51
|
Dorman MJ, Feltwell T, Goulding DA, Parkhill J, Short FL. The Capsule Regulatory Network of Klebsiella pneumoniae Defined by density-TraDISort. mBio 2018; 9:e01863-18. [PMID: 30459193 PMCID: PMC6247091 DOI: 10.1128/mbio.01863-18] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/08/2018] [Indexed: 01/04/2023] Open
Abstract
Klebsiella pneumoniae infections affect infants and the immunocompromised, and the recent emergence of hypervirulent and multidrug-resistant K. pneumoniae lineages is a critical health care concern. Hypervirulence in K. pneumoniae is mediated by several factors, including the overproduction of extracellular capsule. However, the full details of how K. pneumoniae capsule biosynthesis is achieved or regulated are not known. We have developed a robust and sensitive procedure to identify genes influencing capsule production, density-TraDISort, which combines density gradient centrifugation with transposon insertion sequencing. We have used this method to explore capsule regulation in two clinically relevant Klebsiella strains, K. pneumoniae NTUH-K2044 (capsule type K1) and K. pneumoniae ATCC 43816 (capsule type K2). We identified multiple genes required for full capsule production in K. pneumoniae, as well as putative suppressors of capsule in NTUH-K2044, and have validated the results of our screen with targeted knockout mutants. Further investigation of several of the K. pneumoniae capsule regulators identified-ArgR, MprA/KvrB, SlyA/KvrA, and the Sap ABC transporter-revealed effects on capsule amount and architecture, serum resistance, and virulence. We show that capsule production in K. pneumoniae is at the center of a complex regulatory network involving multiple global regulators and environmental cues and that the majority of capsule regulatory genes are located in the core genome. Overall, our findings expand our understanding of how capsule is regulated in this medically important pathogen and provide a technology that can be easily implemented to study capsule regulation in other bacterial species.IMPORTANCE Capsule production is essential for K. pneumoniae to cause infections, but its regulation and mechanism of synthesis are not fully understood in this organism. We have developed and applied a new method for genome-wide identification of capsule regulators. Using this method, many genes that positively or negatively affect capsule production in K. pneumoniae were identified, and we use these data to propose an integrated model for capsule regulation in this species. Several of the genes and biological processes identified have not previously been linked to capsule synthesis. We also show that the methods presented here can be applied to other species of capsulated bacteria, providing the opportunity to explore and compare capsule regulatory networks in other bacterial strains and species.
Collapse
Affiliation(s)
- Matthew J Dorman
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Theresa Feltwell
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - David A Goulding
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Julian Parkhill
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
| | - Francesca L Short
- Wellcome Sanger Institute, Hinxton, Cambridgeshire, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
52
|
Kannan S, Sathasivam G, Marudhamuthu M. Decrease of growth, biofilm and secreted virulence in opportunistic nosocomial Pseudomonas aeruginosa ATCC 25619 by glycyrrhetinic acid. Microb Pathog 2018; 126:332-342. [PMID: 30458255 DOI: 10.1016/j.micpath.2018.11.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 11/27/2022]
Abstract
The present study elucidates the antibiofilm and antivirulent capability of glycyrrhetinic acid (GRA) against Pseudomonas aeruginosa ATCC 25619. The minimum inhibitory concentrations (MIC) and minimum bactericidal concentrations (MBC) of GRA against P. aeruginosa were found to be 160 μg/mL and 420 μg/mL respectively. In an acclimatization resistance analysis using P. aeruginosa, no resistance towards GRA was observed during the habituation period. Adequate penetration of GRA over the biofilm matrix was proposed with the membrane penetration model assembly constructed with the preformed biofilm exhibited the prospective penetration of GRA above the mature biofilm. Furthermore, GRA resulted in the attenuation of virulence factors such as motility, biofilm formation, pyocyanin secretion, secreted proteases with its sub MIC concentrations. The antibiofilm property of GRA was assessed with the light microscopy and high content screening fluorescent imaging system, which clearly demonstrates, the thickness of P. aeruginosa biofilm was reduced to 11.33 ± 2.08 μm from 39 ± 2.51 μm. Transmission Electron Microscopy (TEM) images depicted the morphological changes in cells such as disaggregation of colonies, cell disruption with loss of intracellular material, cytolytic damage, the process of morphological transformation, bacteriolysis indicating the potential effect of GRA.
Collapse
Affiliation(s)
- Suganya Kannan
- Department of Microbial Technology, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625 021, Tamil Nadu, India
| | - Gowtham Sathasivam
- Department of Microbial Technology, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625 021, Tamil Nadu, India
| | - Murugan Marudhamuthu
- Department of Microbial Technology, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625 021, Tamil Nadu, India.
| |
Collapse
|
53
|
Abstract
Bone infection represents greatest challenge in public health care with serious social and economic implications. The efforts of the scientific community are focused in the development of innovative and advanced biomaterials with anti-infective properties related to their non-fouling, bactericidal and/or antibiofilm capabilities. This chapter aims at thoroughly surveying the different approaches based on silica mesoporous materials (SMMs) for bone infection management. Bacteria repelling surfaces by zwitterionization process, bactericidal effect by implantable devices with antimicrobial local delivery agents and antibiofilm effect by more sophisticated systems based on targeted nanocarriers will be considered.
Collapse
Affiliation(s)
- María Vallet-Regí
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| | - Montserrat Colilla
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Isabel Izquierdo-Barba
- Department of Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid and Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
54
|
Vision for medicine: Staphylococcus aureus biofilm war and unlocking key's for anti-biofilm drug development. Microb Pathog 2018; 123:339-347. [DOI: 10.1016/j.micpath.2018.07.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/02/2018] [Accepted: 07/02/2018] [Indexed: 01/28/2023]
|
55
|
Potisap C, Khan MAW, Boonmee A, Rodrigues JLM, Wongratanacheewin S, Sermswan RW. Burkholderia pseudomallei-absent soil bacterial community results in secondary metabolites that kill this pathogen. AMB Express 2018; 8:136. [PMID: 30143892 PMCID: PMC6109036 DOI: 10.1186/s13568-018-0663-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/17/2018] [Indexed: 12/16/2022] Open
Abstract
Burkholderia pseudomallei is a Gram-negative bacterium found in soil and the causative agent of a severe disease in humans and animals known as melioidosis. It is intrinsically resistant to many antibiotics and has been reported resistant to the drugs of choice; ceftazidime. Microbial communities in soil in the presence and absence of B. pseudomallei were investigated using metagenomics approach. The variation in bacterial species diversity was significantly higher in soil samples without B. pseudomallei. Abundances of phyla Actinobacteria and Firmicutes were found significantly higher in B. pseudomallei-negative soils. Bacillus amyloliquefaciens KKU1 in phylum Firmicutes was discovered from negative soil and its secondary metabolites could inhibit clinical, environmental and drug resistant isolates of B. pseudomallei, together with some pathogenic Gram-negative but not Gram-positive bacteria. The antimicrobial activity from KKU 1 against B. pseudomallei was abolished when treated with proteinase K, stable in a wide range of pH and remained active after heating at 100 °C for 15 min. Precipitated proteins from KKU1 were demonstrated to cause lysis and corrugated surfaces of B. pseudomallei. The minimum inhibitory concentrations and minimum bactericidal concentrations of the precipitated proteins from KKU1 against B. pseudomallei were 0.97 μg/ml and 3.9 μg/ml. Interestingly, Native SDS-PAGE showed small active compounds of less than 6 kDa, along with other information collectively suggesting the properties of antimicrobial peptides. For the first time, culture-independent information in melioidosis endemic area could lead to a suspected source of metabolites that may help defense against B. pseudomallei and other pathogenic Gram-negative bacteria.
Collapse
|
56
|
Potent effects of amino acid scanned antimicrobial peptide Feleucin-K3 analogs against both multidrug-resistant strains and biofilms of Pseudomonas aeruginosa. Amino Acids 2018; 50:1471-1483. [PMID: 30136030 DOI: 10.1007/s00726-018-2625-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 07/18/2018] [Indexed: 10/28/2022]
Abstract
Pseudomonas aeruginosa is particularly difficult to treat because it possesses a variety of resistance mechanisms and because it often forms biofilms. Antimicrobial peptides represent promising candidates for future templates of antibiotic-resistant bacterial infections due to their unique mechanism of antimicrobial action. In this study, we first found that the antimicrobial peptide Feleucin-K3 has potent antimicrobial activity against not only the standard strain of P. aeruginosa but also against the multidrug-resistant strains isolated from clinics. Then, the structure-activity relationship of the peptide was investigated using alanine and D-amino acid scanning. Among the analogs synthesized, FK-1D showed much more potent antimicrobial activity, superior stability, and very low toxicity, and it was able to permeabilize bacterial membranes. Furthermore, it exhibited significant anti-biofilm activity. More importantly, FK-1D showed excellent antimicrobial activity in vivo, especially against clinical multidrug-resistant bacteria, in contrast to ceftazidime. Our results suggested that FK-1D could be subjected to fixed-point modification in the first and fourth sites to further optimize its medicinal properties and potential as a lead compound for the treatment of infections caused by multidrug-resistant P. aeruginosa and the associated biofilms.
Collapse
|
57
|
González B, Colilla M, Díez J, Pedraza D, Guembe M, Izquierdo-Barba I, Vallet-Regí M. Mesoporous silica nanoparticles decorated with polycationic dendrimers for infection treatment. Acta Biomater 2018; 68:261-271. [PMID: 29307796 DOI: 10.1016/j.actbio.2017.12.041] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022]
Abstract
This work aims to provide an effective and novel solution for the treatment of infection by using nanovehicles loaded with antibiotics capable of penetrating the bacterial wall, thus increasing the antimicrobial effectiveness. These nanosystems, named "nanoantibiotics", are composed of mesoporous silica nanoparticles (MSNs), which act as nanocarriers of an antimicrobial agent (levofloxacin, LEVO) localized inside the mesopores. To provide the nanosystem of bacterial membrane interaction capability, a polycationic dendrimer, concretely the poly(propyleneimine) dendrimer of third generation (G3), was covalently grafted to the external surface of the LEVO-loaded MSNs. After physicochemical characterization of this nanoantibiotic, the release kinetics of LEVO and the antimicrobial efficacy of each released dosage were evaluated. Besides, internalization studies of the MSNs functionalized with the G3 dendrimer were carried out, showing a high penetrability throughout Gram-negative bacterial membranes. This work evidences that the synergistic combination of polycationic dendrimers as bacterial membrane permeabilization agents with LEVO-loaded MSNs triggers an efficient antimicrobial effect on Gram-negative bacterial biofilm. These positive results open up very promising expectations for their potential application in new infection therapies. STATEMENT OF SIGNIFICANCE Seeking new alternatives to current available treatments of bacterial infections represents a great challenge in nanomedicine. This work reports the design and optimization of a new class of antimicrobial agent, named "nanoantibiotic", based on mesoporous silica nanoparticles (MSNs) decorated with polypropyleneimine dendrimers of third generation (G3) and loaded with levofloxacin (LEVO) antibiotic. The covalently grafting of these G3 dendrimers to MSNs allows an effective internalization in Gram-negative bacteria. Furthermore, the LEVO loaded into the mesoporous cavities is released in a sustained manner at effective antimicrobial dosages. The novelty and originality of this manuscript relies on proving that the synergistic combination of bacteria-targeting and antimicrobial agents into a unique nanosystem provokes a remarkable antimicrobial effect against bacterial biofilm.
Collapse
Affiliation(s)
- Blanca González
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Montserrat Colilla
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Jaime Díez
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Daniel Pedraza
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Marta Guembe
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Isabel Izquierdo-Barba
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain.
| | - María Vallet-Regí
- Departamento de Química Inorgánica y Bioinorgánica, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain.
| |
Collapse
|
58
|
Garcia M, Morello E, Garnier J, Barrault C, Garnier M, Burucoa C, Lecron JC, Si-Tahar M, Bernard FX, Bodet C. Pseudomonas aeruginosa flagellum is critical for invasion, cutaneous persistence and induction of inflammatory response of skin epidermis. Virulence 2018; 9:1163-1175. [PMID: 30070169 PMCID: PMC6086312 DOI: 10.1080/21505594.2018.1480830] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/21/2018] [Indexed: 12/22/2022] Open
Abstract
Pseudomonas aeruginosa, an opportunistic pathogen involved in skin and lung diseases, possesses numerous virulence factors, including type 2 and 3 secretion systems (T2SS and T3SS) and its flagellum, whose functions remain poorly known during cutaneous infection. Using isogenic mutants deleted from genes encoding each or all of these three virulence factors, we investigated their role in induction of inflammatory response and in tissue invasiveness in human primary keratinocytes and reconstructed epidermis. Our results showed that flagellum, but not T2SS and T3SS, is involved in induction of a large panel of cytokine, chemokine, and antimicrobial peptide (AMP) mRNA in the infected keratinocytes. Chemokine secretion and AMP tissular production were also dependent on the presence of the bacterial flagellum. This pro-inflammatory effect was significantly reduced in keratinocytes infected in presence of anti-toll-like receptor 5 (TLR5) neutralizing antibody. Bacterial invasion of human epidermis and persistence in a mouse model of sub-cutaneous infection were dependent on the P. aeruginosa flagellum. We demonstrated that flagellum constitutes the main virulence factor of P. aeruginosa involved not only in early induction of the epidermis inflammatory response but also in bacterial invasion and cutaneous persistence. P. aeruginosa is mainly sensed by TLR5 during the early innate immune response of human primary keratinocytes.
Collapse
Affiliation(s)
- Magali Garcia
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers
, Poitiers, France
| | - Eric Morello
-
Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Université de Tours
, Tours, France
| | | | | | - Martine Garnier
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
| | - Christophe Burucoa
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire de Bactériologie et Hygiène, CHU de Poitiers
, Poitiers, France
| | - Jean-Claude Lecron
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
-
Laboratoire d’Immunologie et Inflammation, CHU de Poitiers
, Poitiers, France
| | - Mustapha Si-Tahar
-
Centre d'Etude des Pathologies Respiratoires, INSERM UMR 1100, Université de Tours
, Tours, France
| | | | - Charles Bodet
-
Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers
, Poitiers, France
| |
Collapse
|
59
|
Baron S, Leulmi Z, Villard C, Olaitan AO, Telke AA, Rolain JM. Inactivation of the arn operon and loss of aminoarabinose on lipopolysaccharide as the cause of susceptibility to colistin in an atypical clinical isolate of proteus vulgaris. Int J Antimicrob Agents 2017; 51:450-457. [PMID: 29203405 DOI: 10.1016/j.ijantimicag.2017.11.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/15/2017] [Accepted: 11/24/2017] [Indexed: 12/27/2022]
Abstract
Colistin has become a last-line antibiotic for the treatment of multidrug-resistant bacterial infections; however, resistance to colistin has emerged in recent years. Some bacteria, such as Proteus and Serratia spp., are intrinsically resistant to colistin although the exact mechanism of resistance is unknown. Here we identified the molecular support for intrinsic colistin resistance in Proteus spp. by comparative genomic, transcriptomic and proteomic analyses of colistin-susceptible (CSUR P1868_S) and colistin-resistant (CSUR P1867_R) strains of an atypical Proteus vulgaris. A significant difference in outer membrane glycoside structures in both strains that was corroborated by MALDI-TOF/MS analysis was found, which showed an absence of 4-amino-4-deoxy-l-arabinose (L-Ara4N) in the outer membrane lipid A moiety of the susceptible strain. Comparative genomic analysis with other resistant strains of P. vulgaris available in a local database found a mutation in the arnBCADTEF operon of the susceptible strain. Transcriptomic analysis of genes belonging to the arnBCADTEF operon showed a significant decrease in mRNA expression level of these genes in the susceptible strain, supporting addition of L-Ara4N in the outer membrane lipid A moiety as an explanation for colistin resistance. Insertion of the arnD gene that was suggested to be altered in the susceptible strain by in silico analysis led to a 16-fold increase of colistin MIC in the susceptible strain, confirming its role in colistin resistance in this species. Here we show that constitutive activation of the arn operon and addition of L-Ara4N is the main molecular mechanism of colistin resistance in P. vulgaris.
Collapse
Affiliation(s)
- Sophie Baron
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Facultés de Médecine et de Pharmacie, 19-21 bd Jean Moulin, Marseille, France
| | - Zineb Leulmi
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Facultés de Médecine et de Pharmacie, 19-21 bd Jean Moulin, Marseille, France
| | - Claude Villard
- Aix-Marseille Université, Plateforme Protéomique et Innovation Technologique, Faculté de Pharmacie, 27 boulevard Jean Moulin, Marseille 13385 CEDEX 05, France
| | - Abiola Olumuyiwa Olaitan
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Facultés de Médecine et de Pharmacie, 19-21 bd Jean Moulin, Marseille, France
| | - Amar A Telke
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Facultés de Médecine et de Pharmacie, 19-21 bd Jean Moulin, Marseille, France
| | - Jean-Marc Rolain
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU-Méditerranée Infection, Facultés de Médecine et de Pharmacie, 19-21 bd Jean Moulin, Marseille, France.
| |
Collapse
|
60
|
Molchanova N, Hansen PR, Franzyk H. Advances in Development of Antimicrobial Peptidomimetics as Potential Drugs. Molecules 2017; 22:E1430. [PMID: 28850098 PMCID: PMC6151827 DOI: 10.3390/molecules22091430] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 01/19/2023] Open
Abstract
The rapid emergence of multidrug-resistant pathogens has evolved into a global health problem as current treatment options are failing for infections caused by pan-resistant bacteria. Hence, novel antibiotics are in high demand, and for this reason antimicrobial peptides (AMPs) have attracted considerable interest, since they often show broad-spectrum activity, fast killing and high cell selectivity. However, the therapeutic potential of natural AMPs is limited by their short plasma half-life. Antimicrobial peptidomimetics mimic the structure and biological activity of AMPs, but display extended stability in the presence of biological matrices. In the present review, focus is on the developments reported in the last decade with respect to their design, synthesis, antimicrobial activity, cytotoxic side effects as well as their potential applications as anti-infective agents. Specifically, only peptidomimetics with a modular structure of residues connected via amide linkages will be discussed. These comprise the classes of α-peptoids (N-alkylated glycine oligomers), β-peptoids (N-alkylated β-alanine oligomers), β³-peptides, α/β³-peptides, α-peptide/β-peptoid hybrids, α/γ N-acylated N-aminoethylpeptides (AApeptides), and oligoacyllysines (OAKs). Such peptidomimetics are of particular interest due to their potent antimicrobial activity, versatile design, and convenient optimization via assembly by standard solid-phase procedures.
Collapse
Affiliation(s)
- Natalia Molchanova
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen, Denmark.
| | - Paul R Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen, Denmark.
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
61
|
Bolt HL, Eggimann GA, Jahoda CAB, Zuckermann RN, Sharples GJ, Cobb SL. Exploring the links between peptoid antibacterial activity and toxicity. MEDCHEMCOMM 2017; 8:886-896. [PMID: 30108804 PMCID: PMC6072100 DOI: 10.1039/c6md00648e] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/27/2017] [Indexed: 12/31/2022]
Abstract
Peptoids are a promising class of antimicrobial agents with reported activities against a range of both Gram-positive and Gram-negative bacteria, fungi and most recently parasites. However, at present the available toxicity data is somewhat limited and as such rationally designing effective antimicrobial peptoids can be challenging. Herein, we present the toxicity profiling of a series of linear peptoids against mammalian cell lines (HaCaT and HepG2). The cytotoxicity of the peptoid library has then been correlated with their antibacterial properties against Gram-positive and Gram-negative bacteria and also to the hydrophobicity of the peptoid sequences. The work presented provides valuable data to aid in the future rational design of antimicrobial peptoids.
Collapse
Affiliation(s)
- H L Bolt
- Department of Chemistry , Durham University , South Road , Durham , DH1 3LE , UK .
| | - G A Eggimann
- Department of Chemistry , Durham University , South Road , Durham , DH1 3LE , UK .
| | - C A B Jahoda
- School of Biological and Biomedical Sciences , Durham University , Durham DH1 3LE , UK .
| | - R N Zuckermann
- Molecular Foundry , Lawrence Berkeley National Laboratory , Berkeley , California , USA
| | - G J Sharples
- School of Biological and Biomedical Sciences , Durham University , Durham DH1 3LE , UK .
| | - S L Cobb
- Department of Chemistry , Durham University , South Road , Durham , DH1 3LE , UK .
| |
Collapse
|
62
|
Sierra JM, Fusté E, Rabanal F, Vinuesa T, Viñas M. An overview of antimicrobial peptides and the latest advances in their development. Expert Opin Biol Ther 2017; 17:663-676. [PMID: 28368216 DOI: 10.1080/14712598.2017.1315402] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The recent dramatic increase in the incidence of antimicrobial resistance has been recognized by organizations such as the United Nations and World Health Organization as well as the governments of the USA and several European countries. A relatively new weapon in the fight against severe infections caused by multi-drug resistant bacteria is antimicrobial peptides (AMPs). These include colistin, currently regarded as the last line of antimicrobial therapy against multi-drug resistant microorganisms. Areas covered: Here, the authors provide an overview of the current research on AMPs. The focus is AMPs currently being developed for the treatment of recalcitrant bacterial infections, the synergies of AMPs and antibiotics, and the activity of AMPs against biofilm. This review also includes a brief introduction into the use of AMPs in infections caused by Mycobacterium, fungi, and parasites. Expert opinion: In research into new antimicrobials, AMPs are gaining increasing attention. While many are natural and are produced by a wide variety of organisms, others are being newly designed and chemically synthesized in the laboratory to achieve novel antimicrobial agents. The same strategy to fight infections in nature is thus being effectively exploited to safeguard human and animal health.
Collapse
Affiliation(s)
- Josep M Sierra
- a Laboratory of Molecular Microbiology and Antimicrobials, Department of Pathology and Experimental Therapeutics, School of Medicine , University of Barcelona , Barcelona , Spain
| | - Ester Fusté
- a Laboratory of Molecular Microbiology and Antimicrobials, Department of Pathology and Experimental Therapeutics, School of Medicine , University of Barcelona , Barcelona , Spain.,b Dept. Public Health, Mental Health and Perinatal Nursing. School of nursing , University of Barcelona , Barcelona , Spain
| | - Francesc Rabanal
- c Department of Organic Chemistry, Faculty of Chemistry , University of Barcelona , Barcelona , Spain
| | - Teresa Vinuesa
- a Laboratory of Molecular Microbiology and Antimicrobials, Department of Pathology and Experimental Therapeutics, School of Medicine , University of Barcelona , Barcelona , Spain
| | - Miguel Viñas
- a Laboratory of Molecular Microbiology and Antimicrobials, Department of Pathology and Experimental Therapeutics, School of Medicine , University of Barcelona , Barcelona , Spain.,d Departament of Biomedicine , Cooperativa de Ensino Superior Politécnico e Universitário, IINFACTS , Gandra , Portugal
| |
Collapse
|
63
|
Abstract
Antimicrobial peptides (AMPs), also known as host defense peptides, are small naturally occurring microbicidal molecules produced by the host innate immune response that function as a first line of defense to kill pathogenic microorganisms by inducing deleterious cell membrane damage. AMPs also possess signaling and chemoattractant activities and can modulate the innate immune response to enhance protective immunity or suppress inflammation. Human pathogens have evolved defense molecules and strategies to counter and survive the AMPs released by host immune cells such as neutrophils and macrophages. Here, we review the various mechanisms used by human bacterial pathogens to resist AMP-mediated killing, including surface charge modification, active efflux, alteration of membrane fluidity, inactivation by proteolytic digestion, and entrapment by surface proteins and polysaccharides. Enhanced understanding of AMP resistance at the molecular level may offer insight into the mechanisms of bacterial pathogenesis and augment the discovery of novel therapeutic targets and drug design for the treatment of recalcitrant multidrug-resistant bacterial infections.
Collapse
|
64
|
Atas A, Seddon AM, Ford DC, Cooper IA, Wren BW, Oyston PCF, Karlyshev AV. YPTB3816 of Yersinia pseudotuberculosis strain IP32953 is a virulence-related metallo-oligopeptidase. BMC Microbiol 2016; 16:282. [PMID: 27887582 PMCID: PMC5124237 DOI: 10.1186/s12866-016-0900-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 11/17/2016] [Indexed: 12/03/2022] Open
Abstract
Background Although bacterial peptidases are known to be produced by various microorganisms, including pathogenic bacteria, their role in bacterial physiology is not fully understood. In particular, oligopeptidases are thought to be mainly involved in degradation of short peptides e.g. leader peptides released during classical protein secretion pathways. The aim of this study was to investigate effects of inactivation of an oligopeptidase encoding gene opdA gene of Yersinia pseudotuberculosis on bacterial properties in vivo and in vitro, and to test dependence of the enzymatic activity of the respective purified enzyme on the presence of different divalent cations. Results In this study we found that oligopeptidase OpdA of Yersinia pseudotuberculosis is required for bacterial virulence, whilst knocking out the respective gene did not have any effect on bacterial viability or growth rate in vitro. In addition, we studied enzymatic properties of this enzyme after expression and purification from E. coli. Using an enzyme depleted of contaminant divalent cations and different types of fluorescently labelled substrates, we found strong dependence of its activity on the presence of particular cations. Unexpectedly, Zn2+ showed stimulatory activity only at low concentrations, but inhibited the enzyme at higher concentrations. In contrast, Co2+, Ca2+ and Mn2+ stimulated activity at all concentrations tested, whilst Mg2+ revealed no effect on the enzyme activity at all concentrations used. Conclusions The results of this study provide valuable contribution to the investigation of bacterial peptidases in general, and that of metallo-oligopeptidases in particular. This is the first study demonstrating that opdA in Yersinia pseudotuberculsosis is required for pathogenicity. The data reported are important for better understanding of the role of OpdA-like enzymes in pathogenesis in bacterial infections. Characterisation of this protein may serve as a basis for the development of novel antibacterials based on specific inhibition of this peptidase activity. Electronic supplementary material The online version of this article (doi:10.1186/s12866-016-0900-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ali Atas
- School of Life Sciences, Pharmacy and Chemistry; Faculty of Science, Engineering and Computing, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, UK
| | - Alan M Seddon
- School of Life Sciences, Pharmacy and Chemistry; Faculty of Science, Engineering and Computing, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, UK
| | - Donna C Ford
- Biomedical Sciences, DSTL Porton Down, Salisbury, Wiltshire, SP4 0JQ, UK
| | - Ian A Cooper
- Biomedical Sciences, DSTL Porton Down, Salisbury, Wiltshire, SP4 0JQ, UK
| | - Brendan W Wren
- Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Petra C F Oyston
- Biomedical Sciences, DSTL Porton Down, Salisbury, Wiltshire, SP4 0JQ, UK
| | - Andrey V Karlyshev
- School of Life Sciences, Pharmacy and Chemistry; Faculty of Science, Engineering and Computing, Kingston University, Penrhyn Road, Kingston upon Thames, KT1 2EE, UK.
| |
Collapse
|
65
|
Pyne A, Pfeil MP, Bennett I, Ravi J, Iavicoli P, Lamarre B, Roethke A, Ray S, Jiang H, Bella A, Reisinger B, Yin D, Little B, Muñoz-García JC, Cerasoli E, Judge PJ, Faruqui N, Calzolai L, Henrion A, Martyna GJ, Grovenor CRM, Crain J, Hoogenboom BW, Watts A, Ryadnov MG. Engineering monolayer poration for rapid exfoliation of microbial membranes. Chem Sci 2016; 8:1105-1115. [PMID: 28451250 PMCID: PMC5369539 DOI: 10.1039/c6sc02925f] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 09/25/2016] [Indexed: 12/04/2022] Open
Abstract
A novel mechanism of monolayer poration leading to the rapid exfoliation and lysis of microbial membranes is reported.
The spread of bacterial resistance to traditional antibiotics continues to stimulate the search for alternative antimicrobial strategies. All forms of life, from bacteria to humans, are postulated to rely on a fundamental host defense mechanism, which exploits the formation of open pores in microbial phospholipid bilayers. Here we predict that transmembrane poration is not necessary for antimicrobial activity and reveal a distinct poration mechanism that targets the outer leaflet of phospholipid bilayers. Using a combination of molecular-scale and real-time imaging, spectroscopy and spectrometry approaches, we introduce a structural motif with a universal insertion mode in reconstituted membranes and live bacteria. We demonstrate that this motif rapidly assembles into monolayer pits that coalesce during progressive membrane exfoliation, leading to bacterial cell death within minutes. The findings offer a new physical basis for designing effective antibiotics.
Collapse
Affiliation(s)
- Alice Pyne
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK . .,London Centre for Nanotechnology and Department of Physics and Astronomy , University College London , London WC1E 6BT , UK
| | - Marc-Philipp Pfeil
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK . .,Department of Biochemistry , University of Oxford , Oxford OX1 3QU , UK
| | - Isabel Bennett
- London Centre for Nanotechnology and Department of Physics and Astronomy , University College London , London WC1E 6BT , UK
| | - Jascindra Ravi
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK .
| | - Patrizia Iavicoli
- European Commission , Joint Research Centre , Institute for Health and Consumer Protection , Ispra (VA) , Italy
| | - Baptiste Lamarre
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK .
| | - Anita Roethke
- Physikalisch-Technische Bundesanstalt , 38116 Braunschweig , Germany
| | - Santanu Ray
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK .
| | - Haibo Jiang
- Centre for Microscopy , Characterisation and Analysis , The University of Western Australia , Crawley , Western Australia 6009 , Australia
| | - Angelo Bella
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK .
| | - Bernd Reisinger
- Physikalisch-Technische Bundesanstalt , 38116 Braunschweig , Germany
| | - Daniel Yin
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK . .,Department of Biochemistry , University of Oxford , Oxford OX1 3QU , UK
| | - Benjamin Little
- School of Physics and Astronomy , University of Edinburgh , Edinburgh EH9 3JZ , UK
| | | | - Eleonora Cerasoli
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK .
| | - Peter J Judge
- Department of Biochemistry , University of Oxford , Oxford OX1 3QU , UK
| | - Nilofar Faruqui
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK .
| | - Luigi Calzolai
- European Commission , Joint Research Centre , Institute for Health and Consumer Protection , Ispra (VA) , Italy
| | - Andre Henrion
- Physikalisch-Technische Bundesanstalt , 38116 Braunschweig , Germany
| | - Glenn J Martyna
- IBM T. J. Watson Research Center , Yorktown Heights , NY 10598 , USA
| | | | - Jason Crain
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK . .,School of Physics and Astronomy , University of Edinburgh , Edinburgh EH9 3JZ , UK
| | - Bart W Hoogenboom
- London Centre for Nanotechnology and Department of Physics and Astronomy , University College London , London WC1E 6BT , UK
| | - Anthony Watts
- Department of Biochemistry , University of Oxford , Oxford OX1 3QU , UK
| | - Maxim G Ryadnov
- National Physical Laboratory , Teddington , Middlesex TW11 0LW , UK .
| |
Collapse
|
66
|
Experimental Induction of Bacterial Resistance to the Antimicrobial Peptide Tachyplesin I and Investigation of the Resistance Mechanisms. Antimicrob Agents Chemother 2016; 60:6067-75. [PMID: 27480861 DOI: 10.1128/aac.00640-16] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 07/21/2016] [Indexed: 12/29/2022] Open
Abstract
Tachyplesin I is a 17-amino-acid cationic antimicrobial peptide (AMP) with a typical cyclic antiparallel β-sheet structure that is a promising therapeutic for infections, tumors, and viruses. To date, no bacterial resistance to tachyplesin I has been reported. To explore the safety of tachyplesin I as an antibacterial drug for wide clinical application, we experimentally induced bacterial resistance to tachyplesin I by using two selection procedures and studied the preliminary resistance mechanisms. Aeromonas hydrophila XS91-4-1, Pseudomonas aeruginosa CGMCC1.2620, and Escherichia coli ATCC 25922 and F41 showed resistance to tachyplesin I under long-term selection pressure with continuously increasing concentrations of tachyplesin I. In addition, P. aeruginosa and E. coli exhibited resistance to tachyplesin I under UV mutagenesis selection conditions. Cell growth and colony morphology were slightly different between control strains and strains with induced resistance. Cross-resistance to tachyplesin I and antimicrobial agents (cefoperazone and amikacin) or other AMPs (pexiganan, tachyplesin III, and polyphemusin I) was observed in some resistant mutants. Previous studies showed that extracellular protease-mediated degradation of AMPs induced bacterial resistance to AMPs. Our results indicated that the resistance mechanism of P. aeruginosa was not entirely dependent on extracellular proteolytic degradation of tachyplesin I; however, tachyplesin I could induce increased proteolytic activity in P. aeruginosa Most importantly, our findings raise serious concerns about the long-term risks associated with the development and clinical use of tachyplesin I.
Collapse
|
67
|
Lam SJ, O'Brien-Simpson NM, Pantarat N, Sulistio A, Wong EHH, Chen YY, Lenzo JC, Holden JA, Blencowe A, Reynolds EC, Qiao GG. Combating multidrug-resistant Gram-negative bacteria with structurally nanoengineered antimicrobial peptide polymers. Nat Microbiol 2016; 1:16162. [PMID: 27617798 DOI: 10.1038/nmicrobiol.2016.162] [Citation(s) in RCA: 545] [Impact Index Per Article: 68.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 08/02/2016] [Indexed: 12/25/2022]
Abstract
With the recent emergence of reports on resistant Gram-negative 'superbugs', infections caused by multidrug-resistant (MDR) Gram-negative bacteria have been named as one of the most urgent global health threats due to the lack of effective and biocompatible drugs. Here, we show that a class of antimicrobial agents, termed 'structurally nanoengineered antimicrobial peptide polymers' (SNAPPs) exhibit sub-μM activity against all Gram-negative bacteria tested, including ESKAPE and colistin-resistant and MDR (CMDR) pathogens, while demonstrating low toxicity. SNAPPs are highly effective in combating CMDR Acinetobacter baumannii infections in vivo, the first example of a synthetic antimicrobial polymer with CMDR Gram-negative pathogen efficacy. Furthermore, we did not observe any resistance acquisition by A. baumannii (including the CMDR strain) to SNAPPs. Comprehensive analyses using a range of microscopy and (bio)assay techniques revealed that the antimicrobial activity of SNAPPs proceeds via a multimodal mechanism of bacterial cell death by outer membrane destabilization, unregulated ion movement across the cytoplasmic membrane and induction of the apoptotic-like death pathway, possibly accounting for why we did not observe resistance to SNAPPs in CMDR bacteria. Overall, SNAPPs show great promise as low-cost and effective antimicrobial agents and may represent a weapon in combating the growing threat of MDR Gram-negative bacteria.
Collapse
Affiliation(s)
- Shu J Lam
- Polymer Science Group, Department of Chemical &Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil M O'Brien-Simpson
- Melbourne Dental School and The Bio21 Institute of Molecular Science and Biotechnology, Oral Health CRC, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Namfon Pantarat
- Melbourne Dental School and The Bio21 Institute of Molecular Science and Biotechnology, Oral Health CRC, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Adrian Sulistio
- Polymer Science Group, Department of Chemical &Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Edgar H H Wong
- Polymer Science Group, Department of Chemical &Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yu-Yen Chen
- Melbourne Dental School and The Bio21 Institute of Molecular Science and Biotechnology, Oral Health CRC, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jason C Lenzo
- Melbourne Dental School and The Bio21 Institute of Molecular Science and Biotechnology, Oral Health CRC, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - James A Holden
- Melbourne Dental School and The Bio21 Institute of Molecular Science and Biotechnology, Oral Health CRC, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anton Blencowe
- Polymer Science Group, Department of Chemical &Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia.,School of Pharmacy and Medical Sciences, Division of Health Sciences, The University of South Australia, Adelaide, South Australia 5000, Australia
| | - Eric C Reynolds
- Melbourne Dental School and The Bio21 Institute of Molecular Science and Biotechnology, Oral Health CRC, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Greg G Qiao
- Polymer Science Group, Department of Chemical &Biomolecular Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
68
|
Santos MRE, Fonseca AC, Mendonça PV, Branco R, Serra AC, Morais PV, Coelho JFJ. Recent Developments in Antimicrobial Polymers: A Review. MATERIALS 2016; 9:ma9070599. [PMID: 28773721 PMCID: PMC5456892 DOI: 10.3390/ma9070599] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 07/01/2016] [Accepted: 07/14/2016] [Indexed: 12/12/2022]
Abstract
Antimicrobial polymers represent a very promising class of therapeutics with unique characteristics for fighting microbial infections. As the classic antibiotics exhibit an increasingly low capacity to effectively act on microorganisms, new solutions must be developed. The importance of this class of materials emerged from the uncontrolled use of antibiotics, which led to the advent of multidrug-resistant microbes, being nowadays one of the most serious public health problems. This review presents a critical discussion of the latest developments involving the use of different classes of antimicrobial polymers. The synthesis pathways used to afford macromolecules with antimicrobial properties, as well as the relationship between the structure and performance of these materials are discussed.
Collapse
Affiliation(s)
- Madson R E Santos
- CEMUC, Department of Chemical Engineering, University of Coimbra, Coimbra 3030-790, Portugal.
| | - Ana C Fonseca
- CEMUC, Department of Chemical Engineering, University of Coimbra, Coimbra 3030-790, Portugal.
| | - Patrícia V Mendonça
- CEMUC, Department of Chemical Engineering, University of Coimbra, Coimbra 3030-790, Portugal.
| | - Rita Branco
- CEMUC, Department of Life Sciences, University of Coimbra, Coimbra 3001-401, Portugal.
| | - Arménio C Serra
- CEMUC, Department of Chemical Engineering, University of Coimbra, Coimbra 3030-790, Portugal.
| | - Paula V Morais
- CEMUC, Department of Life Sciences, University of Coimbra, Coimbra 3001-401, Portugal.
| | - Jorge F J Coelho
- CEMUC, Department of Chemical Engineering, University of Coimbra, Coimbra 3030-790, Portugal.
| |
Collapse
|
69
|
Lüthje P, Brauner A. Novel Strategies in the Prevention and Treatment of Urinary Tract Infections. Pathogens 2016; 5:E13. [PMID: 26828523 PMCID: PMC4810134 DOI: 10.3390/pathogens5010013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/05/2015] [Accepted: 01/21/2016] [Indexed: 01/15/2023] Open
Abstract
Urinary tract infections are one of the most common bacterial infections, especially in women and children, frequently treated with antibiotics. The alarming increase in antibiotic resistance is a global threat to future treatment of infections. Therefore, alternative strategies are urgently needed. The innate immune system plays a fundamental role in protecting the urinary tract from infections. Antimicrobial peptides form an important part of the innate immunity. They are produced by epithelial cells and neutrophils and defend the urinary tract against invading bacteria. Since efficient resistance mechanisms have not evolved among bacterial pathogens, much effort has been put into exploring the role of antimicrobial peptides and possibilities to utilize them in clinical practice. Here, we describe the impact of antimicrobial peptides in the urinary tract and ways to enhance the production by hormones like vitamin D and estrogen. We also discuss the potential of medicinal herbs to be used in the prophylaxis and the treatment of urinary tract infections.
Collapse
Affiliation(s)
- Petra Lüthje
- Department of Microbiology, Tumor and Cell Biology, Division of Clinical Microbiology, Karolinska Institutet and Karolinska University Hospital, Stockholm SE-171 76, Sweden.
| | - Annelie Brauner
- Department of Microbiology, Tumor and Cell Biology, Division of Clinical Microbiology, Karolinska Institutet and Karolinska University Hospital, Stockholm SE-171 76, Sweden.
| |
Collapse
|
70
|
GRISCHKE J, EBERHARD J, STIESCH M. Antimicrobial dental implant functionalization strategies —A systematic review. Dent Mater J 2016; 35:545-58. [DOI: 10.4012/dmj.2015-314] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Jasmin GRISCHKE
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School
| | - Jörg EBERHARD
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School
| | - Meike STIESCH
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School
| |
Collapse
|
71
|
Antimicrobial Peptide Conformation as a Structural Determinant of Omptin Protease Specificity. J Bacteriol 2015; 197:3583-91. [PMID: 26350132 DOI: 10.1128/jb.00469-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/28/2015] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED Bacterial proteases contribute to virulence by cleaving host or bacterial proteins to promote survival and dissemination. Omptins are a family of proteases embedded in the outer membrane of Gram-negative bacteria that cleave various substrates, including host antimicrobial peptides, with a preference for cleaving at dibasic motifs. OmpT, the enterohemorrhagic Escherichia coli (EHEC) omptin, cleaves and inactivates the human cathelicidin LL-37. Similarly, the omptin CroP, found in the murine pathogen Citrobacter rodentium, which is used as a surrogate model to study human-restricted EHEC, cleaves the murine cathelicidin-related antimicrobial peptide (CRAMP). Here, we compared the abilities of OmpT and CroP to cleave LL-37 and CRAMP. EHEC OmpT degraded LL-37 and CRAMP at similar rates. In contrast, C. rodentium CroP cleaved CRAMP more rapidly than LL-37. The different cleavage rates of LL-37 and CRAMP were independent of the bacterial background and substrate sequence specificity, as OmpT and CroP have the same preference for cleaving at dibasic sites. Importantly, LL-37 was α-helical and CRAMP was unstructured under our experimental conditions. By altering the α-helicity of LL-37 and CRAMP, we found that decreasing LL-37 α-helicity increased its rate of cleavage by CroP. Conversely, increasing CRAMP α-helicity decreased its cleavage rate. This structural basis for CroP substrate specificity highlights differences between the closely related omptins of C. rodentium and E. coli. In agreement with previous studies, this difference in CroP and OmpT substrate specificity suggests that omptins evolved in response to the substrates present in their host microenvironments. IMPORTANCE Omptins are recognized as key virulence factors for various Gram-negative pathogens. Their localization to the outer membrane, their active site facing the extracellular environment, and their unique catalytic mechanism make them attractive targets for novel therapeutic strategies. Gaining insights into similarities and variations between the different omptin active sites and subsequent substrate specificities will be critical to develop inhibitors that can target multiple omptins. Here, we describe subtle differences between the substrate specificities of two closely related omptins, CroP and OmpT. This is the first reported example of substrate conformation acting as a structural determinant for omptin activity between OmpT-like proteases.
Collapse
|
72
|
Chen F, Hu H, Li Z, Huang J, Cai X, Wang C, He Q, Cao J. Deletion of HAPS_2096 Increases Sensitivity to Cecropin B in Haemophilus parasuis. J Mol Microbiol Biotechnol 2015; 25:284-91. [PMID: 26304836 DOI: 10.1159/000434752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cecropin B (CB) is a very effective natural antimicrobial peptide that has shown great potential for future antimicrobial drug development. HAPS_2096 is a Haemophilus parasuis gene that encodes the periplasmic substrate-binding protein of an ATP-binding cassette-type amino acid transporter. In this research, we constructed and verified an HAPS_2096 deletion mutant and a complementary HAPS_2096 mutant of H. parasuis JS0135. A bactericidal assay revealed that the HAPS_2096 deletion mutant was significantly more sensitive than the wild-type strain to 0.25-0.5 µg/ml CB. However, the gene complementation alleviated the CB sensitivity of the mutant. Immunoelectron microscopy observation following a 30-min treatment with a sublethal concentration of CB (0.25 μg/ml) revealed more extensive morphological damage in the mutant strain than in the wild-type strain. Hence, our results suggest that the HAPS_2096 gene contributes to H. parasuis resistance to CB.
Collapse
Affiliation(s)
- Fanjie Chen
- Division of Animal Infectious Disease, State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Wuhan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Peptides and Peptidomimetics for Antimicrobial Drug Design. Pharmaceuticals (Basel) 2015; 8:366-415. [PMID: 26184232 PMCID: PMC4588174 DOI: 10.3390/ph8030366] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/27/2015] [Accepted: 06/17/2015] [Indexed: 12/21/2022] Open
Abstract
The purpose of this paper is to introduce and highlight a few classes of traditional antimicrobial peptides with a focus on structure-activity relationship studies. After first dissecting the important physiochemical properties that influence the antimicrobial and toxic properties of antimicrobial peptides, the contributions of individual amino acids with respect to the peptides antibacterial properties are presented. A brief discussion of the mechanisms of action of different antimicrobials as well as the development of bacterial resistance towards antimicrobial peptides follows. Finally, current efforts on novel design strategies and peptidomimetics are introduced to illustrate the importance of antimicrobial peptide research in the development of future antibiotics.
Collapse
|
74
|
Nuri R, Shprung T, Shai Y. Defensive remodeling: How bacterial surface properties and biofilm formation promote resistance to antimicrobial peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:3089-100. [PMID: 26051126 DOI: 10.1016/j.bbamem.2015.05.022] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 11/15/2022]
Abstract
Multidrug resistance bacteria are a major concern worldwide. These pathogens cannot be treated with conventional antibiotics and thus alternative therapeutic agents are needed. Antimicrobial peptides (AMPs) are considered to be good candidates for this purpose. Most AMPs are short and positively charged amphipathic peptides, which are found in all known forms of life. AMPs are known to kill bacteria by binding to the negatively charged bacterial surface, and in most cases cause membrane disruption. Resistance toward AMPs can be developed, by modification of bacterial surface molecules, secretion of protective material and up-regulation or elimination of specific proteins. Because of the general mechanisms of attachment and action of AMPs, bacterial resistance to AMPs often involves biophysical and biochemical changes such as surface rigidity, cell wall thickness, surface charge, as well as membrane and cell wall modification. Here we focus on the biophysical, surface and surrounding changes that bacteria undergo in acquiring resistance to AMPs. In addition we discuss the question of whether bacterial resistance to administered AMPs might compromise our innate immunity to endogenous AMPs. This article is part of a Special Issue entitled: Bacterial Resistance to Antimicrobial Peptides.
Collapse
Affiliation(s)
- Reut Nuri
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tal Shprung
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yechiel Shai
- Department of Biological Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
75
|
Antimicrobial peptide resistance in Neisseria meningitidis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:3026-31. [PMID: 26002321 DOI: 10.1016/j.bbamem.2015.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 05/06/2015] [Accepted: 05/12/2015] [Indexed: 12/26/2022]
Abstract
Antimicrobial peptides (AMPs) play an important role as a host defense against microbial pathogens and are key components of the human innate immune response. Neisseria meningitidis frequently colonizes the human nasopharynx as a commensal but also is a worldwide cause of epidemic meningitis and rapidly fatal sepsis. In the human respiratory tract, the only known reservoir of N. meningitidis, meningococci are exposed to human endogenous AMPs. Thus, it is not surprising that meningococci have evolved effective mechanisms to confer intrinsic and high levels of resistance to the action of AMPs. This article reviews the current knowledge about AMP resistance mechanisms employed by N. meningitidis. Two major resistance mechanisms employed by meningococci are the constitutive modification of the lipid A head groups of lipooligosaccharides by phosphoethanolamine and the active efflux pump mediated excretion of AMPs. Other factors influencing AMP resistance, such as the major porin PorB, the pilin biogenesis apparatus, and capsular polysaccharides, have also been identified. Even with an inherently high intrinsic resistance, several AMP resistance determinants can be further induced upon exposure to AMPs. Many well-characterized AMP resistance mechanisms in other Gram-negative bacteria are not found in meningococci. Thus, N. meningitidis utilizes a limited but highly effective set of molecular mechanisms to mediate antimicrobial peptide resistance. This article is part of a Special Issue entitled: Bacterial Resistance to Antimicrobial Peptides.
Collapse
|
76
|
Reid-Yu SA, Tuinema BR, Small CN, Xing L, Coombes BK. CXCL9 contributes to antimicrobial protection of the gut during citrobacter rodentium infection independent of chemokine-receptor signaling. PLoS Pathog 2015; 11:e1004648. [PMID: 25643352 PMCID: PMC4333760 DOI: 10.1371/journal.ppat.1004648] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/30/2014] [Indexed: 12/20/2022] Open
Abstract
Chemokines have been shown to be effective bactericidal molecules against a variety of bacteria and fungi in vitro. These direct antimicrobial effects are independent of their chemotactic activities involving immunological receptors. However, the direct biological role that these proteins may play in host defense, particularly against intestinal pathogens, is poorly understood. Here, we show that CXCL9, an ELR- chemokine, exhibits direct antimicrobial activity against Citrobacter rodentium, an attaching/effacing pathogen that infects the gut mucosa. Inhibition of this antimicrobial activity in vivo using anti-CXCL9 antibodies increases host susceptibility to C. rodentium infection with pronounced bacterial penetration into crypts, increased bacterial load, and worsened tissue pathology. Using Rag1-/- mice and CXCR3-/- mice, we demonstrate that the role for CXCL9 in protecting the gut mucosa is independent of an adaptive response or its immunological receptor, CXCR3. Finally, we provide evidence that phagocytes function in tandem with NK cells for robust CXCL9 responses to C. rodentium. These findings identify a novel role for the immune cell-derived CXCL9 chemokine in directing a protective antimicrobial response in the intestinal mucosa. Host defense peptides are an essential part of the innate immune response to pathogens, particularly at mucosal surfaces. Some chemokines, previously known for their ability to recruit immune cells to a site of inflammation, have been identified to have direct antimicrobial activity in vitro against a variety of pathogens. Despite this, it was unknown whether chemokines play a role in protecting the gut mucosa against enteric pathogens, independent of their immunological receptors. Using a mouse model of enteric pathogen infection with both wild type mice and genetic knockouts, we showed that the chemokine CXCL9 has direct antimicrobial activity against pathogen infection. This antimicrobial activity prevented the invasion of bacteria into intestinal crypts, thus protecting the host from immunopathology. Neutralization of this CXCL9-dependent antimicrobial activity increased host susceptibility to infection, leading to bacterial penetration into intestinal crypts and increased tissue pathology. These data support the importance of a receptor-independent role for chemokines in host defense at mucosal surfaces and may offer alternative treatment strategies for infections, particularly in regards to organisms that are resistant to conventional antibiotics.
Collapse
Affiliation(s)
- Sarah A. Reid-Yu
- Michael G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Brian R. Tuinema
- Michael G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Cherrie N. Small
- Michael G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Lydia Xing
- Michael G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Brian K. Coombes
- Michael G. DeGroote Institute for Infectious Disease Research, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
77
|
A novel fragment based strategy for membrane active antimicrobials against MRSA. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1023-31. [PMID: 25582665 DOI: 10.1016/j.bbamem.2015.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/29/2014] [Accepted: 01/05/2015] [Indexed: 01/04/2023]
Abstract
Membrane active antimicrobials are a promising new generation of antibiotics that hold the potential to avert antibiotic resistance. However, poor understanding of the action mechanism and the lack of general design principles have impeded their development. Here we extend the concept of fragment based drug design and propose a pharmacophore model based on first principles for the design of membrane active antimicrobials against Gram positive pathogens. Elaborating on a natural xanthone-based hydrophobic scaffold, two derivatives of the pharmacophore model are proposed, and these demonstrate excellent antimicrobial activity. Rigorous molecular dynamics simulations combined with biophysical experiments suggest a three-step mechanism of action (absorption-translocation-disruption) which allows us to identify key factors for the practical optimization of each fragment of the pharmacophore. Moreover, the model matches the structures of several membrane active antimicrobials which are currently in clinical trials. Our model provides a novel and rational approach for the design of bactericidal molecules that target the bacterial membrane.
Collapse
|
78
|
Sumi CD, Yang BW, Yeo IC, Hahm YT. Antimicrobial peptides of the genus Bacillus: a new era for antibiotics. Can J Microbiol 2014; 61:93-103. [PMID: 25629960 DOI: 10.1139/cjm-2014-0613] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The rapid onset of resistance reduces the efficacy of most conventional antimicrobial drugs and is a general cause of concern for human well-being. Thus, there is great demand for a continuous supply of novel antibiotics to combat this problem. Bacteria-derived antimicrobial peptides (AMPs) have long been used as food preservatives; moreover, prior to the development of conventional antibiotics, these AMPs served as an efficient source of antibiotics. Recently, peptides produced by members of the genus Bacillus were shown to have a broad spectrum of antimicrobial activity against pathogenic microbes. Bacillus-derived AMPs can be synthesized both ribosomally and nonribosomally and can be classified according to peptide biosynthesis, structure, and molecular weight. The precise mechanism of action of these AMPs is not yet clear; however, one proposed mechanism is that these AMPs kill bacteria by forming channels in and (or) disrupting the bacterial cell wall. Bacillus-derived AMPs have potential in the pharmaceutical industry, as well as the food and agricultural sectors. Here, we focus on Bacillus-derived AMPs as a novel alternative approach to antibacterial drug development. We also provide an overview of the biosynthesis, mechanisms of action, applications, and effectiveness of different AMPs produced by members of the Bacillus genus, including several recently identified novel AMPs.
Collapse
Affiliation(s)
- Chandra Datta Sumi
- a Department of Systems Biotechnology, Chung-Ang University, 72-1 Nae-Ri, Daeduk-Myun, Anseong-Si, Gyeonggi-Do 456-756, South Korea
| | | | | | | |
Collapse
|
79
|
Dishaw LJ, Cannon JP, Litman GW, Parker W. Immune-directed support of rich microbial communities in the gut has ancient roots. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 47:36-51. [PMID: 24984114 PMCID: PMC4146740 DOI: 10.1016/j.dci.2014.06.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/30/2014] [Accepted: 06/21/2014] [Indexed: 05/12/2023]
Abstract
The animal gut serves as a primary location for the complex host-microbe interplay that is essential for homeostasis and may also reflect the types of ancient selective pressures that spawned the emergence of immunity in metazoans. In this review, we present a phylogenetic survey of gut host-microbe interactions and suggest that host defense systems arose not only to protect tissue directly from pathogenic attack but also to actively support growth of specific communities of mutualists. This functional dichotomy resulted in the evolution of immune systems much more tuned for harmonious existence with microbes than previously thought, existing as dynamic but primarily cooperative entities in the present day. We further present the protochordate Ciona intestinalis as a promising model for studying gut host-bacterial dialogue. The taxonomic position, gut physiology and experimental tractability of Ciona offer unique advantages in dissecting host-microbe interplay and can complement studies in other model systems.
Collapse
Affiliation(s)
- Larry J Dishaw
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA.
| | - John P Cannon
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA
| | - Gary W Litman
- Department of Pediatrics, University of South Florida Morsani College of Medicine, USF/ACH Children's Research Institute, 140 7th Avenue South, St. Petersburg, FL 33701, USA; Department of Molecular Genetics, All Children's Hospital-Johns Hopkins Medicine, 501 6th Avenue South, St. Petersburg, FL 33701, USA
| | - William Parker
- Department of Surgery, Duke University Medical Center, Box 2605, Durham, NC 27710, USA
| |
Collapse
|
80
|
García AN, Ayub ND, Fox AR, Gómez MC, Diéguez MJ, Pagano EM, Berini CA, Muschietti JP, Soto G. Alfalfa snakin-1 prevents fungal colonization and probably coevolved with rhizobia. BMC PLANT BIOLOGY 2014; 14:248. [PMID: 25227589 PMCID: PMC4177055 DOI: 10.1186/s12870-014-0248-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/11/2014] [Indexed: 05/23/2023]
Abstract
BACKGROUND The production of antimicrobial peptides is a common defense strategy of living cells against a wide range of pathogens. Plant snakin peptides inhibit bacterial and fungal growth at extremely low concentrations. However, little is known of their molecular and ecological characteristics, including origin, evolutionary equivalence, specific functions and activity against beneficial microbes. The aim of this study was to identify and characterize snakin-1 from alfalfa (MsSN1). RESULTS Phylogenetic analysis showed complete congruence between snakin-1 and plant trees. The antimicrobial activity of MsSN1 against bacterial and fungal pathogens of alfalfa was demonstrated in vitro and in vivo. Transgenic alfalfa overexpressing MsSN1 showed increased antimicrobial activity against virulent fungal strains. However, MsSN1 did not affect nitrogen-fixing bacterial strains only when these had an alfalfa origin. CONCLUSIONS The results reported here suggest that snakin peptides have important and ancestral roles in land plant innate immunity. Our data indicate a coevolutionary process, in which alfalfa exerts a selection pressure for resistance to MsSN1 on rhizobial bacteria. The increased antimicrobial activity against virulent fungal strains without altering the nitrogen-fixing symbiosis observed in MsSN1-overexpressing alfalfa transgenic plants opens the way to the production of effective legume transgenic cultivars for biotic stress resistance.
Collapse
Affiliation(s)
- Araceli Nora García
- />Instituto de Genética Ewald A. Favret (CICVyA-INTA), De los Reseros S/N, Castelar, C25 (1712) Buenos Aires Argentina
| | - Nicolás Daniel Ayub
- />Instituto de Genética Ewald A. Favret (CICVyA-INTA), De los Reseros S/N, Castelar, C25 (1712) Buenos Aires Argentina
| | - Ana Romina Fox
- />Instituto de Genética Ewald A. Favret (CICVyA-INTA), De los Reseros S/N, Castelar, C25 (1712) Buenos Aires Argentina
| | - María Cristina Gómez
- />Instituto de Genética Ewald A. Favret (CICVyA-INTA), De los Reseros S/N, Castelar, C25 (1712) Buenos Aires Argentina
| | - María José Diéguez
- />Instituto de Genética Ewald A. Favret (CICVyA-INTA), De los Reseros S/N, Castelar, C25 (1712) Buenos Aires Argentina
| | - Elba María Pagano
- />Instituto de Genética Ewald A. Favret (CICVyA-INTA), De los Reseros S/N, Castelar, C25 (1712) Buenos Aires Argentina
| | - Carolina Andrea Berini
- />Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), UBA-CONICET, Paraguay 2155, C1121ABG Ciudad Autónoma de Buenos Aires, Argentina
| | - Jorge Prometeo Muschietti
- />Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, Ciudad Universitaria, Pabellón II, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
- />Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, “Dr. Hector Torres”, (INGEBI-CONICET), Vuelta de Obligado 2490, C1428ADN Ciudad Autónoma de Buenos Aires, Argentina
| | - Gabriela Soto
- />Instituto de Genética Ewald A. Favret (CICVyA-INTA), De los Reseros S/N, Castelar, C25 (1712) Buenos Aires Argentina
| |
Collapse
|
81
|
Dobson AJ, Purves J, Rolff J. Increased survival of experimentally evolved antimicrobial peptide-resistant Staphylococcus aureus in an animal host. Evol Appl 2014; 7:905-12. [PMID: 25469169 PMCID: PMC4211720 DOI: 10.1111/eva.12184] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 05/20/2014] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial peptides (AMPs) have been proposed as new class of antimicrobial drugs, following the increasing prevalence of bacteria resistant to antibiotics. Synthetic AMPs are functional analogues of highly evolutionarily conserved immune effectors in animals and plants, produced in response to microbial infection. Therefore, the proposed therapeutic use of AMPs bears the risk of 'arming the enemy': bacteria that evolve resistance to AMPs may be cross-resistant to immune effectors (AMPs) in their hosts. We used a panel of populations of Staphylococcus aureus that were experimentally selected for resistance to a suite of individual AMPs and antibiotics to investigate the 'arming the enemy' hypothesis. We tested whether the selected strains showed higher survival in an insect model (Tenebrio molitor) and cross-resistance against other antimicrobials in vitro. A population selected for resistance to the antimicrobial peptide iseganan showed increased in vivo survival, but was not more virulent. We suggest that increased survival of AMP-resistant bacteria almost certainly poses problems to immune-compromised hosts.
Collapse
Affiliation(s)
- Adam J Dobson
- Animal & Plant Sciences, University of Sheffield, Western BankSheffield, UK
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College LondonLondon, UK
| | - Joanne Purves
- Animal & Plant Sciences, University of Sheffield, Western BankSheffield, UK
- School of Life Sciences, Centre for Biomolecular Science, University of NottinghamNottingham, UK
| | - Jens Rolff
- Animal & Plant Sciences, University of Sheffield, Western BankSheffield, UK
- Institute of Biology, Free University BerlinBerlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB)Berlin, Germany
| |
Collapse
|
82
|
Zähringer U, Ittig S, Lindner B, Moll H, Schombel U, Gisch N, Cornelis GR. NMR-based structural analysis of the complete rough-type lipopolysaccharide isolated from Capnocytophaga canimorsus. J Biol Chem 2014; 289:23963-76. [PMID: 24993825 DOI: 10.1074/jbc.m114.571489] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We here describe the NMR analysis of an intact lipopolysaccharide (LPS, endotoxin) in water with 1,2-dihexanoyl-sn-glycero-3-phosphocholine as detergent. When HPLC-purified rough-type LPS of Capnocytophaga canimorsus was prepared, (13)C,(15)N labeling could be avoided. The intact LPS was analyzed by homonuclear ((1)H) and heteronuclear ((1)H,(13)C, and (1)H,(31)P) correlated one- and two-dimensional NMR techniques as well as by mass spectrometry. It consists of a penta-acylated lipid A with an α-linked phosphoethanolamine attached to C-1 of GlcN (I) in the hybrid backbone, lacking the 4'-phosphate. The hydrophilic core oligosaccharide was found to be a complex hexasaccharide with two mannose (Man) and one each of 3-deoxy-d-manno-oct-2-ulosonic acid (Kdo), Gal, GalN, and l-rhamnose residues. Position 4 of Kdo is substituted by phosphoethanolamine, also present in position 6 of the branched Man(I) residue. This rough-type LPS is exceptional in that all three negative phosphate residues are "masked" by positively charged ethanolamine substituents, leading to an overall zero net charge, which has so far not been observed for any other LPS. In biological assays, the corresponding isolated lipid A was found to be endotoxically almost inactive. By contrast, the intact rough-type LPS described here expressed a 20,000-fold increased endotoxicity, indicating that the core oligosaccharide significantly contributes to the endotoxic potency of the whole rough-type C. canimorsus LPS molecule. Based on these findings, the strict view that lipid A alone represents the toxic center of LPS needs to be reassessed.
Collapse
Affiliation(s)
- Ulrich Zähringer
- From the Division of Immunochemistry/Bioanalytical Chemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Parkallee 4a, 23845 Borstel, Germany,
| | - Simon Ittig
- Infection Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland, and
| | - Buko Lindner
- From the Division of Immunochemistry/Bioanalytical Chemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Parkallee 4a, 23845 Borstel, Germany
| | - Hermann Moll
- From the Division of Immunochemistry/Bioanalytical Chemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Parkallee 4a, 23845 Borstel, Germany
| | - Ursula Schombel
- From the Division of Immunochemistry/Bioanalytical Chemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Parkallee 4a, 23845 Borstel, Germany
| | - Nicolas Gisch
- From the Division of Immunochemistry/Bioanalytical Chemistry, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Parkallee 4a, 23845 Borstel, Germany
| | - Guy R Cornelis
- Infection Biology, Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland, and the Department of Biology, University of Namur, B5000 Namur, Belgium
| |
Collapse
|
83
|
Host defense peptide resistance contributes to colonization and maximal intestinal pathology by Crohn's disease-associated adherent-invasive Escherichia coli. Infect Immun 2014; 82:3383-93. [PMID: 24866805 DOI: 10.1128/iai.01888-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Host defense peptides secreted by colonocytes and Paneth cells play a key role in innate host defenses in the gut. In Crohn's disease, the burden of tissue-associated Escherichia coli commonly increases at epithelial surfaces where host defense peptides concentrate, suggesting that this bacterial population might actively resist this mechanism of bacterial killing. Adherent-invasive E. coli (AIEC) is associated with Crohn's disease; however, the colonization determinants of AIEC in the inflamed gut are undefined. Here, we establish that host defense peptide resistance contributes to host colonization by Crohn's-associated AIEC. We identified a plasmid-encoded genomic island (called PI-6) in AIEC strain NRG857c that confers high-level resistance to α-helical cationic peptides and α- and β-defensins. Deletion of PI-6 sensitized strain NRG857c to these host defense molecules, reduced its competitive fitness in a mouse model of infection, and attenuated its ability to induce cecal pathology. This phenotype is due to two genes in PI-6, arlA, which encodes a Mig-14 family protein implicated in defensin resistance, and arlC, an OmpT family outer membrane protease. Implicit in these findings are new bacterial targets whose inhibition might limit AIEC burden and disease in the gut.
Collapse
|
84
|
Lu S, Walters G, Parg R, Dutcher JR. Nanomechanical response of bacterial cells to cationic antimicrobial peptides. SOFT MATTER 2014; 10:1806-1815. [PMID: 24652481 DOI: 10.1039/c3sm52801d] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The effectiveness of antimicrobial compounds can be easily screened, however their mechanism of action is much more difficult to determine. Many compounds act by compromising the mechanical integrity of the bacterial cell envelope, and our study introduces an AFM-based creep deformation technique to evaluate changes in the time-dependent mechanical properties of Pseudomonas aeruginosa PAO1 bacterial cells upon exposure to two different but structurally related antimicrobial peptides. We observed a distinctive signature for the loss of integrity of the bacterial cell envelope following exposure to the peptides. Measurements performed before and after exposure, as well as time-resolved measurements and those performed at different concentrations, revealed large changes to the viscoelastic parameters that are consistent with differences in the membrane permeabilizing effects of the peptides. The AFM creep deformation measurement provides new, unique insight into the kinetics and mechanism of action of antimicrobial peptides on bacteria.
Collapse
Affiliation(s)
- Shun Lu
- Department of Physics, University of Guelph, Guelph, N1G 2W1, Ontario, Canada.
| | | | | | | |
Collapse
|
85
|
Derde M, Guérin-Dubiard C, Lechevalier V, Cochet MF, Jan S, Baron F, Gautier M, Vié V, Nau F. Dry-heating of lysozyme increases its activity against Escherichia coli membranes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:1692-1700. [PMID: 24450740 DOI: 10.1021/jf405155p] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
For food as well as for medical applications, there is a growing interest in novel and natural antimicrobial molecules. Lysozyme is a promising candidate for the development of such molecules. This protein is largely studied and known for its muramidase activity against Gram-positive bacteria, but it also shows antimicrobial activity against Gram-negative bacteria, especially when previously modified. In this study, the activity of dry-heated lysozyme (DH-L) against Escherichia coli has been investigated and compared to that of native lysozyme (N-L). Whereas N-L only delays bacterial growth, DH-L causes an early-stage population decrease. The accompanying membrane permeabilization suggests that DH-L induces either larger pores or more pores in the outer membrane as compared to N-L, as well as more ion channels in the inner membrane. The strong morphological modifications observed by optical microscopy and atomic force microscopy when E. coli cells are treated with DH-L are consistent with the suggested disturbances of membrane integrity. The higher hydrophobicity, surface activity, and positive charge induced by dry-heating could be responsible for the increased activity of DH-L on the E. coli membranes.
Collapse
Affiliation(s)
- Melanie Derde
- Agrocampus Ouest, UMR1253 Science et technologie du lait et de l'œuf , F-35042 Rennes, France
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Thomassin JL, Lee MJ, Brannon JR, Sheppard DC, Gruenheid S, Le Moual H. Both group 4 capsule and lipopolysaccharide O-antigen contribute to enteropathogenic Escherichia coli resistance to human α-defensin 5. PLoS One 2013; 8:e82475. [PMID: 24324796 PMCID: PMC3853201 DOI: 10.1371/journal.pone.0082475] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/02/2013] [Indexed: 12/18/2022] Open
Abstract
Enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC) are food-borne pathogens that colonize the small intestine and colon, respectively. To cause disease, these pathogens must overcome the action of different host antimicrobial peptides (AMPs) secreted into these distinct niches. We have shown previously that EHEC expresses high levels of the OmpT protease to inactivate the human cathelicidin LL-37, an AMP present in the colon. In this study, we investigate the mechanisms used by EPEC to resist human α-defensin 5 (HD-5), the most abundant AMP in the small intestine. Quantitative PCR was used to measure transcript levels of various EPEC surface structures. High transcript levels of gfcA, a gene required for group 4 capsule (G4C) production, were observed in EPEC, but not in EHEC. The unencapsulated EPEC ∆gfcA and EHEC wild-type strains were more susceptible to HD-5 than EPEC wild-type. Since the G4C is composed of the same sugar repeats as the lipopolysaccharide O-antigen, an -antigen ligase (waaL) deletion mutant was generated in EPEC to assess its role in HD-5 resistance. The ∆waaL EPEC strain was more susceptible to HD-5 than both the wild-type and ∆gfcA strains. The ∆gfcA∆waaL EPEC strain was not significantly more susceptible to HD-5 than the ∆waaL strain, suggesting that the absence of -antigen influences G4C formation. To determine whether the G4C and -antigen interact with HD-5, total polysaccharide was purified from wild-type EPEC and added to the ∆gfcA∆waaL strain in the presence of HD-5. The addition of exogenous polysaccharide protected the susceptible strain against HD-5 killing in a dose-dependent manner, suggesting that HD-5 binds to the polysaccharides present on the surface of EPEC. Altogether, these findings indicate that EPEC relies on both the G4C and the -antigen to resist the bactericidal activity of HD-5.
Collapse
Affiliation(s)
- Jenny-Lee Thomassin
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Mark J. Lee
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - John R. Brannon
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Donald C. Sheppard
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Microbiome and Disease Tolerance Centre, McGill University, Montreal, Quebec, Canada
| | - Samantha Gruenheid
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Microbiome and Disease Tolerance Centre, McGill University, Montreal, Quebec, Canada
| | - Hervé Le Moual
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Microbiome and Disease Tolerance Centre, McGill University, Montreal, Quebec, Canada
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
87
|
Volzing K, Borrero J, Sadowsky MJ, Kaznessis YN. Antimicrobial peptides targeting Gram-negative pathogens, produced and delivered by lactic acid bacteria. ACS Synth Biol 2013; 2:643-50. [PMID: 23808914 DOI: 10.1021/sb4000367] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We present results of tests with recombinant Lactococcus lactis that produce and secrete heterologous antimicrobial peptides with activity against Gram-negative pathogenic Escherichia coli and Salmonella . In an initial screening, the activities of numerous candidate antimicrobial peptides, made by solid state synthesis, were assessed against several indicator pathogenic E. coli and Salmonella strains. Peptides A3APO and Alyteserin were selected as top performers based on high antimicrobial activity against the pathogens tested and on significantly lower antimicrobial activity against L. lactis . Expression cassettes containing the signal peptide of the protein Usp45 fused to the codon-optimized sequence of mature A3APO and Alyteserin were cloned under the control of a nisin-inducible promoter PnisA and transformed into L. lactis IL1403. The resulting recombinant strains were induced to express and secrete both peptides. A3APO- and Alyteserin-containing supernatants from these recombinant L. lactis inhibited the growth of pathogenic E. coli and Salmonella by up to 20-fold, while maintaining the host's viability. This system may serve as a model for the production and delivery of antimicrobial peptides by lactic acid bacteria to target Gram-negative pathogenic bacteria populations.
Collapse
Affiliation(s)
- Katherine Volzing
- Department of Chemical
Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Juan Borrero
- Department of Chemical
Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Michael J. Sadowsky
- Department of Soil,
Water, and Climate, University of Minnesota, St. Paul, Minnesota 55108, United States
- BioTechnology Institute, University of Minnesota, St. Paul, Minnesota 55108,
United States
| | - Yiannis N. Kaznessis
- Department of Chemical
Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- BioTechnology Institute, University of Minnesota, St. Paul, Minnesota 55108,
United States
| |
Collapse
|
88
|
Xu ZQ, Flavin MT, Flavin J. Combating multidrug-resistant Gram-negative bacterial infections. Expert Opin Investig Drugs 2013; 23:163-82. [PMID: 24215473 DOI: 10.1517/13543784.2014.848853] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Multidrug-resistant (MDR) bacterial infections, especially those caused by Gram-negative pathogens, have emerged as one of the world's greatest health threats. The development of novel antibiotics to treat MDR Gram-negative bacteria has, however, stagnated over the last half century. AREAS COVERED This review provides an overview of recent R&D activities in the search for novel antibiotics against MDR Gram-negatives. It provides emphasis in three key areas. First, the article looks at new analogs of existing antibiotic molecules such as β-lactams, tetracyclines, and aminoglycoside as well as agents against novel bacterial targets such as aminoacyl-tRNA synthetase and peptide deformylase. Second, it also examines alternative strategies to conventional approaches including cationic antimicrobial peptides, siderophores, efflux pump inhibitors, therapeutic antibodies, and renewed interest in abandoned treatments or those with limited indications. Third, the authors aim to provide an update on the current clinical development status for each drug candidate. EXPERT OPINION The traditional analog approach is insufficient to meet the formidable challenge brought forth by MDR superbugs. With the disappointing results of the genomics approach for delivering novel targets and drug candidates, alternative strategies to permeate the bacterial cell membrane, enhance influx, disrupt efflux, and target specific pathogens via therapeutic antibodies are attractive and promising. Coupled with incentivized business models, governmental policies, and a clarified regulatory pathway, it is hoped that the antibiotic pipeline will be filled with an effective armamentarium to safeguard global health.
Collapse
Affiliation(s)
- Ze-Qi Xu
- SynChem, Inc. , 1400 Chase Avenue, Elk Grove Village, IL 60007 , USA +1 847 298 2436 ;
| | | | | |
Collapse
|
89
|
Locock KES, Michl TD, Valentin JDP, Vasilev K, Hayball JD, Qu Y, Traven A, Griesser HJ, Meagher L, Haeussler M. Guanylated Polymethacrylates: A Class of Potent Antimicrobial Polymers with Low Hemolytic Activity. Biomacromolecules 2013; 14:4021-31. [DOI: 10.1021/bm401128r] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Katherine E. S. Locock
- CSIRO
Materials Science and Engineering, Bayview Avenue, Clayton, Victoria 3168, Australia
| | - Thomas D. Michl
- Ian
Wark Research Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Jules D. P. Valentin
- Mawson
Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Krasimir Vasilev
- Mawson
Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - John D. Hayball
- Sansom
Institute, School of Pharmacy and Medical Sciences, University of South Australia, City East, South Australia 5000, Australia
| | - Yue Qu
- Department
of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Ana Traven
- Department
of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Hans J. Griesser
- Ian
Wark Research Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Laurence Meagher
- CSIRO
Materials Science and Engineering, Bayview Avenue, Clayton, Victoria 3168, Australia
| | - Matthias Haeussler
- CSIRO
Materials Science and Engineering, Bayview Avenue, Clayton, Victoria 3168, Australia
| |
Collapse
|
90
|
Dobson AJ, Purves J, Kamysz W, Rolff J. Comparing selection on S. aureus between antimicrobial peptides and common antibiotics. PLoS One 2013; 8:e76521. [PMID: 24204634 PMCID: PMC3799789 DOI: 10.1371/journal.pone.0076521] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 09/01/2013] [Indexed: 11/18/2022] Open
Abstract
With a diminishing number of effective antibiotics, there has been interest in developing antimicrobial peptides (AMPs) as drugs. However, any new drug faces potential bacterial resistance evolution. Here, we experimentally compare resistance evolution in Staphylococcus aureus selected by three AMPs (from mammals, amphibians and insects), a combination of two AMPs, and two antibiotics: the powerful last-resort vancomycin and the classic streptomycin. We find that resistance evolves readily against single AMPs and against streptomycin, with no detectable fitness cost. However the response to selection from our combination of AMPs led to extinction, in a fashion qualitatively similar to vancomycin. This is consistent with the hypothesis that simultaneous release of multiple AMPs during immune responses is a factor which constrains evolution of AMP resistant pathogens.
Collapse
Affiliation(s)
- Adam J. Dobson
- Animal & Plant Sciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
- Department of Entomology, Cornell University, Ithaca, New York, United States of America
| | - Joanne Purves
- Animal & Plant Sciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
- School of Molecular Medical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Wojciech Kamysz
- Faculty of Pharmacy, Gdansk University of Medicine, Gdansk, Poland
| | - Jens Rolff
- Animal & Plant Sciences, University of Sheffield, Western Bank, Sheffield, United Kingdom
- Institute of Biology, Free University Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
91
|
O'Connell KMG, Hodgkinson JT, Sore HF, Welch M, Salmond GPC, Spring DR. Die Bekämpfung multiresistenter Bakterien: aktuelle Strategien zur Entdeckung neuer Antibiotika. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201209979] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
92
|
O'Connell KMG, Hodgkinson JT, Sore HF, Welch M, Salmond GPC, Spring DR. Combating Multidrug-Resistant Bacteria: Current Strategies for the Discovery of Novel Antibacterials. Angew Chem Int Ed Engl 2013; 52:10706-33. [DOI: 10.1002/anie.201209979] [Citation(s) in RCA: 310] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 02/25/2013] [Indexed: 11/07/2022]
|
93
|
Ostaff MJ, Stange EF, Wehkamp J. Antimicrobial peptides and gut microbiota in homeostasis and pathology. EMBO Mol Med 2013; 5:1465-83. [PMID: 24039130 PMCID: PMC3799574 DOI: 10.1002/emmm.201201773] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/19/2013] [Accepted: 07/04/2013] [Indexed: 12/17/2022] Open
Abstract
We survive because we adapted to a world of microorganisms. All our epithelial surfaces participate in keeping up an effective barrier against microbes while not initiating ongoing inflammatory processes and risking collateral damage to the host. Major players in this scenario are antimicrobial peptides (AMPs). Such broad-spectrum innate antibiotics are in part produced by specialized cells but also widely sourced from all epithelia as well as circulating inflammatory cells. AMPs belong to an ancient defense system found in all organisms and participated in a preservative co-evolution with a complex microbiome. Particularly interesting interactions between host barrier and microbiota can be found in the gut. The intestinal cell lining not only has to maintain a tightly regulated homeostasis during its high-throughput regeneration, but also a balanced relationship towards an extreme number of mutualistic or commensal inhabitants. Recent research suggests that advancing our understanding of the circumstances of such balanced and sometimes imbalanced interactions between gut microbiota and host AMPs should have therapeutic implications for different intestinal disorders.
Collapse
Affiliation(s)
- Maureen J Ostaff
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tuebingen, Germany
| | | | | |
Collapse
|
94
|
Dennison SR, Harris F, Mura M, Morton LHG, Zvelindovsky A, Phoenix DA. A novel form of bacterial resistance to the action of eukaryotic host defense peptides, the use of a lipid receptor. Biochemistry 2013; 52:6021-9. [PMID: 23895279 DOI: 10.1021/bi400719j] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Host defense peptides show great potential for development as new antimicrobial agents with novel mechanisms of action. However, a small number of resistance mechanisms to their action are known, and here, we report a novel bacterial resistance mechanism mediated by a lipid receptor. Maximin H5 from Bombina maxima bound anionic and zwitterionic membranes with low affinity (Kd > 225 μM) while showing a strong ability to lyse (>55%) and penetrate (π > 6.0 mN m(-1)) these membranes. However, the peptide bound Escherichia coli and 1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine (DMPE) membranes with higher affinity (Kd < 65 μM) and showed a very low ability for bilayer lysis (<8%) and partitioning (π > 1.0 mN m(-1)). Increasing levels of membrane DMPE correlated with enhanced binding by the peptide (R(2) = 0.96) but inversely correlated with its lytic ability (R(2) = 0.98). Taken with molecular dynamic simulations, these results suggest that maximin H5 possesses membranolytic activity, primarily involving bilayer insertion of its strongly hydrophobic N-terminal region. However, this region was predicted to form multiple hydrogen bonds with phosphate and ammonium groups within PE head-groups, which in concert with charge-charge interactions anchor the peptide to the surface of E. coli membranes, inhibiting its membranolytic action.
Collapse
Affiliation(s)
- Sarah R Dennison
- School of Pharmacy and Biomedical Sciences, †School of Forensic and Investigative Sciences, and ‡School of Computing, Engineering and Physical Sciences, University of Central Lancashire , Preston, PR1 2HE, U.K
| | | | | | | | | | | |
Collapse
|
95
|
Differential adaptive responses of Staphylococcus aureus to in vitro selection with different antimicrobial peptides. Antimicrob Agents Chemother 2013; 57:5134-7. [PMID: 23856775 DOI: 10.1128/aac.00780-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
We subjected Staphylococcus aureus ATCC 29213 to serial passage in the presence of subinhibitory concentrations of magainin 2 and gramicidin D for several hundred generations. We obtained S. aureus strains with induced resistance to magainin 2 (strain 55MG) and gramicidin D (strain 55GR) that showed different phenotypic changes in membrane properties. Both exhibited a change in membrane phospholipid content and an increase in membrane rigidity, while an alteration in net charge compared to that of the control occurred only in the case of 55MG.
Collapse
|
96
|
Brannon JR, Thomassin JL, Desloges I, Gruenheid S, Le Moual H. Role of uropathogenicEscherichia coliOmpT in the resistance against human cathelicidin LL-37. FEMS Microbiol Lett 2013; 345:64-71. [DOI: 10.1111/1574-6968.12185] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 11/30/2022] Open
Affiliation(s)
- John R. Brannon
- Department of Microbiology and Immunology; McGill University; Montreal; QC; Canada
| | - Jenny-Lee Thomassin
- Department of Microbiology and Immunology; McGill University; Montreal; QC; Canada
| | - Isabelle Desloges
- Department of Microbiology and Immunology; McGill University; Montreal; QC; Canada
| | - Samantha Gruenheid
- Department of Microbiology and Immunology; McGill University; Montreal; QC; Canada
| | | |
Collapse
|
97
|
Confer AW, Ayalew S. The OmpA family of proteins: Roles in bacterial pathogenesis and immunity. Vet Microbiol 2013; 163:207-22. [DOI: 10.1016/j.vetmic.2012.08.019] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/23/2012] [Indexed: 10/27/2022]
|
98
|
Laurencin M, Amor M, Fleury Y, Baudy-Floc’h M. De Novo Cyclic Pseudopeptides Containing Aza-β3-amino Acids Exhibiting Antimicrobial Activities. J Med Chem 2012; 55:10885-95. [DOI: 10.1021/jm3009037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Mathieu Laurencin
- Université de Rennes 1, ICMV, UMR CNRS 6226, 263 Av. du Général
Leclerc, F-35042 Rennes Cedex, France
| | - Mosbah Amor
- Université de Rennes 1, ICMV, UMR CNRS 6226, 263 Av. du Général
Leclerc, F-35042 Rennes Cedex, France
| | - Yannick Fleury
- Université de Bretagne Occidentale, Laboratoire Universitaire de
Biodiversité et d’Ecologie Microbienne, EA 3882, F-29000
Quimper, France
| | - Michèle Baudy-Floc’h
- Université de Rennes 1, ICMV, UMR CNRS 6226, 263 Av. du Général
Leclerc, F-35042 Rennes Cedex, France
| |
Collapse
|
99
|
Thomassin JL, Brannon JR, Kaiser J, Gruenheid S, Le Moual H. Enterohemorrhagic and enteropathogenic Escherichia coli evolved different strategies to resist antimicrobial peptides. Gut Microbes 2012; 3:556-61. [PMID: 22895086 PMCID: PMC3495793 DOI: 10.4161/gmic.21656] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Enterohemorrhagic and enteropathogenic Escherichia coli (EHEC and EPEC) are enteric human pathogens that colonize the large and small intestines, respectively. To establish infection EHEC and EPEC must overcome innate host defenses, such as antimicrobial peptides (AMPs) produced by the intestinal epithelium. Gram-negative pathogens have evolved different mechanisms to resist AMPs, including outer-membrane proteases that degrade AMPs. We showed that the protease OmpT degrades the human AMP LL-37 more rapidly in EHEC than in EPEC. Promoter-swap experiments showed that this is due to differences in the promoters of the two genes, leading to greater ompT expression and subsequently greater levels of OmpT in EHEC. Here, we propose that the different ompT expression in EHEC and EPEC reflects the varying levels of LL-37 throughout the human intestinal tract. These data suggest that EHEC and EPEC adapted to their specific niches by developing distinct AMP-specific resistance mechanisms.
Collapse
Affiliation(s)
- Jenny-Lee Thomassin
- Department of Microbiology and Immunology; McGill University, Montreal, QC Canada
| | - John R. Brannon
- Department of Microbiology and Immunology; McGill University, Montreal, QC Canada
| | - Julienne Kaiser
- Department of Microbiology and Immunology; McGill University, Montreal, QC Canada
| | - Samantha Gruenheid
- Department of Microbiology and Immunology; McGill University, Montreal, QC Canada,Correspondence to: Samantha Gruenheid, and Hervé Le Moual,
| | - Hervé Le Moual
- Department of Microbiology and Immunology; McGill University, Montreal, QC Canada,Faculty of Dentistry; McGill University, Montreal, QC Canada,Correspondence to: Samantha Gruenheid, and Hervé Le Moual,
| |
Collapse
|
100
|
Biofilm formation in Staphylococcus implant infections. A review of molecular mechanisms and implications for biofilm-resistant materials. Biomaterials 2012; 33:5967-82. [PMID: 22695065 DOI: 10.1016/j.biomaterials.2012.05.031] [Citation(s) in RCA: 675] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 05/15/2012] [Indexed: 02/07/2023]
Abstract
Implant infections in orthopaedics, as well as in many other medical fields, are chiefly caused by staphylococci. The ability of growing within a biofilm enhances the chances of staphylococci to protect themselves from host defences, antibiotic therapies, and biocides. Advances in scientific knowledge on structural molecules (exopolysaccharide, proteins, teichoic acids, and the most recently described extracellular DNA), on the synthesis and genetics of staphylococcal biofilms, and on the complex network of signal factors that intervene in their control are here presented, also reporting on the emerging strategies to disrupt or inhibit them. The attitude of polymorphonuclear neutrophils and macrophages to infiltrate and phagocytise biofilms, as well as the ambiguous behaviour exhibited by these innate immune cells in biofilm-related implant infections, are here discussed. Research on anti-biofilm biomaterials is focused, reviewing materials loaded with antibacterial substances, or coated with anti-adhesive/anti-bacterial immobilized agents, or surfaced with nanostructures. Latter approaches appear promising, since they avoid the spread of antibacterial substances in the neighbouring tissues with the consequent risk of inducing bacterial resistance.
Collapse
|