Open Access

Melittin, a honeybee venom‑derived antimicrobial peptide, may target methicillin‑resistant Staphylococcus aureus

  • Authors:
    • Ji Hae Choi
    • A Yeung Jang
    • Shunmei Lin
    • Sangyong Lim
    • Dongho Kim
    • Kyungho Park
    • Sang‑Mi Han
    • Joo‑Hong Yeo
    • Ho Seong Seo
  • View Affiliations

  • Published online on: September 1, 2015     https://doi.org/10.3892/mmr.2015.4275
  • Pages: 6483-6490
  • Copyright: © Choi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Methicillin‑resistant Staphylococcus aureus (MRSA) is difficult to treat using available antibiotic agents. Honeybee venom has been widely used as an oriental treatment for several inflammatory diseases and bacterial infections. The venom contains predominantly biologically active compounds, however, the therapeutic effects of such materials when used to treat MRSA infections have not been investigated extensively. The present study evaluated bee venom and its principal active component, melittin, in terms of their antibacterial activities and in vivo protection against MRSA infections. In vitro, bee venom and melittin exhibited comparable levels of antibacterial activity, which was more marked against MRSA strains, compared with other Gram‑positive bacteria. When MRSA‑infected mice were treated with bee venom or melittin, only the latter animals were successfully rescued from MRSA‑ induced bacteraemia or exhibited recovery from MRSA‑infected skin wounds. Together, the data of the present study demonstrated for the first time, to the best of our knowledge, that melittin may be used as a promising antimicrobial agent to enhance the healing of MRSA‑induced wounds.

Introduction

Staphylococcus aureus is a significant human pathogen causing healthcare-associated and community-acquired infections (1). Antibiotics effectively treat these infections, however, the emergence of methicillin-resistant S. aureus (MRSA) currently presents a challenge to healthcare systems worldwide (2). Globally, ~2,000,000,000 MRSA carriers exist, of whom as many as 53,000,000 suffer from overt MRSA infections. In addition, Staphylococcus aureus clones resistant to the antibiotic vancomycin have been identified; and vancomycin is the last known drug to which earlier strains had been uniformly sensitive (3). These organisms are termed vancomycin-intermediate-resistant Staphylococcus aureus and vancomycin-resistant Staphylococcus aureus (4,5). Therefore, it is becoming difficult to treat staphylococcal infections with current chemotherapeutic agents (6).

Honeybee (Apis mellifera L.) venom contains a complex mixture of therapeutic compounds, including antimicrobial peptides, allowing bees to defend their hives against predators and external threats (7). Several biological and pharmacological studies have examined bee venom components for use as potential pain relievers and treatments for inflammatory diseases (810). In addition, the antibacterial activities of venom against several human and animal pathogens have been evaluated (11). However, as venom contains certain complex toxic components, its human therapeutic applications have been limited. Previously, the majority of bee venom components have been individually purified and their specific pharmacological activities investigated.

The melittin peptide, the predominant component of bee venom (40–48%, w/w), has been investigated substantially, and exhibits potent cytolytic and antimicrobial activities (12). Potential actions against bacteria, viruses and cancer cells have been extensively examined in vitro, although the antimicrobial molecular mechanism remains to be elucidated (13,14). However, to date, few investigations of the in vivo antimicrobial activities of melittin have been performed. The present study investigated the antimicrobial activity of melittin from bee venom, and examined whether it can inhibit MRSA infections in vitro and in vivo.

Materials and methods

Ethical statement

All animal investigations were performed in accordance with the Guidelines for the Care and Use of Laboratory Animals of the Ministry of Food and Drug Safety of Korea, and were approved by the Animal Care and Use Committee of the Korea Atomic Energy Research Institute (Jeongeup Si, Korea; IACUC protocol no. 2014–023).

Bacterial strains and reagents

The bacterial strains examined in the present study are listed in Table I. The streptococcal and staphylococcal strains were grown at 37°C in Todd-Hewitt broth (BD Biosciences, Franklin Lakes, NJ, USA) supplemented with 0.5% (w/v) yeast extract and Tryptic-soy broth (BD Biosciences), respectively. Purified melittin was purchased from Sigma-Aldrich (St. Louis, MO, USA). Synthetic melittin (GIGAVLKVLTTGLPALISWIKRKRQQ) was chemically synthesised by A&PEP Co., Inc. (DaeJeon, Korea).

Table I

Bacterial strains examined in the present study.

Table I

Bacterial strains examined in the present study.

Bacterial strainDescriptionSource
Streptococcus agalactiae CNCTC 10/84Clinical isolate, serotype V(18)
Streptococcus gordonii M99Endocarditis clinical isolate(21)
Streptococcus pneumonia TIGR4Laboratory strain, serotype IV(22)
Streptococcus epidermidis RP62aClinical isolatePresent study
Streptococcus bovis NEM760Clinical isolate, biotype IIPresent study
Staphylococcus aureus USA300 (LAC) Methicillin-resistant clinical isolate(23)
Staphylococcus aureus Newman Methicillin-resistant clinical isolate(23)
Staphylococcus aureus MW2 Methicillin-resistant clinical isolate(23)
Staphylococcus aureus MRSA1 Methicillin-resistant clinical isolatePresent study
Staphylococcus aureus MRSA2 Methicillin-resistant clinical isolatePresent study
Staphylococcus aureus ISP4790Clinical isolate(23)
Staphylococcus aureus MU50Clinical isolate(23)
Purification of bee venom

Controlled colonies of natural honeybees (Apis mellifera L.) were maintained at room temperature at the National Academy of Agricultural Science (Suwon, Korea). In brief, a bee venom collector apparatus (Chunglin Biotech, Ansan, Korea) was placed on the hive, and the bees that landed on the apparatus were subjected to an electric shock sufficient to cause the bees to 'sting' a glass plate from which dried bee venom was harvested. The collected venom was dissolved in distilled water, centrifuged at 12,000 × g for 10 min to remove insoluble materials, and stored in a refrigerator until further use (1517).

Bactericidal assay

Bacteria were harvested at the early log phase (A600=0.5) and suspended in phosphate-buffered saline (PBS) at ~108 to 1010 CFU/ml. Subsequently, the bacterial samples were incubated with the indicated concentrations of bee venom or melittin at 25°C for 30 min, and surviving bacteria were evaluated using a plate counting method, as described previously (18). Briefly, samples were serially diluted in PBS and plated onto blood agar (Kisan Bio, Suwon, Korea). Following a 16 h incubation at 37°C, the number of surviving bacteria was counted.

Determination of the minimum inhibitory concentration

To determine the minimum inhibitory concentration (MIC), the present study used a micro-dilution broth method, according to the recommendations of the National Committee for Clinical Laboratory Standards (19). In brief, the cells of the experimental bacterial strains were collected in the logarithmic phase of growth, suspended in 30 mM phosphate buffer (pH 7.0) with 60 mM NaCl, and adjusted to an A600 of 0.3 arbitrary units (1×105 cells/ml). The bee venom and the melittin samples were dissolved in 10 mM phosphate buffer (pH 6.0) with 130 mM NaCl and 0.2% (w/v) bovine serum albumin prior to serial dilution. Sample aliquots (10 µl) were mixed with the diluted bacterial suspensions (190 µl) followed by incubation for 20 h at 37°C. Bacterial growth was determined by measurement of the A650 levels using a VICTOR™ X3 ELISA reader (PerkinElmer, Inc., Waltham MA, USA).

Cytotoxicity assays

The cytotoxic effects of bee venom and melittin on cultured MCF7 cells were evaluated using a Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Gaithersburg, MD, USA). The cells were seeded at a density of 5×103 cells/200 µl/well into wells of 96-well round-bottomed plates and allowed to grow for 24 h at 37°C, followed by incubation with bee venom or purified synthetic melittin for 6 h at 37°C. The culture supernatants (100 µl quantities) were harvested and mixed with 10 µl aliquots of CCK-8 solution. Following 3 h incubation at 37°C, the optical densities at A450 were measured using the VICTOR™ X3 ELISA reader (PerkinElmer, Inc.).

Mouse intraperitoneal infection

Mouse infection with Staphylococcus aureus was performed, as described previously (20). Bacteria of the USA300 strain (American Type Culture Collection, Manassas, VA, USA) were spectrophotometrically (OPTIZEN POP; Mecasys Co., Ltd., Daejeon, Korea) adjusted to the desired concentration prior to injection, and bacterial numbers were confirmed via serial dilution and Tryptic soy agar plating. The cultured USA300 bacteria were pelleted, washed and suspended in PBS at 0.5×108 CFU/ml. Mice (7-week-old males) of the CD1 strain were obtained from Oriental Bio, Inc. (Seongnam, Korea), with 10 animals per treatment group. The mice were infected with the USA300 strain (200 µl) via intraperitoneal (i.p.) injection, followed by i.p. injection of 100 µl bee venom or purified melittin 1 h later. The infected animals were monitored every 3 h for up to 36 h. The mice were housed in controlled conditions: Temperature, 23±2°C; humidity 55±10%; light between 07:00 and 19:00. Each group was housed seperately. All animal experiments in the present study adhered to institutional guidelines upon review of the experimental protocol, and were approved by the Institutional Biosafety Committee and the Institutional Animal Care and Use Committee of Korea Atomic Energy Research Institute.

Mouse skin infection

CD1 mice (7-week old; 3 mice/group) were used to examine skin infection. Following the induction of general anesthesia, the dorsal hair was electrically shaved and the skin was cleaned with 70% (v/v) ethanol. Skin infection was induced via subcutaneous inoculation of 50 µl volumes of USA300 suspension (106 CFU/ml) in PBS. Subsequently, bee venom, melittin (purified or synthetic; 100 µg in 80 µl PBS), or sterile PBS was applied once daily to each surface lesion. Lesion progression was monitored at 24 h intervals for 10 days by measuring the lesion dimensions with callipers (Jeung Do B&P Co., Ltd., Seoul, Korea), and capturing images using a digital camera (WB5500; Samsung, Seoul, Korea).

Statistical analysis

Data are presented as the mean ± standard deviation. Statistical analysis was conducted using GraphPad InStat software version 5 (GraphPad Software, Inc., La Jolla, CA, USA). The statistical significance of between-group differences was evaluated using two-tailed Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Bee venom exhibits a broad specrtum of antimicrobial activity

The present study examined the antibacterial activities of bee venom against the Streptococcus agalactiae, Streptococcus gordonii, Streptococcus pneumonia, Streptococcus epidermidis, Streptococcus bovis and Staphylococcus aureus Gram-positive bacteria. As shown in Fig. 1, when all the bacterial strains were treated with the indicated concentrations of bee venom for 30 min, concentration-dependent death of the bacteria was evident. At venom concentrations between 1.25 and 12.5 µg/ml, bacterial viability decreased by >90%. The MIC values of the bee venom ranged between 1.56 and 12.5 µg/ml (Table II). Notably, the USA300 antibiotic-resistant Staphylococcus strain had the lowest observed MIC (1.56 µg/ml).

Table II

MIC of bee venom towards bacterial strains.

Table II

MIC of bee venom towards bacterial strains.

Bacterial strainMIC (µg/ml)
Streptococcus agalactiae CNCTC 10/846.25
Streptococcus gordonii M996.25
Streptococcus pneumonia TIGR43.12
Streptococcus epidermidis RP62a0.78
Streptococcus bovis NEM7601.56
Staphylococcus aureus USA300 (LAC)0.78
Staphylococcus aureus Newman0.78
Staphylococcus aureus MW21.56
Staphylococcus aureus MRSA13.12
Staphylococcus aureus MRSA21.56
Staphylococcus aureus ISP47906.25
Staphylococcus aureus MU506.25

[i] MIC is defined as the lowest concentration of bee venom required to cause the optical density (OD)600 value to remain constant between 0 and 18 h. MIC, minimum inhibitory concentration.

The present study further examined the antibacterial activities of bee venom against three MRSA clinical isolates. As shown in Fig. 2, the viabilities of all three strains decreased markedly upon treatment with bee venom for 30 min, and no bacteria survived incubation with 100 µg/ml venom. The MIC values for the three MRSA strains ranged between 0.78 and 3.13 µg/ml (Table II). Notably, the methicillin-sensitive Staphylococcus aureus strains (Mu50, ISP479C, PS735, PS736 and PS737) were less susceptible to bee venom (MIC=3.13–12.5 µg/ml), compared with the MRSA strains (Table II), suggesting that bee venom contains antimicrobial molecules, which specifically target MRSA strains.

Bee venom protects against staphylococcal infection

To measure the cytotoxicity of bee venom, human epithelial cells were incubated with venom for 24 h and cell viabilities were measured using an MTT assay. As shown in Fig. 3, bee venom was not cellulotoxic at a concentration of 0.4 µg/ml. In addition, the administration of bee venom in vivo at up to 20 mg/kg i.p., caused no signs or symptoms of toxicity in the CD1 mice (data not shown).

The i.p injection of 1×108 CFU of the USA300 strain into mice caused bacteraemia and mortality rates of 100% within 18 h. When the USA300-infected mice were administered with 1.25 or 2.5 mg/kg bee venom at the time of infection, no protective effect was evident (data not shown). A low dose of USA300 (1×107 CFU per mouse) was injected 1 h following the administration of PBS or bee venom. Notably, all the mice died 18 h following the injection of USA300 with bee venom, whereas only five mice of the control group had died by 24 h post-infection (Fig. 4A). These data demonstrated that, although bee venom exhibited a marked antimicrobial effect in vitro, in vivo administration enhanced MRSA propagation and infection.

In addition, the present study examined the protective effect of bee venom in a staphylococcal skin infection model (Fig. 4B). When USA300 was inoculated intradermally and the areas of infected skin treated with PBS or bee venom (10 µg) once daily, the abscesses formed by USA300 were 21.3±4.8 and 18.8±6.8 mm in diameter in the PBS and bee venom groups, respectively, by day 5, and no significant difference was observed even following 10 days of venom treatment.

Melittin is the major antimicrobial component of bee venom

Bee venom is a complex mixture of proteins, peptides and low-molecular-weight materials. The principal components of the venom are phospholipase A2 (PLA2; 10–12%, w/w) and the melittin peptide (40–48%, w/w). The results of the present study confirmed and extended the previous results, demonstrating that melittin and PLA2 induced death in a broad range of bacteria, including MRSA strains. As shown in Fig. 5A, treatment of the USA300 and MRSA2 strains with PLA2 did not affect cell viability, whereas the viabilities of the MRSA strains treated with purified melittin decreased to levels comparable to those observed when bee venom was used. To examine whether melittin and PLA2 acted synergistically, two MRSA strains were treated with melittin admixed with PLA2 at various concentrations. When the USA300 and MRSA2 strains were treated with melittin alone (25 µg/ml), the total number of bacteria decreased by ~2.5–3 log CFU (Fig. 5B). However, when the cells were treated with melittin (25 µg/ml) in combination with various concentrations of PLA2, similar results were observed, indicating that PLA2 did not act synergistically with melittin to cause bacterial cell death.

Subsequently, the present study confirmed that synthetic melittin exhibited an antimicrobial activity similar to that of purified melittin. Initially, the toxicities of the two forms of melittin towards human epithelial cells were determined, as described above. As shown in Fig. 6A, synthetic melittin (99.2% pure) was ~25% less toxic than the 'purified' melittin (93% pure). However, the antibacterial activities of the two preparations against the MRSAs were comparable (Fig. 6B).

Protection from staphylococcal infection by melittin

The present study also investigated whether melittin can protect against MRSA skin infections. USA300 bacteria (1×107 CFU/mouse) were injected intradermally into CD1 mice, which were administered with either PBS, or purified or synthetic melittin (10 µg) 1 h post-infection. As shown in Fig. 7, abscesses in the PBS-treated group gradually increased in size to attain a diameter of 22±6.3 mm by day 5. When the infected areas were treated with purified or synthetic melittin once daily for 4 days, the diameters of the abscesses were significantly lower than those measured in the control group.

In addition, the protective effect of melittin was investigated in a model of MRSA bacteraemia (Fig. 8). When a high dose of USA300 was injected i.p., all the mice died following treatment with either PBS or 2.5 mg/kg melittin after 24 h. However, when the infected mice were injected with 5 mg/kg melittin 1 h post-infection, 50% of the mice survived >24 h.

Discussion

Staphylococcus aureus is an important human pathogen, which is responsible for the majority of bacterial soft skin tissue infections and life-threatening infections, including pneumonia, abscesses, endocarditis and infections of surgical sites (2). The rapid spread of MRSA strains is cause for alarm. The rates of MSRA infections are increasing, and MRSA has become the leading cause of invasive illness, resulting in a high rate of mortality worldwide (2426). Thus, the development of novel therapeutic methods is essential to treat chronic wounds or systemic infections caused by MRSA. In the present study, the in vitro anti-MRSA activities of the natural antimicrobial components of bee venom were investigated.

Bee venom contains several potential antibacterial toxins, including melittin, PLA2, adolpanin, dopamine and hyaluronidase (27). Each component may exert selective and specific actions on human cells and/or bacteria (16,28). Although the bee venom isolated in the present study exhibited potential antimicrobial activities against all the Gram-positive bacteria assessed in vitro, as has been reported in several previous studies (9,11,29), the i.p. administration of venom into MRSA-infected mice caused the a higher mortality rate, compared with that observed in the venom-free controls, suggesting that bee venom actually facilitated MRSA infection. Notably, the PLA2 of bee venom is central to the proinflammatory cascade by activating several physiological and pathogenic immune activities (30,31). In addition, certain hypervirulent bacteria produce and secrete PLA2, which significantly potentiates early-stage infection and inflammation (3235). The present study also found that, although PLA2 exhibited minimal antibacterial activity, i.p. injection of MRSA-infected mice with PLA2 caused 100% mortality, whereas only 50% mortality was observed in the control animals by 24 h, which was also true of the bee venom-treated mice (data not shown). Thus, it is reasonable to suggest that PLA2 increased the susceptibility of at-risk hosts to bacterial infection.

Melittin is the principal component (40–48%, w/w) of honeybee venom (12), being a small linear peptide of 26 amino acids forming an amphipathic helix with a hydrophobic amino- and hydrophilic carboxyl-terminus. The antibacterial effects of melittin have been widely investigated in vitro (36). In the present study, synthetic melittin exhibited anti-MRSA toxicity in vitro, which was comparable to that of purified melittin. However, the synthetic melittin was less toxic towards human epithelial cells, suggesting that the purified melittin (93% pure) in the present study contained an uncharacterized component, which is either toxic and/or enhances the toxicity of melittin. Following acquisition of these in vitro results, the present study examined the protective effects of melittin in MRSA-infected mice. Unlike bee venom, melittin exhibited significantly higher protective effects in vivo in the models of bacteraemia and skin infection. Although melittin directly affects microbes by damaging or destabilising cell membranes, the material appears to potentiate the innate immune and anti-inflammatory responses, preventing the development of MRSA systemic infections and facilitating wound healing around infected sites (14,3739). Melittin exerts anti-inflammatory effects on several types of cell (38,40,41). Melittin suppresses innate immune signaling, including that mediated by nuclear factor-κB via Toll-like receptor and mitogen activated protein kinase; the synthesis of cyclooxygenase-2; and the expression of inducible nitric oxide synthase (38,39). In addition, melittin stimulates pyrin domain-containing inflammasomes to activate caspase-1 and interleukin1β, which crucially recruit neutrophils to sites of expression (14,40,42). Thus, melittin may inhibit MRSA infections by several mechanisms, including the direct induction of MRSA cell death, the downregulation of the innate immune response induced by MRSA and the acceleration of neutrophil recruitment to sites of infection.

Together, the results of the present study demonstrated that bee venom, which is intrinsically toxic, exerts negative effects when used as an anti-MRSA therapy. However, the principal component of bee venom, melittin, exhibits antibacterial effects with minimal toxicity in vitro and in vivo. To the best of our knowledge, the present study is the first to demonstrate that melittin may exert a possible therapeutic role in the treatment of MRSA infections. The mechanism of this effect requires further investigation.

Acknowledgments

This study was supported by grants from the Nuclear R&D program of the Ministry of Science, ICT and Future planning (grant no. 523330) to Dr Sangyong Lim and the Next BioGreen21 Program, Rural Development Administration, Republic of Korea (grant. no. PJ009534) to Dr Joo-Hong Yeo.

References

1 

Rasigade JP and Vandenesch F: Staphylococcus aureus: A pathogen with still unresolved issues. Infect Genet Evol. 21:510–514. 2014. View Article : Google Scholar

2 

Taylor AR: Methicillin-resistant Staphylococcus aureus infections. Prim Care. 40:637–654. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Limbago BM, Kallen AJ, Zhu W, Eggers P, McDougal LK and Albrecht VS: Report of the 13th vancomycin-resistant Staphylococcus aureus isolate from the United States. J Clin Microbiol. 52:998–1002. 2014. View Article : Google Scholar :

4 

Corey GR: Staphylococcus aureus bloodstream infections: Definitions and treatment. Clin Infect Dis. 48(Suppl 4): S254–S259. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Gould IM: VRSA-doomsday superbug or damp squib? Lancet Infect Dis. 10:816–818. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Bassetti M, Merelli M, Temperoni C and Astilean A: New antibiotics for bad bugs: Where are we? Ann Clin Microbiol Antimicrob. 12(22)2013. View Article : Google Scholar : PubMed/NCBI

7 

Annila I: Bee venom allergy. Clin Exp Allergy. 30:1682–1687. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Son DJ, Lee JW, Lee YH, Song HS, Lee CK and Hong JT: Therapeutic application of anti-arthritis, pain-releasing and anti-cancer effects of bee venom and its constituent compounds. Pharmacol Ther. 115:246–270. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Kim JY, Lee WR, Kim KH, An HJ, Chang YC, Han SM, Park YY, Pak SC and Park KK: Effects of bee venom against Propionibacterium acnes-induced inflammation in human keratinocytes and monocytes. Int J Mol Med. 35:1651–1656. 2015.PubMed/NCBI

10 

Lee H, Lee EJ, Kim H, Lee G, Um EJ, Kim Y, Lee BY and Bae H: Bee venom-associated Th1/Th2 immunoglobulin class switching results in immune tolerance of NZB/W F1 murine lupus nephritis. Am J Nephrol. 34:163–172. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Perumal Samy R, Gopalakrishnakone P, Thwin MM, Chow TK, Bow H, Yap EH and Thong TW: Antibacterial activity of snake, scorpion and bee venoms: A comparison with purified venom phospholipase A2 enzymes. J Appl Microbiol. 102:650–659. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Gajski G and Garaj-Vrhovac V: Melittin: A lytic peptide with anticancer properties. Environ Toxicol Pharmacol. 36:697–705. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Adade CM, Oliveira IR, Pais JA and Souto-Padron T: Melittin peptide kills Trypanosoma cruzi parasites by inducing different cell death pathways. Toxicon. 69:227–239. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Jo M, Park MH, Kollipara PS, An BJ, Song HS, Han SB, Kim JH, Song MJ and Hong JT: Anti-cancer effect of bee venom toxin and melittin in ovarian cancer cells through induction of death receptors and inhibition of JAK2/STAT3 pathway. Toxicol Appl Pharmacol. 258:72–81. 2012. View Article : Google Scholar

15 

Han SM, Lee GG and Park KK: Acute dermal toxicity study of bee venom (Apis mellifera L.) in rats. Toxicol Res. 28:99–102. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Han SM, Lee KG, Park KK and Pak SC: Skin sensitization study of bee venom (Apis mellifera L.) in guinea pigs and rats. Cutan Ocul Toxicol. 32:27–30. 2013. View Article : Google Scholar

17 

Han SM, Lee GG and Park KK: Skin sensitization study of bee venom (Apis mellifera L.) in guinea pigs. Toxicol Res. 28:1–4. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Seo HS, Mu R, Kim BJ, Doran KS and Sullam PM: Binding of glycoprotein Srr1 of Streptococcus agalactiae to fibrinogen promotes attachment to brain endothelium and the eevelopment of meningitis. PLoS Pathog. 8:e10029472012. View Article : Google Scholar

19 

Clinical and Laboratory Standards Institute: M100-S16, Performance standards for antimicrobial susceptibility testing; 16th informational supplement. Clinical and Laboratory Standards Institute; Wayne, PA: 2007

20 

Ganesh VK, Rivera JJ, Smeds E, Ko YP, Bowden MG, Wann ER, Gurusiddappa S, Fitzgerald JR and Höök M: A structural model of the Staphylococcus aureus ClfA-fibrinogen interaction opens new avenues for the design of anti-staphylococcal therapeutics. PLoS Pathog. 4:e10002262008. View Article : Google Scholar : PubMed/NCBI

21 

Bensing BA, Gibson BW and Sullam PM: The Streptococcus gordonii platelet binding protein GspB undergoes glycosylation independently of export. J Bacteriol. 186:638–645. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Seo HS, Cartee RT, Pritchard DG and Nahm MH: A new model of pneumococcal lipoteichoic acid structure resolves biochemical, biosynthetic and serologic inconsistencies of the current model. J Bacteriol. 190:2379–2387. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Qian Z, Yin Y, Zhang Y, Lu L, Li Y and Jiang Y: Genomic char-acterization of ribitol teichoic acid synthesis in Staphylococcus aureus: Genes, genomic organization and gene duplication. BMC Genomics. 7(74)2006. View Article : Google Scholar

24 

Goldrick BA: MRSA, VRE, and VRSA: How do we control them in nursing homes? Am J Nurs. 104:50–51. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Hebert C and Weber SG: Common approaches to the control of multidrug-resistant organisms other than methicillin-resistant Staphylococcus aureus (MRSA). Infect Dis Clin North Am. 25:181–200. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Todd B: Beyond MRSA: VISA and VRSA: What will ward off these pathogens in health care facilities? Am J Nurs. 106:28–30. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Park D, Jung JW, Lee MO, Lee SY, Kim B, Jin HJ, Kim J, Ahn YJ, Lee KW, Song YS, et al: Functional characterization of naturally occurring melittin peptide isoforms in two honey bee species, Apis mellifera and Apis cerana. Peptides. 53:185–193. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Palm NW and Medzhitov R: Role of the inflammasome in defense against venoms. Proc Natl Acad Sci USA. 110:1809–1814. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Fennell JF, Shipman WH and Cole LJ: Antibacterial action of a bee venom fraction (melittin) against a penicillin-resistant staphylococcus and other microorganisms. USNRDL-TR-67-101. Res Dev Tech Rep. 5:1–13. 1967.

30 

Putz T, Ramoner R, Gander H, Rahm A, Bartsch G, Bernardo K, Ramsay S and Thurnher M: Bee venom secretory phospholipase A2 and phosphatidylinositol-homologues cooperatively disrupt membrane integrity, abrogate signal transduction and inhibit proliferation of renal cancer cells. Cancer Immunol Immunother. 56:627–640. 2007. View Article : Google Scholar

31 

Carballido JM, Carballido-Perrig N, Schwärzler C and Lametschwandtner G: Regulation of human T helper cell differentiation by antigen-presenting cells: The bee venom phospholipase A2 model. Chem Immunol Allergy. 91:147–158. 2006. View Article : Google Scholar

32 

Lapointe S, Brkovic A, Cloutier I, Tanguay JF, Arm JP and Sirois MG: Group V secreted phospholipase A2 contributes to LPS-induced leukocyte recruitment. J Cell Physiol. 224:127–134. 2010.PubMed/NCBI

33 

Sitkiewicz I, Stockbauer KE and Musser JM: Secreted bacterial phospholipase A2 enzymes: Better living through phospholipolysis. Trends Microbiol. 15:63–69. 2007. View Article : Google Scholar

34 

Hunt CL, Nauseef WM and Weiss JP: Effect of D-alanylation of (lipo) teichoic acids of Staphylococcus aureus on host secretory phospholipase A2 action before and after phagocytosis by human neutrophils. J Immunol. 176:4987–4994. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Koprivnjak T, Peschel A, Gelb MH, Liang NS and Weiss JP: Role of charge properties of bacterial envelope in bactericidal action of human group IIA phospholipase A2 against Staphylococcus aureus. J Biol Chem. 277:47636–47644. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Fennell JF, Shipman WH and Cole LJ: Antibacterial action of melittin, a polypeptide from bee venom. Proc Soc Exp Biol Med. 127:707–710. 1968. View Article : Google Scholar : PubMed/NCBI

37 

Park JH, Kim KH, Lee WR, Han SM and Park KK: Protective effect of melittin on inflammation and apoptosis in acute liver failure. Apoptosis. 17:61–69. 2012. View Article : Google Scholar

38 

Park HJ, Lee HJ, Choi MS, Son DJ, Song HS, Song MJ, Lee JM, Han SB, Kim Y and Hong JT: JNK pathway is involved in the inhibition of inflammatory target gene expression and NF-kappaB activation by melittin. J Inflamm (Lond). 5:72008. View Article : Google Scholar

39 

Moon DO, Park SY, Choi YH, Kim ND, Lee C and Kim GY: Melittin induces Bcl-2 and caspase-3-dependent apoptosis through downregulation of Akt phosphorylation in human leukemic U937 cells. Toxicon. 51:112–120. 2008. View Article : Google Scholar

40 

Sommer A, Fries A, Cornelsen I, Speck N, Koch-Nolte F, Gimpl G, Andrä J, Bhakdi S and Reiss K: Melittin modulates keratinocyte function through P2 receptor-dependent ADAM activation. J Biol Chem. 287:23678–23689. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Dempsey CE: The actions of melittin on membranes. Biochim Biophys Acta. 1031:143–161. 1990. View Article : Google Scholar : PubMed/NCBI

42 

Kim SJ, Park JH, Kim KH, Lee WR, Kim KS and Park KK: Melittin inhibits atherosclerosis in LPS/high-fat treated mice through athero-protective actions. J Atheroscler Thromb. 18:1117–1126. 2011. View Article : Google Scholar

Related Articles

Journal Cover

November-2015
Volume 12 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Choi JH, Jang AY, Lin S, Lim S, Kim D, Park K, Han SM, Yeo JH and Seo HS: Melittin, a honeybee venom‑derived antimicrobial peptide, may target methicillin‑resistant Staphylococcus aureus. Mol Med Rep 12: 6483-6490, 2015
APA
Choi, J.H., Jang, A.Y., Lin, S., Lim, S., Kim, D., Park, K. ... Seo, H.S. (2015). Melittin, a honeybee venom‑derived antimicrobial peptide, may target methicillin‑resistant Staphylococcus aureus. Molecular Medicine Reports, 12, 6483-6490. https://doi.org/10.3892/mmr.2015.4275
MLA
Choi, J. H., Jang, A. Y., Lin, S., Lim, S., Kim, D., Park, K., Han, S., Yeo, J., Seo, H. S."Melittin, a honeybee venom‑derived antimicrobial peptide, may target methicillin‑resistant Staphylococcus aureus". Molecular Medicine Reports 12.5 (2015): 6483-6490.
Chicago
Choi, J. H., Jang, A. Y., Lin, S., Lim, S., Kim, D., Park, K., Han, S., Yeo, J., Seo, H. S."Melittin, a honeybee venom‑derived antimicrobial peptide, may target methicillin‑resistant Staphylococcus aureus". Molecular Medicine Reports 12, no. 5 (2015): 6483-6490. https://doi.org/10.3892/mmr.2015.4275