Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2012 Jan;56(1):378–390. doi: 10.1128/AAC.00689-11

The Oligo-Acyl Lysyl Antimicrobial Peptide C12K-2β12 Exhibits a Dual Mechanism of Action and Demonstrates Strong In Vivo Efficacy against Helicobacter pylori

Morris O Makobongo a, Hanan Gancz a, Beth M Carpenter a, Dennis P McDaniel b, D Scott Merrell a,
PMCID: PMC3256018  PMID: 22064541

Abstract

Helicobacter pylori has developed antimicrobial resistance to virtually all current antibiotics. Thus, there is a pressing need to develop new anti-H. pylori therapies. We recently described a novel oligo-acyl-lysyl (OAK) antimicrobial peptidomimetic, C12K-2β12, that shows potent in vitro bactericidal activity against H. pylori. Herein, we define the mechanism of action and evaluate the in vivo efficacy of C12K-2β12 against H. pylori after experimental infection of Mongolian gerbils. We demonstrate using a 1-N-phenylnaphthylamine (fluorescent probe) uptake assay and electron microscopy that C12K-2β12 rapidly permeabilizes the bacterial membrane and creates pores that cause bacterial cell lysis. Furthermore, using nucleic acid binding assays, Western blots, and confocal microscopy, we show that C12K-2β12 can cross the bacterial membranes into the cytoplasm and tightly bind to bacterial DNA, RNA, and proteins, a property that may result in inhibition of enzymatic activities and macromolecule synthesis. To define the in vivo efficacy of C12K-2β12, H. pylori-infected gerbils were orogastrically treated with increasing doses and concentrations of C12K-2β12 1 day or 1 week postinfection. The efficacy of C12K-2β12 was strongest in animals that received the largest number of doses at the highest concentration, indicating dose-dependent activity of the peptide (P < 0.001 by analysis of variance [ANOVA]) regardless of the timing of the treatment with C12K-2β12. Overall, our results demonstrate a dual mode of action of C12K-2β12 against the H. pylori membrane and cytoplasmic components. Moreover, and consistent with the previously reported in vitro efficacy, C12K-2β12 shows significant in vivo efficacy against H. pylori when used as monotherapy. Therefore, OAK peptides may be a valuable resource for therapeutic treatment of H. pylori infection.

INTRODUCTION

Helicobacter pylori is a spiral-shaped, flagellated, Gram-negative bacterium that has adapted to infect the human gastric mucosa. Persistent infection leads to the development of gastroduodenal diseases that include chronic gastritis, peptic ulcer disease, and gastric cancer (31). H. pylori is carried by half of the world's population, and its prevalence is estimated to exceed 90% in some developing countries. In the United States, it is estimated that 30 to 40% of the population is infected with H. pylori (8). In addition, individuals infected with H. pylori are at a greater risk of developing gastric cancer (33). This fact led the World Health Organization (WHO) International Agency for Research on Cancer to classify H. pylori as a group I carcinogen (9). Given the association with gastric disease, together with complicated diagnosis procedures and aggregate health care expenditures, the annual health care costs attributed to H. pylori infection are estimated at about $10 billion in the United States alone (8, 66). Thus, H. pylori infection presents a major global public health burden, and eradication of the bacterium from infected symptomatic individuals is still the preferred choice of treatment and management.

In the 3 decades since the discovery of the bacterium, treatment strategies for H. pylori infection have undergone tremendous evolutionary changes that span from monotherapy to multidrug therapy for extended periods of treatment (15). Though initially favored, monotherapy and dual therapy for 4 to 7 days were the first treatments to be shown to result in eradication failure (21). As a result, treatment strategies currently involve combination therapies administered for 10 to 14 days and include triple, quadruple, and sequential therapies.

Despite complex treatment strategies, during the last 4 to 6 years, the efficacy of triple and quadruple therapies has been reduced dramatically, and eradication rates of below 50 to 75% have been reported in some areas (21, 25). Eradication failure is mainly attributed to the development of antimicrobial resistance and to noncompliance by patients who do not adhere to the complicated and sometimes side effect-inducing multidrug therapy. Given the immense challenge in rising antimicrobial resistance (36), there is a need for new antibiotics for the treatment of H. pylori infection. Moreover, given the complicated nature of the H. pylori treatment regimens, it is envisioned that drugs that could be used as monotherapy or in combination with new classes of antibiotics would be the most helpful.

Recently, antimicrobial peptides (AMPs) have been scrutinized as potential novel antimicrobial agents against human pathogens (63, 68). AMPs are usually small molecules comprised of 12 to 60 amino acids with molecular masses of 10 kDa or less. However, much bigger polypeptides have recently been discovered; examples are lactoffericin (12), kinocidins (microbicidal chemokines) (55), and complement-derived peptides (82). AMPs display immense diversity in sequence, secondary structural motifs, charge, and/or the abundance of certain specific amino acids (72). However, the formation of amphipathic structures and the presence of multiple basic amino acid residues are highly conserved (14, 74) and are the characteristic features that endow cationicity and enable the AMPs to interact selectively with anionic bacterial membranes.

The use of AMPs as potential therapeutics has been particularly attractive because of their ability to be promptly synthesized by the host and their capacity to lyse the cell membranes of pathogens via direct interaction. However, several studies have demonstrated that pathogens are able to elude the effects of natural AMPs by production of proteolytic peptidases (24, 75), by recognition and extracellular capture of the AMPs, by active extrusion of AMPs from the bacterial cell wall (35), or by reduction of the net anionic charge of the bacterial cell envelope (1, 59, 61). Thus, research has turned to the production of synthetic AMPs in order to circumvent the possibility of microbial resistance. Oligomers of acylated lysines (oligo-acyl-lysyls [OAKs]) are novel synthetic peptidomimetics that consist of alternating amino acyl chains and cationic amino acids that are arranged to create an optimal molecular charge and hydrophobicity for enhanced potency (65, 71). Like most natural AMPs, the OAKs are cationic and form amphipathic structures that associate with one another to protect the hydrophobic side and expose the cations to interact with the bacterial membrane (87). However, in contrast to natural AMPs, the OAK design allows for the formation of a simple structure, which allows for the establishment of structure-activity relationships and prevents the formation of secondary structures (65). These properties are in turn associated with the lack of development of antimicrobial resistance (37, 65). In addition, the OAK peptides are resistant to bacterial enzymatic degradation (13, 67) and exhibit no or minimal cytotoxicity to mammalian cells (64, 67). Furthermore, their structures can be fine tuned to optimize antibacterial activity (67), and they have been shown to possess significant in vivo efficacy against Escherichia coli (65, 67) and Staphylococcus aureus (43, 70) in mouse models of infection. Thus, OAKs display characteristic features that are attractive for the development of a potent new class of therapeutic drugs.

We recently demonstrated the strong in vitro efficacy of the OAK C12K-2β12 against H. pylori (47). C12K-2β12 showed significant postantibiotic effects (PAE) and significant thermal and pH stability, suggesting that it might be a potential new therapeutic agent. Given the strong in vitro efficacy we previously reported, here we addressed two major questions. (i) How does C12K-2β12 exert its effect on the H. pylori cell? (ii) Could monotreatment with C12K-2β12 be used to decrease the H. pylori colonization load in an animal model of infection? Collectively, our data demonstrate that C12K-2β12 exerts its bactericidal effects through a dual mode of action; at low concentrations, C12K-2β12 binds nucleic acids and proteins, while at higher concentrations, the OAK extensively disrupts the bacterial membrane leading to cell lysis by pore formation. Furthermore, we found that monotherapeutic dosing with C12K-2β12 resulted in significant clearance of H. pylori in the gerbil model of infection. These results suggest that C12K-2β12 and other OAK peptides may represent a potential new class of antibiotics for treatment of H. pylori infection.

MATERIALS AND METHODS

Peptide synthesis, reagents, and antibiotics.

The C12K-2β12 and K-5α12 oligo-acyl-lysyls (OAKs) used in this study were synthesized as previously described (47, 6365). K-5α12 is a control OAK peptide that has no antibacterial activity (data not shown). The composition of purified peptides was confirmed using amino acid analysis and electrospray mass spectrometry. Peptides were stored as lyophilized powder at −20°C. Before each experiment, a fresh solution containing the appropriate concentration of peptide was reconstituted from a stock solution (1 mg/ml) that was prepared by two cycles, with one cycle consisting of 2 min of sonication in a water bath followed by brief vortexing. The fresh peptide solution was centrifuged and then used in subsequent efficacy studies and mechanism of action experiments. Amoxicillin (Sigma, St. Louis, MO) and vancomycin (USB Corporation, Cleveland, OH) were reconstituted according to the manufacturer's instructions and used at the indicated concentrations.

Bacterial strains and growth conditions.

H. pylori strains G27 (17) and 7.13 (18) were used to determine the C12K-2β12 mechanism of action and to test for in vivo efficacy, respectively. Both strains were cryopreserved at −80°C in brain heart infusion medium supplemented with 20% glycerol and 10% fetal bovine serum (FBS). Bacteria were cultured under microaerobic and medium conditions as previously described (47), and the cultures were maintained with shaking in brucella broth (Neogen-Acumedia) containing 10 μg/ml vancomycin (Sigma) and 10% fetal bovine serum (Invitrogen Gibco-BRL, Carlsbad, CA) in a microaerobic environment. An Anoxomat (Spiral Biotech, Norwood, MA) was used to achieve the microaerobic atmospheric condition of 5% O2, 10% CO2, and 85% N2 gas mixture.

Determination of membrane perturbation and translocation into bacterial cytoplasm by C12K-2β12. (i) NPN membrane permeation assay.

The ability of C12K-2β12 to permeabilize the bacterial membrane was determined using intact H. pylori cells and 1-N-phenylnaphthylamine (NPN). NPN (Sigma) is a membrane potential-sensitive fluorescent probe that shows increased fluorescence upon exposure to a hydrophobic environment (44). Briefly, a starter culture of H. pylori G27 was grown for 16 to 18 h (optical density at 600 nm [OD600] of 0.4 to 0.5) as described above. At this point, the starter culture was used to inoculate independent test cultures to an OD600 of 0.05. These cultures were then incubated for 6 to 8 h in the presence or absence of C12K-2β12 (5 μM, 10 μM, 20 μM, 40 μM, and 80 μM) or amoxicillin (100 μM). Approximately 5 × 106 bacterial cells were then sampled from each culture to determine NPN uptake at 15 min, 30 min, 1 h, 4 h, and 6 h. The cells were washed and resuspended in PBS (pH 7.2) to achieve a final volume of 250 μl in the presence of 22 μg/ml of NPN. Cells grown in the absence of drugs and 80 μM C12K-2β12 resuspended in phosphate-buffered saline (PBS) alone in the absence of H. pylori cells were used as controls. The NPN fluorescence intensity emitted (excitation λ [λex] of 350 nm, emission λ [λem] of 420 nm, and a slit width of 5 nm) was recorded after 3 min as arbitrary units (AU) on a Spectramax M2 spectrofluorimeter (Molecular Devices, Sunnyvale, CA) and was considered a correlate of membrane potential and permeation. The NPN assays were performed at room temperature and were repeated three times. Additionally, to determine the kinetics of H. pylori death in relation to membrane permeation, samples exposed to 20 μM C12K-2β12 were plated to determine the viable CFU in relation to NPN fluorescence.

(ii) Electron microscopy studies.

To investigate whether C12K-2β12 affected the overall cell morphology and to visualize the bacterial membrane, we studied bacterial ultrastructure using transmission electron microscopy (TEM). Briefly, a starter culture of H. pylori G27 was grown for 16 to 18 h (OD600 of 0.4 to 0.5) in liquid medium as described above. At this point, the starter culture was used to inoculate independent test cultures at an OD600 of 0.05. These cultures were then incubated with 20 μM K-5α12 (control peptide that has no antibacterial activity; data not shown), 20 μM C12K-2β12, 100 μM amoxicillin, or PBS as an untreated control. At 2 h and 16 h of culture, approximately 1.5 × 107 bacterial cells were collected and subsequently washed twice in PBS by centrifugation and resuspension. The cells were then fixed and processed for TEM using previously described methods (50, 85) with minor modifications. Briefly, the cells were exposed to dehydration in a graduated series of ethanol followed by resin polymerization at 65°C for 3 days. A Leica EM UC6 ultramicrotome was used to slice thin sections of 70- to 80-nm thickness from the polymerized blocks, which were then loaded on 400-mesh copper grids. The sections were poststained with 2% uranyl acetate for 15 min and Reynold's lead citrate for 5 min before being mounted for observation on a Philips CM100 transmission electron microscope (FEI Company, Hillsboro, OR) operating at 80 keV. Images were recorded with a 4MP SPOT Insight charge-coupled-device (CCD) camera (Diagnostic Instruments Inc., Sterling Heights, MI) using SPOT Insight software (Diagnostic Instruments). The morphology of the cells was observed at low-power magnification (×13,500), and the cell wall ultrastructure was observed at high-power magnification (×64,000 or ×130,000). Using the digital camera, images were acquired in the thin sections randomly across multiple grids. Each grid was examined in a similar systematic manner.

(iii) Confocal microscopy studies.

In order to determine whether C12K-2β12 had the ability to traverse the bacterial membrane and enter the cytoplasm, DyLight 488 fluorescent dye (Thermo Fisher Scientific-Pierce Biotechnology, Rockford, IL) was conjugated to C12K-2β12. Labeled peptide was purified to remove unreacted dye according to the manufacturer's protocol in the DyLight 488 antibody labeling kit. It is noteworthy that fluorescent labeling did not affect the bactericidal activity of the peptide. Bacterial samples were inoculated into 2-ml liquid cultures at an OD600 of 0.05 and then were incubated with 20 μM, 40 μM, or 80 μM DyLight 488-conjugated C12K-2β12 for 30 min. Bacterial cell samples incubated with unconjugated DyLight 488 or PBS were used as controls. The bacterial cells were washed twice in PBS (pH 6.5) and resuspended in 2% paraformaldehyde–PBS fixative followed by incubation in the dark for 30 min on ice. The bacterial cells were then washed twice to remove traces of fixative, and the DNA was stained with 4′,6′-diamidino-2-phenylindole (DAPI) for 20 min to mark the bacterial cytoplasm. The cells were then resuspended in PBS and spotted onto gelatin-coated slides treated with vector-shield mounting reagent. Fluorescent images of bacterial cells were visualized at a magnification of ×100, and confocal images were taken with a Zeiss LSM 710 confocal microscope (Carl Zeiss MicroImaging, LLC, Thornwood, NY) using two lasers with peak wavelengths allowing excitation of green and blue fluorochromes. The images were processed and analyzed with Zeiss LSM Image 5 Browser software.

Determination of C12K-2β12 interaction with bacterial nucleic acids and proteins.

All experiments for peptide-nucleic acid interactions were performed with peptides that were preheated to 95°C.

(i) Binding to DNA.

The ability of C12K-2β12 to bind DNA was investigated by incubation of the peptide with 32P-labeled (end-labeled) DNA probes, followed by analysis of subsequent DNA mobility using electrophoretic mobility shift assay (EMSA) gels as previously described (5, 20). Briefly, regions of the oorD and rpoB promoters were PCR amplified from the H. pylori G27 strain, and the PCR products were purified by gel extraction. Additionally, the E. coli chloramphenicol acetyltransferase gene and human antibody light-chain DNA fragment were included to test the specificity of the binding.

For the EMSA, 1 ng of an end-labeled DNA fragment was mixed with C12K-2β12 in a final volume of 20 μl binding buffer (24% glycerol, 40 mM Tris [pH 8.0], 150 mM KCl, 2 mM dithiothreitol [DTT], 600 μg/ml bovine serum albumin, 200 μM EDTA, and 0.1 mg/ml sheared salmon sperm DNA). Final OAK peptide concentrations of 1.25 μM, 2.5 μM, 5 μM, 10 μM, and 20 μM C12K-2β12 were used. A tube containing no peptide and a tube containing 20 μM K-5α12 control peptide were included as control reactions. All reaction mixtures were incubated at 37°C for 1 h. Following incubation, the reaction products were separated on a 5% polyacrylamide gel (5% 19:1 acrylamide, Tris-glycine buffer, 2.5% glycerol) at 100 V in Tris-glycine buffer. The gels were then exposed to a phosphor screen and scanned on a FLA-5100 multifunctional phosphorimager (Fuji Medical Systems, Hanover Park, IL). Analysis was performed using Multi-Gauge version 3.0 software (Fujifilm Medical Systems). The reported results are representative of five independent experiments.

(ii) Binding to RNA.

Similar to binding to DNA, the ability of C12K-2β12 to bind to RNA was determined by examining changes in the electrophoretic mobility of ureA or oorD RNA probes that were preincubated with C12K-2β12. RNA riboprobes labeled with [α-32P]UTP (Perkin Elmer) were synthesized by in vitro transcription as previously described (6, 78). Briefly, 1 μg of ureA or oorD RNA riboprobes were mixed with C12K-2β12 (0 μg, 1 μg, or 5 μg) or with 5 μg of K-5α12 in a final volume of 50 μl. The reaction mixtures were incubated at 37°C for 1 h. The mixtures were then resolved on 5% acrylamide–Tris-borate-EDTA–8 M urea denaturing gels to determine the mobility of the RNA riboprobes. The gels were exposed to phosphor screens, which were subsequently scanned using a FLA-5100 multifunctional scanner (Fujifilm Medical Systems). The images were analyzed using Multi-Gauge software version 3.0 (Fujifilm Medical Systems). The reported results are representative of three independent experiments.

(iii) Binding to H. pylori proteins by SDS-PAGE and Western blot analysis.

The ability of the C12K-2β12 peptide to bind to H. pylori proteins was investigated using a modified Western blot protocol where blots were probed with DyLight 488-conjugated C12K-2β12. Briefly, bacterial cell lysates were prepared from liquid cultured bacterial pellets using lysis buffer (150 mM NaCl, 50 mM Tris-HCl [pH 8.0], 5 mM EDTA, 1% sodium dodecyl sulfate [SDS], 10% glycerol containing protease inhibitor cocktail [Roche]). Proteins from these lysates, as well as purified recombinant H. pylori Fur protein, were separated via SDS-polyacrylamide gel electrophoresis (PAGE) using a 4% stacking gel and an 18% separating gel and were then electrotransferred onto a nitrocellulose membrane by semidry blotting (Owl separation system; ThermoScientific). After transfer, residual proteins in the gel were detected with Coomassie brilliant blue R250. The membranes were washed in PBS, blocked for 1 h in PBS containing 3% bovine serum albumin, and then washed again 3 times for 5 min each time in PBS containing 0.05% Tween 20 (PBS-T). The membranes were then probed with 20 μM DyLight 488-conjugated C12K-2β12 (ThermoScientific/Pierce), and direct protein–C12K-2β12 binding was revealed by fluorescence analysis using a LAS-3000 instrument with LAS-3000 Lite capture software (FujiFilm).

Functional inhibition of restriction digestion by C12K-2β12.

The ability of C12K-2β12 to block restriction enzyme activity via DNA binding was examined using the 10,031-bp endogenous plasmid from H. pylori G27 and five restriction enzymes predicted to cut multiple times within the plasmid sequence. Briefly, the H. pylori G27 endogenous plasmid was isolated using the QIAprep spin miniprep kit (Qiagen). For each restriction enzyme examined (MfeI, HindIII, EcoRI, SpeI, and XmnI [New England BioLabs]), two reaction mixtures containing 500 ng plasmid were preincubated in the presence or absence of 40 μM C12K-2β12 at 37°C for 1 h in a total volume of 20 μl. Plasmids preincubated with 40 μM or 80 μM K-5α12 were also included as controls. Following incubation, the appropriate restriction enzyme buffer was added, and the reaction mixtures were split such that one half of each tube was exposed to 5,000 U of MfeI, HindIII, EcoRI, SpeRI, or XmnI, and the second half of each tube was treated as an undigested native control. The digestion reaction mixtures were incubated at 37°C for 1 h, and the plasmid DNA was analyzed on a 1% agarose gel stained with 5 μg/ml ethidium bromide (Sigma). The experiments were repeated three times.

Determination of the in vivo efficacy of C12K-2β12 in H. pylori-infected gerbils.

A total of 90 animals that had been allowed water but no food for 12 h were inoculated via intragastric gavage with approximately 8 × 108 CFU of H. pylori. Five animals were then assigned to a total of 18 possible groups. Briefly, animals were grouped into two major categories: (i) animals treated with C12K-2β12 1 day after H. pylori infection (n = 45) and (ii) animals treated with C12K-2β12 7 days postinfection (n = 45). Each of these two categories was further subdivided into three dose treatment groups based on the following C12K-2β12 dose schedule: (i) those given a single dose (n = 15), (ii) those given three sequential once daily doses (n = 15), and (iii) those given five sequential once daily doses of the peptide (n = 15). Each dose treatment group was additionally divided into three regimen treatment groups: (i) those treated with 1 mg C12K-2β12/kg body weight (n = 5), (ii) those treated with 4 mg C12K-2β12/kg body weight (n = 5), and (iii) those treated with PBS rather than the peptide. The animals were weighed immediately before dosing, and all treatments were orogastrically administered following a 12-hour fasting period during which animals were allowed water. One week after the last treatment, the animals were sacrificed, and their stomachs were harvested, weighed, and homogenized. Stomach homogenates were then plated to determine H. pylori CFU per gram of stomach tissue. Animal experiments were carried out according to strict guidelines and recommendations in the Guide for the Care and Use of Laboratory Animals (54a), and the study protocols were approved by the Uniformed Services University of Health Sciences Institutional Animal Care and Use Committee (approved protocol numbers MIC-10-774 and MIC-07-503).

Statistical analysis.

To determine whether a significant difference in the number of colonizing bacteria existed after C12K-2β12 treatment of H. pylori-challenged gerbils, colonization levels across various peptide treatment groups were analyzed using one-way analysis of variance (ANOVA) analysis. The ANOVA analyses were completed using SPSS 16.0 statistical software (SPSS Inc., IBM Corporation, Somers, NY). Where significant treatment effects were observed (P < 0.05), the least significant difference (LSD) analyses were computed to permit separation and comparison of the means of the C12K-2β12-treated groups and the corresponding PBS control. A P value of <0.05 was considered a significant difference.

RESULTS

C12K-2β12 rapidly permeabilizes the H. pylori membrane and triggers bactericidal effects.

The mechanisms by which antimicrobial agents act are varied and may involve membrane disruption or functional inhibition of key pathways that are essential for microbial growth and/or survival. Cationic antimicrobial peptides have been shown to target and disrupt bacterial membranes and/or interact with nucleic acids in a manner that could potentially block macromolecule synthesis (19, 57, 69). Thus, to determine whether the effect of the C12K-2β12 OAK peptide on H. pylori involved disruption of the cell membrane, we assessed membrane integrity using 1-N-phenylnaphthylamine (NPN). NPN is a neutral hydrophobic fluorescent probe whose quantum yield increases in hydrophobic environments compared to aqueous environments (44). Normally, NPN is excluded by the intact bacterial cell membrane. However, when the outer membrane (OM) structure is damaged, NPN can partition into the hydrophobic interior of the OM or plasma membrane, leading to a dramatic increase in fluorescence. Therefore, we used changes in NPN fluorescence intensity as a direct indicator of cell membrane permeability (26).

Incubation of H. pylori with 5 μM C12K-2β12, which is below the previously demonstrated MIC of 6.5 to 7.0 μM for the G27 strain, resulted in a minor change in NPN fluorescence that was similar to the control H. pylori exposed to PBS (Fig. 1A). However, utilization of concentrations of C12K-2β12 above the MIC resulted in significant changes in fluorescence as early as 15 min after exposure to the peptide. This response was dose dependent, since increasing concentrations of C12K-2β12 (5 μM, 10 μM, 20 μM, 40 μM, and 80 μM) resulted in a more rapid increase in NPN fluorescence, and it was specific to C12K-2β12 interaction with H. pylori, since utilization of related OAK peptides that do not affect H. pylori growth or survival, 2β12 (47) and K-5α12 (data not shown), did not affect NPN fluorescence. Incubation of C12K-2β12 alone without H. pylori cells also did not affect NPN fluorescence, indicating no or minimal C12K-2β12 autofluorescence. However, the response could be mimicked through the addition of amoxicillin (AMX), which acts by inhibiting cell wall mucopeptide synthesis and is known to disrupt bacterial membrane integrity through inhibition of peptidoglycan polymerization (2). It is noteworthy that 10 μM C12K-2β12 showed greater and more rapid membrane permeabilization effects than a 10-fold higher concentration of AMX (100 μM), consistent with the previously reported superior effects of C12K-2β12 over AMX (47). To determine whether membrane permeation was coincident with or a consequence of bacterial death, the NPN assay was repeated, and the bacteria were monitored by plating. As shown in Fig. 1B, treatment of H. pylori with 20 μM C12K-2β12 resulted in dramatic increases in NPN fluorescence prior to significant killing; therefore, membrane permeation precedes cell death. Overall, these data suggest that C12K-2β12 has the ability to rapidly disrupt the bacterial outer membrane and indicate that our previously demonstrated in vitro killing of H. pylori is likely due to induction of bacterial membrane damage.

Fig 1.

Fig 1

C12K-2β12 rapidly permeabilizes the H. pylori membrane in a dose-dependent manner that correlates with bactericidal activity. (A) Membrane permeabilization was measured by increased fluorescence due to uptake of 1-N-phenylnaphthylamine (NPN), and (B) a parallel bactericidal assay was performed with 20 μM C12K-2β12. H. pylori (Hp) G27 was cultured for 24 h in the presence of C12K-2β12 (5 μM, 10 μM, 20 μM, 40 μM, and 80 μM) or amoxicillin (100 μM). Cells grown in the absence of drugs but exposed to PBS or 80 μM C12K-2β12 alone (without cells) were used to determine background fluorescence. Approximately 5 × 106 bacterial cells were sampled at 15 min, 30 min, 1 h, 4 h, and 6 h to determine NPN uptake and bactericidal activity. NPN fluorescence intensity emitted (excitation λ of 350 nm and emission λ of 420 nm) was recorded in arbitrary units (AU) as a correlate of membrane permeation. Bactericidal activity was expressed as percent survival of starting CFU. The data shown are representative of five independent experiments. In panel B, NPN fluorescence is shown on the left y axis and depicted by lines, while percent survival is shown on the right y axis and indicated by the bars.

C12K-2β12 dramatically disrupts H. pylori membranes, causes lysis through pore formation, and traverses into the cytoplasm.

The dose-dependent increase in NPN fluorescence (Fig. 1) suggested that C12K-2β12 was inducing membrane damage. To further confirm and more thoroughly investigate this possibility, we next examined the ultrastructural morphology of the bacterial cell after C12K-2β12 exposure using transmission electron microscopy (TEM). PBS-treated cells, cells treated with the OAK peptide K-5α12 (47), and cells treated with amoxicillin were included as controls. H. pylori cells were exposed to the indicated treatments for 2 or 16 h. The cells were then harvested, fixed, stained with uranyl acetate, and observed as thin sections (70 to 80 nm). As shown in electron micrographs taken at low magnification (×13,500) 16 h posttreatment (Fig. 2), no dramatic differences in cell morphology were observed for PBS-treated bacterial cells (Fig. 2A) or those exposed to the K-5α12 control peptide (Fig. 2C). However, amoxicillin-treated H. pylori cells developed an enlarged coccoid morphology (Fig. 2B). These enlarged cells were typified by clear separation of the plasma membrane from the outer membrane. Additionally, 5 to 10% of the cells appeared as ghost cells, which showed a complete loss of cytoplasmic contents and appeared as empty cell membrane outlines similar to previously described H. pylori ghost cells (56). Save for the appearance of the ghost cells, these data are consistent with the previously reported effects of amoxicillin on H. pylori (2); the minor difference is likely due to amoxicillin concentration differences.

Fig 2.

Fig 2

Thin-section transmission electron micrograph of H. pylori G27 reveals dramatic changes in morphology following 16-h treatment with C12K-2β12. Approximately 1.5 × 107 H. pylori cells were sampled following 16 h of exposure to PBS (A), 100 μM amoxicillin (B), 20 μM K-5α12 (C), or 20 μM C12K-2β12 (D). The cells were examined at a low magnification (×13,500) on a transmission electron microscope (TEM) equipped with a 4MP SPOT Insight CCD camera. (A and C) Normal spiral- or comma-shaped rod morphology of H. pylori cells and intact cell membranes following treatment with PBS and the K-5α12 control peptide (negative controls), respectively. (B) Amoxicillin-treated cells show characteristically swollen cells (white arrows) and detachment of the inner membrane from the outer membrane (white arrowhead). (D) C12K-2β12-treated cells show the appearance of numerous ghost cells (white arrows) and the formation of electron-dense structures (white arrowhead) within the extracellular medium and inside the cells. The data are representative images from two independent experiments.

H. pylori samples that were exposed to the C12K-2β12 OAK peptide (Fig. 2D) showed a significantly altered morphological phenotype compared to control cells. Approximately 20 to 35% of the C12K-2β12-treated cells appeared as ghost cells, and the media and intact cells became filled with extracellular electron-dense structures. The appearance of the electron-dense structures, which were absent in PBS- or K-5α12-treated samples, is consistent with extravasation of the bacterial cytoplasmic contents due to cell lysis and/or the presence of membrane pores. Furthermore, the appearance of the electron-dense material in nonghost intact H. pylori cells suggested translocation of C12K-2β12 into the bacterial cytoplasm and the subsequent formation of aggregate molecules with the cytoplasmic contents. Additionally, treatment of cells with C12K-2β12 resulted in changes in overall bacterial shape; the typical spiral and comma rod-shaped bacterial cells seen in the controls (Fig. 2A and C) were not observed in C12K-2β12-treated cells (Fig. 2D). Instead, consistent with modification of the cell wall and/or cytoskeleton structure (2), these cells became coccoid. The notable lack of intracellular and extracellular electron-dense structures in amoxicillin-treated control H. pylori cells and the presence of significantly higher numbers of ghost cells in the C12K-2β12-treated samples suggest differences in the modes of action of the two drugs. In order to better visualize and analyze the ultrastructural morphology of the cell wall and cytoplasmic changes observed at lower magnification (×13,500; Fig. 2), we additionally examined each sample at higher magnification following treatment of bacteria with the peptide for 16 h. We observed extensive damage of the cell wall, and these results are presented as Fig. S1 in the supplemental material.

The formation of ghost cells by the C12K-2β12-treated H. pylori samples suggested that the OAK peptide was inducing the formation of pores in the bacterial membrane that allowed cellular contents to leak out. However, no visible pores in the cell walls were observed at the 16-hour time point (see Fig. S1 in the supplemental material). Given the fast killing rate of C12K-2β12, the observation that no viable bacteria were evident after treatment with C12K-2β12 for 6 to 8 h (47), and the rapid permeation of the membrane demonstrated by the NPN assays (Fig. 1), we hypothesized that the extent of cell damage was too great at the late time point to see changes in the membrane. Thus, in order to investigate the early events related to treatment of H. pylori with C12K-2β12, we next used TEM to examine the ultrastructure of the cells harvested 2 h posttreatment with the OAK peptide. Analysis of low-magnification (×13,500) images revealed that 4 to 8% of the H. pylori samples appeared as ghost cells at this early time point (Fig. 3A). Additionally, we visualized the presence of intracellular and extracellular electron-dense structures, as well as less dramatic changes in cell morphology. The C12K-2β12 effects at this early time point are consistent with the observed NPN membrane permeabilization within 15 to 30 min (Fig. 1) and our previously reported in vitro killing kinetics of 90% of the bacterial population within 2 to 3 h of OAK treatment (47). At higher magnifications (×64,000 and ×130,000), we visualized extensive membrane perturbation of H. pylori following two hours of exposure to C12K-2β12 (Fig. 3B, C, and D). Consistent with the appearance of ghost cells, cell lysis, and leakage of cellular contents observed at later time points (Fig. 2D), membrane damage could be clearly visualized by 2 h after exposure to the OAK peptide. Outer membrane damage was evidenced as the formation of pore-like structures with a width measured between 50 nm (Fig. 3B, black arrows) and over 400 nm (Fig. 3C, black arrowheads). In addition, we observed large areas of membrane sloughing (S), which led to complete separation and peeling away of the OM from the plasma membrane (Fig. 3D). Taken en masse, these data indicate that the C12K-2β12 OAK peptide induces H. pylori cell death through dramatic disruption of the bacterial cell membrane, which in turn leads to leakage of the cytoplasmic contents and bacterial cell lysis.

Fig 3.

Fig 3

Distinct classical pores and sloughing of the outer membrane of C12K-2β12-treated H. pylori cells as visualized with a TEM. H. pylori G27 was incubated with 20 μM C12K-2β12 for 2 h. (A) TEM analysis at a low magnification (×13,500) shows morphological changes at this early time point. (B to D) Representative images showing H. pylori cells observed at a higher magnification (×130,000). The black arrows in panel B indicate the loss of a small section of outer membrane of the bacterium, while the black arrowheads in panel C indicate the borders of a large 400-nm section of missing membrane. In panel D, sloughing (S) of a large section of the outer membrane and electron-dense structures (E) inside the cell cytoplasm are indicated. The data are representative results from two independent experiments.

There is evidence that several cationic antimicrobial peptides exert their effects through interaction with intracellular nucleic acids (4, 32). Moreover, it has been reported that the hexamer OAK peptide C12K-5α8 is able to traverse the bacterial membrane and to bind to bacterial DNA without overt disruption of the cell wall (67). Given that we saw virtually no change in NPN fluorescence at low concentrations of C12K-2β12 (Fig. 1A), we next considered the possibility that at lower concentrations, C12K-2β12 might have the ability to permeate the bacterial cell membrane into the cytoplasm. Since we observed little immediate killing of samples treated with 20 μM C12K-2β12 (Fig. 1B), we utilized confocal microscopy and this concentration of fluorescently labeled C12K-2β12 to study localization of the peptide 30 min after exposure. Analysis of single-plane sections through the center of the bacteria (Fig. 4A) and series of z-stack images taken from the surface of the bacterial cell moving through the interior of the cell (Fig. 4B) revealed that little C12K-2β12 was visible near the cell surface but was instead colocalized with bacterial cytoplasmic DNA prior to cell death. These data indicate that the peptide has the ability to cross the bacterial membrane and enter the cytosol.

Fig 4.

Fig 4

DyLight 488-conjugated C12K-2β12 rapidly colocalizes with DNA in the bacterial cytoplasm upon treatment of H. pylori with the peptide. Bacterial cells were treated with DyLight 488-conjugated C12K-2β12 (green) for 30 min. The cells were washed and fixed, and cytoplasmic DNA was stained with DAPI (blue). The cells were then visualized by confocal microscopy. Images represent a single plane taken through the center of the bacterium (A) or six sequential z-stack slices starting near the surface of bacteria and moving toward the middle (B). Both verify the presence of C12K-2β12 in the bacterial cytoplasm, as significant green fluorescence is seen only within the cell center. Differential interference contrast (DIC) and overlay images are included for clarity. All images were taken with a magnification of ×400. The images are representative of three experiments.

C12K-2β12 binds to bacterial nucleic acids (DNA and RNA) and proteins.

Given that C12K-2β12 had the ability to traverse into the cytoplasm and appeared to colocalize with DNA, we hypothesized that C12K-2β12 may directly interact with cytoplasmic nucleic acids and proteins. To determine whether C12K-2β12 could interact with DNA, we first investigated whether preincubation of DNA with the peptide could prevent restriction enzyme cleavage of the H. pylori G27 endogenous plasmid. Undigested plasmid and plasmid preincubated with K-5α12 were included as negative controls. As shown in Fig. 5, undigested native plasmid ran as two bands (nicked and supercoiled species) (lane 2), and digestion with the MfeI restriction enzyme (lane 4) resulted in the appearance of linearized bands. The overall pattern of plasmid migration was slightly affected by preincubation with C12K-2β12 (Fig. 5, lane 3); some DNA failed to migrate into the gel, and bands appeared less distinct, suggesting the formation of DNA aggregates. Moreover, MfeI digestion was completely blocked upon preincubation with C12K-2β12 (lane 5). The blockage of enzymatic activity was specific to C12K-2β12, since preincubation with the control peptide K-5α12 (lanes 6 and 7) resulted in a digestion pattern similar to that of nontreated DNA (lane 4). Similar results were also obtained using HindIII, EcoRI, SpeI, and XmnI restriction enzymes (data not shown). Taken together, these results suggest that C12K-2β12 is able to tightly bind to bacterial nucleic acid and that this binding inhibits restriction enzyme digestion.

Fig 5.

Fig 5

Functional inhibition of restriction enzyme activity by C12K-2β12. The H. pylori G27 strain endogenous plasmid, which contains MfeI restriction enzyme-cut sites, was incubated for 1 h at 37°C in the presence (+) or absence (−) of 40 or 80 μM K-5α12 (control peptide) or 40 μM C12K-2β12 prior to digestion with MfeI (+). Agarose gel electrophoresis (1%) was then used to analyze the digestion. Lane 1, molecular weight markers; lane 2, native undigested plasmid; lane 3, C12K-2β12-treated native plasmid; lane 4, MfeI-digested plasmid; lane 5, C12K-2β12-treated MfeI-digested plasmid; lane 6, 80 μM K-5α12-treated MfeI-digested plasmid; lane 7, 40 μM K-5α12-treated MfeI-digested plasmid. Since the banding patterns for native plasmid (lane 2), native plasmid incubated with C12K-2β12 (lane 3), and C12K-2β12-treated MfeI-digested plasmid (lane 5) are similar, C12K-2β12 incubation protected the plasmid DNA from restriction enzyme digestion. This gel is a representative of the gels in three independent experiments.

To conclusively prove interaction of C12K-2β12 with DNA and to rule out the possibility that enzymatic activity was inhibited via direct interaction of C12K-2β12 with the restriction enzymes, we also investigated the ability of the peptide to directly bind to DNA. To this end, the ability of the peptide to change the electrophoretic mobility of DNA was examined by electrophoretic mobility shift assay (EMSA). As shown in Fig. 6A, EMSA analysis of a labeled PCR product representing a portion of the H. pylori oorD gene (377 bp) demonstrated that in the absence of C12K-2β12 (lane 2), oorD migrated as a single 377-bp band. However, upon the addition of C12K-2β12 (lane 3 to 7), this pattern was altered such that the oorD fragment was no longer able to migrate into the gel. Similar results were obtained when a labeled fragment of the H. pylori rpoB gene (142 bp) (Fig. 6A), the E. coli chloramphenicol acetyltransferase gene (data not shown), or human antibody light-chain DNA fragment (data not shown) was used as the DNA target. This suggests that C12K-2β12 is able to efficiently bind to DNA in a sequence-independent manner. DNA binding was specific to C12K-2β12, since incubation of all of the fragments with the K-5α12 control peptide (Fig. 6A, lanes 1 for both DNA fragments and data not shown) resulted in no change in DNA migration. Thus, the ability of C12K-2β12 to bind DNA may be due to specific structure-function relationships to the target and not merely electrostatic attraction to anionic DNA.

Fig 6.

Fig 6

Gel retardation and modified Western blot assays demonstrate the ability of C12K-2β12 to bind H. pylori nucleic acids and proteins. (A) oorD or rpoB DNA was end labeled with [γ-32P]ATP according to standard procedures (see Materials and Methods) and was incubated for 1 h at 37°C in the presence of different concentrations (0 μM [−], 1.3 μM, 2.5 μM, 5 μM, 10 μM, and 20 μM) of C12K-2β12. Twenty micromolar K-5α12, an OAK peptide that has no activity against H. pylori, was included as a control (lane 1) for each DNA sample. (B) ureA or oorD mRNA was end labeled with [α-32P]UTP, according to standard procedures (see Materials and Methods), and was incubated for 1 h at 37°C in the presence of increasing amounts of C12K-2β12 (0 μg, 1.0 μg, or 5 μg). Five micrograms of K-5α12 was included as a control. Increased retention of DNA and RNA in the wells corresponding to increasing concentrations of C12K-2β12 demonstrate binding of the peptide to the nucleic acids. (C) Analysis of H. pylori proteins in Coomassie blue-stained SDS-polyacrylamide gel (left panel) and modified Western blot (right panel). Following separation and transfer of proteins, the blot was probed with fluorescently labeled C12K-2β12 (DyLight 488) to identify binding to purified H. pylori Fur and lysate protein dilutions of 1:2, 1:4, and 1:8. PBS containing no protein was used as a negative control. These are representative gels and blots from three independent experiments.

Because C12K-2β12 strongly interacted with DNA, we next sought to determine whether C12K-2β12 could also bind to RNA. Thus, the above-described assays were repeated using in vitro-transcribed ureA and oorD riboprobes instead of PCR products. As shown in Fig. 6B, approximately 60% of the ureA RNA probe was efficiently shifted by the addition of C12K-2β12 at a peptide/RNA ratio (wt/wt) of 1:1 (lane 3). Nearly 90% of the RNA was bound when the ratio (wt/wt) was increased to 5:1 (lane 4), thus indicating concentration-dependent binding of C12K-2β12 to the RNA target. Similar patterns were observed when the oorD RNA probe was used (Fig. 6B). Importantly, the addition of the K-5α12 control peptide resulted in no change in migration of either riboprobes (lanes 2 of both RNA fragments in Fig. 6B). Taken en masse, these results suggest that in addition to bacterial lysis due to membrane pore formation, at low concentrations, C12K-2β12 may also exert an antimicrobial effect by traversing the membrane and binding to both DNA and RNA.

Finally, we monitored the ability of C12K-2β12 to bind H. pylori proteins by using a modified Western blot method and fluorescently labeled peptide. The separation of purified H. pylori Fur protein or total cell lysates on an SDS-polyacrylamide gel followed by probing with fluorescent C12K-2β12 revealed that the peptide binds numerous bacterial proteins (Fig. 6C). Given that proteins and nucleic acids are key molecules involved in replication and synthesis of micro- and macromolecules, we speculate that the binding of C12K-2β12 to nucleic acid and proteins may also have significant implications for survival of H. pylori.

C12K-2β12 shows strong in vivo efficacy against H. pylori in the gerbil model of infection.

Having previously demonstrated strong in vitro efficacy of C12K-2β12 against H. pylori (47) and having shown two possible mechanisms of action of the peptide (described above), we next sought to determine the in vivo efficacy of C12K-2β12 as a therapeutic agent against H. pylori. To this end, we utilized the Mongolian gerbil model of infection, which has become a popular animal model to study H. pylori colonization and disease due to the similarity of clinical pathophysiology found in this model compared to infected humans (18). Figure 7 presents a schematic of the challenge and treatment schedule followed during the study and shows the sampling times of the animal stomach tissue for determination of viable CFU. Animals infected with H. pylori strain 7.13 (18) were treated with one, three, or five doses of C12K-2β12 orogastrically either 1 day (Fig. 8A) or 1 week (Fig. 8B) postinfection. Two concentrations of drug were administered by applying the following dosing regimen: 1 mg C12K-2β12/kg body weight or 4 mg C12K-2β12/kg body weight. The animals were sacrificed 1 week after the end of treatment, and the density of H. pylori colonization was determined by plating and enumeration of viable CFU. The CFU per gram of stomach in the C12K-2β12-treated gerbils was then compared to counts determined from the corresponding control animals that were administered PBS instead of the C12K-2β12 peptide.

Fig 7.

Fig 7

Gerbil infection and treatment schedule utilized to investigate in vivo efficacy of C12K-2β12 against H. pylori. All gerbils were infected with approximately 8 × 108 CFU of H. pylori strain 7.13 on day 0 (black arrowhead) and divided into two categories: (i) animals that received treatment 1 day postinfection (schedule shown above the black line in the middle of the figure) and (ii) animals that received treatment 1 week postinfection (schedule shown below the black line in the middle of the figure). Each category consisted of three subgroups of dose treatments: (i) single dose, (ii) three doses, or (iii) five doses. In each dose treatment subgroup, there were three groups of animals based on treatment regimen; animals were orogastrically administered 4 mg C12K-2β12/kg body weight or 1 mg C12K-2β12/kg body weight or PBS as a placebo control. All treatment doses were administered sequentially once daily by oral gavage. The initiation and completion of treatment in all groups for each dose treatment are shown (long thin black arrows). Shorter thicker black arrows represent 1 week after completion of treatment and represent the day gerbils were sacrificed for stomach tissue sampling for surviving CFU.

Fig 8.

Fig 8

Therapeutic efficacy of C12K-2β12 against H. pylori in the gerbil model of colonization. Ninety gerbils divided into 18 groups of five gerbils per group were orogastrically infected with approximately 8 × 108 CFU of H. pylori strain 7.13. The gerbils were then randomly assigned into two equal categories based on whether treatment was initiated 1 day postinfection (A) or 1 week postinfection (B) as described in the legend to Fig. 7. Animals were administered a single dose, 3 doses, or 5 doses of 1 mg C12K-2β12/kg body weight or 4 mg C12K-2β12/kg body weight or PBS as a placebo control. One week posttreatment, the gerbils were sacrificed. The stomachs of the gerbils were harvested, weighed, and homogenized, and the homogenates were plated to determine the CFU of H. pylori colonization. Data are expressed as CFU/gram of stomach, and each symbol represents the value for a single animal. The geometric mean for each group is plotted for comparison (short black bar). The P values are from one-way ANOVA analyses of H. pylori colonization levels across each dose treatment group. P values greater than 0.05 were considered nonsignificant difference (NS) in the means of bacterial colonization levels as determined by comparison of C12K-2β12-treated group and the corresponding PBS-treated control animals.

Figure 8A presents colonization levels for animals whose treatment was initiated 1 day postinfection. Animals that received a single dose of either 1 mg/kg body weight or 4 mg/kg body weight C12K-2β12 showed significantly lower levels of H. pylori colonization compared to PBS-treated control animals (P = 0.034 and P = 0.023 by one-way ANOVA, respectively). Similarly, we observed significantly lower levels of colonization in the stomachs of gerbils treated with 5 doses of 4 mg/kg body weight and 1 mg/kg body weight C12K-2β12 compared to PBS-treated control gerbils (P = 0.0001 and P = 0.01 by one-factor ANOVA, respectively) (Fig. 8A). Although the differences in the bacterial colonization density of gerbils that received three doses of either regimens of C12K-2β12 (1 mg/kg or 4 mg/kg) were not statistically significant, the trends were the same as the other dosing regimens; there was a reduction in the bacterial load in the C12K-2β12-treated animals compared to the PBS-treated animal control.

In order to more closely imitate a clinical situation in which H. pylori infection is established before commencement of treatment, we next challenged animals 1 week postinfection with H. pylori; at this point, the bacterial infection is established in the stomach, and H. pylori has typically achieved a near-maximal bacterial load within the gerbil gastric mucosa (51). In contrast to the treatment 1 day postinfection, C12K-2β12 showed no effect on H. pylori colonization density in animals that received a single dose of 1 mg/kg or 4 mg/kg of the peptide (Fig. 8B). Similarly, treatment of gerbils with three doses of 1 mg of C12K-2β12/kg of body weight showed no significant effect on colonization levels compared to the PBS-treated control gerbils (Fig. 8B). However, comparison of bacterial colonization levels in animals that received three doses of 4 mg/kg of C12K-2β12 with the corresponding PBS-treated animal controls revealed a significant reduction in the H. pylori colonization density (P < 0.043 by one-factor ANOVA) (Fig. 8B). Notably, animals that received 5 doses of either 1 mg/kg or 4 mg/kg of C12K-2β12/kg showed a significant reduction in H. pylori colonization compared to PBS-treated control animals (P = 0.021 and P = 0.001 by one-way ANOVA, respectively) (Fig. 8B). Taken together, these data indicate that therapeutic treatment of gerbils with five doses of C12K-2β12 showed the greatest effect in reduction of H. pylori colonization whether the treatment was initiated 1 day (Fig. 8A) or 1 week (Fig. 8B) postinfection. Similarly, administration of more concentrated doses (4 mg/kg) of C12K-2β12 resulted in lower H. pylori colonization than lower doses, regardless of the number of treatments. Thus, the results are consistent with dose- and concentration-dependent effects and demonstrate strong in vivo efficacy of C12K-2β12 against H. pylori in the gerbil model of infection.

DISCUSSION

In the past half century, only three new classes of antibiotics have entered the clinics, lipopeptides, oxazolidinones, and streptogramins (11, 48, 52), and each of these drugs targets Gram-positive bacterial infections. This trend signifies the slow pace of development of new drugs and highlights the dire lack of novel drugs against Gram-negative bacteria such as H. pylori. Indeed, H. pylori has developed resistance to nearly all conventional antibiotics currently used in the clinic for treatment of the infection. Furthermore, the requirement for the use of multiple drugs for extended periods of time represents an increased cost to the patient and often results in negative antibiotic-associated side effects; collectively, these factors often lead to noncompliance by patients. Therefore, H. pylori clearly represents a bacterium that demands that new antimicrobial strategies be explored for the effective future treatment of the infection. To this end, we recently screened a panel of peptidomimetic antimicrobial peptides and found that the OAK peptide C12K-2β12 demonstrated strong in vitro efficacy against numerous strains of H. pylori (47). In vitro, C12K-2β12 was effective at low pH and against nonreplicating or slow-growing bacteria. Furthermore, the OAK peptide demonstrated strong irreversible PAE and bactericidal effects against H. pylori (47). As an extension of that previous work, we sought to characterize the mechanism of action of C12K-2β12 and to determine the in vivo efficacy of the OAK peptide. Herein, we show that the bactericidal effect of the drug is exerted through the induction of distinct classical pores in the H. pylori cell membrane. Furthermore, at lower concentrations, the peptide is able to cross bacterial membranes, enter the cytoplasm, and tightly bind to bacterial nucleic acids and proteins, which we speculate may block bacte-rial replication, transcription, and translation. To our knowledge, this study is the first to demonstrate action of an OAK peptide against numerous bacterial targets.

Transmission electron microscopy revealed the presence of pores, ghost cells, membrane blebs, and areas of sloughing of the outer membranes of H. pylori cells treated with C12K-2β12. Interestingly, these results are similar to the reported effects of αs2 casein-derived peptide on E. coli (45) and suggest that pore formation and cell lysis may be a general mechanism of action of antimicrobial peptides. Furthermore, these observations largely explain the previously described irreversible PAE and rapid in vitro bactericidal effects of C12K-2β12 (47).

Broadly speaking, one of the most attractive advantages of antimicrobial peptides is their ability to exert antibacterial activity extremely rapidly, which is not the case for many of the currently used antibiotics. For instance, the beta-lactam antibiotic amoxicillin targets the bacterial cell wall by inhibiting synthesis of peptidoglycan. However, the drug requires active replication to be the most effective. While amoxicillin is effective against replicating H. pylori (73), it lacks significant PAE against the bacterium (27, 47). Moreover, it exhibits relatively slow antibacterial activity against H. pylori (22, 29, 47), which is likely due to the fact that the in vitro division time of H. pylori is long, approximately 3 to 7 h (23, 62). In vivo, not only is the doubling rate likely longer, but amoxicillin efficacy is further compromised by the gastric acidic pH (7, 16). Thus, the long division time combined with the requirement for active replication likely helps explain the long lag phase in activity. Similarly, during stationary phase, relatively slower activity is also evidenced for antibiotics such as penicillins and vancomycin, which target the bacterial cell wall (53) via mechanisms involving peptidases and peptidoglycans, respectively, that must be synthesized before they can be properly targeted (77, 83).

As a potential class of new drugs, antimicrobial peptides do not suffer from some of these limitations due to the fact that they typically have a broader more nonspecific target within the bacterial cell. Indeed, C12K-2β12 permeation of the bacterial membrane was very rapid; significant dose-dependent membrane disruption was observed within 15 min and reached a maximum within 30 min at the higher concentrations (Fig. 1). These permeation kinetics correlate with our previously reported in vitro killing kinetics of C12K-2β12 (47). Moreover, compared to amoxicillin, C12K-2β12 showed superior ability to permeate the bacterial membrane; similar membrane permeation levels by amoxicillin required 10-fold the concentration of C12K-2β12, supporting differential modes of action.

In many cases, the activity of antimicrobial peptides is dependent on charge, hydrophobicity, and amphipathic structure (4). Indeed, cationic peptides such as cecropin A and magainin 2 have been shown to utilize electrostatic attractions between the peptide and the negatively charged molecules on the surface of bacterial membrane as a means of facilitating activity (76, 88). Given the cationic nature of the C12K-2β12 OAK peptide, a similar mechanism is likely used in interaction with the bacterial membrane; phospholipids and acidic lipopolysaccharide (LPS) molecules are likely targeted (46). However, the fact that we observed a dose-dependent response in the permeation of the bacterial cell wall by C12K-2β12 (Fig. 1) suggests that a critical ratio between peptide molecules and target cell membrane molecules exists. C12K-2β12, like other cationic peptides, is amphipathic, a property that likely allows the peptide molecules to orient and insert themselves into the bacterial membrane (60, 86). Typically, this insertion is thought to be responsible for the formation of transmembrane pores via the interaction of hydrophobic regions of the peptide with lipid components of the membrane and association of hydrophilic regions of the peptide with phospholipid head groups within the membrane (4). Thus, perhaps a particular concentration of C12K-2β12 is required to effectively facilitate these types of interactions and to induce pore formation within the H. pylori membrane. Though the exact mechanism by which antimicrobial peptides interact with target membranes is not clearly understood, electrostatic interactions between the cationic peptide and anionic bacterial cell membrane are no doubt crucial. Furthermore, the exact amino acid composition, amphipathicity, size, and cationic charge of the peptide no doubt affect how an individual peptide would attach to and insert into a membrane bilayer to form transmembrane pores (38).

Permeation of the bacterial cell membranes was previously believed to be the sole mechanism of action of antibacterial peptides. However, recent studies have demonstrated that some antimicrobial peptides, including certain OAK peptides, achieve their effects through alternative modes of action (34, 58, 67). Some peptides have the ability to traverse to the bacterial cytoplasm and to bind to intracellular targets such as nucleic acids (DNA and RNA) (67) and proteins (58); these interactions may result in bacterial death (3, 4, 57). Indeed, human defensin HNP-1 (human neutrophil peptide 1), cathelicidin, magainin 2, and buforin II, a 21-amino-acid peptide derived from a natural peptide isolated from the Asian toad, have the ability to permeate membranes without significant disruption and traverse into the cytoplasm to bind bacterial nucleic acids (49, 57). We found that C12K-2β12 tightly bound to DNA and RNA (Fig. 6), whereas similar OAK peptides that were not active against H. pylori, K-5α12 (Fig. 5 and 6) and 2β12 (data not shown), did not bind either nucleic acid. This dramatic difference between OAKs perhaps suggests that protein and DNA/RNA binding is not a mere consequence of electrostatic interactions between the positively charged peptide and the polyanionic DNA or RNA. Conversely, binding may be attributed to a specific structure-activity relationship of each of the OAKs and is consistent with the structures of C12K-2β12, K-5α12, and 2β12 (47, 64). Equally consistent with this idea is lactoferricin B, a 25-amino-acid-residue lactoferrin-derived peptide with a net charge of +8 that does not bind to DNA (80, 81), whereas liver-expressed antimicrobial peptide 2 (LEAP-2), an antibacterial peptide with a net charge of +9 binds efficiently to DNA (30). Thus, a positive charge is not sufficient, and perhaps distinct differences in overall structural attributes are required to promote nucleic acid binding. That overall peptide structure might be more important than net charge and perhaps hydrophobicity for a functional mode of action of the OAK peptides is also evident when one contrasts the biophysical and chemical properties of C12K-2β12 and 2β12. Both are linear OAK peptides that display similar net charges and hydrophobicities (+5 and 51 for C12K-2β12 and +5 and 38.1 for 2β12, respectively). However, despite this similarity, 2β12 lacks antimicrobial activity against H. pylori (47). Regardless of the mechanism of interaction, the ability of C12K-2β12 to bind H. pylori nucleic acids and proteins at low concentrations suggests that this OAK peptide may have the ability to inhibit DNA, RNA, and protein synthesis, which would in turn lead to inhibition of bacterial growth and compromise bacterial survival similar to what has been seen with indolicidin-treated S. aureus (54). Indeed, the cytoplasmic electron-dense structures we observed (Fig. 2 and 3) may be the result of C12K-2β12-induced aggregation of nucleic acids and protein. The fact that C12K-2β12 binding to DNA showed no sequence specificity (Fig. 6) suggests that the peptide may have broad-spectrum activity and will likely affect the normal gut flora. Conversely, binding of the peptide to human DNA should not be a concern due to the selective nature of amphipathic peptides against anionic bacterial membranes; they exhibit no or low affinity for the zwitterionic membranes of mammalian cells (74).

In vivo testing of the efficacy of the C12K-2β12 OAK peptide against H. pylori in the gerbil model of infection demonstrated that therapeutic monotreatment of animals with C12K-2β12 significantly reduced H. pylori colonization (Fig. 8). This suggests that, despite the presence of a physical mucous barrier and the harsh acidic and proteolytic environment of the stomach, which are commonly associated with reduced bioavailability of some antibiotics, C12K-2β12 is stable and active in this environment. Furthermore, C12K-2β12 must be able to penetrate the mucous layer to reach the mucosal surface where the bacterium resides; however, the mechanism by which this is accomplished is not currently clear. Indeed, orogastrically administered C12K-2β12 could reach and eventually interact with the bacteria in vivo following absorption via two possible ways: (i) directly within the stomach where the peptide simply diffuses across the mucous layer to reach the gastric epithelium and/or (ii) indirectly via absorption in the intestine and permeation through the vasculature back to the gastric epithelium. Regardless of the mechanism by which it reaches the bacteria, C12K-2β12 is clearly effective in vivo.

Interestingly, therapeutic treatment of animals 1 week postinfection with 3 doses of 4 mg C12K-2β12/kg body weight resulted in significant reduction in stomach colonization (P = 0.043 by one-factor ANOVA) (Fig. 8B). However, the same dose and concentration administered 1 day postinfection lacked this magnitude of therapeutic effect; this may suggest interplay of C12K-2β12 with the immune response similar to what has been reported with other AMPs (10, 40, 41, 79). Indeed, antimicrobial peptides play a multifactorial role in the body, including modulation of the immune response. Cathelicidins and β-defensins have been shown to have the ability to recruit leukocytes directly or to stimulate the production of cytokines such as interleukin 8 (IL-8), monocyte chemotactic peptide 1 (MCP-1), and gamma interferon (IFN-γ), thereby indirectly recruiting other effector cells (28, 41, 42). We postulate that allowing infection to establish for 1 week before the initiation of C12K-2β12 treatment may have resulted in increased bacterial numbers that resulted in the induction of an immune response that acted synergistically with the effects of C12K-2β12 to further reduce colonization. Indeed, our explanation is corroborated by previous studies which demonstrated the appearance of serum immunoglobulin G immune response to H. pylori antigens at 1 week postinfection of Mongolian gerbils (39, 84). In humans, H. pylori typically colonizes for extended periods prior to clinical symptoms. Thus, it would be of interest to determine whether chronic colonization in the face of inflammation results in adaptation of the bacteria to become more resistant to conventional antibiotics and C12K-2β12.

One of the significant limitations in the development of AMPs for use in the clinic is cytotoxicity to host cells. We observed no mortality or obvious observable side effects due to administration of C12K-2β12, suggesting no or minimal physiological toxicity. Furthermore, these data agree with our previously reported in vitro results in which C12K-2β12 showed nonhemolytic properties (47). However, clearly detailed toxicology, pharmacokinetic, and pharmacodynamic studies are required to determine systemic or gastrointestinal tract bioavailability and potential safety parameters of the peptide.

In conclusion, our data demonstrate a dual mechanism of action of C12K-2β12 against H. pylori and suggest that the previously observed in vitro bactericidal effect of the peptide against H. pylori (47) is largely due to bacterial lysis as a result of pore formation within the bacterial membrane as well as binding of the peptide to bacterial proteins and nucleic acids, which in turn may inhibit critical cellular processes that may be important for bacterial survival. The data further demonstrate strong in vivo efficacy of C12K-2β12 when used as monotherapy against H. pylori in the gerbil model of infection. En masse, these results support the potential development of C12K-2β12 as a novel class of anti-H. pylori therapeutics.

Supplementary Material

Supplemental material

ACKNOWLEDGMENTS

We are grateful to Jeannette Whitmire and Thomas Baginski for scientific/technical help, Cara Olsen for assistance with statistical analysis, Bang Vu and Roshan D. Yedery for providing human DNA reagents, and Oscar Quijada-Pich for helpful criticisms, discussions, and suggestions. Peptides were produced and kindly provided by Amram Mor and Tchelet Kovachi.

Research in the laboratory of D. Scott Merrell is supported by grants R073PW from the Uniformed Services University, 60393-300411-7.20 from the United States Military Cancer Institute, and R01AI065529 from the National Institutes of Health (NIH). We have no conflict of interest.

The contents of this article are the sole responsibility of the authors and do not necessarily represent the official views of the NIH, DOD, USUHS, or federal government.

Footnotes

Published ahead of print 7 November 2011

Supplemental material for this article may be found at http://aac.asm.org/.

REFERENCES

  • 1. Abachin E, et al. 2002. Formation of D-alanyl-lipoteichoic acid is required for adhesion and virulence of Listeria monocytogenes. Mol. Microbiol. 43:1–14 [DOI] [PubMed] [Google Scholar]
  • 2. Armstrong JA, Wee SH, Goodwin CS, Wilson DH. 1987. Response of Campylobacter pyloridis to antibiotics, bismuth and an acid-reducing agent in vitro—an ultrastructural study. J. Med. Microbiol. 24:343–350 [DOI] [PubMed] [Google Scholar]
  • 3. Boman HG, Agerberth B, Boman A. 1993. Mechanisms of action on Escherichia coli of cecropin P1 and PR-39, two antibacterial peptides from pig intestine. Infect. Immun. 61:2978–2984 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Brogden KA. 2005. Antimicrobial peptides: pore formers or metabolic inhibitors in bacteria? Nat. Rev. Microbiol. 3:238–250 [DOI] [PubMed] [Google Scholar]
  • 5. Carpenter BM, et al. 2009. A single nucleotide change affects fur-dependent regulation of sodB in H. pylori. PLoS One 4:e5369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Carpenter BM, et al. 2007. Expanding the Helicobacter pylori genetic toolbox: modification of an endogenous plasmid for use as a transcriptional reporter and complementation vector. Appl. Environ. Microbiol. 73:7506–7514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Cederbrant G, Kahlmeter G, Schalen C, Kamme C. 1994. Additive effect of clarithromycin combined with 14-hydroxy clarithromycin, erythromycin, amoxycillin, metronidazole or omeprazole against Helicobacter pylori. J. Antimicrob. Chemother. 34:1025–1029 [DOI] [PubMed] [Google Scholar]
  • 8. Chey WD, Wong BC. 2007. American College of Gastroenterology guideline on the management of Helicobacter pylori infection. Am. J. Gastroenterol. 102:1808–1825 [DOI] [PubMed] [Google Scholar]
  • 9. Covacci A, Telford JL, Del Giudice G, Parsonnet J, Rappuoli R. 1999. Helicobacter pylori virulence and genetic geography. Science 284:1328–1333 [DOI] [PubMed] [Google Scholar]
  • 10. De Yang, et al. 2000. LL-37, the neutrophil granule- and epithelial cell-derived cathelicidin, utilizes formyl peptide receptor-like 1 (FPRL1) as a receptor to chemoattract human peripheral blood neutrophils, monocytes, and T cells. J. Exp. Med. 192:1069–1074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Diekema DI, Jones RN. 2000. Oxazolidinones: a review. Drugs 59:7–16 [DOI] [PubMed] [Google Scholar]
  • 12. Ellison RT, III, Giehl TJ, LaForce FM. 1988. Damage of the outer membrane of enteric gram-negative bacteria by lactoferrin and transferrin. Infect. Immun. 56:2774–2781 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Epand RF, Sarig H, Mor A, Epand RM. 2009. Cell-wall interactions and the selective bacteriostatic activity of a miniature oligo-acyl-lysyl. Biophys. J. 97:2250–2257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Epand RM, Vogel HJ. 1999. Diversity of antimicrobial peptides and their mechanisms of action. Biochim. Biophys. Acta 1462:11–28 [DOI] [PubMed] [Google Scholar]
  • 15. Felga GE, et al. 2008. Quadruple therapy with furazolidone for retreatment in patients with peptic ulcer disease. World J. Gastroenterol. 14:6224–6227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Fiese EF, Steffen SH. 1990. Comparison of the acid stability of azithromycin and erythromycin A. J. Antimicrob. Chemother. 25(Suppl. A):39–47 [DOI] [PubMed] [Google Scholar]
  • 17. Figura N, et al. 2004. Inactivation of Helicobacter pylori cagA gene affects motility. Helicobacter 9:185–193 [DOI] [PubMed] [Google Scholar]
  • 18. Franco AT, et al. 2005. Activation of beta-catenin by carcinogenic Helicobacter pylori. Proc. Natl. Acad. Sci. U. S. A. 102:10646–10651 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Friedrich CL, Rozek A, Patrzykat A, Hancock RE. 2001. Structure and mechanism of action of an indolicidin peptide derivative with improved activity against gram-positive bacteria. J. Biol. Chem. 276:24015–24022 [DOI] [PubMed] [Google Scholar]
  • 20. Gancz H, Censini S, Merrell DS. 2006. Iron and pH homeostasis intersect at the level of Fur regulation in the gastric pathogen Helicobacter pylori. Infect. Immun. 74:602–614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Gisbert JP, et al. 2008. Empirical rescue therapy after Helicobacter pylori treatment failure: a 10-year single-centre study of 500 patients. Aliment. Pharmacol. Ther. 27:346–354 [DOI] [PubMed] [Google Scholar]
  • 22. Graham DY, Shiotani A. 2008. New concepts of resistance in the treatment of Helicobacter pylori infections. Nat. Clin. Pract. Gastroenterol. Hepatol. 5:321–331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Graham JE, Peek RM, Jr, Krishna U, Cover TL. 2002. Global analysis of Helicobacter pylori gene expression in human gastric mucosa. Gastroenterology 123:1637–1648 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Guina T, Yi EC, Wang H, Hackett M, Miller SI. 2000. A PhoP-regulated outer membrane protease of Salmonella enterica serovar typhimurium promotes resistance to alpha-helical antimicrobial peptides. J. Bacteriol. 182:4077–4086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Gumurdulu Y, et al. 2004. Low eradication rate of Helicobacter pylori with triple 7-14 days and quadriple therapy in Turkey. World J. Gastroenterol. 10:668–671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Hancock RE, Wong PG. 1984. Compounds which increase the permeability of the Pseudomonas aeruginosa outer membrane. Antimicrob. Agents Chemother. 26:48–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Hassan IJ, Stark RM, Greenman J, Millar MR. 1998. Absence of a post-antibiotic effect (PAE) of beta-lactams against Helicobacter pylori NCTC 11637. J. Antimicrob. Chemother. 42:661–663 [DOI] [PubMed] [Google Scholar]
  • 28. Heilborn JD, et al. 2003. The cathelicidin anti-microbial peptide LL-37 is involved in re-epithelialization of human skin wounds and is lacking in chronic ulcer epithelium. J. Investig. Dermatol. 120:379–389 [DOI] [PubMed] [Google Scholar]
  • 29. Hirschl AM, Rotter ML. 1996. Amoxicillin for the treatment of Helicobacter pylori infection. J. Gastroenterol. 31(Suppl. 9):44–47 [PubMed] [Google Scholar]
  • 30. Hocquellet A, et al. 2010. Structure-activity relationship of human liver-expressed antimicrobial peptide 2. Peptides 31:58–66 [DOI] [PubMed] [Google Scholar]
  • 31. Hopkins RJ, Girardi LS, Turney EA. 1996. Relationship between Helicobacter pylori eradication and reduced duodenal and gastric ulcer recurrence: a review. Gastroenterology 110:1244–1252 [DOI] [PubMed] [Google Scholar]
  • 32. Hsu CH, et al. 2005. Structural and DNA-binding studies on the bovine antimicrobial peptide, indolicidin: evidence for multiple conformations involved in binding to membranes and DNA. Nucleic Acids Res. 33:4053–4064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Jang S, et al. 2010. Epidemiological link between gastric disease and polymorphisms in VacA and CagA. J. Clin. Microbiol. 48:559–567 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Jenssen H, Hamill P, Hancock RE. 2006. Peptide antimicrobial agents. Clin. Microbiol. Rev. 19:491–511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Jin T, et al. 2004. Staphylococcus aureus resists human defensins by production of staphylokinase, a novel bacterial evasion mechanism. J. Immunol. 172:1169–1176 [DOI] [PubMed] [Google Scholar]
  • 36. Jones KR, Cha JH, Merrell DS. 2008. Who's winning the war? Molecular mechanisms of antibiotic resistance in Helicobacter pylori. Curr. Drug Ther. 3:190–203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Khrapunov S, Cheng H, Hegde S, Blanchard J, Brenowitz M. 2008. Solution structure and refolding of the Mycobacterium tuberculosis pentapeptide repeat protein MfpA. J. Biol. Chem. 283:36290–36299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Kohanski MA, Dwyer DJ, Hayete B, Lawrence CA, Collins JJ. 2007. A common mechanism of cellular death induced by bactericidal antibiotics. Cell 130:797–810 [DOI] [PubMed] [Google Scholar]
  • 39. Kumagai T, et al. 2001. Serum immunoglobulin G immune response to Helicobacter pylori antigens in Mongolian gerbils. J. Clin. Microbiol. 39:1283–1288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Kurosaka K, Chen Q, Yarovinsky F, Oppenheim JJ, Yang D. 2005. Mouse cathelin-related antimicrobial peptide chemoattracts leukocytes using formyl peptide receptor-like 1/mouse formyl peptide receptor-like 2 as the receptor and acts as an immune adjuvant. J. Immunol. 174:6257–6265 [DOI] [PubMed] [Google Scholar]
  • 41. Lai Y, et al. 2010. Activation of TLR2 by a small molecule produced by Staphylococcus epidermidis increases antimicrobial defense against bacterial skin infections. J. Investig. Dermatol. 130:2211–2221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Liu AY, et al. 2002. Human beta-defensin-2 production in keratinocytes is regulated by interleukin-1, bacteria, and the state of differentiation. J. Investig. Dermatol. 118:275–281 [DOI] [PubMed] [Google Scholar]
  • 43. Livne L, et al. 2009. Design and characterization of a broad-spectrum bactericidal acyl-lysyl oligomer. Chem. Biol. 16:1250–1258 [DOI] [PubMed] [Google Scholar]
  • 44. Loh B, Grant C, Hancock RE. 1984. Use of the fluorescent probe 1-N-phenylnaphthylamine to study the interactions of aminoglycoside antibiotics with the outer membrane of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 26:546–551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Lopez-Exposito I, Amigo L, Recio I. 2008. Identification of the initial binding sites of alpha-S2-casein f(183-207) and effect on bacterial membranes and cell morphology. Biochim. Biophys. Acta 1778:2444–2449 [DOI] [PubMed] [Google Scholar]
  • 46. Lugtenberg B, Van Alphen L. 1983. Molecular architecture and functioning of the outer membrane of Escherichia coli and other gram-negative bacteria. Biochim. Biophys. Acta 737:51–115 [DOI] [PubMed] [Google Scholar]
  • 47. Makobongo MO, Kovachi T, Gancz H, Mor A, Merrell DS. 2009. In vitro antibacterial activity of acyl-lysyl oligomers against Helicobacter pylori. Antimicrob. Agents Chemother. 53:4231–4239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Marchese A, Schito GC. 2001. The oxazolidinones as a new family of antimicrobial agent. Clin. Microbiol. Infect. 7(Suppl. 4):66–74 [DOI] [PubMed] [Google Scholar]
  • 49. Matsuzaki K, Murase O, Fujii N, Miyajima K. 1995. Translocation of a channel-forming antimicrobial peptide, magainin 2, across lipid bilayers by forming a pore. Biochemistry 34:6521–6526 [DOI] [PubMed] [Google Scholar]
  • 50. McDaniel DP, Roberson RW. 2000. Microtubules are required for motility and positioning of vesicles and mitochondria in hyphal tip cells of Allomyces macrogynus. Fungal Genet. Biol. 31:233–244 [DOI] [PubMed] [Google Scholar]
  • 51. Miles S, et al. 2010. Detailed in vivo analysis of the role of Helicobacter pylori Fur in colonization and disease. Infect. Immun. 78:3073–3082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Mukhtar TA, Koteva KP, Wright GD. 2005. Chimeric streptogramin-tyrocidine antibiotics that overcome streptogramin resistance. Chem. Biol. 12:229–235 [DOI] [PubMed] [Google Scholar]
  • 53. Mutschler H, Gebhardt M, Shoeman RL, Meinhart A. 2011. A novel mechanism of programmed cell death in bacteria by toxin-antitoxin systems corrupts peptidoglycan synthesis. PLoS Biol. 9:e1001033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Nan YH, Bang JK, Shin SY. 2009. Design of novel indolicidin-derived antimicrobial peptides with enhanced cell specificity and potent anti-inflammatory activity. Peptides 30:832–838 [DOI] [PubMed] [Google Scholar]
  • 54a. National Research Council 1996. Guide for the care and use of laboratory animals. National Academy Press, Washington, DC [Google Scholar]
  • 55. Nordahl EA, et al. 2004. Activation of the complement system generates antibacterial peptides. Proc. Natl. Acad. Sci. U. S. A. 101:16879–16884 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Panthel K, et al. 2003. Generation of Helicobacter pylori ghosts by PhiX protein E-mediated inactivation and their evaluation as vaccine candidates. Infect. Immun. 71:109–116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Park CB, Kim HS, Kim SC. 1998. Mechanism of action of the antimicrobial peptide buforin II: buforin II kills microorganisms by penetrating the cell membrane and inhibiting cellular functions. Biochem. Biophys. Res. Commun. 244:253–257 [DOI] [PubMed] [Google Scholar]
  • 58. Park CB, Yi KS, Matsuzaki K, Kim MS, Kim SC. 2000. Structure-activity analysis of buforin II, a histone H2A-derived antimicrobial peptide: the proline hinge is responsible for the cell-penetrating ability of buforin II. Proc. Natl. Acad. Sci. U. S. A. 97:8245–8250 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Peschel A, et al. 2001. Staphylococcus aureus resistance to human defensins and evasion of neutrophil killing via the novel virulence factor MprF is based on modification of membrane lipids with l-lysine. J. Exp. Med. 193:1067–1076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Powers JP, Hancock RE. 2003. The relationship between peptide structure and antibacterial activity. Peptides 24:1681–1691 [DOI] [PubMed] [Google Scholar]
  • 61. Poyart C, et al. 2003. Attenuated virulence of Streptococcus agalactiae deficient in D-alanyl-lipoteichoic acid is due to an increased susceptibility to defensins and phagocytic cells. Mol. Microbiol. 49:1615–1625 [DOI] [PubMed] [Google Scholar]
  • 62. Psakis G, et al. 2009. The sodium-dependent D-glucose transport protein of Helicobacter pylori. Mol. Microbiol. 71:391–403 [DOI] [PubMed] [Google Scholar]
  • 63. Radzishevsky I, Krugliak M, Ginsburg H, Mor A. 2007. Antiplasmodial activity of lauryl-lysine oligomers. Antimicrob. Agents Chemother. 51:1753–1759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Radzishevsky IS, et al. 2008. Structure-activity relationships of antibacterial acyl-lysine oligomers. Chem. Biol. 15:354–362 [DOI] [PubMed] [Google Scholar]
  • 65. Radzishevsky IS, et al. 2007. Improved antimicrobial peptides based on acyl-lysine oligomers. Nat. Biotechnol. 25:657–659 [DOI] [PubMed] [Google Scholar]
  • 66. Ramakrishnan K, Salinas RC. 2007. Peptic ulcer disease. Am. Fam. Physician 76:1005–1012 [PubMed] [Google Scholar]
  • 67. Rotem S, et al. 2008. Analogous oligo-acyl-lysines with distinct antibacterial mechanisms. FASEB J. 22:2652–2661 [DOI] [PubMed] [Google Scholar]
  • 68. Salvatore M, et al. 2007. Alpha-defensin inhibits influenza virus replication by cell-mediated mechanism(s). J. Infect. Dis. 196:835–843 [DOI] [PubMed] [Google Scholar]
  • 69. Sarig H, Goldfeder Y, Rotem S, Mor A. 2011. Mechanisms mediating bactericidal properties and conditions that enhance the potency of a broad-spectrum oligo-acyl-lysyl. Antimicrob. Agents Chemother. 55:688–695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Sarig H, et al. 2010. A miniature mimic of host defense peptides with systemic antibacterial efficacy. FASEB J. 24:1904–1913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Sarig H, Rotem S, Ziserman L, Danino D, Mor A. 2008. Impact of self-assembly properties on antibacterial activity of short acyl-lysine oligomers. Antimicrob. Agents Chemother. 52:4308–4314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Schibli DJ, Epand RF, Vogel HJ, Epand RM. 2002. Tryptophan-rich antimicrobial peptides: comparative properties and membrane interactions. Biochem. Cell Biol. 80:667–677 [DOI] [PubMed] [Google Scholar]
  • 73. Scott D, Weeks D, Melchers K, Sachs G. 1998. The life and death of Helicobacter pylori. Gut 43(Suppl. 1):S56–S60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Shai Y. 1999. Mechanism of the binding, insertion and destabilization of phospholipid bilayer membranes by alpha-helical antimicrobial and cell non-selective membrane-lytic peptides. Biochim. Biophys. Acta 1462:55–70 [DOI] [PubMed] [Google Scholar]
  • 75. Sieprawska-Lupa M, et al. 2004. Degradation of human antimicrobial peptide LL-37 by Staphylococcus aureus-derived proteinases. Antimicrob. Agents Chemother. 48:4673–4679 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Silvestro L, Gupta K, Weiser JN, Axelsen PH. 1997. The concentration-dependent membrane activity of cecropin A. Biochemistry 36:11452–11460 [DOI] [PubMed] [Google Scholar]
  • 77. Spratt BG, Cromie KD. 1988. Penicillin-binding proteins of gram-negative bacteria. Rev. Infect. Dis. 10:699–711 [DOI] [PubMed] [Google Scholar]
  • 78. Thompson LJ, et al. 2003. Gene expression profiling of Helicobacter pylori reveals a growth-phase-dependent switch in virulence gene expression. Infect. Immun. 71:2643–2655 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Tjabringa GS, et al. 2003. The antimicrobial peptide LL-37 activates innate immunity at the airway epithelial surface by transactivation of the epidermal growth factor receptor. J. Immunol. 171:6690–6696 [DOI] [PubMed] [Google Scholar]
  • 80. Ulvatne H, Haukland HH, Samuelsen O, Kramer M, Vorland LH. 2002. Proteases in Escherichia coli and Staphylococcus aureus confer reduced susceptibility to lactoferricin B. J. Antimicrob. Chemother. 50:461–467 [DOI] [PubMed] [Google Scholar]
  • 81. Ulvatne H, Samuelsen O, Haukland HH, Kramer M, Vorland LH. 2004. Lactoferricin B inhibits bacterial macromolecular synthesis in Escherichia coli and Bacillus subtilis. FEMS Microbiol. Lett. 237:377–384 [DOI] [PubMed] [Google Scholar]
  • 82. Viejo-Diaz M, Andres MT, Fierro JF. 2005. Different anti-Candida activities of two human lactoferrin-derived peptides, Lfpep and kaliocin-1. Antimicrob. Agents Chemother. 49:2583–2588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Williams DH. 1996. The glycopeptide story–how to kill the deadly ‘superbugs’. Nat. Prod. Rep. 13:469–477 [DOI] [PubMed] [Google Scholar]
  • 84. Wirth HP, Beins MH, Yang M, Tham KT, Blaser MJ. 1998. Experimental infection of Mongolian gerbils with wild-type and mutant Helicobacter pylori strains. Infect. Immun. 66:4856–4866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Xie M, et al. 2010. Rostrocaudal analysis of corpus callosum demyelination and axon damage across disease stages refines diffusion tensor imaging correlations with pathological features. J. Neuropathol. Exp. Neurol. 69:704–716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Yeaman MR, Yount NY. 2003. Mechanisms of antimicrobial peptide action and resistance. Pharmacol. Rev. 55:27–55 [DOI] [PubMed] [Google Scholar]
  • 87. Zasloff M. 2002. Antimicrobial peptides of multicellular organisms. Nature 415:389–395 [DOI] [PubMed] [Google Scholar]
  • 88. Zhao H, Mattila JP, Holopainen JM, Kinnunen PK. 2001. Comparison of the membrane association of two antimicrobial peptides, magainin 2 and indolicidin. Biophys. J. 81:2979–2991 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental material

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES