201
|
He Y, Jin Y, Ying X, Wu Q, Yao S, Li Y, Liu H, Ma G, Wang X. Development of an antimicrobial peptide-loaded mineralized collagen bone scaffold for infective bone defect repair. Regen Biomater 2020; 7:515-525. [PMID: 33149940 PMCID: PMC7597801 DOI: 10.1093/rb/rbaa015] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/21/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022] Open
Abstract
The repair of infective bone defects is a great challenge in clinical work. It is of vital importance to develop a kind of bone scaffold with good osteogenic properties and long-term antibacterial activity for local anti-infection and bone regeneration. A porous mineralized collagen (MC) scaffold containing poly(d,l-lactide-co-glycolic acid) (PLGA) microspheres loaded with two antibacterial synthetic peptides, Pac-525 or KSL-W was developed and characterized via scanning electron microscopy (SEM), porosity measurement, swelling and mechanical tests. The results showed that the MC scaffold embedded with smooth and compact PLGA microspheres had a positive effect on cell growth and also had antibacterial properties. Through toxicity analysis, cell morphology and proliferation analysis and alkaline phosphatase evaluation, the antibacterial scaffolds showed excellent biocompatibility and osteogenic activity. The antibacterial property evaluated with Staphylococcus aureus and Escherichia coli suggested that the sustained release of Pac-525 or KSL-W from the scaffolds could inhibit the bacterial growth aforementioned in the long term. Our results suggest that the antimicrobial peptides-loaded MC bone scaffold has good antibacterial and osteogenic activities, thus providing a great promise for the treatment of infective bone defects.
Collapse
Affiliation(s)
- Yuzhu He
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Zhongguancun Street, Haidian District, Beijing 100084, China.,Department of Oral Prosthodontics, School of Stomatology, Dalian Medical University, Lvshun South Road, Lushunkou District, Dalian 116044, China
| | - Yahui Jin
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Zhongguancun Street, Haidian District, Beijing 100084, China.,Department of Oral Prosthodontics, School of Stomatology, Dalian Medical University, Lvshun South Road, Lushunkou District, Dalian 116044, China.,Department of Stomatology, Zhejiang Provincial Hospital of Chinese Medicine, The 9th Street, Economic and Technological Development Zone, Hangzhou 310018, China
| | - Xiaoxia Ying
- Department of Oral Prosthodontics, School of Stomatology, Dalian Medical University, Lvshun South Road, Lushunkou District, Dalian 116044, China
| | - Qiong Wu
- School of Life Sciences, Tsinghua University, Zhongguancun Street, Haidian District, Beijing 100084, China
| | - Shenglian Yao
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Zhongguancun Street, Haidian District, Beijing 100084, China
| | - Yuanyuan Li
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Zhongguancun Street, Haidian District, Beijing 100084, China.,Department of Oral Prosthodontics, School of Stomatology, Dalian Medical University, Lvshun South Road, Lushunkou District, Dalian 116044, China
| | - Huiying Liu
- Department of Oral Prosthodontics, School of Stomatology, Dalian Medical University, Lvshun South Road, Lushunkou District, Dalian 116044, China
| | - Guowu Ma
- Department of Oral Prosthodontics, School of Stomatology, Dalian Medical University, Lvshun South Road, Lushunkou District, Dalian 116044, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Zhongguancun Street, Haidian District, Beijing 100084, China
| |
Collapse
|
202
|
Liu Y, Sun Y, Li S, Liu M, Qin X, Chen X, Lin Y. Tetrahedral Framework Nucleic Acids Deliver Antimicrobial Peptides with Improved Effects and Less Susceptibility to Bacterial Degradation. NANO LETTERS 2020; 20:3602-3610. [PMID: 32272018 DOI: 10.1021/acs.nanolett.0c00529] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yuhao Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Songhang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mengting Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Qin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
203
|
Synthetic molecular evolution of host cell-compatible, antimicrobial peptides effective against drug-resistant, biofilm-forming bacteria. Proc Natl Acad Sci U S A 2020; 117:8437-8448. [PMID: 32241895 DOI: 10.1073/pnas.1918427117] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Novel classes of antibiotics and new strategies to prevent and treat infections are urgently needed because the rapid rise in drug-resistant bacterial infections in recent decades has been accompanied by a parallel decline in development of new antibiotics. Membrane permeabilizing antimicrobial peptides (AMPs) have long been considered a potentially promising, novel class of antibiotic, especially for wound protection and treatment to prevent the development of serious infections. Yet, despite thousands of known examples, AMPs have only infrequently proceeded as far as clinical trials, especially the chemically simple, linear examples. In part, this is due to impediments that often limit their applications in vivo. These can include low solubility, residual toxicity, susceptibility to proteolysis, and loss of activity due to host cell, tissue, and protein binding. Here we show how synthetic molecular evolution can be used to evolve potentially advantageous antimicrobial peptides that lack these impediments from parent peptides that have at least some of them. As an example of how the antibiotic discovery pipeline can be populated with more promising candidates, we evolved and optimized one family of linear AMPs into a new generation with high solubility, low cytotoxicity, potent broad-spectrum sterilizing activity against a panel of gram-positive and gram-negative ESKAPE pathogens, and antibiofilm activity against gram-positive and gram-negative biofilms. The evolved peptides have these activities in vitro even in the presence of concentrated host cells and also in vivo in the complex, cell- and protein-rich environment of a purulent animal wound model infected with drug-resistant bacteria.
Collapse
|
204
|
Mücke PA, Maaß S, Kohler TP, Hammerschmidt S, Becher D. Proteomic Adaptation of Streptococcus pneumoniae to the Human Antimicrobial Peptide LL-37. Microorganisms 2020; 8:E413. [PMID: 32183275 PMCID: PMC7143398 DOI: 10.3390/microorganisms8030413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 01/13/2023] Open
Abstract
Secreted antimicrobial peptides (AMPs) are an important part of the human innate immune system and prevent local and systemic infections by inhibiting bacterial growth in a concentration-dependent manner. In the respiratory tract, the cationic peptide LL-37 is one of the most abundant AMPs and capable of building pore complexes in usually negatively charged bacterial membranes, leading to the destruction of bacteria. However, the adaptation mechanisms of several pathogens to LL-37 are already described and are known to weaken the antimicrobial effect of the AMP, for instance, by repulsion, export or degradation of the peptide. This study examines proteome-wide changes in Streptococcus pneumoniae D39, the leading cause of bacterial pneumonia, in response to physiological concentrations of LL-37 by high-resolution mass spectrometry. Our data indicate that pneumococci may use some of the known adaptation mechanisms to reduce the effect of LL-37 on their physiology, too. Additionally, several proteins seem to be involved in resistance to AMPs which have not been related to this process before, such as the teichoic acid flippase TacF (SPD_1128). Understanding colonization- and infection-relevant adaptations of the pneumococcus to AMPs, especially LL-37, could finally uncover new drug targets to weaken the burden of this widespread pathogen.
Collapse
Affiliation(s)
- Pierre-Alexander Mücke
- Department of Microbial Proteomics, Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany; (P.-A.M.); (S.M.)
| | - Sandra Maaß
- Department of Microbial Proteomics, Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany; (P.-A.M.); (S.M.)
| | - Thomas P. Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany; (T.P.K.); (S.H.)
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany; (T.P.K.); (S.H.)
| | - Dörte Becher
- Department of Microbial Proteomics, Institute of Microbiology, Center for Functional Genomics of Microbes, University of Greifswald, Felix-Hausdorff-Str. 8, 17489 Greifswald, Germany; (P.-A.M.); (S.M.)
| |
Collapse
|
205
|
Kapach G, Nuri R, Schmidt C, Danin A, Ferrera S, Savidor A, Gerlach RG, Shai Y. Loss of the Periplasmic Chaperone Skp and Mutations in the Efflux Pump AcrAB-TolC Play a Role in Acquired Resistance to Antimicrobial Peptides in Salmonella typhimurium. Front Microbiol 2020; 11:189. [PMID: 32210923 PMCID: PMC7075815 DOI: 10.3389/fmicb.2020.00189] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/27/2020] [Indexed: 01/01/2023] Open
Abstract
Bacterial resistance to antibiotics is a major concern worldwide, leading to an extensive search for alternative drugs. Promising candidates are antimicrobial peptides (AMPs), innate immunity molecules, shown to be highly efficient against multidrug resistant bacteria. Therefore, it is essential to study bacterial resistance mechanisms against them. For that purpose, we used experimental evolution, and isolated a Salmonella enterica serovar typhimurium-resistant line to the AMP 4DK5L7. This AMP displayed promising features including widespread activity against Gram-negative bacteria and protection from proteolytic degradation. However, the resistance that evolved in the isolated strain was particularly high. Whole genome sequencing revealed that five spontaneous mutations had evolved. Of these, three are novel in the context of acquired AMP resistance. Two mutations are related to the AcrAB-TolC multidrug efflux pump. One occurred in AcrB, the substrate-binding domain of the system, and the second in RamR, a transcriptional regulator of the system. Together, the mutations increased the minimal inhibitory concentration (MIC) by twofold toward this AMP. Moreover, the mutation in AcrB induced hypersusceptibility toward ampicillin and colistin. The last mutation occurred in Skp, a periplasmic chaperone that participates in the biogenesis of outer membrane proteins (OMPs). This mutation increased the MIC by twofold to 4DK5L7 and by fourfold to another AMP, seg5D. Proteomic analysis revealed that the mutation abolished Skp expression, reduced OMP abundance, and increased DegP levels. DegP, a protease that was reported to have an additional chaperone activity, escorts OMPs through the periplasm along with Skp, but is also associated with AMP resistance. In conclusion, our data demonstrate that both loss of Skp and manipulation of the AcrAB-TolC system are alternative strategies of AMP acquired resistance in Salmonella typhimurium and might represent a common mechanism in other Gram-negative bacteria.
Collapse
Affiliation(s)
- Gal Kapach
- Departmant of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Reut Nuri
- Departmant of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Adi Danin
- Departmant of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shir Ferrera
- Departmant of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Roman G Gerlach
- Project Group 5, Robert Koch Institute, Wernigerode, Germany
| | - Yechiel Shai
- Departmant of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
206
|
Anionic food color tartrazine enhances antibacterial efficacy of histatin-derived peptide DHVAR4 by fine-tuning its membrane activity. Q Rev Biophys 2020; 53:e5. [PMID: 32115014 DOI: 10.1017/s0033583520000013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Here it is demonstrated how some anionic food additives commonly used in our diet, such as tartrazine (TZ), bind to DHVAR4, an antimicrobial peptide (AMP) derived from oral host defense peptides, resulting in significantly fostered toxic activity against both Gram-positive and Gram-negative bacteria, but not against mammalian cells. Biophysical studies on the DHVAR4-TZ interaction indicate that initially large, positively charged aggregates are formed, but in the presence of lipid bilayers, they rather associate with the membrane surface. In contrast to synergistic effects observed for mixed antibacterial compounds, this is a principally different mechanism, where TZ directly acts on the membrane-associated AMP promoting its biologically active helical conformation. Model vesicle studies show that compared to dye-free DHVAR4, peptide-TZ complexes are more prone to form H-bonds with the phosphate ester moiety of the bilayer head-group region resulting in more controlled bilayer fusion mechanism and concerted severe cell damage. AMPs are considered as promising compounds to combat formidable antibiotic-resistant bacterial infections; however, we know very little on their in vivo actions, especially on how they interact with other chemical agents. The current example illustrates how food dyes can modulate AMP activity, which is hoped to inspire improved therapies against microbial infections in the alimentary tract. Results also imply that the structure and function of natural AMPs could be manipulated by small compounds, which may also offer a new strategic concept for the future design of peptide-based antimicrobials.
Collapse
|
207
|
Kepiro IE, Marzuoli I, Hammond K, Ba X, Lewis H, Shaw M, Gunnoo SB, De Santis E, Łapińska U, Pagliara S, Holmes MA, Lorenz CD, Hoogenboom BW, Fraternali F, Ryadnov MG. Engineering Chirally Blind Protein Pseudocapsids into Antibacterial Persisters. ACS NANO 2020; 14:1609-1622. [PMID: 31794180 DOI: 10.1021/acsnano.9b06814] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Antimicrobial resistance stimulates the search for antimicrobial forms that may be less subject to acquired resistance. Here we report a conceptual design of protein pseudocapsids exhibiting a broad spectrum of antimicrobial activities. Unlike conventional antibiotics, these agents are effective against phenotypic bacterial variants, while clearing "superbugs" in vivo without toxicity. The design adopts an icosahedral architecture that is polymorphic in size, but not in shape, and that is available in both l and d epimeric forms. Using a combination of nanoscale and single-cell imaging we demonstrate that such pseudocapsids inflict rapid and irreparable damage to bacterial cells. In phospholipid membranes they rapidly convert into nanopores, which remain confined to the binding positions of individual pseudocapsids. This mechanism ensures precisely delivered influxes of high antimicrobial doses, rendering the design a versatile platform for engineering structurally diverse and functionally persistent antimicrobial agents.
Collapse
Affiliation(s)
- Ibolya E Kepiro
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
| | - Irene Marzuoli
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
- Randall Centre for Cell and Molecular Biophysics , King's College London , London , SE1 1UL , U.K
| | - Katharine Hammond
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
- Department of Physics and Astronomy , University College London , London , WC1E 6BT , U.K
- London Centre for Nanotechnology , University College London , London , WC1H 0AH , U.K
| | - Xiaoliang Ba
- Department of Veterinary Medicine , University of Cambridge , Cambridge , CB3 0ES , U.K
| | - Helen Lewis
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
| | - Michael Shaw
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
- Department of Computer Science , University College London , London , WC1 6BT , U.K
| | - Smita B Gunnoo
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
| | - Emiliana De Santis
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
| | - Urszula Łapińska
- Living Systems Institute , University of Exeter , Exeter , EX4 4QD , U.K
| | - Stefano Pagliara
- Living Systems Institute , University of Exeter , Exeter , EX4 4QD , U.K
| | - Mark A Holmes
- Department of Veterinary Medicine , University of Cambridge , Cambridge , CB3 0ES , U.K
| | - Christian D Lorenz
- Department of Physics , King's College London , Strand Lane , London , WC2R 2LS , U.K
| | - Bart W Hoogenboom
- Department of Physics and Astronomy , University College London , London , WC1E 6BT , U.K
- London Centre for Nanotechnology , University College London , London , WC1H 0AH , U.K
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biophysics , King's College London , London , SE1 1UL , U.K
| | - Maxim G Ryadnov
- National Physical Laboratory , Hampton Road , Teddington , TW11 0LW , U.K
- Department of Physics , King's College London , Strand Lane , London , WC2R 2LS , U.K
| |
Collapse
|
208
|
Molecular mechanisms of collateral sensitivity to the antibiotic nitrofurantoin. PLoS Biol 2020; 18:e3000612. [PMID: 31986134 PMCID: PMC7004380 DOI: 10.1371/journal.pbio.3000612] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/06/2020] [Accepted: 01/06/2020] [Indexed: 12/20/2022] Open
Abstract
Antibiotic resistance increasingly limits the success of antibiotic treatments, and physicians require new ways to achieve efficient treatment despite resistance. Resistance mechanisms against a specific antibiotic class frequently confer increased susceptibility to other antibiotic classes, a phenomenon designated collateral sensitivity (CS). An informed switch of antibiotic may thus enable the efficient treatment of resistant strains. CS occurs in many pathogens, but the mechanisms that generate hypersusceptibility are largely unknown. We identified several molecular mechanisms of CS against the antibiotic nitrofurantoin (NIT). Mutants that are resistant against tigecycline (tetracycline), mecillinam (β-lactam), and protamine (antimicrobial peptide) all show CS against NIT. Their hypersusceptibility is explained by the overexpression of nitroreductase enzymes combined with increased drug uptake rates, or increased drug toxicity. Increased toxicity occurs through interference of the native drug-response system for NIT, the SOS response, with growth. A mechanistic understanding of CS will help to develop drug switches that combat resistance. Resistance mechanisms against a specific antibiotic class frequently often confer negative cross-resistance to other antibiotic classes, a phenomenon known as collateral sensitivity. This study shows that collateral sensitivity in bacteria can be explained by a combination of several mechanisms that can be exploited to develop drug switches that combat resistance.
Collapse
|
209
|
Sundaramoorthy NS, Suresh P, Selva Ganesan S, GaneshPrasad A, Nagarajan S. Restoring colistin sensitivity in colistin-resistant E. coli: Combinatorial use of MarR inhibitor with efflux pump inhibitor. Sci Rep 2019; 9:19845. [PMID: 31882661 PMCID: PMC6934491 DOI: 10.1038/s41598-019-56325-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023] Open
Abstract
Antibiotics like colistin are the last resort to deal with infections by carbapenem-resistant Enterobacteriaceae (CREB). Resistance to colistin severely restricts therapeutic options. To tackle this dire situation, urgent measures to restore colistin sensitivity are needed. In this study, whole-genome sequencing of colistin-resistant E. coli strain was performed and the genome analysis revealed that the strain belonged to the sequence type ST405. Multiple mutations were observed in genes implicated in colistin resistance, especially those related to the L-Ara-4-N pathway but mgrB was unmutated and mcr1-9 genes were missing. MarR inhibitor salicylate was used to re-sensitize this strain to colistin, which increased the negative charge on the cell surface especially in colistin resistant E. coli (U3790 strain) and thereby facilitated a decrease in colistin MIC by 8 fold. It is indeed well known that MarR inhibition by salicylate triggers the expression of AcrAB efflux pumps through MarA. So, in order to fully restore colistin sensitivity, a potent efflux pump inhibitor (BC1), identified earlier by this group was employed. The combination of colistin with both salicylate and BC1 caused a remarkable 6 log reduction in cell counts of U3790 in time-kill assay. Infection of muscle tissue of zebrafish with U3790 followed by various treatments showed that the combination of colistin + salicylate + BC1 was highly effective in reducing bioburden in infected muscle tissue by 4 log fold. Thus, our study shows that a combination of MarR inhibitor to enhance colistin binding and efflux pump inhibitor to reduce colistin extrusion was highly effective in restoring colistin sensitivity in colistin-resistant clinical isolate of E. coli in vitro and in vivo.
Collapse
Affiliation(s)
- Niranjana Sri Sundaramoorthy
- Center for Research on Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - Pavithira Suresh
- Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - Subramaniapillai Selva Ganesan
- Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - ArunKumar GaneshPrasad
- Center for Research on Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India
| | - Saisubramanian Nagarajan
- Center for Research on Infectious Diseases, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, India.
| |
Collapse
|
210
|
Manteghi R, Pallagi E, Olajos G, Csóka I. Pegylation and formulation strategy of Anti-Microbial Peptide (AMP) according to the quality by design approach. Eur J Pharm Sci 2019; 144:105197. [PMID: 31862311 DOI: 10.1016/j.ejps.2019.105197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 12/08/2019] [Accepted: 12/16/2019] [Indexed: 01/10/2023]
Abstract
Antimicrobial resistance is one of the main global threats according to the World Health Organization's (WHO) report (World Health Organization 2014), therefore there is a need for the development of other agents, such as antimicrobial peptides (AMPs). Although AMPs are considered as major candidates for next-generation antibiotics, several challenges including low bioavailability, high manufacturing cost and toxicity are still to be solved for their practical use in therapeutic applications. Novel chemical modification approaches as well as strategies for their delivery offer several opportunities to overcome these barriers and develop more stable and cost-effective synthetic peptides with efficient delivery to the target site. The integration of the Quality by Design (QbD) approach in the early pharmaceutical developments supports researchers in optimizing the targeted product by a risk based manner. Peptide modifications and formulation of peptide delivery systems are challenging tasks and hide several risks. Understanding and evaluating the cause - effect relations within the initial Risk Assessment (RA) step in case of all attributes give the basis for the experimental design as the next step, and aids the formulation development in order to get the final product in the targeted quality range. This study presents a Quality by Design based antimicrobial peptide modification and formulation design. Analyses the potential risks in the AMP PEGylation process through the example of PGLa. The QbD based initial RA screened and evaluated the risk factors in this AMP modification procedure. The critical quality and process related factors were defined and their ranking was performed due to their estimated critical effect on the PEGylated AMP. This pre-formulation design study highlights the critical risk factors as decision points for the further steps.
Collapse
Affiliation(s)
- Reihaneh Manteghi
- University of Szeged, Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, Szeged, Hungary
| | - Edina Pallagi
- University of Szeged, Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, Szeged, Hungary
| | - Gábor Olajos
- University of Szeged, Faculty of Medicine, Department of Medical Chemistry, Szeged, Hungary
| | - Ildikó Csóka
- University of Szeged, Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, Szeged, Hungary.
| |
Collapse
|
211
|
Kintses B, Jangir PK, Fekete G, Számel M, Méhi O, Spohn R, Daruka L, Martins A, Hosseinnia A, Gagarinova A, Kim S, Phanse S, Csörgő B, Györkei Á, Ari E, Lázár V, Nagy I, Babu M, Pál C, Papp B. Chemical-genetic profiling reveals limited cross-resistance between antimicrobial peptides with different modes of action. Nat Commun 2019; 10:5731. [PMID: 31844052 PMCID: PMC6915728 DOI: 10.1038/s41467-019-13618-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/14/2019] [Indexed: 11/09/2022] Open
Abstract
Antimicrobial peptides (AMPs) are key effectors of the innate immune system and promising therapeutic agents. Yet, knowledge on how to design AMPs with minimal cross-resistance to human host-defense peptides remains limited. Here, we systematically assess the resistance determinants of Escherichia coli against 15 different AMPs using chemical-genetics and compare to the cross-resistance spectra of laboratory-evolved AMP-resistant strains. Although generalizations about AMP resistance are common in the literature, we find that AMPs with different physicochemical properties and cellular targets vary considerably in their resistance determinants. As a consequence, cross-resistance is prevalent only between AMPs with similar modes of action. Finally, our screen reveals several genes that shape susceptibility to membrane- and intracellular-targeting AMPs in an antagonistic manner. We anticipate that chemical-genetic approaches could inform future efforts to minimize cross-resistance between therapeutic and human host AMPs.
Collapse
Affiliation(s)
- Bálint Kintses
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
- HCEMM-BRC Translational Microbiology Lab, Szeged, Hungary.
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary.
| | - Pramod K Jangir
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Gergely Fekete
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Mónika Számel
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Orsolya Méhi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Réka Spohn
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ana Martins
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Ali Hosseinnia
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Alla Gagarinova
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sunyoung Kim
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Sadhna Phanse
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Bálint Csörgő
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Department of Microbiology and Immunology, University of California, San Francisco, USA
| | - Ádám Györkei
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Eszter Ari
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Viktória Lázár
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - István Nagy
- Sequencing Platform, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, Saskatchewan, Canada
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary.
| |
Collapse
|
212
|
Raheem N, Straus SK. Mechanisms of Action for Antimicrobial Peptides With Antibacterial and Antibiofilm Functions. Front Microbiol 2019; 10:2866. [PMID: 31921046 PMCID: PMC6927293 DOI: 10.3389/fmicb.2019.02866] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/27/2019] [Indexed: 12/14/2022] Open
Abstract
The antibiotic crisis has led to a pressing need for alternatives such as antimicrobial peptides (AMPs). Recent work has shown that these molecules have great potential not only as antimicrobials, but also as antibiofilm agents, immune modulators, anti-cancer agents and anti-inflammatories. A better understanding of the mechanism of action (MOA) of AMPs is an important part of the discovery of more potent and less toxic AMPs. Many models and techniques have been utilized to describe the MOA. This review will examine how biological assays and biophysical methods can be utilized in the context of the specific antibacterial and antibiofilm functions of AMPs.
Collapse
Affiliation(s)
- Nigare Raheem
- Department of Chemistry, The University of British Columbia, Vancouver, BC, Canada
| | - Suzana K Straus
- Department of Chemistry, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
213
|
Solstad RG, Johansen C, Stensvåg K, Strøm MB, Haug T. Structure‐activity relationship studies of shortened analogues of the antimicrobial peptide EeCentrocin 1 from the sea urchin
Echinus esculentus. J Pept Sci 2019; 26:e3233. [DOI: 10.1002/psc.3233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Runar G. Solstad
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT ‐ The Arctic University of Norway Tromsø Norway
- Nofima – The Norwegian Institute of Food, Fisheries and Aquaculture Research Tromsø Norway
| | - Cecilie Johansen
- Department of Pharmacy, Faculty of Health Sciences, UiT ‐ The Arctic University of Norway Tromsø Norway
| | - Klara Stensvåg
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT ‐ The Arctic University of Norway Tromsø Norway
| | - Morten B. Strøm
- Department of Pharmacy, Faculty of Health Sciences, UiT ‐ The Arctic University of Norway Tromsø Norway
| | - Tor Haug
- The Norwegian College of Fishery Science, Faculty of Biosciences, Fisheries and Economics, UiT ‐ The Arctic University of Norway Tromsø Norway
| |
Collapse
|
214
|
Der Torossian Torres M, de la Fuente-Nunez C. Reprogramming biological peptides to combat infectious diseases. Chem Commun (Camb) 2019; 55:15020-15032. [PMID: 31782426 DOI: 10.1039/c9cc07898c] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
With the rapid spread of resistance among parasites and bacterial pathogens, antibiotic-resistant infections have drawn much attention worldwide. Consequently, there is an urgent need to develop new strategies to treat neglected diseases and drug-resistant infections. Here, we outline several new strategies that have been developed to counter pathogenic microorganisms by designing and constructing antimicrobial peptides (AMPs). In addition to traditional discovery and design mechanisms guided by chemical biology, synthetic biology and computationally-based approaches offer useful tools for the discovery and generation of bioactive peptides. We believe that the convergence of such fields, coupled with systematic experimentation in animal models, will help translate biological peptides into the clinic. The future of anti-infective therapeutics is headed towards specifically designed molecules whose form is driven by computer-based frameworks. These molecules are selective, stable, and active at therapeutic doses.
Collapse
Affiliation(s)
- Marcelo Der Torossian Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, and Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, and Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
215
|
Hu C, Zhang F, Kong Q, Lu Y, Zhang B, Wu C, Luo R, Wang Y. Synergistic Chemical and Photodynamic Antimicrobial Therapy for Enhanced Wound Healing Mediated by Multifunctional Light-Responsive Nanoparticles. Biomacromolecules 2019; 20:4581-4592. [PMID: 31697486 DOI: 10.1021/acs.biomac.9b01401] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recently, rapid acquisition of antibiotic resistance, increased prevalence of antibiotic-resistant bacterial infections, and slow healing of infected wound have led to vast difficulties in developing innovative antimicrobial agents to obliterate pathogenic bacteria and simultaneously accelerate wound healing. To effectively solve this problem, we designed light-responsive multifunctional nanoparticles with conjugation of quaternary ammonium chitosan and photosensitizer chlorin e6 (Ce6) to merge chemical and photodynamic therapy to efficient antibacteria. The Mg/(-)-epigallocatechin-3-gallate (EGCG) complex rapidly responded to light irradiation under 660 nm with release of magnesium ions, which effectively accelerated wound healing without toxicity to mammalian cells. Notably, positively charged nanoparticles could efficiently adhere to the bacterial surface, and reactive oxygen species (ROS) produced under laser irradiation destroyed the membrane structure of the bacteria, which is irreversible, ultimately leading to bacteria death. Thus, multifunctional nanoparticles with a combination of chemical and photodynamic antimicrobial therapy would offer guidance to rational predicted and designed new effective antimicrobial nanomaterials. Most importantly, it may represent a promising class of antimicrobial strategy for potential clinical translation.
Collapse
Affiliation(s)
- Cheng Hu
- National Engineering Research Center for Biomaterials , Sichuan University , Chengdu , Sichuan 610064 , People's Republic of China
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials , Sichuan University , Chengdu , Sichuan 610064 , People's Republic of China
| | - Qunshou Kong
- National Engineering Research Center for Biomaterials , Sichuan University , Chengdu , Sichuan 610064 , People's Republic of China
| | - Yuhui Lu
- National Engineering Research Center for Biomaterials , Sichuan University , Chengdu , Sichuan 610064 , People's Republic of China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials , Sichuan University , Chengdu , Sichuan 610064 , People's Republic of China
| | - Can Wu
- National Engineering Research Center for Biomaterials , Sichuan University , Chengdu , Sichuan 610064 , People's Republic of China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials , Sichuan University , Chengdu , Sichuan 610064 , People's Republic of China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials , Sichuan University , Chengdu , Sichuan 610064 , People's Republic of China
| |
Collapse
|
216
|
Bazan EL, Ruan L, Zhou C. Improving the antimicrobial efficacy against resistant Staphylococcus aureus by a combined use of conjugated oligoelectrolytes. PLoS One 2019; 14:e0224816. [PMID: 31730663 PMCID: PMC6857938 DOI: 10.1371/journal.pone.0224816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023] Open
Abstract
Two membrane-intercalating conjugated oligoelectrolytes (COEs), namely COE-D8 and COE-S6, were combined to achieve enhanced antimicrobial efficacy. COE-D8 has a shorter molecular length than COE-S6 and is typical of effective antimicrobial COE molecules, presumably due to its prominent membrane disrupting function. In contrast, COE-D6 exhibits lower efficacy against bacteria and lower toxicity toward mammalian cells. Surprisingly, after supplementing 8 μM COE-S6, the minimum inhibitory concentration (MIC) of COE-D8 against methicillin-resistant Staphylococcus aureus (MRSA) was improved 8-fold, from 0.5 μM to 0.063 μM (0.050 μg mL-1). No increased toxicity toward mammalian cells was observed by the combination of COEs, as indicated by cytotoxicity measurements using the 3T3 cell line. Indeed, there is an extended ratio between the half maximal inhibitory concentration based on 3T3 cells to MIC against MRSA from 12 to greater than 256. Biophysical experiments using liposome models suggest that COE-S6 promotes the interactions between COE-D8 and lipid bilayers, which is in agreement with damages of cellular permeability and morphology, as observed by confocal microscopy and scanning electron microscopy. The application of a combined mixture of COEs further demonstrates their promising potential as a new class of antimicrobial agents with high efficacy and selectivity.
Collapse
Affiliation(s)
- Elias L. Bazan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Lin Ruan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Cheng Zhou
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
- Singapore Centre on Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore
- * E-mail:
| |
Collapse
|
217
|
Simpson DH, Hapeshi A, Rogers NJ, Brabec V, Clarkson GJ, Fox DJ, Hrabina O, Kay GL, King AK, Malina J, Millard AD, Moat J, Roper DI, Song H, Waterfield NR, Scott P. Metallohelices that kill Gram-negative pathogens using intracellular antimicrobial peptide pathways. Chem Sci 2019; 10:9708-9720. [PMID: 32015803 PMCID: PMC6977464 DOI: 10.1039/c9sc03532j] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
A range of new water-compatible optically pure metallohelices - made by self-assembly of simple non-peptidic organic components around Fe ions - exhibit similar architecture to some natural cationic antimicrobial peptides (CAMPs) and are found to have high, structure-dependent activity against bacteria, including clinically problematic Gram-negative pathogens. A key compound is shown to freely enter rapidly dividing E. coli cells without significant membrane disruption, and localise in distinct foci near the poles. Several related observations of CAMP-like mechanisms are made via biophysical measurements, whole genome sequencing of tolerance mutants and transcriptomic analysis. These include: high selectivity for binding of G-quadruplex DNA over double stranded DNA; inhibition of both DNA gyrase and topoisomerase I in vitro; curing of a plasmid that contributes to the very high virulence of the E. coli strain used; activation of various two-component sensor/regulator and acid response pathways; and subsequent attempts by the cell to lower the net negative charge of the surface. This impact of the compound on multiple structures and pathways corresponds with our inability to isolate fully resistant mutant strains, and supports the idea that CAMP-inspired chemical scaffolds are a realistic approach for antimicrobial drug discovery, without the practical barriers to development that are associated with natural CAMPS.
Collapse
Affiliation(s)
- Daniel H Simpson
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - Alexia Hapeshi
- Warwick Medical School , University of Warwick , Coventry , CV4 7AL , UK
| | - Nicola J Rogers
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - Viktor Brabec
- The Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Guy J Clarkson
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - David J Fox
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - Ondrej Hrabina
- The Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
- Department of Biophysics , Palacky University , Slechtitelu 27 , 783 71 Olomouc , Czech Republic
| | - Gemma L Kay
- Warwick Medical School , University of Warwick , Coventry , CV4 7AL , UK
| | - Andrew K King
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | - Jaroslav Malina
- The Czech Academy of Sciences , Institute of Biophysics , Kralovopolska 135 , CZ-61265 Brno , Czech Republic
| | - Andrew D Millard
- Warwick Medical School , University of Warwick , Coventry , CV4 7AL , UK
| | - John Moat
- School of Life Sciences , University of Warwick , Gibbet Hill Campus , Coventry , CV4 7AL , UK
| | - David I Roper
- School of Life Sciences , University of Warwick , Gibbet Hill Campus , Coventry , CV4 7AL , UK
| | - Hualong Song
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| | | | - Peter Scott
- Department of Chemistry , University of Warwick , Gibbet Hill Road , Coventry , CV4 7AL , UK .
| |
Collapse
|
218
|
Peng J, Wu Z, Liu W, Long H, Zhu G, Guo G, Wu J. Antimicrobial functional divergence of the cecropin antibacterial peptide gene family in Musca domestica. Parasit Vectors 2019; 12:537. [PMID: 31727142 PMCID: PMC6857134 DOI: 10.1186/s13071-019-3793-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 11/05/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND It has been reported that there are more than ten antimicrobial peptides (AMPs) belonging to the cecropin family in Musca domestica; however, few of them have been identified, and the functions of the other molecules are poorly understood. METHODS Sequences of the M. domestica cecropin family of genes were cloned from cDNA template, which was reverse-transcribed from total mRNA isolated from third-instar larvae of M. domestica that were challenged with pathogens. Sequence analysis was performed using DNAMAN comprehensive analysis software, and a molecular phylogenetic tree of the cecropin family was constructed using the Neighbour-Joining method in MEGA v.5.0 according to the mature peptide sequences. Antibacterial activity of the synthetic M. domestica cecropin protein was detected and the minimum inhibitory concentration (MIC) values were determined using broth microdilution techniques. Time-killing assays were performed on the Gram-negative bacteria, Acinetobacter baumannii, at the logarithmic or stabilizing stages of growth, and its morphological changes when treated with Cec4 were assessed by scanning electron microscopy (SEM) and detection of leakage of 260 nm absorbing material. RESULTS Eleven cecropin family genes, namely Cec01, Cec02 and Cec1-9, show homology to the Cec form in a multigene family on the Scaffold18749 of M. domestica. In comparing the encoded cecropin protein sequences, most of them have the basic characteristics of the cecropin family, containing 19 conservative amino acid residues. To our knowledge, this is the first experimental demonstration that most genes in the Cec family are functional. Cec02, Cec1, Cec2, Cec5 and Cec7 have similar antibacterial spectra and antibacterial effects against Gram-negative bacteria, while Cec4 displays a more broad-spectrum of antimicrobial activity and has a very strong effect on A. baumannii. Cec4 eliminated A. baumannii in a rapid and concentration-dependent manner, with antibacterial effects within 24 h at 1× MIC and 2× MIC. Furthermore, SEM analysis and the leakage of 260 nm absorbing material detection indicated that Cec4 sterilized the bacteria through the disruption of cell membrane integrity. CONCLUSIONS Although there are more than ten cecropin genes related to M. domestica, some of them have no preferred antibacterial activity other than Cec4 against A. baumannii.
Collapse
Affiliation(s)
- Jian Peng
- Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, People's Republic of China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550004, People's Republic of China.,The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Zhaoying Wu
- Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Weiwei Liu
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Huiling Long
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Guiming Zhu
- Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Guo Guo
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550004, People's Republic of China.
| | - Jianwei Wu
- The Key and Characteristic Laboratory of Modern Pathogen Biology, Guizhou Medical University, Guiyang, 550004, People's Republic of China.
| |
Collapse
|
219
|
Xu Z, Li M, Li Y, Cao H, Miao L, Xu Z, Higuchi Y, Yamasaki S, Nishino K, Woo PC, Xiang H, Yan A. Native CRISPR-Cas-Mediated Genome Editing Enables Dissecting and Sensitizing Clinical Multidrug-Resistant P. aeruginosa. Cell Rep 2019; 29:1707-1717.e3. [DOI: 10.1016/j.celrep.2019.10.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 09/09/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
|
220
|
Wang Y, Feng T, Li H, Yu Y, Han Y, Zhang J, Li X, Li Y, Zhang XH. A novel heterologous expression strategy for the quorum-quenching enzyme MomL in Lysobacter enzymogenes to the inhibit pathogenicity of Pectobacterium. Appl Microbiol Biotechnol 2019; 103:8889-8898. [PMID: 31656979 DOI: 10.1007/s00253-019-10166-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/20/2019] [Accepted: 09/28/2019] [Indexed: 01/28/2023]
Abstract
Quorum-quenching (QQ) enzymes can block the quorum-sensing (QS) system and prevent the expression of QS-controlled pathogenic factors in bacteria. However, the low expression levels of QQ proteins in the original host bacteria have affected their widespread application. In this study, we heterologously expressed momL, encoding a QQ enzyme with high activity, in Lysobacter enzymogenes. A "yellow-to-white" selection marker and the high-constitutive-expression promoter PgroEL were used in this novel heterologous expression system. In addition, we optimized the spacer between the SD sequence and the initiator to improve the efficiency of the expression system by 1.54-fold. The engineered strain LeMomL degraded the AHL molecule and the virulence factors of Pectobacterium carotovorum subsp. carotovora (Pcc). Additionally, LeMomL significantly decreased the disease caused by Pcc in Chinese cabbages and carrot root tissues. In conclusion, this novel and facile L. enzymogenes expression strategy has good prospects and is an ideal approach for foreign protein expression.
Collapse
Affiliation(s)
- Yan Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China. .,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China. .,Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
| | - Tao Feng
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Hui Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yameng Yu
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yong Han
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA
| | - Jingjing Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Xiaoyu Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yaoyao Li
- Key Laboratory of Chemical Biology, School of Pharmaceutical Sciences, Shandong University, Jinan, 250100, China
| | - Xiao-Hua Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China. .,Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China. .,Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
221
|
Shruti SR, Rajasekaran R. Identification of therapeutic peptide scaffold from tritrpticin family for urinary tract infections using in silico techniques. J Biomol Struct Dyn 2019; 38:4407-4417. [DOI: 10.1080/07391102.2019.1680437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- S. R. Shruti
- Department of Biotechnology, School of Biosciences and Technology, VIT (Deemed to Be University), Vellore, India
| | - R. Rajasekaran
- Department of Biotechnology, School of Biosciences and Technology, VIT (Deemed to Be University), Vellore, India
| |
Collapse
|
222
|
The Ancestral N-Terminal Domain of Big Defensins Drives Bacterially Triggered Assembly into Antimicrobial Nanonets. mBio 2019; 10:mBio.01821-19. [PMID: 31641083 PMCID: PMC6805989 DOI: 10.1128/mbio.01821-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
β-Defensins are host defense peptides controlling infections in species ranging from humans to invertebrates. However, the antimicrobial activity of most human β-defensins is impaired at physiological salt concentrations. We explored the properties of big defensins, the β-defensin ancestors, which have been conserved in a number of marine organisms, mainly mollusks. By focusing on a big defensin from oyster (Cg-BigDef1), we showed that the N-terminal domain lost during evolution toward β-defensins confers bactericidal activity to Cg-BigDef1, even at high salt concentrations. Cg-BigDef1 killed multidrug-resistant human clinical isolates of Staphylococcus aureus. Moreover, the ancestral N-terminal domain drove the assembly of the big defensin into nanonets in which bacteria are entrapped and killed. This discovery may explain why the ancestral N-terminal domain has been maintained in diverse marine phyla and creates a new path of discovery to design β-defensin derivatives active at physiological and high salt concentrations. Big defensins, ancestors of β-defensins, are composed of a β-defensin-like C-terminal domain and a globular hydrophobic ancestral N-terminal domain. This unique structure is found in a limited number of phylogenetically distant species, including mollusks, ancestral chelicerates, and early-branching cephalochordates, mostly living in marine environments. One puzzling evolutionary issue concerns the advantage for these species of having maintained a hydrophobic domain lost during evolution toward β-defensins. Using native ligation chemistry, we produced the oyster Crassostrea gigas BigDef1 (Cg-BigDef1) and its separate domains. Cg-BigDef1 showed salt-stable and broad-range bactericidal activity, including against multidrug-resistant human clinical isolates of Staphylococcus aureus. We found that the ancestral N-terminal domain confers salt-stable antimicrobial activity to the β-defensin-like domain, which is otherwise inactive. Moreover, upon contact with bacteria, the N-terminal domain drives Cg-BigDef1 assembly into nanonets that entrap and kill bacteria. We speculate that the hydrophobic N-terminal domain of big defensins has been retained in marine phyla to confer salt-stable interactions with bacterial membranes in environments where electrostatic interactions are impaired. Those remarkable properties open the way to future drug developments when physiological salt concentrations inhibit the antimicrobial activity of vertebrate β-defensins.
Collapse
|
223
|
Spohn R, Daruka L, Lázár V, Martins A, Vidovics F, Grézal G, Méhi O, Kintses B, Számel M, Jangir PK, Csörgő B, Györkei Á, Bódi Z, Faragó A, Bodai L, Földesi I, Kata D, Maróti G, Pap B, Wirth R, Papp B, Pál C. Integrated evolutionary analysis reveals antimicrobial peptides with limited resistance. Nat Commun 2019; 10:4538. [PMID: 31586049 PMCID: PMC6778101 DOI: 10.1038/s41467-019-12364-6] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/27/2019] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial peptides (AMPs) are promising antimicrobials, however, the potential of bacterial resistance is a major concern. Here we systematically study the evolution of resistance to 14 chemically diverse AMPs and 12 antibiotics in Escherichia coli. Our work indicates that evolution of resistance against certain AMPs, such as tachyplesin II and cecropin P1, is limited. Resistance level provided by point mutations and gene amplification is very low and antibiotic-resistant bacteria display no cross-resistance to these AMPs. Moreover, genomic fragments derived from a wide range of soil bacteria confer no detectable resistance against these AMPs when introduced into native host bacteria on plasmids. We have found that simple physicochemical features dictate bacterial propensity to evolve resistance against AMPs. Our work could serve as a promising source for the development of new AMP-based therapeutics less prone to resistance, a feature necessary to avoid any possible interference with our innate immune system.
Collapse
Affiliation(s)
- Réka Spohn
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Viktória Lázár
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ana Martins
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Fanni Vidovics
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Gábor Grézal
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Orsolya Méhi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Bálint Kintses
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Mónika Számel
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Pramod K Jangir
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Bálint Csörgő
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- University of California, San Francisco, Department of Microbiology and Immunology, San Francisco, CA, USA
| | - Ádám Györkei
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Zoltán Bódi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Anikó Faragó
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - László Bodai
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, University of Szeged, Szeged, Hungary
| | - Diána Kata
- Department of Laboratory Medicine, University of Szeged, Szeged, Hungary
| | - Gergely Maróti
- Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Bernadett Pap
- Institute of Plant Biology, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Roland Wirth
- Department of Biotechnology, University of Szeged, Szeged, Hungary
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
224
|
Maltas J, Wood KB. Pervasive and diverse collateral sensitivity profiles inform optimal strategies to limit antibiotic resistance. PLoS Biol 2019; 17:e3000515. [PMID: 31652256 PMCID: PMC6834293 DOI: 10.1371/journal.pbio.3000515] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 11/06/2019] [Accepted: 10/07/2019] [Indexed: 11/19/2022] Open
Abstract
Evolved resistance to one antibiotic may be associated with "collateral" sensitivity to other drugs. Here, we provide an extensive quantitative characterization of collateral effects in Enterococcus faecalis, a gram-positive opportunistic pathogen. By combining parallel experimental evolution with high-throughput dose-response measurements, we measure phenotypic profiles of collateral sensitivity and resistance for a total of 900 mutant-drug combinations. We find that collateral effects are pervasive but difficult to predict because independent populations selected by the same drug can exhibit qualitatively different profiles of collateral sensitivity as well as markedly different fitness costs. Using whole-genome sequencing of evolved populations, we identified mutations in a number of known resistance determinants, including mutations in several genes previously linked with collateral sensitivity in other species. Although phenotypic drug sensitivity profiles show significant diversity, they cluster into statistically similar groups characterized by selecting drugs with similar mechanisms. To exploit the statistical structure in these resistance profiles, we develop a simple mathematical model based on a stochastic control process and use it to design optimal drug policies that assign a unique drug to every possible resistance profile. Stochastic simulations reveal that these optimal drug policies outperform intuitive cycling protocols by maintaining long-term sensitivity at the expense of short-term periods of high resistance. The approach reveals a new conceptual strategy for mitigating resistance by balancing short-term inhibition of pathogen growth with infrequent use of drugs intended to steer pathogen populations to a more vulnerable future state. Experiments in laboratory populations confirm that model-inspired sequences of four drugs reduce growth and slow adaptation relative to naive protocols involving the drugs alone, in pairwise cycles, or in a four-drug uniform cycle.
Collapse
Affiliation(s)
- Jeff Maltas
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kevin B. Wood
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Physics, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
225
|
Bacterial cross-resistance to anti-infective compounds. Is it a real problem? Curr Opin Pharmacol 2019; 48:76-81. [DOI: 10.1016/j.coph.2019.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 12/30/2022]
|
226
|
Qu J, Huang Y, Lv X. Crisis of Antimicrobial Resistance in China: Now and the Future. Front Microbiol 2019; 10:2240. [PMID: 31611863 PMCID: PMC6777638 DOI: 10.3389/fmicb.2019.02240] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/12/2019] [Indexed: 02/05/2023] Open
Abstract
The crisis of antimicrobial resistance is worsening and has become a major public safety problem in China, seriously endangering human and animal health and ecological environment. Gram-negative bacterial resistance in China is severe: the related pathogens mainly include carbapenem-resistant Acinetobacter, Pseudomonas aeruginosa and Klebsiella pneumoniae. Surging antimicrobial consumption and irrational use of antimicrobials are the main causes of resistance. In China, a variety of strategies are implemented to control the antimicrobial resistance in hospitals, agriculture and environment. However, there is still a long way to go to strengthen the drug resistance surveillance, to reduce the emergence of drug-resistant bacteria, and to find new antimicrobials and therapies for drug-resistant bacteria. Controlling the antimicrobial resistance crisis takes great efforts from the whole society.
Collapse
Affiliation(s)
- Junyan Qu
- Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Yimei Huang
- College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Xiaoju Lv
- Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
227
|
Mohan NM, Zorgani A, Jalowicki G, Kerr A, Khaldi N, Martins M. Unlocking NuriPep 1653 From Common Pea Protein: A Potent Antimicrobial Peptide to Tackle a Pan-Drug Resistant Acinetobacter baumannii. Front Microbiol 2019; 10:2086. [PMID: 31620099 PMCID: PMC6759681 DOI: 10.3389/fmicb.2019.02086] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/23/2019] [Indexed: 12/22/2022] Open
Abstract
While the antibiotic era has come and gone, antimicrobial peptides (AMPs) hold promise as novel therapies to treat multidrug resistant (MDR) pathogens in an age where the threat of multidrug resistance escalates worldwide. Here, we report the bactericidal properties of NuriPep 1653, a novel 22 mer and non-modified peptide. NuriPep 1653 was identified within the sequence of the non-antimicrobial P54 protein, which is involved in nutrient reservoir activity in Pisum sativum. Total bacterial clearance of Acinetobacter baumannii cells (1 × 108 cells/mL) was observed using only 4 × MIC (48 μg/mL) of NuriPep 1653 after just 20 min of treatment. We uncovered a synergistic interaction between NuriPep 1653 and another antimicrobial peptide, colistin. The MIC of NuriPep 1653 and colistin dropped from 12 and 8 μg/mL to 2 and 1 μg/mL, respectively, when they were combined. NuriPep 1653 exhibits no cytotoxicity in different human cell lines and has a low propensity to induce bacterial resistance in a colistin resistant clinical isolate of A. baumannii. The existence of these peptides embedded in proteins unearths potentially new classes of antimicrobials with activity against clinically relevant pathogens. Our findings push the boundaries of traditional peptide discovery and represent a leading edge for natural bioactive compounds which may have a common existence in nature but remain unexposed.
Collapse
Affiliation(s)
- Niamh Maire Mohan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, University of Dublin, Dublin, Ireland
- Nuritas Limited, Dublin, Ireland
| | | | | | | | | | - Marta Martins
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
228
|
Salazar VA, Arranz-Trullén J, Prats-Ejarque G, Torrent M, Andreu D, Pulido D, Boix E. Insight into the Antifungal Mechanism of Action of Human RNase N-terminus Derived Peptides. Int J Mol Sci 2019; 20:ijms20184558. [PMID: 31540052 PMCID: PMC6770517 DOI: 10.3390/ijms20184558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
Candida albicans is a polymorphic fungus responsible for mucosal and skin infections. Candida cells establish themselves into biofilm communities resistant to most currently available antifungal agents. An increase of severe infections ensuing in fungal septic shock in elderly or immunosuppressed patients, along with the emergence of drug-resistant strains, urge the need for the development of alternative antifungal agents. In the search for novel antifungal drugs our laboratory demonstrated that two human ribonucleases from the vertebrate-specific RNaseA superfamily, hRNase3 and hRNase7, display a high anticandidal activity. In a previous work, we proved that the N-terminal region of the RNases was sufficient to reproduce most of the parental protein bactericidal activity. Next, we explored their potency against a fungal pathogen. Here, we have tested the N-terminal derived peptides that correspond to the eight human canonical RNases (RN1-8) against planktonic cells and biofilms of C. albicans. RN3 and RN7 peptides displayed the most potent inhibitory effect with a mechanism of action characterized by cell-wall binding, membrane permeabilization and biofilm eradication activities. Both peptides are able to eradicate planktonic and sessile cells, and to alter their gene expression, reinforcing its role as a lead candidate to develop novel antifungal and antibiofilm therapies.
Collapse
Affiliation(s)
- Vivian A Salazar
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| | - Javier Arranz-Trullén
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| | - Guillem Prats-Ejarque
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| | - Marc Torrent
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| | - David Andreu
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - David Pulido
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| |
Collapse
|
229
|
Agbale CM, Sarfo JK, Galyuon IK, Juliano SA, Silva GGO, Buccini DF, Cardoso MH, Torres MDT, Angeles-Boza AM, de la Fuente-Nunez C, Franco OL. Antimicrobial and Antibiofilm Activities of Helical Antimicrobial Peptide Sequences Incorporating Metal-Binding Motifs. Biochemistry 2019; 58:3802-3812. [PMID: 31448597 DOI: 10.1021/acs.biochem.9b00440] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antimicrobial peptides (AMPs) represent alternative strategies to combat the global health problem of antibiotic resistance. However, naturally occurring AMPs are generally not sufficiently active for use as antibiotics. Optimized synthetic versions incorporating additional design principles are needed. Here, we engineered amino-terminal Cu(II) and Ni(II) (ATCUN) binding motifs, which can enhance biological function, into the native sequence of two AMPs, CM15 and citropin1.1. The incorporation of metal-binding motifs modulated the antimicrobial activity of synthetic peptides against a panel of carbapenem-resistant enterococci (CRE) bacteria, including carbapenem-resistant Klebsiella pneumoniae (KpC+) and Escherichia coli (KpC+). Activity modulation depended on the type of ATCUN variant utilized. Membrane permeability assays revealed that the in silico selected lead template, CM15, and its ATCUN analogs increased bacterial cell death. Mass spectrometry, circular dichroism, and molecular dynamics simulations indicated that coordinating ATCUN derivatives with Cu(II) ions did not increase the helical tendencies of the AMPs. CM15 ATCUN variants, when combined with Meropenem, streptomycin, or chloramphenicol, showed synergistic effects against E. coli (KpC+ 1812446) biofilms. Motif addition also reduced the hemolytic activity of the wild-type AMP and improved the survival rate of mice in a systemic infection model. The dependence of these bioactivities on the particular amino acids of the ATCUN motif highlights the possible use of size, charge, and hydrophobicity to fine-tune AMP biological function. Our data indicate that incorporating metal-binding motifs into peptide sequences leads to synthetic variants with modified biological properties. These principles may be applied to augment the activities of other peptide sequences.
Collapse
Affiliation(s)
- Caleb M Agbale
- S-INOVA Biotech, Programa de Pós-Graduação em Biotecnologia , Universidade Católica Dom Bosco , Campo Grande , Mato Grosso Do Sul, MS 79117-900 , Brazil.,Department of Biochemistry, School of Biological Sciences, College of Agriculture and Natural Sciences , University of Cape Coast , Cape Coast , Ghana.,Department of Molecular Biology and Biotechnology, School of Biological Sciences, College of Agriculture and Natural Sciences , University of Cape Coast , Cape Coast , Ghana
| | - Justice K Sarfo
- S-INOVA Biotech, Programa de Pós-Graduação em Biotecnologia , Universidade Católica Dom Bosco , Campo Grande , Mato Grosso Do Sul, MS 79117-900 , Brazil
| | - Isaac K Galyuon
- Department of Molecular Biology and Biotechnology, School of Biological Sciences, College of Agriculture and Natural Sciences , University of Cape Coast , Cape Coast , Ghana
| | - Samuel A Juliano
- Department of Chemistry , University of Connecticut , Storrs , Connecticut 06269 , United States
| | - Gislaine G O Silva
- S-INOVA Biotech, Programa de Pós-Graduação em Biotecnologia , Universidade Católica Dom Bosco , Campo Grande , Mato Grosso Do Sul, MS 79117-900 , Brazil
| | - Danieli F Buccini
- S-INOVA Biotech, Programa de Pós-Graduação em Biotecnologia , Universidade Católica Dom Bosco , Campo Grande , Mato Grosso Do Sul, MS 79117-900 , Brazil
| | - Marlon H Cardoso
- S-INOVA Biotech, Programa de Pós-Graduação em Biotecnologia , Universidade Católica Dom Bosco , Campo Grande , Mato Grosso Do Sul, MS 79117-900 , Brazil.,Centro de Análises de Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia , Universidade Católica de Brasília , Brasília , DF 70790-160 , Brazil.,Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina , Universidade de Brasília , Brasília , DF 70910-900 , Brazil
| | - Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Perelman School of Medicine, and Department of Bioengineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Alfredo M Angeles-Boza
- Department of Chemistry , University of Connecticut , Storrs , Connecticut 06269 , United States.,Institute of Materials Science , University of Connecticut , Storrs , Connecticut 06269 , United States
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Perelman School of Medicine, and Department of Bioengineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Octavio L Franco
- S-INOVA Biotech, Programa de Pós-Graduação em Biotecnologia , Universidade Católica Dom Bosco , Campo Grande , Mato Grosso Do Sul, MS 79117-900 , Brazil.,Centro de Análises de Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia , Universidade Católica de Brasília , Brasília , DF 70790-160 , Brazil.,Programa de Pós-Graduação em Patologia Molecular, Faculdade de Medicina , Universidade de Brasília , Brasília , DF 70910-900 , Brazil
| |
Collapse
|
230
|
Rapid Evolution of Reduced Susceptibility against a Balanced Dual-Targeting Antibiotic through Stepping-Stone Mutations. Antimicrob Agents Chemother 2019; 63:AAC.00207-19. [PMID: 31235632 DOI: 10.1128/aac.00207-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/14/2019] [Indexed: 11/20/2022] Open
Abstract
Multitargeting antibiotics, i.e., single compounds capable of inhibiting two or more bacterial targets, are generally considered to be a promising therapeutic strategy against resistance evolution. The rationale for this theory is that multitargeting antibiotics demand the simultaneous acquisition of multiple mutations at their respective target genes to achieve significant resistance. The theory presumes that individual mutations provide little or no benefit to the bacterial host. Here, we propose that such individual stepping-stone mutations can be prevalent in clinical bacterial isolates, as they provide significant resistance to other antimicrobial agents. To test this possibility, we focused on gepotidacin, an antibiotic candidate that selectively inhibits both bacterial DNA gyrase and topoisomerase IV. In a susceptible organism, Klebsiella pneumoniae, a combination of two specific mutations in these target proteins provide an >2,000-fold reduction in susceptibility, while individually, none of these mutations affect resistance significantly. Alarmingly, strains with decreased susceptibility against gepotidacin are found to be as virulent as the wild-type Klebsiella pneumoniae strain in a murine model. Moreover, numerous pathogenic isolates carry mutations which could promote the evolution of clinically significant reduction of susceptibility against gepotidacin in the future. As might be expected, prolonged exposure to ciprofloxacin, a clinically widely employed gyrase inhibitor, coselected for reduced susceptibility against gepotidacin. We conclude that extensive antibiotic usage could select for mutations that serve as stepping-stones toward resistance against antimicrobial compounds still under development. Our research indicates that even balanced multitargeting antibiotics are prone to resistance evolution.
Collapse
|
231
|
Peng J, Long H, Liu W, Wu Z, Wang T, Zeng Z, Guo G, Wu J. Antibacterial mechanism of peptide Cec4 against Acinetobacter baumannii. Infect Drug Resist 2019; 12:2417-2428. [PMID: 31496754 PMCID: PMC6689099 DOI: 10.2147/idr.s214057] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/11/2019] [Indexed: 01/23/2023] Open
Abstract
Background A case of Acinetobacter baumannii (A. baumannii), known as gram-negative bacteria, causes a range of nosocomial infections. Due to the continuous detection of multi-drug resistant A. baumannii in the clinic, there is an urgent need to find alternative therapies, including broad-spectrum antibacterial peptides (AMP). Recently it has been found that the peptide Cec4 has good antibacterial activity against A. baumannii, but the antibacterial mechanism remains elusive. Materials and methods The basic structure of Cec4 was analyzed by circular dichroism (CD) spectroscopy, and the potential antibacterial mechanism of Cec4 was detected by flow cytometry, transmission electron microscopy, fluorescence and confocal microscopy. The minimum inhibitory concentration (MIC) of antimicrobial peptides against various A. baumannii was determinated with broth microdilution techniques. The biofilm formation and the sensitivity detection of biofilms to antimicrobial peptides were detected by crystal violet staining. Results In this study, the main secondary structure of the antibacterial peptide Cec4 is α-helix (99.7%) in the hydrophobic environment. Furthermore, after the treatment with Cec4, an amount of leakage of A. baumannii and the destruction of its cell membrane were detected. Moreover, it was observed that FITC-Cec4 can enter the cell, and more cells were held in the G1 phase with peptide Cec4. However, the DNA binding assay of the peptide Cec4 indicates that the peptide does not target DNA. In addition, peptide Cec4 was superior in reducing adherent biofilms of A. baumannii compared to conventional antibiotics and has no cytotoxicity. Conclusion It is apparent that the antibacterial peptide Cec4 may achieve rapid sterilization by multi-target interaction and presents an attractive therapeutic option for the prevention and control of A. baumannii infections.
Collapse
Affiliation(s)
- Jian Peng
- Key Laboratory of Biology and Medical Engineering, Department of Biotechnology, School of Biology & Engineering, Guizhou Medical University, Guiyang 550004, People's Republic of China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550004, People's Republic of China.,Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Human Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Huiling Long
- Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Human Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Weiwei Liu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550004, People's Republic of China.,Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Human Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Zhaoying Wu
- Key Laboratory of Biology and Medical Engineering, Department of Biotechnology, School of Biology & Engineering, Guizhou Medical University, Guiyang 550004, People's Republic of China.,Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Human Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Tao Wang
- Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Human Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Zhu Zeng
- Key Laboratory of Biology and Medical Engineering, Department of Biotechnology, School of Biology & Engineering, Guizhou Medical University, Guiyang 550004, People's Republic of China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550004, People's Republic of China.,Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Human Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Guo Guo
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550004, People's Republic of China.,Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Human Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, People's Republic of China
| | - Jianwei Wu
- Key and Characteristic Laboratory of Modern Pathogen Biology, Department of Human Parasitology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550004, People's Republic of China
| |
Collapse
|
232
|
Talpin A, Kattah MG, Advincula R, Fadrosh D, Lynch K, LaMere B, Fujimura KE, Nagalingam NA, Malynn BA, Lynch SV, Ma A. A20 in dendritic cells restrains intestinal anti-bacterial peptide expression and preserves commensal homeostasis. PLoS One 2019; 14:e0218999. [PMID: 31295268 PMCID: PMC6622485 DOI: 10.1371/journal.pone.0218999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/13/2019] [Indexed: 12/24/2022] Open
Abstract
Microbial dysbiosis commonly occurs in patients with inflammatory bowel diseases (IBD). Exogenous causes of dysbiosis such as antibiotics and diet are well described, but host derived causes are understudied. A20 is a potent regulator of signals triggered by microbial pattern molecules, and A20 regulates susceptibility to intestinal inflammation in mice and in humans. We now report that mice lacking A20 expression in dendritic cells, A20FL/FL CD11c-Cre mice (or A20dDC mice), spontaneously develop colitogenic intestinal dysbiosis that is evident upon weaning and precedes the onset of colitis. Intestines from A20dDC mice express increased amounts of Reg3β and Reg3γ, but not Ang4. A20 deficient DCs promote gut microbiota perturbation in the absence of adaptive lymphocytes. Moreover, A20 deficient DCs directly induce expression of Reg3β and Reg3γ but not Ang 4 in normal intestinal epithelial cell enteroid cultures in the absence of other cell types. These findings reveal a pathophysiological pathway in which defective expression of an IBD susceptibility gene in DCs drives aberrant expression of anti-bacterial peptides and luminal dysbiosis that in turn confers host susceptibility to intestinal inflammation.
Collapse
Affiliation(s)
- Alice Talpin
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Michael G. Kattah
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Rommel Advincula
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Douglas Fadrosh
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Kole Lynch
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Brandon LaMere
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Kei E. Fujimura
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Nabeetha A. Nagalingam
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Barbara A. Malynn
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Susan V. Lynch
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
233
|
Lu L, Arranz-Trullén J, Prats-Ejarque G, Pulido D, Bhakta S, Boix E. Human Antimicrobial RNases Inhibit Intracellular Bacterial Growth and Induce Autophagy in Mycobacteria-Infected Macrophages. Front Immunol 2019; 10:1500. [PMID: 31312205 PMCID: PMC6614385 DOI: 10.3389/fimmu.2019.01500] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
The development of novel treatment against tuberculosis is a priority global health challenge. Antimicrobial proteins and peptides offer a multifaceted mechanism suitable to fight bacterial resistance. Within the RNaseA superfamily there is a group of highly cationic proteins secreted by innate immune cells with anti-infective and immune-regulatory properties. In this work, we have tested the human canonical members of the RNase family using a spot-culture growth inhibition assay based mycobacteria-infected macrophage model for evaluating their anti-tubercular properties. Out of the seven tested recombinant human RNases, we have identified two members, RNase3 and RNase6, which were highly effective against Mycobacterium aurum extra- and intracellularly and induced an autophagy process. We observed the proteins internalization within macrophages and their capacity to eradicate the intracellular mycobacterial infection at a low micro-molar range. Contribution of the enzymatic activity was discarded by site-directed mutagenesis at the RNase catalytic site. The protein induction of autophagy was analyzed by RT-qPCR, western blot, immunofluorescence, and electron microscopy. Specific blockage of auto-phagosome formation and maturation reduced the protein's ability to eradicate the infection. In addition, we found that the M. aurum infection of human THP1 macrophages modulates the expression of endogenous RNase3 and RNase6, suggesting a function in vivo. Overall, our data anticipate a biological role for human antimicrobial RNases in host response to mycobacterial infections and set the basis for the design of novel anti-tubercular drugs.
Collapse
Affiliation(s)
- Lu Lu
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Javier Arranz-Trullén
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Mycobacteria Research Laboratory, Department of Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, London, United Kingdom
| | - Guillem Prats-Ejarque
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - David Pulido
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Sanjib Bhakta
- Mycobacteria Research Laboratory, Department of Biological Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, London, United Kingdom
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
234
|
Abstract
Rebecca S. Shapiro studies antimicrobial resistance and genetic interaction networks. In this mSphere of Influence article, she reflects on how the papers "Bacterial evolution of antibiotic hypersensitivity" by Lázár et al. (V. Lázár, G. Pal Singh, R. Spohn, I. Nagy, et al., Mol Syst Biol 9:700, 2013, https://doi.org/10.1038/msb.2013.57) and "Use of collateral sensitivity networks to design drug cycling protocols that avoid resistance development" by L. Imamovic and M. O. A. Sommer (Sci Transl Med 5:204ra132, 2013, https://doi.org/10.1126/scitranslmed.3006609) impacted her thinking about multigene interaction effects on drug resistance.
Collapse
|
235
|
Prats-Ejarque G, Li J, Ait-Ichou F, Lorente H, Boix E. Testing a Human Antimicrobial RNase Chimera Against Bacterial Resistance. Front Microbiol 2019; 10:1357. [PMID: 31275278 PMCID: PMC6594349 DOI: 10.3389/fmicb.2019.01357] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 05/31/2019] [Indexed: 12/11/2022] Open
Abstract
The emergence of bacterial resistance to the most commonly used antibiotics encourages the design of novel antimicrobial drugs. Antimicrobial proteins and peptides (AMPs) are the key players in host innate immunity. They exert a rapid and multifaceted action that reduces the development of bacterial adaptation mechanisms. Human antimicrobial RNases belonging to the vertebrate specific RNase A superfamily participate in the maintenance of tissue and body fluid sterility. Among the eight human canonical RNases, RNase 3 stands out as the most cationic and effective bactericidal protein against Gram-negative species. Its enhanced ability to disrupt the bacterial cell wall has evolved in detriment of its catalytic activity. Based on structure-functional studies we have designed an RNase 3/1 hybrid construct that combines the high catalytic activity of RNase 1 with RNase 3 bactericidal properties. Next, we have explored the ability of this hybrid RNase to target the development of bacterial resistance on an Acinetobacter baumannii cell culture. Synergy assays were performed in combination with colistin, a standard antimicrobial peptide used as an antibiotic to treat severe infections. Positive synergism was observed between colistin and the RNase 3/1 hybrid protein. Subsequently, using an in vitro experimental evolution assay, by exposure of a bacterial culture to colistin at incremental doses, we demonstrated the ability of the RNase 3/1 construct to reduce the emergence of bacterial antimicrobial resistance. The results advance the potential applicability of RNase-based drugs as antibiotic adjuvants.
Collapse
Affiliation(s)
| | | | | | | | - Ester Boix
- Faculty of Biosciences, Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
236
|
De Novo Design and In Vitro Testing of Antimicrobial Peptides against Gram-Negative Bacteria. Pharmaceuticals (Basel) 2019; 12:ph12020082. [PMID: 31163671 PMCID: PMC6631481 DOI: 10.3390/ph12020082] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/26/2019] [Accepted: 05/30/2019] [Indexed: 12/13/2022] Open
Abstract
Antimicrobial peptides (AMPs) have been identified as a potentially new class of antibiotics to combat bacterial resistance to conventional drugs. The design of de novo AMPs with high therapeutic indexes, low cost of synthesis, high resistance to proteases and high bioavailability remains a challenge. Such design requires computational modeling of antimicrobial properties. Currently, most computational methods cannot accurately calculate antimicrobial potency against particular strains of bacterial pathogens. We developed a tool for AMP prediction (Special Prediction (SP) tool) and made it available on our Web site (https://dbaasp.org/prediction). Based on this tool, a simple algorithm for the design of de novo AMPs (DSP) was created. We used DSP to design short peptides with high therapeutic indexes against gram-negative bacteria. The predicted peptides have been synthesized and tested in vitro against a panel of gram-negative bacteria, including drug resistant ones. Predicted activity against Escherichia coli ATCC 25922 was experimentally confirmed for 14 out of 15 peptides. Further improvements for designed peptides included the synthesis of D-enantiomers, which are traditionally used to increase resistance against proteases. One synthetic D-peptide (SP15D) possesses one of the lowest values of minimum inhibitory concentration (MIC) among all DBAASP database short peptides at the time of the submission of this article, while being highly stable against proteases and having a high therapeutic index. The mode of anti-bacterial action, assessed by fluorescence microscopy, shows that SP15D acts similarly to cell penetrating peptides. SP15D can be considered a promising candidate for the development of peptide antibiotics. We plan further exploratory studies with the SP tool, aiming at finding peptides which are active against other pathogenic organisms.
Collapse
|
237
|
Abstract
Bacterial antibiotic resistance modulation by small signaling molecules is an emerging mechanism that has been increasingly reported in recent years. Several studies indicate that indole, an interkingdom signaling molecule, increases bacterial antibiotic resistance. However, the mechanism through which indole reduces antibiotic resistance is largely unknown. In this study, we demonstrated a novel mechanism for indole-mediated reversal of intrinsic antibiotic resistance in Lysobacter This reversal was facilitated by a novel BtuD-associated dual-function importer that can transfer both vitamin B12 and antibiotics. Indole stimulated btuD overexpression and promoted efficient absorption of extracellular vitamin B12; meanwhile, the weak selectivity of the importer caused cells to take up excessive doses of antibiotics that resulted in cell death. Consistently, btuD deletion and G48Y/K49D substitution led to marked reductions in the uptake of both antibiotics and vitamin B12 This novel mechanism is common across multiple bacterial species, among which the Q-loop amino acid of BtuD proteins is Glu (E) instead of Gln (Q). Interestingly, the antibiotic resistance of Lysobacter spp. can be restored by another small quorum sensing signaling factor, 13-methyltetradecanoic acid, designated LeDSF, in response to bacterial population density. This work highlights the mechanisms underlying dynamic regulation of bacterial antibiotic resistance by small signaling molecules and suggests that the effectiveness of traditional antibiotics could be increased by coupling them with appropriate signaling molecules.IMPORTANCE Recently, signaling molecules were found to play a role in mediating antibiotic resistance. In this study, we demonstrated that indole reversed the intrinsic antibiotic resistance (IRAR) of multiple bacterial species by promoting the expression of a novel dual-function importer. In addition, population-dependent behavior induced by 13-methyltetradecanoic acid, a quorum sensing signal molecule designated LeDSF, was involved in the IRAR process. This study highlights the dynamic regulation of bacterial antibiotic resistance by small signaling molecules and provides direction for new therapeutic strategies using traditional antibiotics in combination with signaling molecules.
Collapse
|
238
|
Gold‐Ions‐Mediated Diproline Peptide Nanocarpets and Their Inhibition of Bacterial Growth. ChemistrySelect 2019. [DOI: 10.1002/slct.201900847] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
239
|
Torres MDT, de la Fuente-Nunez C. Toward computer-made artificial antibiotics. Curr Opin Microbiol 2019; 51:30-38. [PMID: 31082661 DOI: 10.1016/j.mib.2019.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
Abstract
Merging concepts from synthetic biology and computational biology may yield antibiotics that are less likely to elicit resistance than existing drugs and that yet can fight drug-resistant infections. Indeed, computer-guided strategies coupled with massively parallel high-throughput experimental methods represent a new paradigm for antibiotic discovery. Infections caused by multidrug-resistant microorganisms are increasingly deadly. In the current post-antibiotic era, many of these infections cannot be treated with our existing antimicrobial arsenal. Furthermore, we may have already exhausted the category of large molecules produced in nature having antimicrobial activity: the antibiotic scaffolds we have discovered so far may represent the majority of those that exist. The rise in drug-resistant bacteria and lack of new antibiotic classes clearly call for out-of-the-box strategies. Recent advances in computational synthetic biology have enabled the development of antimicrobials. New molecular descriptors and genetic and pattern recognition algorithms are powerful tools that bring us a step closer to developing efficient antibiotics. We review several computational tools for drug design and a number of recently generated antibiotic candidates, with an emphasis on peptide-based molecules. Design strategies can generate a diversity of synthetic antimicrobial peptides, which may help to mitigate the spread of resistance and combat multidrug-resistant microorganisms.
Collapse
Affiliation(s)
- Marcelo Der Torossian Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America.
| |
Collapse
|
240
|
Hong J, Lu X, Deng Z, Xiao S, Yuan B, Yang K. How Melittin Inserts into Cell Membrane: Conformational Changes, Inter-Peptide Cooperation, and Disturbance on the Membrane. Molecules 2019; 24:molecules24091775. [PMID: 31067828 PMCID: PMC6539814 DOI: 10.3390/molecules24091775] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 01/27/2023] Open
Abstract
Antimicrobial peptides (AMPs), as a key component of the immune defense systems of organisms, are a promising solution to the serious threat of drug-resistant bacteria to public health. As one of the most representative and extensively studied AMPs, melittin has exceptional broad-spectrum activities against microorganisms, including both Gram-positive and Gram-negative bacteria. Unfortunately, the action mechanism of melittin with bacterial membranes, especially the underlying physics of peptide-induced membrane poration behaviors, is still poorly understood, which hampers efforts to develop melittin-based drugs or agents for clinical applications. In this mini-review, we focus on recent advances with respect to the membrane insertion behavior of melittin mostly from a computational aspect. Membrane insertion is a prerequisite and key step for forming transmembrane pores and bacterial killing by melittin, whose occurrence is based on overcoming a high free-energy barrier during the transition of melittin molecules from a membrane surface-binding state to a transmembrane-inserting state. Here, intriguing simulation results on such transition are highlighted from both kinetic and thermodynamic aspects. The conformational changes and inter-peptide cooperation of melittin molecules, as well as melittin-induced disturbances to membrane structure, such as deformation and lipid extraction, are regarded as key factors influencing the insertion of peptides into membranes. The associated intermediate states in peptide conformations, lipid arrangements, membrane structure, and mechanical properties during this process are specifically discussed. Finally, potential strategies for enhancing the poration ability and improving the antimicrobial performance of AMPs are included as well.
Collapse
Affiliation(s)
- Jiajia Hong
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, China.
| | - Xuemei Lu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, China.
| | - Zhixiong Deng
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, China.
| | - Shufeng Xiao
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, China.
| | - Bing Yuan
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, China.
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, China.
| |
Collapse
|
241
|
Lu X, Liu J, Gou L, Li J, Yuan B, Yang K, Ma Y. Designing Melittin-Graphene Hybrid Complexes for Enhanced Antibacterial Activity. Adv Healthc Mater 2019; 8:e1801521. [PMID: 30866165 DOI: 10.1002/adhm.201801521] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/28/2019] [Indexed: 01/11/2023]
Abstract
Antimicrobial peptides (AMPs) promise a fundamental solution to the devastating threat of drug-resistant bacteria. However, drawbacks of AMPs (e.g., poor cell membrane penetration efficiency) seriously block their clinical use. In this work, rational design of a hybrid complex of melittin (as a representative AMP) and graphene or graphene oxide (Gra or GO) nanosheets for enhanced antibacterial ability is achieved, via combining in-silico prediction and in-tube test. In comparison to pristine melittin, the specifically designed AMP-Gra (/GO) complex exhibits remarkable efficiency in transmembrane perforation with an over tenfold decrease in the threshold working concentration of peptide; moreover, it has an up to 20-fold enhancement in antibacterial activity against both Gram-negative and Gram-positive bacteria. Such improvement is ascribed to the synergetic insertion of nanosheets and melittin due to similarity in antibacterial mechanism between them and is further regulated by the structural factors of the complex, including the intersheet spacing and surface functionalization of the Gra/GO sheets, etc. These results provide practical guidelines to engineer AMPs with nanotechnology for improved antimicrobial performances, especially based on targeted functionalization of the Gra/GO nanosheets.
Collapse
Affiliation(s)
- Xuemei Lu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and TechnologySoochow University Suzhou 215006 P. R. China
| | - Jiaojiao Liu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and TechnologySoochow University Suzhou 215006 P. R. China
| | - Lu Gou
- Shaanxi Province Key Laboratory of Quantum Information and Quantum Optoelectronic DevicesSchool of ScienceXi'an Jiaotong University Xi'an 710049 P. R. China
| | - Jingliang Li
- Institute for Frontier MaterialsDeakin University Geelong 3216 Australia
| | - Bing Yuan
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and TechnologySoochow University Suzhou 215006 P. R. China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and TechnologySoochow University Suzhou 215006 P. R. China
| | - Yuqiang Ma
- Center for Soft Condensed Matter Physics and Interdisciplinary Research and School of Physical Science and TechnologySoochow University Suzhou 215006 P. R. China
- National Laboratory of Solid State Microstructures and Department of PhysicsNanjing University Nanjing 210093 P. R. China
| |
Collapse
|
242
|
Al Tall Y, Abualhaijaa A, Alsaggar M, Almaaytah A, Masadeh M, Alzoubi KH. Design and characterization of a new hybrid peptide from LL-37 and BMAP-27. Infect Drug Resist 2019; 12:1035-1045. [PMID: 31118709 PMCID: PMC6503343 DOI: 10.2147/idr.s199473] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 03/13/2019] [Indexed: 01/11/2023] Open
Abstract
Background and purpose: The world is heading to a post-antibiotic era where the treatment of bacterial infections will not be possible even with well-known last-line antibiotics. Unfortunately, the emergence of multidrug resistant bacterial strains is uncontrollable, and the humanity will face a life-threatening fate unless new antimicrobial agents with new bacterial target sites are promptly developed. Herein, we design a hybrid antimicrobial peptide (B1) from helical parts taken from the parent peptides: LL-37 and BMAP-27. The purpose of this design is to improve the potency and enhance the toxicity profile of the parent peptides. Methods: Rational design was used to hybridize two antimicrobial peptides, in which two helical parts from the bovine analog BMAP-27, and the human cathelicidin LL-37 were used to generate a novel peptide (B1). The physicochemical properties were checked using in silico methods. The antimicrobial activities were tested against nine control and resistant strains of Gram-positive and Gram-negative bacteria. On the other hand, the antibiofilm activities were tested against four resistant strains. The cytotoxicity on mammalian cells was tested using HEK293, and the hemolysis activity was also investigated on human blood. Finally, synergistic studies were performed with four conventional antibiotics against four resistant strains of Gram-positive and Gram-negative bacteria. Results: The new peptide B1 exhibited broad-spectrum activities against all tested strains. The concentration against planktonic cells ranged between 10 and 20 µM. However, 40-60 µM were needed to eradicate the biofilms. B1 showed reduced toxicity toward mammalian cells with minimal hemolysis risk. On the other hand, the synergistic studies showed improved activities for the combined conventional antibiotics with a huge reduction in their minimum inhibitory concentration values. The concentrations of B1 peptide combined with the tested antibiotics were also decreased markedly down to 0.5 µM in some cases. Conclusion: B1 is a hybrid peptide from two cathelicidin peptides. It showed an improved activity compared to parent peptides. The hybridization was successful in this study. It generated a new potent broad-spectrum antimicrobial. The toxicity profile was improved, and the synergism with the convention antibiotics showed promising results.
Collapse
Affiliation(s)
| | | | | | | | | | - Karem H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
243
|
Zheng Y, Saitou A, Wang CM, Toyoda A, Minakuchi Y, Sekiguchi Y, Ueda K, Takano H, Sakai Y, Abe K, Yokota A, Yabe S. Genome Features and Secondary Metabolites Biosynthetic Potential of the Class Ktedonobacteria. Front Microbiol 2019; 10:893. [PMID: 31080444 PMCID: PMC6497799 DOI: 10.3389/fmicb.2019.00893] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/08/2019] [Indexed: 12/30/2022] Open
Abstract
The prevalence of antibiotic resistance and the decrease in novel antibiotic discovery in recent years necessitates the identification of potentially novel microbial resources to produce natural products. Ktedonobacteria, a class of deeply branched bacterial lineage in the ancient phylum Chloroflexi, are ubiquitous in terrestrial environments and characterized by their large genome size and complex life cycle. These characteristics indicate Ktedonobacteria as a potential active producer of bioactive compounds. In this study, we observed the existence of a putative "megaplasmid," multiple copies of ribosomal RNA operons, and high ratio of hypothetical proteins with unknown functions in the class Ktedonobacteria. Furthermore, a total of 104 antiSMASH-predicted putative biosynthetic gene clusters (BGCs) for secondary metabolites with high novelty and diversity were identified in nine Ktedonobacteria genomes. Our investigation of domain composition and organization of the non-ribosomal peptide synthetase and polyketide synthase BGCs further supports the concept that class Ktedonobacteria may produce compounds structurally different from known natural products. Furthermore, screening of bioactive compounds from representative Ktedonobacteria strains resulted in the identification of broad antimicrobial activities against both Gram-positive and Gram-negative tested bacterial strains. Based on these findings, we propose the ancient, ubiquitous, and spore-forming Ktedonobacteria as a versatile and promising microbial resource for natural product discovery.
Collapse
Affiliation(s)
- Yu Zheng
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ayana Saitou
- Faculty of Agriculture, Tohoku University, Sendai, Japan
| | - Chiung-Mei Wang
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Atsushi Toyoda
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima, Japan
| | - Yohei Minakuchi
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima, Japan
| | - Yuji Sekiguchi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Kenji Ueda
- Life Science Research Center, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Hideaki Takano
- Life Science Research Center, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Yasuteru Sakai
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Keietsu Abe
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Akira Yokota
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shuhei Yabe
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- Hazaka Plant Research Center, Kennan Eisei Kogyo Co., Ltd., Miyagi, Japan
| |
Collapse
|
244
|
Martín-Escolano R, Cebrián R, Martín-Escolano J, Rosales MJ, Maqueda M, Sánchez-Moreno M, Marín C. Insights into Chagas treatment based on the potential of bacteriocin AS-48. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2019; 10:1-8. [PMID: 30953804 PMCID: PMC6447751 DOI: 10.1016/j.ijpddr.2019.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 03/26/2019] [Indexed: 02/01/2023]
Abstract
Chagas disease caused by the protozoan parasite Trypanosoma cruzi represents a significant public health problem in Latin America, affecting around 8 million cases worldwide. Nowadays is urgent the identification of new antichagasic agents as the only therapeutic options available, Nifurtimox and Benznidazole, are in use for >40 years, and present high toxicity, limited efficacy and frequent treatment failures in the chronic phase of the disease. Recently, it has been described the antiparasitic effect of AS-48, a bacteriocin produced by Enterococcus faecalis, against Trypanosoma brucei and Leishmania spp. In this work, we have demonstrated the in vitro potential of the AS-48 bacteriocin against T. cruzi. Interesting, AS-48 was more effective against the three morphological forms of different T. cruzi strains, and displayed lower cytotoxicity than the reference drug Benznidazole. In addition, AS-48 combines the criteria established as a potential antichagasic agent, resulting in a promising therapeutic alternative. According to the action mechanism, AS-48 trypanocidal activity could be explained in a mitochondrion-dependent manner through a reactive oxygen species production and mitochondrial depolarization, causing a fast and severe bioenergetic collapse.
Collapse
Affiliation(s)
- Rubén Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa S/n, E-18071, Granada, Spain
| | - Rubén Cebrián
- Department of Microbiology, Faculty of Sciences. C/ Fuentenueva S/n. University of Granada, Severo Ochoa /n, 18071, Granada, Spain
| | - Javier Martín-Escolano
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa S/n, E-18071, Granada, Spain
| | - Maria J Rosales
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa S/n, E-18071, Granada, Spain
| | - Mercedes Maqueda
- Department of Microbiology, Faculty of Sciences. C/ Fuentenueva S/n. University of Granada, Severo Ochoa /n, 18071, Granada, Spain
| | - Manuel Sánchez-Moreno
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa S/n, E-18071, Granada, Spain
| | - Clotilde Marín
- Department of Parasitology, Instituto de Investigación Biosanitaria (ibs.Granada), Hospitales Universitarios De Granada/University of Granada, Severo Ochoa S/n, E-18071, Granada, Spain.
| |
Collapse
|
245
|
Wendler J, Schroeder BO, Ehmann D, Koeninger L, Mailänder-Sánchez D, Lemberg C, Wanner S, Schaller M, Stange EF, Malek NP, Weidenmaier C, LeibundGut-Landmann S, Wehkamp J. Proteolytic Degradation of reduced Human Beta Defensin 1 generates a Novel Antibiotic Octapeptide. Sci Rep 2019; 9:3640. [PMID: 30842543 PMCID: PMC6403363 DOI: 10.1038/s41598-019-40216-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/07/2019] [Indexed: 01/06/2023] Open
Abstract
Microbial resistance against clinical used antibiotics is on the rise. Accordingly, there is a high demand for new innovative antimicrobial strategies. The host-defense peptide human beta-defensin 1 (hBD-1) is produced continuously by epithelial cells and exhibits compelling antimicrobial activity after reduction of its disulphide bridges. Here we report that proteolysis of reduced hBD-1 by gastrointestinal proteases as well as human duodenal secretions produces an eight-amino acid carboxy-terminal fragment. The generated octapeptide retains antibiotic activity, yet with distinct characteristics differing from the full-length peptide. We modified the octapeptide by stabilizing its termini and by using non-natural D-amino acids. The native and modified peptide variants showed antibiotic activity against pathogenic as well as antibiotic-resistant microorganisms, including E. coli, P. aeruginosa and C. albicans. Moreover, in an in vitro C. albicans infection model the tested peptides demonstrated effective amelioration of C. albicans infection without showing cytotoxity on human cells. In summary, protease degradation of hBD-1 provides a yet unknown mechanism to broaden antimicrobial host defense, which could be used to develop defensin-derived therapeutic applications.
Collapse
Affiliation(s)
- Judith Wendler
- Department of Internal Medicine 1, University Hospital Tuebingen, Tuebingen, Germany
| | - Bjoern O Schroeder
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Tuebingen, Germany.,Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Dirk Ehmann
- Department of Internal Medicine 1, University Hospital Tuebingen, Tuebingen, Germany
| | - Louis Koeninger
- Department of Internal Medicine 1, University Hospital Tuebingen, Tuebingen, Germany
| | | | - Christina Lemberg
- Institute of Dermatology, University Hospital Tuebingen, Tuebingen, Germany.,Institute of Immunology, Vetsuisse Faculty, University of Zürich, Zurich, Switzerland
| | - Stephanie Wanner
- Institute of Dermatology, University Hospital Tuebingen, Tuebingen, Germany.,Institute of Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
| | - Martin Schaller
- Institute of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Eduard F Stange
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Tuebingen, Germany
| | - Nisar P Malek
- Department of Internal Medicine 1, University Hospital Tuebingen, Tuebingen, Germany
| | - Christopher Weidenmaier
- Institute of Medical Microbiology and Hygiene, University Hospital Tuebingen, Tuebingen, Germany
| | | | - Jan Wehkamp
- Department of Internal Medicine 1, University Hospital Tuebingen, Tuebingen, Germany.
| |
Collapse
|
246
|
Halami PM. Sublichenin, a new subtilin-like lantibiotics of probiotic bacterium Bacillus licheniformis MCC 2512T with antibacterial activity. Microb Pathog 2019; 128:139-146. [DOI: 10.1016/j.micpath.2018.12.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 11/16/2022]
|
247
|
Dual Action of the PN159/KLAL/MAP Peptide: Increase of Drug Penetration across Caco-2 Intestinal Barrier Model by Modulation of Tight Junctions and Plasma Membrane Permeability. Pharmaceutics 2019; 11:pharmaceutics11020073. [PMID: 30744154 PMCID: PMC6410202 DOI: 10.3390/pharmaceutics11020073] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/26/2019] [Accepted: 02/05/2019] [Indexed: 12/16/2022] Open
Abstract
The absorption of drugs is limited by the epithelial barriers of the gastrointestinal tract. One of the strategies to improve drug delivery is the modulation of barrier function by the targeted opening of epithelial tight junctions. In our previous study the 18-mer amphiphilic PN159 peptide was found to be an effective tight junction modulator on intestinal epithelial and blood–brain barrier models. PN159, also known as KLAL or MAP, was described to interact with biological membranes as a cell-penetrating peptide. In the present work we demonstrated that the PN159 peptide as a penetration enhancer has a dual action on intestinal epithelial cells. The peptide safely and reversibly enhanced the permeability of Caco-2 monolayers by opening the intercellular junctions. The penetration of dextran molecules with different size and four efflux pump substrate drugs was increased several folds. We identified claudin-4 and -7 junctional proteins by docking studies as potential binding partners and targets of PN159 in the opening of the paracellular pathway. In addition to the tight junction modulator action, the peptide showed cell membrane permeabilizing and antimicrobial effects. This dual action is not general for cell-penetrating peptides (CPPs), since the other three CPPs tested did not show barrier opening effects.
Collapse
|
248
|
Haney EF, Straus SK, Hancock REW. Reassessing the Host Defense Peptide Landscape. Front Chem 2019; 7:43. [PMID: 30778385 PMCID: PMC6369191 DOI: 10.3389/fchem.2019.00043] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/15/2019] [Indexed: 12/18/2022] Open
Abstract
Current research has demonstrated that small cationic amphipathic peptides have strong potential not only as antimicrobials, but also as antibiofilm agents, immune modulators, and anti-inflammatories. Although traditionally termed antimicrobial peptides (AMPs) these additional roles have prompted a shift in terminology to use the broader term host defense peptides (HDPs) to capture the multi-functional nature of these molecules. In this review, we critically examined the role of AMPs and HDPs in infectious diseases and inflammation. It is generally accepted that HDPs are multi-faceted mediators of a wide range of biological processes, with individual activities dependent on their polypeptide sequence. In this context, we explore the concept of chemical space as it applies to HDPs and hypothesize that the various functions and activities of this class of molecule exist on independent but overlapping activity landscapes. Finally, we outline several emerging functions and roles of HDPs and highlight how an improved understanding of these processes can potentially be leveraged to more fully realize the therapeutic promise of HDPs.
Collapse
Affiliation(s)
- Evan F Haney
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Suzana K Straus
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
249
|
Meng Q, Li Y, Shen C. Antibacterial Coatings of Biomedical Surfaces by Polydextran Aldehyde/Polyethylenimine Nanofibers. ACS APPLIED BIO MATERIALS 2018; 2:562-569. [DOI: 10.1021/acsabm.8b00708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Qin Meng
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310027, P.R. China
| | - Yingjun Li
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310027, P.R. China
| | - Chong Shen
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou Zhejiang 310027, P.R. China
| |
Collapse
|
250
|
Panteleev PV, Bolosov IA, Kalashnikov AÀ, Kokryakov VN, Shamova OV, Emelianova AA, Balandin SV, Ovchinnikova TV. Combined Antibacterial Effects of Goat Cathelicidins With Different Mechanisms of Action. Front Microbiol 2018; 9:2983. [PMID: 30555455 PMCID: PMC6284057 DOI: 10.3389/fmicb.2018.02983] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022] Open
Abstract
Being essential components of innate immune system, animal antimicrobial peptides (AMPs) also known as host-defense peptides came into sharp focus as possible alternatives to conventional antibiotics due to their high efficacy against a broad range of MDR pathogens and low rate of resistance development. Mammalian species can produce a set of co-localized AMPs with different structures and mechanisms of actions. Here we examined the combined antibacterial effects of cathelicidins, structurally diverse family of host-defense peptides found in vertebrate species. As a model we have used structurally distinct cathelicidins expressed in the leukocytes of goat Capra hircus. The recombinant analogs of natural peptides were obtained by heterologous expression in bacterial system and biological activities as well as the major mechanisms of antibacterial action of the peptides were investigated. As the result, the marked synergistic effect against wide panel of bacterial strains including extensively drug-resistant ones was observed for the pair of membranolytic α-helical amphipathic peptide ChMAP-28 and Pro-rich peptide mini-ChBac7.5Nα targeting a bacterial ribosome. ChMAP-28 was shown to damage the outer bacterial membrane at sub-inhibitory concentrations that could facilitate Pro-rich peptide translocation into the cell. Finally, resistance changes under a long-term continuous selective pressure of each individual peptide and the synergistic combination of both peptides were tested against Escherichia coli strains. The combination was shown to keep a high activity after the 26-days selection experiment in contrast to mini-ChBac7.5Nα used alone and the reference antibiotic polymyxin B. We identified the point mutation leading to amino acid substitution V102E in the membrane transport protein SbmA of the mini-ChBac7.5Nα-resistant strain obtained by selection. The experiments revealed that the presence of sub-inhibitory concentrations of ChMAP-28 restored the activity of mini-ChBac7.5Nα against this strain and clinical isolate with a weak sensitivity to mini-ChBac7.5Nα. The obtained results suggest a potential medical application of synergistic combinations of natural cathelicidins, which allows using a lower therapeutic dose and minimizes the risk of resistance development.
Collapse
Affiliation(s)
- Pavel V Panteleev
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ilia A Bolosov
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexander À Kalashnikov
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Olga V Shamova
- Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Anna A Emelianova
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sergey V Balandin
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Tatiana V Ovchinnikova
- M.M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|