in the field of different cancers as well as gastrointestinal cancers, that decrease the survival rate of patients, there is an urgent need to find new effective therapeutic approaches without the adverse effects of the traditional agents. Antimicrobial peptides (AMPs) attract much attention and are well known for their role in innate immunity. These peptides, in addition to their antimicrobial activity, exhibit strong anticancer potential against various types of malignancy. AMPs specifically target tumor cells and have selective toxicity for these cells without affecting normal cells. Here we aim to comprehensively overview the current knowledge in the field of using AMPs as novel therapeutic agents for gastrointestinal cancer.
Collapse
Affiliation(s)
- Xiaoxia Yang
- Heping Hospital Attached to Changzhi Medical College, Changzhi, 046000, China
| | - Cui Hua
- Tangshan Fengnan District Traditional Chinese Medicine Hospital, Tangshan, 063000, China.
| | - Lin Lin
- Tangshan Hongci Hospital, Tangshan, 063000, China
| | - Zhao Ganting
- Heping Hospital Attached to Changzhi Medical College, Changzhi, 046000, China
| |
Collapse
|
45
|
Brown AN, Anderson MT, Smith SN, Bachman MA, Mobley HLT. Conserved metabolic regulator ArcA responds to oxygen availability, iron limitation, and cell envelope perturbations during bacteremia. mBio 2023; 14:e0144823. [PMID: 37681955 PMCID: PMC10653796 DOI: 10.1128/mbio.01448-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 09/09/2023] Open
Abstract
IMPORTANCE Infections of the bloodstream are life-threatening and can result in sepsis. Gram-negative bacteria cause a significant portion of bloodstream infections, which is also referred to as bacteremia. The long-term goal of our work is to understand how such bacteria establish and maintain infection during bacteremia. We have previously identified the transcription factor ArcA, which promotes fermentation in bacteria, as a likely contributor to the growth and survival of bacteria in this environment. Here, we study ArcA in the Gram-negative species Citrobacter freundii, Klebsiella pneumoniae, and Serratia marcescens. Our findings aid in determining how these bacteria sense their environment, utilize nutrients, and generate energy while countering the host immune system. This information is critical for developing better models of infection to inform future therapeutic development.
Collapse
Affiliation(s)
- Aric N. Brown
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mark T. Anderson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sara N. Smith
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Michael A. Bachman
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Harry L. T. Mobley
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
46
|
Huang LH, Rau CS, Liu YW, Lin HP, Wu YC, Tsai CW, Chien PC, Wu CJ, Huang CY, Hsieh TM, Hsieh CH. Cathelicidin Antimicrobial Peptide Acts as a Tumor Suppressor in Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:15652. [PMID: 37958632 PMCID: PMC10647698 DOI: 10.3390/ijms242115652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is associated with high rates of metastasis and recurrence, and is one of the most common causes of cancer-associated death worldwide. This study examined the protein changes within circulating exosomes in patients with HCC against those in healthy people using isobaric tags for a relative or absolute quantitation (iTRAQ)-based quantitative proteomics analysis. The protein levels of von Willebrand factor (VWF), cathelicidin antimicrobial peptide (CAMP), and proteasome subunit beta type-2 (PSMB2) were altered in HCC. The increased levels of VWF and PSMB2 but decreased CAMP levels in the serum of patients with HCC were validated by enzyme-linked immunosorbent assays. The level of CAMP (the only cathelicidin found in humans) also decreased in the circulating exosomes and buffy coat of the HCC patients. The serum with reduced levels of CAMP protein in the HCC patients increased the cell proliferation of Huh-7 cells; this effect was reduced following the addition of CAMP protein. The depletion of CAMP proteins in the serum of healthy people enhances the cell proliferation of Huh-7 cells. In addition, supplementation with synthetic CAMP reduces cell proliferation in a dose-dependent manner and significantly delays G1-S transition in Huh-7 cells. This implies that CAMP may act as a tumor suppressor in HCC.
Collapse
Affiliation(s)
- Lien-Hung Huang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (L.-H.H.); (C.-S.R.)
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (L.-H.H.); (C.-S.R.)
| | - Yueh-Wei Liu
- Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Hui-Ping Lin
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-P.L.); (Y.-C.W.); (C.-W.T.); (P.-C.C.); (C.-J.W.); (C.-Y.H.); (T.-M.H.)
| | - Yi-Chan Wu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-P.L.); (Y.-C.W.); (C.-W.T.); (P.-C.C.); (C.-J.W.); (C.-Y.H.); (T.-M.H.)
| | - Chia-Wen Tsai
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-P.L.); (Y.-C.W.); (C.-W.T.); (P.-C.C.); (C.-J.W.); (C.-Y.H.); (T.-M.H.)
| | - Peng-Chen Chien
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-P.L.); (Y.-C.W.); (C.-W.T.); (P.-C.C.); (C.-J.W.); (C.-Y.H.); (T.-M.H.)
| | - Chia-Jung Wu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-P.L.); (Y.-C.W.); (C.-W.T.); (P.-C.C.); (C.-J.W.); (C.-Y.H.); (T.-M.H.)
| | - Chun-Ying Huang
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-P.L.); (Y.-C.W.); (C.-W.T.); (P.-C.C.); (C.-J.W.); (C.-Y.H.); (T.-M.H.)
| | - Ting-Min Hsieh
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (H.-P.L.); (Y.-C.W.); (C.-W.T.); (P.-C.C.); (C.-J.W.); (C.-Y.H.); (T.-M.H.)
| | - Ching-Hua Hsieh
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
47
|
Cafaro V, Bosso A, Di Nardo I, D’Amato A, Izzo I, De Riccardis F, Siepi M, Culurciello R, D’Urzo N, Chiarot E, Torre A, Pizzo E, Merola M, Notomista E. The Antimicrobial, Antibiofilm and Anti-Inflammatory Activities of P13#1, a Cathelicidin-like Achiral Peptoid. Pharmaceuticals (Basel) 2023; 16:1386. [PMID: 37895857 PMCID: PMC10610514 DOI: 10.3390/ph16101386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Cationic antimicrobial peptides (CAMPs) are powerful molecules with antimicrobial, antibiofilm and endotoxin-scavenging activities. These properties make CAMPs very attractive drugs in the face of the rapid increase in multidrug-resistant (MDR) pathogens, but they are limited by their susceptibility to proteolytic degradation. An intriguing solution to this issue could be the development of functional mimics of CAMPs with structures that enable the evasion of proteases. Peptoids (N-substituted glycine oligomers) are an important class of peptidomimetics with interesting benefits: easy synthetic access, intrinsic proteolytic stability and promising bioactivities. Here, we report the characterization of P13#1, a 13-residue peptoid specifically designed to mimic cathelicidins, the best-known and most widespread family of CAMPs. P13#1 showed all the biological activities typically associated with cathelicidins: bactericidal activity over a wide spectrum of strains, including several ESKAPE pathogens; the ability to act in combination with different classes of conventional antibiotics; antibiofilm activity against preformed biofilms of Pseudomonas aeruginosa, comparable to that of human cathelicidin LL-37; limited toxicity; and an ability to inhibit LPS-induced proinflammatory effects which is comparable to that of "the last resource" antibiotic colistin. We further studied the interaction of P13#1 with SDS, LPSs and bacterial cells by using a fluorescent version of P13#1. Finally, in a subcutaneous infection mouse model, it showed antimicrobial and anti-inflammatory activities comparable to ampicillin and gentamicin without apparent toxicity. The collected data indicate that P13#1 is an excellent candidate for the formulation of new antimicrobial therapies.
Collapse
Affiliation(s)
- Valeria Cafaro
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Andrea Bosso
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Ilaria Di Nardo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Assunta D’Amato
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, 84084 Fisciano, Italy; (A.D.); (I.I.); (F.D.R.)
| | - Irene Izzo
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, 84084 Fisciano, Italy; (A.D.); (I.I.); (F.D.R.)
| | - Francesco De Riccardis
- Department of Chemistry and Biology “A. Zambelli”, University of Salerno, 84084 Fisciano, Italy; (A.D.); (I.I.); (F.D.R.)
| | - Marialuisa Siepi
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Rosanna Culurciello
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Nunzia D’Urzo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | | | | | - Elio Pizzo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Marcello Merola
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| | - Eugenio Notomista
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (V.C.); (A.B.); (I.D.N.); (M.S.); (R.C.); (N.D.); (E.P.)
| |
Collapse
|
48
|
Wei Z, Rolle MW, Camesano TA. Characterization of LL37 Binding to Collagen through Peptide Modification with a Collagen-Binding Domain. ACS OMEGA 2023; 8:35370-35381. [PMID: 37779975 PMCID: PMC10536065 DOI: 10.1021/acsomega.3c05328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023]
Abstract
Collagen-based biomaterials loaded with antimicrobial peptides (AMPs) present a promising approach for promoting wound healing while providing protection against infections. In our previous work, we modified the AMP LL37 by incorporating a collagen-binding domain (cCBD) as an anchoring unit for collagen-based wound dressings. We demonstrated that cCBD-modified LL37 (cCBD-LL37) exhibited improved retention on collagen after washing with PBS. However, the binding mechanism of cCBD-LL37 to collagen remained to be elucidated. In this study, we found that cCBD-LL37 showed a slightly higher affinity for collagen compared to LL37. Our results indicated that cCBD inhibited cCBD-LL37 binding to collagen but did not fully eliminate the binding. This suggests that cCBD-LL37 binding to collagen may involve more than just one-site-specific binding through the collagen-binding domain, with non-specific interactions also playing a role. Electrostatic studies revealed that both LL37 and cCBD-LL37 interact with collagen via long-range electrostatic forces, initiating low-affinity binding that transitions to close-range or hydrophobic interactions. Circular dichroism analysis showed that cCBD-LL37 exhibited enhanced structural stability compared to LL37 under varying ionic strengths and pH conditions, implying potential improvements in antimicrobial activity. Moreover, we demonstrated that the release of LL37 and cCBD-LL37 into the surrounding medium was influenced by the electrostatic environment, but cCBD could enhance the retention of peptide on collagen scaffolds. Collectively, these results provide important insights into cCBD-modified AMP-binding mechanisms and suggest that the addition of cCBD may enhance peptide structural stability and retention under varying electrostatic conditions.
Collapse
Affiliation(s)
- Ziqi Wei
- Department
of Chemical Engineering, Worcester Polytechnic
Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| | - Marsha W. Rolle
- Department
of Biomedical Engineering, Worcester Polytechnic
Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| | - Terri A. Camesano
- Department
of Chemical Engineering, Worcester Polytechnic
Institute, 100 Institute Road, Worcester, Massachusetts 01609, United States
| |
Collapse
|
49
|
Zhang H, Dong M, Xu H, Li H, Zheng A, Sun G, Jin W. Recombinant Lactococcus lactis Expressing Human LL-37 Prevents Deaths from Viral Infections in Piglets and Chicken. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10155-6. [PMID: 37743432 DOI: 10.1007/s12602-023-10155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 09/26/2023]
Abstract
Novel antibiotic substitutes are increasingly in demand in the animal husbandry industry. An oral recombinant Lactococcus lactis (L. lactis) expressing human LL-37 (oral LL-37) was developed and its safety and antiviral effectiveness in vivo was tested. In addition to impairing liposome integrity, LL-37 polypeptide from recombinant L. lactis could prevent the host cell infection by a variety of viruses, including recombinant SARS, SARS-CoV-2, Ebola virus, and vesicular stomatitis virus G. Subchronic toxicity studies performed on Sprague-Dawley rats showed that no cumulative toxicity was found during short-term intervention. Oral LL-37 treatment after the onset of fever could reduce mortality in piglets infected with porcine reproductive and respiratory syndrome virus. Moreover, body weight gain of piglets receiving treatment was progressively restored, and nucleic acid positive rebound was not undetected after discontinuation. Oral LL-37 consistently increased the lifespan of chickens infected with Newcastle viruses. These findings suggested a potential use of recombinantly modified microorganisms in veterinary medicine.
Collapse
Affiliation(s)
- Hanlin Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Huihui Xu
- Jilin Yuanheyuan Bioengineering Co., Ltd. Changchun, Jilin Province, 130000, China
| | - Hongyue Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Aihua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Gang Sun
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
50
|
Abd El-Aal AAA, Jayakumar FA, Lahiri C, Tan KO, Reginald K. Novel cationic cryptides in Penaeus vannamei demonstrate antimicrobial and anti-cancer activities. Sci Rep 2023; 13:14673. [PMID: 37673929 PMCID: PMC10482825 DOI: 10.1038/s41598-023-41581-9] [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: 03/04/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023] Open
Abstract
Cryptides are a subfamily of bioactive peptides that exist in all living organisms. They are latently encrypted in their parent sequences and exhibit a wide range of biological activities when decrypted via in vivo or in vitro proteases. Cationic cryptides tend to be drawn to the negatively charged membranes of microbial and cancer cells, causing cell death through various mechanisms. This makes them promising candidates for alternative antimicrobial and anti-cancer therapies, as their mechanism of action is independent of gene mutations. In the current study, we employed an in silico approach to identify novel cationic cryptides with potential antimicrobial and anti-cancer activities in atypical and systematic strategy by reanalysis of a publicly available RNA-seq dataset of Pacific white shrimp (Penaus vannamei) in response to bacterial infection. Out of 12 cryptides identified, five were selected based on their net charges and potential for cell penetration. Following chemical synthesis, the cryptides were assayed in vitro to test for their biological activities. All five cryptides demonstrated a wide range of selective activity against the tested microbial and cancer cells, their anti-biofilm activities against mature biofilms, and their ability to interact with Gram-positive and negative bacterial membranes. Our research provides a framework for a comprehensive analysis of transcriptomes in various organisms to uncover novel bioactive cationic cryptides. This represents a significant step forward in combating the crisis of multi-drug-resistant microbial and cancer cells, as these cryptides neither induce mutations nor are influenced by mutations in the cells they target.
Collapse
Affiliation(s)
- Amr Adel Ahmed Abd El-Aal
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
- Marine Microbiology Lab., National Institute of Oceanography and Fisheries (NIOF), Alexandria, 84511, Egypt
| | - Fairen Angelin Jayakumar
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chandrajit Lahiri
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
- Department of Biotechnology, Atmiya University, Rajkot, Gujarat, 360005, India
| | - Kuan Onn Tan
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia
| | - Kavita Reginald
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|