Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Appl Environ Microbiol ; 90(2): e0084223, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38259077

ABSTRACT

Diverse influenza A viruses (IAVs) circulate in wild birds, including highly pathogenic strains that infect poultry and humans. Consequently, surveillance of IAVs in wild birds is a cornerstone of agricultural biosecurity and pandemic preparedness. Surveillance is traditionally done by testing wild birds directly, but obtaining these specimens is labor intensive, detection rates can be low, and sampling is often biased toward certain avian species. As a result, local incursions of dangerous IAVs are rarely detected before outbreaks begin. Testing environmental specimens from wild bird habitats has been proposed as an alternative surveillance strategy. These specimens are thought to contain diverse IAVs deposited by a broad range of avian hosts, including species that are not typically sampled by surveillance programs. To enable this surveillance strategy, we developed a targeted genomic sequencing method for characterizing IAVs in these challenging environmental specimens. It combines custom hybridization probes, unique molecular index-based library construction, and purpose-built bioinformatic tools, allowing IAV genomic material to be enriched and analyzed with single-fragment resolution. We demonstrated our method on 90 sediment specimens from wetlands around Vancouver, Canada. We recovered 2,312 IAV genome fragments originating from all eight IAV genome segments. Eleven hemagglutinin subtypes and nine neuraminidase subtypes were detected, including H5, the current global surveillance priority. Our results demonstrate that targeted genomic sequencing of environmental specimens from wild bird habitats could become a valuable complement to avian influenza surveillance programs.IMPORTANCEIn this study, we developed genome sequencing tools for characterizing avian influenza viruses in sediment from wild bird habitats. These tools enable an environment-based approach to avian influenza surveillance. This could improve early detection of dangerous strains in local wild birds, allowing poultry producers to better protect their flocks and prevent human exposures to potential pandemic threats. Furthermore, we purposefully developed these methods to contend with viral genomic material that is diluted, fragmented, incomplete, and derived from multiple strains and hosts. These challenges are common to many environmental specimens, making these methods broadly applicable for genomic pathogen surveillance in diverse contexts.


Subject(s)
Influenza A virus , Influenza in Birds , Animals , Animals, Wild , Birds , Genomics , Influenza A virus/genetics , Influenza in Birds/epidemiology , Phylogeny , Poultry , Wetlands
2.
EBioMedicine ; 33: 211-217, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29936135

ABSTRACT

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA), typified by the pulse-field type USA300, is an emerging endemic pathogen that is spreading rapidly among healthy people. CA-MRSA causes skin and soft tissue infections, life-threatening necrotizing pneumonia and sepsis, and is remarkably resistant to many antibiotics. Here we show that engineered liposomes composed of naturally occurring sphingomyelin were able to sequester cytolytic toxins secreted by USA300 and prevent necrosis of human erythrocytes, peripheral blood mononuclear cells and bronchial epithelial cells. Mass spectrometric analysis revealed the capture by liposomes of phenol-soluble modulins, α-hemolysin and other toxins. Sphingomyelin liposomes prevented hemolysis induced by pure phenol-soluble modulin-α3, one of the main cytolytic components in the USA300 secretome. In contrast, sphingomyelin liposomes harboring a high cholesterol content (66 mol/%) were unable to protect human cells from phenol-soluble modulin-α3-induced lysis, however these liposomes efficiently sequestered the potent staphylococcal toxin α-hemolysin. In a murine cutaneous abscess model, a single dose of either type of liposomes was sufficient to significantly decrease tissue dermonecrosis. Our results provide further insights into the promising potential of tailored liposomal therapy in the battle against infectious diseases.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Methicillin-Resistant Staphylococcus aureus/metabolism , Sphingomyelins/administration & dosage , Staphylococcal Skin Infections/therapy , Animals , Cell Line , Community-Acquired Infections , Disease Models, Animal , Hemolysin Proteins/antagonists & inhibitors , Humans , Liposomes , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Necrosis , Sphingomyelins/pharmacology , Treatment Outcome
3.
PLoS Pathog ; 14(6): e1007084, 2018 06.
Article in English | MEDLINE | ID: mdl-29928049

ABSTRACT

With the antibiotic development pipeline running dry, many fear that we might soon run out of treatment options. High-density infections are particularly difficult to treat due to their adaptive multidrug-resistance and currently there are no therapies that adequately address this important issue. Here, a large-scale in vivo study was performed to enhance the activity of antibiotics to treat high-density infections caused by multidrug-resistant Gram-positive and Gram-negative bacteria. It was shown that synthetic peptides can be used in conjunction with the antibiotics ciprofloxacin, meropenem, erythromycin, gentamicin, and vancomycin to improve the treatment outcome of murine cutaneous abscesses caused by clinical hard-to-treat pathogens including all ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter cloacae) pathogens and Escherichia coli. Promisingly, combination treatment often showed synergistic effects that significantly reduced abscess sizes and/or improved clearance of bacterial isolates from the infection site, regardless of the antibiotic mode of action. In vitro data suggest that the mechanisms of peptide action in vivo include enhancement of antibiotic penetration and potential disruption of the stringent stress response.


Subject(s)
Abscess/drug therapy , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/pathogenicity , Peptide Fragments/pharmacology , Skin Diseases, Bacterial/drug therapy , Abscess/microbiology , Animals , Biofilms/growth & development , Drug Synergism , Female , Injections, Subcutaneous , Mice , Microbial Viability , Skin Diseases, Bacterial/microbiology
4.
Sci Rep ; 8(1): 1871, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382854

ABSTRACT

Biofilms represent a multicellular growth state of bacteria that are intrinsically resistant to conventional antibiotics. It was recently shown that a synthetic immunomodulatory cationic peptide, 1018 (VRLIVAVRIWRR-NH2), exhibits broad-spectrum antibiofilm activity but the sequence determinants of antibiofilm peptides have not been systematically studied. In the present work, a peptide library consisting of 96 single amino acid substituted variants of 1018 was SPOT-synthesized on cellulose arrays and evaluated against methicillin resistant Staphylococcus aureus (MRSA) biofilms. This dataset was used to establish quantitative structure-activity relationship (QSAR) models relating the antibiofilm activity of these peptides to hundreds of molecular descriptors derived from their sequences. The developed 3D QSAR models then predicted the probability that a peptide would possess antibiofilm activity from a library of 100,000 virtual peptide sequences in silico. A subset of these variants were SPOT-synthesized and their activity assessed, revealing that the QSAR models resulted in ~85% prediction accuracy. Notably, peptide 3002 (ILVRWIRWRIQW-NH2) was identified that exhibited an 8-fold increased antibiofilm potency in vitro compared to 1018 and proved effective in vivo, significantly reducing abscess size in a chronic MRSA mouse infection model. This study demonstrates that QSAR modeling can successfully be used to identify antibiofilm specific peptides with therapeutic potential.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Computer-Aided Design , Drug Discovery , Peptide Fragments/pharmacology , Skin Diseases, Bacterial/drug therapy , Staphylococcal Infections/drug therapy , Abscess/drug therapy , Abscess/microbiology , Animals , Biofilms/growth & development , Disease Models, Animal , Female , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Microbial Sensitivity Tests , Quantitative Structure-Activity Relationship , Skin Diseases, Bacterial/microbiology , Staphylococcal Infections/microbiology
5.
ACS Infect Dis ; 4(2): 93-106, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29280609

ABSTRACT

Microbial biofilms, which are elaborate and highly resistant microbial aggregates formed on surfaces or medical devices, cause two-thirds of infections and constitute a serious threat to public health. Immunocompromised patients, individuals who require implanted devices, artificial limbs, organ transplants, or external life support and those with major injuries or burns, are particularly prone to become infected. Antibiotics, the mainstay treatments of bacterial infections, have often proven ineffective in the fight against microbes when growing as biofilms, and to date, no antibiotic has been developed for use against biofilm infections. Antibiotic resistance is rising, but biofilm-mediated multidrug resistance transcends this in being adaptive and broad spectrum and dependent on the biofilm growth state of organisms. Therefore, the treatment of biofilms requires drug developers to start thinking outside the constricted "antibiotics" box and to find alternative ways to target biofilm infections. Here, we highlight recent approaches for combating biofilms focusing on the eradication of preformed biofilms, including electrochemical methods, promising antibiofilm compounds and the recent progress in drug delivery strategies to enhance the bioavailability and potency of antibiofilm agents.


Subject(s)
Anti-Infective Agents/pharmacology , Biofilms/drug effects , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Drug Delivery Systems , Drug Resistance, Bacterial , Electrochemical Techniques , Humans
6.
Eur J Med Chem ; 138: 590-601, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28709125

ABSTRACT

As bacterial biofilms display extreme tolerance to conventional antibiotic treatments, it has become imperative to develop new antibacterial strategies with alternative mechanisms of action. Herein, we report the synthesis of a series of ciprofloxacin-nitroxide conjugates and their corresponding methoxyamine derivatives in high yield. This was achieved by linking various nitroxides or methoxyamines to the secondary amine of the piperazine ring of ciprofloxacin using amide bond coupling. Biological evaluation of the prepared compounds on preformed P. aeruginosa biofilms in flow cells revealed substantial dispersal with ciprofloxacin-nitroxide hybrid 25, and virtually complete killing and removal (94%) of established biofilms in the presence of ciprofloxacin-nitroxide hybrid 27. Compounds 25-28 were shown to be non-toxic in both human embryonic kidney 293 (HEK 293) cells and human muscle rhabdomyosarcoma (RD) cells at concentrations up to 40 µM. Significantly, these hybrids demonstrate the potential of antimicrobial-nitroxide agents to overcome the resistance of biofilms to antimicrobials via stimulation of biofilm dispersal or through direct cell killing.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Ciprofloxacin/pharmacology , Nitrogen Oxides/pharmacology , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cell Line, Tumor , Ciprofloxacin/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Microbial Sensitivity Tests , Molecular Structure , Nitrogen Oxides/chemistry , Structure-Activity Relationship
7.
mBio ; 8(1)2017 02 28.
Article in English | MEDLINE | ID: mdl-28246361

ABSTRACT

Only a few, relatively cumbersome animal models enable long-term Gram-negative bacterial infections that mimic human situations, where untreated infections can last for weeks. Here, we describe a simple murine cutaneous abscess model that enables chronic or progressive infections, depending on the subcutaneously injected bacterial strain. In this model, Pseudomonas aeruginosa cystic fibrosis epidemic isolate LESB58 caused localized high-density skin and soft tissue infections and necrotic skin lesions for up to 10 days but did not disseminate in either CD-1 or C57BL/6 mice. The model was adapted for use with four major Gram-negative nosocomial pathogens, Acinetobacter baumannii, Klebsiella pneumoniae, Enterobacter cloacae, and Escherichia coli This model enabled noninvasive imaging and tracking of lux-tagged bacteria, the influx of activated neutrophils, and production of reactive oxygen-nitrogen species at the infection site. Screening antimicrobials against high-density infections showed that local but not intravenous administration of gentamicin, ciprofloxacin, and meropenem significantly but incompletely reduced bacterial counts and superficial tissue dermonecrosis. Bacterial RNA isolated from the abscess tissue revealed that Pseudomonas genes involved in iron uptake, toxin production, surface lipopolysaccharide regulation, adherence, and lipase production were highly upregulated whereas phenazine production and expression of global activator gacA were downregulated. The model was validated for studying virulence using mutants of more-virulent P. aeruginosa strain PA14. Thus, mutants defective in flagella or motility, type III secretion, or siderophore biosynthesis were noninvasive and suppressed dermal necrosis in mice, while a strain with a mutation in the bfiS gene encoding a sensor kinase showed enhanced invasiveness and mortality in mice compared to controls infected with wild-type P. aeruginosa PA14.IMPORTANCE More than two-thirds of hospital infections are chronic or high-density biofilm infections and difficult to treat due to adaptive, multidrug resistance. Unfortunately, current models of chronic infection are technically challenging and difficult to track without sacrificing animals. Here we describe a model of chronic subcutaneous infection and abscess formation by medically important nosocomial Gram-negative pathogens that is simple and can be used for tracking infections by imaging, examining pathology and immune responses, testing antimicrobial treatments suitable for high-density bacterial infections, and studying virulence. We propose that this mouse model can be a game changer for modeling hard-to-treat Gram-negative bacterial chronic and skin infections.


Subject(s)
Abscess/pathology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Gram-Negative Bacteria/pathogenicity , Gram-Negative Bacterial Infections/pathology , Host-Pathogen Interactions , Skin Diseases, Bacterial/pathology , Abscess/drug therapy , Abscess/microbiology , Animals , Chronic Disease , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Mice , Mice, Inbred C57BL , Skin Diseases, Bacterial/drug therapy , Skin Diseases, Bacterial/microbiology , Treatment Outcome
8.
Emerg Top Life Sci ; 1(1): 41-53, 2017 Apr 21.
Article in English | MEDLINE | ID: mdl-33525815

ABSTRACT

Biofilms represent an adaptive lifestyle where microbes grow as structured aggregates in many different environments, e.g. on body surfaces and medical devices. They are a profound threat in medical (and industrial) settings and cause two-thirds of all infections. Biofilm bacteria are especially recalcitrant to common antibiotic treatments, demonstrating adaptive multidrug resistance. For this reason, novel methods to eradicate or prevent biofilm infections are greatly needed. Recent advances have been made in exploring alternative strategies that affect biofilm lifestyle, inhibit biofilm formation, degrade biofilm components and/or cause dispersal. As such, naturally derived compounds, molecules that interfere with bacterial signaling systems, anti-biofilm peptides and phages show great promise. Their implementation as either stand-alone drugs or complementary therapies has the potential to eradicate resilient biofilm infections. Additionally, altering the surface properties of indwelling medical devices through bioengineering approaches has been examined as a method for preventing biofilm formation. There is also a need for improving current biofilm detection methods since in vitro methods often do not accurately measure live bacteria in biofilms or mimic in vivo conditions. We propose that the design and development of novel compounds will be enabled by the improvement and use of appropriate in vitro and in vivo models.

9.
Methods Mol Biol ; 1548: 3-22, 2017.
Article in English | MEDLINE | ID: mdl-28013493

ABSTRACT

The "golden era" of antibiotic discovery has long passed, but the need for new antibiotics has never been greater due to the emerging threat of antibiotic resistance. This urgency to develop new antibiotics has motivated researchers to find new methods to combat pathogenic microorganisms resulting in a surge of research focused around antimicrobial peptides (AMPs; also termed host defense peptides) and their potential as therapeutics. During the past few decades, more than 2000 AMPs have been identified from a diverse range of organisms (animals, fungi, plants, and bacteria). While these AMPs share a number of common features and a limited number of structural motifs; their sequences, activities, and targets differ considerably. In addition to their antimicrobial effects, AMPs can also exhibit immunomodulatory, anti-biofilm, and anticancer activities. These diverse functions have spurred tremendous interest in research aimed at understanding the activity of AMPs, and various protocols have been described to assess different aspects of AMP function including screening and evaluating the activities of natural and synthetic AMPs, measuring interactions with membranes, optimizing peptide function, and scaling up peptide production. Here, we provide a general overview of AMPs and introduce some of the methodologies that have been used to advance AMP research.


Subject(s)
Anti-Infective Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Host-Pathogen Interactions , Immunomodulation , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biofilms/drug effects , Drug Design , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunologic Factors/chemistry , Immunologic Factors/genetics , Immunologic Factors/metabolism , Immunologic Factors/pharmacology , Immunomodulation/drug effects , Models, Molecular , Protein Conformation , Protein Domains , Protein Engineering , Structure-Activity Relationship
10.
EBioMedicine ; 12: 219-226, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27658736

ABSTRACT

Cutaneous abscess infections are difficult to treat with current therapies and alternatives to conventional antibiotics are needed. Understanding the regulatory mechanisms that govern abscess pathology should reveal therapeutic interventions for these recalcitrant infections. Here we demonstrated that the stringent stress response employed by bacteria to cope and adapt to environmental stressors was essential for the formation of lesions, but not bacterial growth, in a methicillin resistant Staphylococcus aureus (MRSA) cutaneous abscess mouse model. To pharmacologically confirm the role of the stringent response in abscess formation, a cationic peptide that causes rapid degradation of the stringent response mediator, guanosine tetraphosphate (ppGpp), was employed. The therapeutic application of this peptide strongly inhibited lesion formation in mice infected with Gram-positive MRSA and Gram-negative Pseudomonas aeruginosa. Overall, we provide insights into the mechanisms governing abscess formation and a paradigm for treating multidrug resistant cutaneous abscesses.


Subject(s)
Abscess/metabolism , Abscess/microbiology , Stress, Physiological , Abscess/drug therapy , Abscess/pathology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Bacterial Toxins/metabolism , Biofilms/drug effects , Disease Models, Animal , Female , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Microbial Sensitivity Tests , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases/metabolism
11.
Molecules ; 21(7)2016 Jun 27.
Article in English | MEDLINE | ID: mdl-27355936

ABSTRACT

As bacterial biofilms are often refractory to conventional antimicrobials, the need for alternative and/or novel strategies for the treatment of biofilm related infections has become of paramount importance. Herein, we report the synthesis of novel hybrid molecules comprised of two different hindered nitroxides linked to the piperazinyl secondary amine of ciprofloxacin via a tertiary amine linker achieved utilising reductive amination. The corresponding methoxyamine derivatives were prepared alongside their radical-containing counterparts as controls. Subsequent biological evaluation of the hybrid compounds on preformed P. aeruginosa flow cell biofilms divulged significant dispersal and eradication abilities for ciprofloxacin-nitroxide hybrid compound 10 (up to 95% eradication of mature biofilms at 40 µM). Importantly, these hybrids represent the first dual-action antimicrobial-nitroxide agents, which harness the dispersal properties of the nitroxide moiety to circumvent the well-known resistance of biofilms to treatment with antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Ciprofloxacin/chemistry , Ciprofloxacin/pharmacology , Nitric Oxide/chemistry , Anti-Bacterial Agents/chemical synthesis , Ciprofloxacin/chemical synthesis , Microbial Sensitivity Tests , Molecular Structure , Pseudomonas aeruginosa/drug effects
12.
Peptides ; 71: 276-85, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25836992

ABSTRACT

The recent observation that certain cationic peptides possess potent antibiofilm activity demonstrated that small peptides could be used to treat biofilm-associated infections. Other so-called innate defense regulator peptides possess potent immunomodulatory properties such as leukocyte recruitment and suppression of harmful inflammation. A peptide that directly targets biofilm cells while favorably modulating the immune response would be particularly advantageous for treating serious skin infections caused by Staphylococcus aureus. In the present work, using SPOT-synthesized peptide arrays on cellulose membranes, we outline a strategy for systematically assessing the antibiofilm activity of hundreds of IDR-1002 (VQRWLIVWRIRK-NH2) and IDR-HH2 (VQLRIRVAVIRA-NH2) peptide variants against MRSA biofilms. In addition, the ability of these peptides to stimulate production of a monocyte chemoattractant protein (MCP-1) and suppress LPS-induced interleukin (IL)-1ß production in human peripheral blood mononuclear cells (PBMCs) was evaluated. These results informed the synthesis of second-generation peptides resulting in a new peptide, IDR-2009 (KWRLLIRWRIQK-NH2), with enhanced MCP-1 stimulatory activity, favorable IL-1ß suppression characteristics and strong antibiofilm activity against MRSA and Pseudomonas aeruginosa biofilms. This work provides a proof-of-concept that multiple peptide activities can be optimized simultaneously to generate novel sequences that possess a variety of biological properties.


Subject(s)
Antimicrobial Cationic Peptides , Biofilms/drug effects , Immunologic Factors , Leukocytes, Mononuclear/immunology , Methicillin-Resistant Staphylococcus aureus/physiology , Pseudomonas aeruginosa/physiology , Antimicrobial Cationic Peptides/chemical synthesis , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Chemokine CCL2/immunology , Drug Evaluation, Preclinical , Humans , Immunologic Factors/chemical synthesis , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology
13.
Chem Biol ; 22(2): 196-205, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25699603

ABSTRACT

In many infections, bacteria form surface-associated communities known as biofilms that are substantially more resistant to antibiotics than their planktonic counterparts. Based on the design features of active antibiofilm peptides, we made a series of related 12-amino acid L-, D- and retro-inverso derivatives. Specific D-enantiomeric peptides were the most potent at inhibiting biofilm development and eradicating preformed biofilms of seven species of wild-type and multiply antibiotic-resistant Gram-negative pathogens. Moreover, these peptides showed strong synergy with conventional antibiotics, reducing the antibiotic concentrations required for complete biofilm inhibition by up to 64-fold. As shown previously for 1018, these D-amino acid peptides targeted the intracellular stringent response signal (p)ppGpp. The most potent peptides DJK-5 and DJK-6 protected invertebrates from lethal Pseudomonas aeruginosa infections and were considerably more active than a previously described L-amino acid peptide 1018. Thus, the protease-resistant peptides produced here were more effective both in vitro and in vivo.


Subject(s)
Anti-Infective Agents/chemistry , Oligopeptides/chemistry , Peptides/chemistry , Pseudomonas aeruginosa/physiology , Amino Acid Sequence , Animals , Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Biofilms/drug effects , Caenorhabditis elegans/drug effects , Drug Resistance, Bacterial/drug effects , Microbial Sensitivity Tests , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Peptides/pharmacology , Peptides/therapeutic use , Pseudomonas Infections/prevention & control , Pseudomonas Infections/veterinary , Stereoisomerism
14.
J Pept Sci ; 21(5): 323-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25358509

ABSTRACT

Host defense (antimicrobial) peptides, produced by all complex organisms, typically contain an abundance of positively charged and hydrophobic amino acid residues. A small synthetic peptide termed innate defense regulator (IDR-)1018 was derived by substantial modification of the bovine neutrophil host defense peptide bactenecin. Here, we review its intriguing properties that include anti-infective, anti-inflammatory, wound healing, and anti-biofilm activities. It was initially developed as an immune modulator with an ability to selectively enhance chemokine production and polarize cellular differentiation while suppressing/balancing the pro-inflammatory response. In this regard, it has demonstrated in vivo activity in murine models including enhancement of wound healing and an ability to protect against Staphylococcus aureus, multidrug resistant Mycobacterium tuberculosis, herpes virus, and inflammatory disorders, including cerebral malaria and neuronal damage in a pre-term birth model. More recently, IDR-1018 was shown, in a broad-spectrum fashion, to selectively target bacterial biofilms, which are adaptively resistant to many antibiotics and represent the most common growth state of bacteria in human infections. Furthermore, IDR-1018 demonstrated synergy with conventional antibiotics to both prevent biofilm formation and treat pre-existing biofilms. These data are consistent with a strong potential as an adjunctive therapy against antibiotic-resistant infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Biofilms/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Animals , Bacterial Infections/drug therapy , Cattle , Drug Synergism , Humans , Wound Healing/drug effects
15.
Trends Immunol ; 35(9): 443-50, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25113635

ABSTRACT

Although first studied for their antimicrobial activity, host defense peptides (HDPs) are now widely recognized for their multifunctional roles in both the innate and adaptive immune responses. Their diverse immunomodulatory capabilities include the modulation of pro- and anti-inflammatory responses, chemoattraction, enhancement of extracellular and intracellular bacterial killing, cellular differentiation and activation of the innate and adaptive compartments, wound-healing, and modulation of autophagy as well as apoptosis and pyroptosis. We review the various immunomodulatory roles of HDPs and their synthetic analogs, the innate defense regulators (IDRs). We discuss their potential as host-directed therapies, the hurdles they face in clinical development, and propose ways forward.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Immunomodulation , Molecular Targeted Therapy , Adaptive Immunity , Animals , Apoptosis , Autophagy , Chemotaxis , Humans , Immunity, Innate
16.
Antibiotics (Basel) ; 3(4): 509-26, 2014.
Article in English | MEDLINE | ID: mdl-26221537

ABSTRACT

Cystic fibrosis (CF) patients often acquire chronic respiratory tract infections due to Pseudomonas aeruginosa and Burkholderia cepacia complex (Bcc) species. In the CF lung, these bacteria grow as multicellular aggregates termed biofilms. Biofilms demonstrate increased (adaptive) resistance to conventional antibiotics, and there are currently no available biofilm-specific therapies. Using plastic adherent, hydroxyapatite and flow cell biofilm models coupled with confocal and scanning electron microscopy, it was demonstrated that an anti-biofilm peptide 1018 prevented biofilm formation, eradicated mature biofilms and killed biofilms formed by a wide range of P. aeruginosa and B. cenocepacia clinical isolates. New peptide derivatives were designed that, compared to their parent peptide 1018, showed similar or decreased anti-biofilm activity against P. aeruginosa biofilms, but increased activity against biofilms formed by the Gram-positive bacterium methicillin resistant Staphylococcus aureus. In addition, some of these new peptide derivatives retained the immunomodulatory activity of 1018 since they induced the production of the chemokine monocyte chemotactic protein-1 (MCP-1) and suppressed lipopolysaccharide-mediated tumor necrosis factor-α (TNF-α) production by human peripheral blood mononuclear cells (PBMC) and were non-toxic towards these cells. Peptide 1018 and its derivatives provide promising leads for the treatment of chronic biofilm infections and hyperinflammatory lung disease in CF patients.

SELECTION OF CITATIONS
SEARCH DETAIL
...