1
|
Lennard PR, Hiemstra PS, Dorin JR, Nibbering PH. SAAP-148 and halicin exhibit synergistic antimicrobial activity against antimicrobial-resistant bacteria in skin but not airway epithelial culture models. JAC Antimicrob Resist 2025; 7:dlaf050. [PMID: 40224359 PMCID: PMC11986330 DOI: 10.1093/jacamr/dlaf050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Background The escalating global threat of antimicrobial resistance (AMR) necessitates the development of novel antimicrobial agents, innovative strategies, and representative infection models to combat AMR bacterial infections. Host defence peptides (HDPs) and their derivatives have been proposed as complements to conventional antibiotics due to their antibacterial activity and modulation of the immune response. Objectives This study investigated the novel use of the HDP-derived synthetic antibacterial and anti-biofilm peptide (SAAP)-148 as a pretreatment in epithelial tissue models to prevent colonization by AMR bacteria. The combined activities of SAAP-148 pretreatment with post-infection halicin to treat infections were also explored. Methods Employing cultured human skin equivalents (HSEs) and primary bronchial epithelial cells (PBECs) as models of tissue infection, we examined the prophylactic and therapeutic effects of SAAP-148, both singularly and in combination with the repurposed antibiotic halicin, against AMR bacteria. We additionally interrogated the response of HSE and PBEC cultures to SAAP-148 treatment via confocal microscopy and quantitative PCR of native HDPs and inflammatory cytokine genes. Results Our findings demonstrated that pretreatment with SAAP-148 significantly reduces colonization of HSEs and PBECs by AMR Staphylococcus aureus and Pseudomonas aeruginosa. Confocal microscopy revealed differential uptake and localization of SAAP-148 in these tissues, correlating with its distinct activity in these tissues. SAAP-148 exposure temporarily increased expression of the HDPs cathelicidin (CAMP) and β-defensin 1 (DEFB1), and the cytokine IL-8 (CXCL8), which did not correlate with the transient antibacterial activity observed. Sequential treatment with SAAP-148 prior to infection with AMR S. aureus and post-infection halicin treatment demonstrated synergistic activity in HSEs, whereas this combined activity was indifferent in PBEC cultures. Conclusions These results support SAAP-148 as a candidate for pre-infection prophylaxis and synergistic antibiotic therapy with halicin in skin, broadening the potential of both agents to address AMR bacterial infection.
Collapse
Affiliation(s)
- Patrick R Lennard
- PulmoScience Laboratory, Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
- Laboratory of Infectious Diseases, Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Pieter S Hiemstra
- PulmoScience Laboratory, Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Julia R Dorin
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Peter H Nibbering
- Laboratory of Infectious Diseases, Leiden University Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
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
|
3
|
van Gent ME, Klodzinska SN, Drijfhout JW, Nielsen HM, Nibbering PH. Encapsulation in oleyl-modified hyaluronic acid nanogels substantially improves the clinical potential of the antimicrobial peptides SAAP-148 and Ab-Cath. Eur J Pharm Biopharm 2023; 193:254-261. [PMID: 37944710 DOI: 10.1016/j.ejpb.2023.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/23/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
Antimicrobial peptides (AMPs) are promising alternatives to antibiotics for treatment of antimicrobial resistant (AMR) bacterial infections. However, their narrow therapeutic window due to in vivo toxicity and limited stability hampers their clinical use. Here, we evaluated encapsulation of two amphiphilic AMPs, SAAP-148 and snake cathelicidin Ab-Cath, into oleyl-modified hyaluronic acid (OL-HA) nanogels to improve their selectivity index. The AMP-loaded OL-HA nanogels ranged 181-206 nm in size with a PDI of 0.2, highly negative surface charge (-47 to -48 mV) and moderate encapsulation efficiency (53-63%). The AMP-loaded OL-HA nanogels displayed similar activity in vitro as AMP solutions against AMR Staphylococcus aureus and Acinetobacter baumannii, with a dose-dependent effect over time. Importantly, the AMP-loaded OL-HA nanogels showed decreased cytotoxicity towards human erythrocytes and primary skin fibroblast, thereby improving the selectivity index of SAAP-148 and Ab-Cath by 2- and 16.8-fold, respectively. Particularly, the selectivity of Ab-Cath-loaded OL-HA nanogels has great clinical potential, with an index that reached ≥ 300 for S. aureus and ≥ 3000 for A. baumannii. These findings indicate that OL-HA nanogels are a promising drug delivery system to reduce the cytotoxicity of AMPs without substantially affecting their antimicrobial activity, thereby increasing their selectivity index and potential as therapeutics to combat AMR bacterial infections.
Collapse
Affiliation(s)
- Miriam E van Gent
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, the Netherlands.
| | - Sylvia N Klodzinska
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Jan Wouter Drijfhout
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| | - Hanne M Nielsen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Peter H Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, the Netherlands
| |
Collapse
|
4
|
Ali M, van Gent ME, de Waal AM, van Doodewaerd BR, Bos E, Koning RI, Cordfunke RA, Drijfhout JW, Nibbering PH. Physical and Functional Characterization of PLGA Nanoparticles Containing the Antimicrobial Peptide SAAP-148. Int J Mol Sci 2023; 24:2867. [PMID: 36769188 PMCID: PMC9918011 DOI: 10.3390/ijms24032867] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Synthetic antimicrobial and antibiofilm peptide (SAAP-148) commits significant antimicrobial activities against antimicrobial resistant (AMR) planktonic bacteria and biofilms. However, SAAP-148 is limited by its low selectivity index, i.e., ratio between cytotoxicity and antimicrobial activity, as well as its bioavailability at infection sites. We hypothesized that formulation of SAAP-148 in PLGA nanoparticles (SAAP-148 NPs) improves the selectivity index due to the sustained local release of the peptide. The aim of this study was to investigate the physical and functional characteristics of SAAP-148 NPs and to compare the selectivity index of the formulated peptide with that of the peptide in solution. SAAP-148 NPs displayed favorable physiochemical properties [size = 94.1 ± 23 nm, polydispersity index (PDI) = 0.08 ± 0.1, surface charge = 1.65 ± 0.1 mV, and encapsulation efficiency (EE) = 86.7 ± 0.3%] and sustained release of peptide for up to 21 days in PBS at 37 °C. The antibacterial and cytotoxicity studies showed that the selectivity index for SAAP-148 NPs was drastically increased, by 10-fold, regarding AMR Staphylococcus aureus and 20-fold regarding AMR Acinetobacter baumannii after 4 h. Interestingly, the antibiofilm activity of SAAP-148 NPs against AMR S. aureus and A. baumannii gradually increased overtime, suggesting a dose-effect relationship based on the peptide's in vitro release profile. Using 3D human skin equivalents (HSEs), dual drug SAAP-148 NPs and the novel antibiotic halicin NPs provided a stronger antibacterial response against planktonic and cell-associated bacteria than SAAP-148 NPs but not halicin NPs after 24 h. Confocal laser scanning microscopy revealed the presence of SAAP-148 NPs on the top layers of the skin models in close proximity to AMR S. aureus at 24 h. Overall, SAAP-148 NPs present a promising yet challenging approach for further development as treatment against bacterial infections.
Collapse
Affiliation(s)
- Muhanad Ali
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Miriam E. van Gent
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Amy M. de Waal
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Bjorn R. van Doodewaerd
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Erik Bos
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Roman I. Koning
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Robert A. Cordfunke
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
5
|
van Gent ME, van Baaren T, Kłodzińska SN, Ali M, Dolezal N, van Doodewaerd BR, Bos E, de Waal AM, Koning RI, Drijfhout JW, Nielsen HM, Nibbering PH. Encapsulation of SAAP-148 in Octenyl Succinic Anhydride-Modified Hyaluronic Acid Nanogels for Treatment of Skin Wound Infections. Pharmaceutics 2023; 15:pharmaceutics15020429. [PMID: 36839751 PMCID: PMC9967827 DOI: 10.3390/pharmaceutics15020429] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Chronic wound infections colonized by bacteria are becoming more difficult to treat with current antibiotics due to the development of antimicrobial resistance (AMR) as well as biofilm and persister cell formation. Synthetic antibacterial and antibiofilm peptide (SAAP)-148 is an excellent alternative for treatment of such infections but suffers from limitations related to its cationic peptidic nature and thus instability and possible cytotoxicity, resulting in a narrow therapeutic window. Here, we evaluated SAAP-148 encapsulation in nanogels composed of octenyl succinic anhydride (OSA)-modified hyaluronic acid (HA) to circumvent these limitations. SAAP-148 was efficiently (>98%) encapsulated with high drug loading (23%), resulting in monodispersed anionic OSA-HA nanogels with sizes ranging 204-253 nm. Nanogel lyophilization in presence of polyvinyl alcohol maintained their sizes and morphology. SAAP-148 was sustainedly released from lyophilized nanogels (37-41% in 72 h) upon reconstitution. Lyophilized SAAP-148-loaded nanogels showed similar antimicrobial activity as SAAP-148 against planktonic and biofilm-residing AMR Staphylococcus aureus and Acinetobacter baumannii. Importantly, formulated SAAP-148 showed reduced cytotoxicity against human erythrocytes, primary human skin fibroblasts and human keratinocytes. Additionally, lyophilized SAAP-148-loaded nanogels eradicated AMR S. aureus and A. baumannii colonizing a 3D human epidermal model, without inducing any cytotoxicity in contrast to SAAP-148. These findings indicate that OSA-HA nanogels increase SAAP-148's therapeutic potential for treatment of skin wound infections.
Collapse
Affiliation(s)
- Miriam E. van Gent
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Correspondence:
| | - Tom van Baaren
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sylvia N. Kłodzińska
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Muhanad Ali
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Natasja Dolezal
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Bjorn R. van Doodewaerd
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Erik Bos
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Amy M. de Waal
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Roman I. Koning
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Hanne Mørck Nielsen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
6
|
Xiang W, Clemenza P, Klousnitzer J, Chen J, Qin W, Tristram-Nagle S, Doi Y, Di YP, Deslouches B. Rational Framework for the Design of Trp- and Arg-Rich Peptide Antibiotics Against Multidrug-Resistant Bacteria. Front Microbiol 2022; 13:889791. [PMID: 35694289 PMCID: PMC9186412 DOI: 10.3389/fmicb.2022.889791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
The threat of antibiotic resistance warrants the discovery of agents with novel antimicrobial mechanisms. Antimicrobial peptides (AMPs) directly disrupting bacterial membranes may overcome resistance to traditional antibiotics. AMP development for clinical use has been mostly limited to topical application to date. We developed a rational framework for systematically addressing this challenge using libraries composed of 86 novel Trp- and Arg-rich engineered peptides tested against clinical strains of the most common multidrug-resistant bacteria known as ESKAPE pathogens. Structure-function correlations revealed minimum lengths (as low as 16 residues) and Trp positioning for maximum antibacterial potency with mean minimum inhibitory concentration (MIC) of 2–4 μM and corresponding negligible toxicity to mammalian cells. Twelve peptides were selected based on broad-spectrum activity against both gram-negative and -positive bacteria and <25% toxicity to mammalian cells at maximum test concentrations. Most of the selected PAX remained active against the colistin-resistant clinical strains. Of the selected peptides, the shortest (the 16-residue E35) was further investigated for antibacterial mechanism and proof-of-concept in vivo efficacy. E35 killed an extensively-resistant isolate of Pseudomonas aeruginosa (PA239 from the CDC, also resistant to colistin) by irreversibly disrupting the cell membranes as shown by propidium iodide incorporation, using flow cytometry and live cell imaging. As proof of concept, in vivo toxicity studies showed that mice tolerated a systemic dose of up to 30 mg/kg peptide and were protected with a single 5 mg/kg intravenous (IV) dose against an otherwise lethal intraperitoneal injection of PA239. Efficacy was also demonstrated in an immune-compromised Klebsiella pneumoniae infection model using a daily dose of 4mg/kg E35 systemically for 2 days. This framework defines the determinants of efficacy of helical AMPs composed of only cationic and hydrophobic amino acids and provides a path for a potential departure from the restriction to topical use of AMPs toward systemic application.
Collapse
Affiliation(s)
- Wenyu Xiang
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Patrice Clemenza
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jessie Klousnitzer
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jespar Chen
- Biological Physics Group, Department of Physics, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Weiheng Qin
- Biological Physics Group, Department of Physics, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Stephanie Tristram-Nagle
- Biological Physics Group, Department of Physics, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Yohei Doi
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Y Peter Di
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Berthony Deslouches
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
7
|
Therapeutic Potential of Antimicrobial Peptides for Wound Healing. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-021-10350-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
8
|
Current Advances in Lipid and Polymeric Antimicrobial Peptide Delivery Systems and Coatings for the Prevention and Treatment of Bacterial Infections. Pharmaceutics 2021; 13:pharmaceutics13111840. [PMID: 34834254 PMCID: PMC8618997 DOI: 10.3390/pharmaceutics13111840] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Bacterial infections constitute a threat to public health as antibiotics are becoming less effective due to the emergence of antimicrobial resistant strains and biofilm and persister formation. Antimicrobial peptides (AMPs) are considered excellent alternatives to antibiotics; however, they suffer from limitations related to their peptidic nature and possible toxicity. The present review critically evaluates the chemical characteristics and antibacterial effects of lipid and polymeric AMP delivery systems and coatings that offer the promise of enhancing the efficacy of AMPs, reducing their limitations and prolonging their half-life. Unfortunately, the antibacterial activities of these systems and coatings have mainly been evaluated in vitro against planktonic bacteria in less biologically relevant conditions, with only some studies focusing on the antibiofilm activities of the formulated AMPs and on the antibacterial effects in animal models. Further improvements of lipid and polymeric AMP delivery systems and coatings may involve the functionalization of these systems to better target the infections and an analysis of the antibacterial activities in biologically relevant environments. Based on the available data we proposed which polymeric AMP delivery system or coatings could be profitable for the treatment of the different hard-to-treat infections, such as bloodstream infections and catheter- or implant-related infections.
Collapse
|
9
|
Shukla SK, Sharma AK, Gupta V, Kalonia A, Shaw P. Challenges with Wound Infection Models in Drug Development. Curr Drug Targets 2021; 21:1301-1312. [PMID: 32116189 DOI: 10.2174/1389450121666200302093312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/06/2020] [Accepted: 02/06/2020] [Indexed: 01/05/2023]
Abstract
Wound research is an evolving science trying to unfold the complex untold mechanisms behind the wound healing cascade. In particular, interest is growing regarding the role of microorganisms in both acute and chronic wound healing. Microbial burden plays an important role in the persistence of chronic wounds, ultimately resulting in delayed wound healing. It is therefore important for clinicians to understand the evolution of infection science and its various etiologies. Therefore, to understand the role of bacterial biofilm in chronic wound pathogenesis, various in vitro and in vivo models are required to investigate biofilms in wound-like settings. Infection models should be refined comprising an important signet of biofilms. These models are eminent for translational research to obtain data for designing an improved wound care formulation. However, all the existing models possess limitations and do not fit properly in the model frame for developing wound care agents. Among various impediments, one of the major drawbacks of such models is that the wound they possess does not mimic the wound a human develops. Therefore, a novel wound infection model is required which can imitate the human wounds. This review article mainly discusses various in vitro and in vivo models showing microbial colonization, their advantages and challenges. Apart from these models, there are also present ex vivo wound infection models, but this review mainly focused on various in vitro and in vivo models available for studying wound infection in controlled conditions. This information might be useful in designing an ideal wound infection model for developing an effective wound healing formulation.
Collapse
Affiliation(s)
- Sandeep K Shukla
- Institute of Nuclear Medicine & Allied Sciences, Defence Research and Development Organization, SK Mazumdar Marg, Timarpur, Delhi-110054, India
| | - Ajay K Sharma
- Institute of Nuclear Medicine & Allied Sciences, Defence Research and Development Organization, SK Mazumdar Marg, Timarpur, Delhi-110054, India
| | - Vanya Gupta
- Graphic Era deemed to be University, Dehradun, India
| | - Aman Kalonia
- Institute of Nuclear Medicine & Allied Sciences, Defence Research and Development Organization, SK Mazumdar Marg, Timarpur, Delhi-110054, India
| | - Priyanka Shaw
- Institute of Nuclear Medicine & Allied Sciences, Defence Research and Development Organization, SK Mazumdar Marg, Timarpur, Delhi-110054, India
| |
Collapse
|
10
|
Lakshmaiah Narayana J, Golla R, Mishra B, Wang X, Lushnikova T, Zhang Y, Verma A, Kumar V, Xie J, Wang G. Short and Robust Anti-Infective Lipopeptides Engineered Based on the Minimal Antimicrobial Peptide KR12 of Human LL-37. ACS Infect Dis 2021; 7:1795-1808. [PMID: 33890759 DOI: 10.1021/acsinfecdis.1c00101] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This study aims to push the frontier of the engineering of human cathelicidin LL-37, a critical antimicrobial innate immune peptide that wards off invading pathogens. By sequential truncation of the smallest antibacterial peptide (KR12) of LL-37 and conjugation with fatty acids, with varying chain lengths, a library of lipopeptides is generated. These peptides are subjected to antibacterial activity and hemolytic assays. Candidates (including both forms made of l- and d-amino acids) with the optimal cell selectivity are subsequently fed to the second layer of in vitro filters, including salts, pH, serum, and media. These practices lead to the identification of a miniature LL-37 like peptide (d-form) with selectivity, stability, and robust antimicrobial activity in vitro against both Gram-positive and negative bacteria. Proteomic studies reveal far fewer serum proteins that bind to the d-form than the l-form peptide. C10-KR8d targets bacterial membranes to become helical, making it difficult for bacteria to develop resistance in a multiple passage experiment. In vivo, C10-KR8d is able to reduce bacterial burden of methicillin-resistant Staphylococcus aureus (MRSA) USA300 LAC in neutropenic mice. In addition, this designer peptide prevents bacterial biofilm formation in a catheter-associated mouse model. Meanwhile, C10-KR8d also recruits cytokines to the vicinity of catheters to clear infection. Thus, based on the antimicrobial region of LL-37, this study succeeds in identifying the smallest anti-infective peptide C10-KR8d with both robust antimicrobial, antibiofilm, and immune modulation activities.
Collapse
Affiliation(s)
- Jayaram Lakshmaiah Narayana
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Radha Golla
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Biswajit Mishra
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Xiuqing Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Tamara Lushnikova
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Yingxia Zhang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Atul Verma
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| | - Vikas Kumar
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska 68130, United States
| | - Guangshun Wang
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, Nebraska 68198-5900, United States
| |
Collapse
|
11
|
Hou Z, Zhang B, She P, Wu Y, Zhang R. Anti -planktonic and anti -biofilm effects of two synthetic anti -microbial peptides against Staphylococcus epidermidis. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:481-487. [PMID: 34148884 PMCID: PMC10930219 DOI: 10.11817/j.issn.1672-7347.2021.200360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVES Staphylococcus epidermidis is one of the most common Gram-positive cocci in nosocomial infection, which could adhere to the surface of medical apparatus and causes biofilm-related infections. In the present study, we aim to explore the antimicrobial effects of GH12 and SAAP-148 against Staphylococcus epidermidis. METHODS Micro-dilution methods were used to detect the minimal inhibitory/bactericidal concentration of peptides on Staphylococcus epidermidis. Biofilm formation positive type strain was used to determine the antibiofilm effects of the peptides. Biofilms were built on the cover slides and fluorescent dye SYTO9 and laser confocal microscope were used to observe the effects of peptides on the three-dimensional structure of Staphylococcus epidermidis biofilms. The cell membrane permeability of Staphylococcus epidermidis was detected by flow cytometry. Expressions of icaA and icaDgenes were analyzed by real-time reverse transcription PCR. RESULTS The minimal inhibitory concentrations of GH12 and SAAP-148 against Staphylococcus epidermidis were 8 and 16 μg/mL, respectively, and the minimal bactericidal concentration was 64 μg/mL. GH12 and SAAP-148 significantly inhibited the biofilm formation of Staphylococcus epidermidis at the concentration of 8 μg/mL (t=7.193, P<0.05) and 16 μg/mL (t=7.808, P<0.05), respectively. Similarly, the GH12 and SAAP-148 significantly eradicated the pre-formed biofilms at the concentration of 16 μg/mL (t=5.369, P<0.05) and 32 μg/mL (t=4.474, P<0.05) in a dose-response manner, respectively. Meanwhile, the two peptides broke the structure of biofims and reduce the total biomass. GH12 and SAAP-148 at the concentration of minimal inhibitory concentration significantly disrupted the cell membrane of Staphylococcus epidermidis. The expressions of icaA and icaDgenes were significantly inhibited by antimicrobial peptides at the 1×minimal inhibitory concentration. CONCLUSIONS GH12 and SAAP-148 show significantly antimicrobial and anti-biofilm effects against Staphylococcus epidermidis by disruption of cell membrane and inhibition of icaAand icaDgene expression.
Collapse
Affiliation(s)
- Zhengli Hou
- Hunan Normal University School of Medicine, Changsha 410013.
- Department of Clinical Laboratory, Fourth Hospital of Changsha, Changsha 410006.
| | - Biming Zhang
- Department of Clinical Laboratory, Fourth Hospital of Changsha, Changsha 410006
| | - Pengfei She
- Department of Clinical Laboratory, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yuan Wu
- Department of Clinical Laboratory, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ran Zhang
- Hunan Normal University School of Medicine, Changsha 410013.
| |
Collapse
|
12
|
Dijksteel GS, Ulrich MMW, Vlig M, Sobota A, Middelkoop E, Boekema BKHL. Safety and bactericidal efficacy of cold atmospheric plasma generated by a flexible surface Dielectric Barrier Discharge device against Pseudomonas aeruginosa in vitro and in vivo. Ann Clin Microbiol Antimicrob 2020; 19:37. [PMID: 32814573 PMCID: PMC7439657 DOI: 10.1186/s12941-020-00381-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cold atmospheric plasma (CAP), which is ionized gas produced at atmospheric pressure, could be a novel and potent antimicrobial therapy for the treatment of infected wounds. Previously we have shown that CAP generated with a flexible surface Dielectric Barrier Discharge (sDBD) is highly effective against bacteria in vitro and in ex vivo burn wound models. In the current paper, we determined the in vitro and in vivo safety and efficacy of CAP generated by this sDBD device. Methods The effect of CAP on DNA mutations of V79 fibroblasts was measured using a hypoxanthine–guanine-phosphoribosyltransferase (HPRT) assay. Furthermore, effects on cell proliferation, apoptosis and DNA damage in ex vivo burn wound models (BWMs) were assessed using immunohistochemistry. Next, 105 colony forming units (CFU) P. aeruginosa strain PAO1 were exposed to CAP in a 3D collagen-elastin matrix environment to determine the number of surviving bacteria in vitro. Finally, rat excision wounds were inoculated with 107 CFU PAO1 for 24 h. The wounds received a single CAP treatment, repeated treatments on 4 consecutive days with CAP, 100 µL of 1% (wt/wt) silver sulfadiazine or no treatment. Wound swabs and punch biopsies were taken to determine the number of surviving bacteria. Results Exposure of V79 fibroblasts to CAP did not increase the numbers of mutated colonies. Additionally, the number of proliferative, apoptotic and DNA damaged cells in the BWMs was comparable to that of the unexposed control. Exposure of PAO1 to CAP for 2 min resulted in the complete elimination of bacteria in vitro. Contrarily, CAP treatment for 6 min of rat wounds colonized with PAO1 did not effectively reduce the in vivo bacterial count. Conclusions CAP treatment was safe but showed limited efficacy against PAO1 in our rat wound infection model.
Collapse
Affiliation(s)
- Gabrielle S Dijksteel
- Association of Dutch Burn Centres, Zeestraat 29, 1941 AJ, Beverwijk, The Netherlands. .,Dept. of Plastic, Reconstructive & Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Magda M W Ulrich
- Association of Dutch Burn Centres, Zeestraat 29, 1941 AJ, Beverwijk, The Netherlands.,Dept. of Plastic, Reconstructive & Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.,Dept. of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marcel Vlig
- Association of Dutch Burn Centres, Zeestraat 29, 1941 AJ, Beverwijk, The Netherlands
| | - Ana Sobota
- Dept. of Applied Physics, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Esther Middelkoop
- Association of Dutch Burn Centres, Zeestraat 29, 1941 AJ, Beverwijk, The Netherlands.,Dept. of Plastic, Reconstructive & Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Bouke K H L Boekema
- Association of Dutch Burn Centres, Zeestraat 29, 1941 AJ, Beverwijk, The Netherlands
| |
Collapse
|