Next Article in Journal
Antimicrobial Activity of Xanthohumol and Its Selected Structural Analogues
Next Article in Special Issue
Fusarium Toxins in Cereals: Occurrence, Legislation, Factors Promoting the Appearance and Their Management
Previous Article in Journal
Preparation of Pd/Bacterial Cellulose Hybrid Nanofibers for Dopamine Detection
Previous Article in Special Issue
Military Importance of Natural Toxins and Their Analogs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Inflammatory Applications of Melittin, a Major Component of Bee Venom: Detailed Mechanism of Action and Adverse Effects

Department of Physiology, College of Korean Medicine, Kyung Hee University, 1 Hoeki-Dong, Dongdaemoon-gu, Seoul 130-701, Korea
*
Author to whom correspondence should be addressed.
Molecules 2016, 21(5), 616; https://doi.org/10.3390/molecules21050616
Submission received: 10 March 2016 / Revised: 18 April 2016 / Accepted: 9 May 2016 / Published: 11 May 2016
(This article belongs to the Special Issue Natural Toxins)

Abstract

:
Inflammation is a pervasive phenomenon triggered by the innate and adaptive immune systems to maintain homeostasis. The phenomenon normally leads to recovery from infection and healing, but when not properly phased, inflammation may cause immune disorders. Bee venom is a toxin that bees use for their protection from enemies. However, for centuries it has been used in the Orient as an anti-inflammatory medicine for the treatment of chronic inflammatory diseases. Bee venom and its major component, melittin, are potential means of reducing excessive immune responses and provide new alternatives for the control of inflammatory diseases. Recent experimental studies show that the biological functions of melittin could be applied for therapeutic use in vitro and in vivo. Reports verifying the therapeutic effects of melittin are accumulating in the literature, but the cellular mechanism(s) of the anti-inflammatory effects of melittin are not fully elucidated. In the present study, we review the current knowledge on the therapeutic effects of melittin and its detailed mechanisms of action against several inflammatory diseases including skin inflammation, neuroinflammation, atherosclerosis, arthritis and liver inflammation, its adverse effects as well as future prospects regarding the use of melittin.

1. Introduction

Inflammation is a primary process of the immune response that is triggered by any stimulus such as infection, injury, and exposure to contaminants that poses a real or perceived threat to homeostasis [1]. It is a protective process for the body, however chronic inflammation can cause the development of different diseases like rheumatoid arthritis, cardiovascular disease, diabetes, obesity, inflammatory bowel disease, asthma, and CNS related diseases such as Parkinson’s disease and Amyotrophic Lateral Sclerosis (ALS) [2].
While bee venom is a toxin which bees use for their protection from enemies, a number of recent studies regarding the beneficial roles of bee venom report that it possesses radioprotective [3], anti-mutagenic [4], anti-nociceptive [5,6,7], anti-cancer [8,9,10,11,12,13,14] and anti-inflammatory [15,16,17,18,19,20] activities. Melittin is the principal constituent of bee (Apis mellifera) venom, accounting for approximately 50% by weight of dried bee venom [21]. This amphiphilic peptide has a linear structure consists of 26 amino acids (NH2-GIGAVLKVLTTGLPALISWIKRKRQQ-CONH2) [22]. In high doses, melittin may cause itching, inflammation, and local pain; on the other hand, small doses of melittin produce broad anti-inflammatory effects [23]. There are several reviews available in the literature that focus on diverse functions as well as the pharmacological aspects of melittin [23,24,25,26,27,28,29,30,31,32]. However, reviews of the anti-inflammatory role of melittin alone are currently not available. Numerous recent reports point to several anti-inflammatory mechanisms of melittin in different types of disease models. Herein, we highlight the newest findings on the beneficial role and mechanisms of melittin against inflammatory disorders (Table 1, Figure 1). We also summarize possible adverse effects of melittin and attempts to overcome them.

2. Therapeutic Applications of the Anti-Inflammatory Effects of Melittin

2.1. Application for Skin Inflammation

Acne vulgaris is a long term skin disorder of the hair follicle in the face and upper trunk, and Propionibacterium acnes (P. acnes) is the main cause of the inflammation of acne [33]. Toll-like receptor 2 signaling activated by P. acnes causes keratinocytes and monocytes to secrete pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-8 [34,35]. A wide range of agents, including antibiotics, is used to suppress inflammation in acne vulgaris, but antibiotics can cause side effects [36].
The protective effects of melittin on P. acnes-induced inflammatory responses in vitro and in vivo were reported by Lee et al. [37]. They investigated the anti-inflammatory effects of melittin treatment in heat-killed P. acnes-treated HaCaT cells. Melittin treatment attenuated the increased phosphorylation of IKK, IκB, NF-κB as well as p38 by heat-killed P. acnes in HaCaT cells. These data show that melittin treatment abrogates P. acnes-induced inflammatory cytokine production through blocking NF-κB signaling as well as p38 MAPK signaling in HaCaT cells. In addition, the anti-inflammatory effect of melittin was examined in a live P. acnes-induced inflammatory skin disease animal model. In the animal model, melittin-treated ears show markedly reduced P. acnes-injected swelling and granulomatous responses, as compared with ears injected with live P. acnes alone.
Another study showed the inhibitory action of melittin against heat-killed P. acnes-induced apoptosis and inflammation in human THP-1 monocytic cells [38]. Melittin treatment suppressed the cleavage of the caspase-3, -8, and PARP in heat-killed P. acnes-treated THP-1 monocytic cells; in addition, administration of melittin significantly decreased the expression of TNF-α and IL-1β.

2.2. Application for Neurodegenerative Diseases

In vitro assays have revealed the potential of melittin as an agent for the prevention of neurodegenerative diseases. Moon et al. showed that melittin has a potent suppressive effect on pro-inflammatory responses of BV2 microglia and suggested that melittin may have potential to treat neurodegenerative diseases accompanied with microglial activation [39]. Melittin suppresses expression of NO and iNOS by blocking LPS-induced activation of NF-κB in BV2 microglial cell line. These results indicate that melittin suppresses COX-2/PGE2 expression resulting in anti-inflammatory properties. Meanwhile, Han et al. investigated the anti-apoptotic effects of melittin in a H2O2-induced cytotoxicity model using the SH-SY5Y human neuroblastoma cell line [40]. Melittin treatment increased cell viability and decreased apoptotic DNA fragmentation. Melittin inhibited the H2O2-induced decrease of anti-apoptotic factor Bcl-2 expression and increase of pro-apoptotic factor Bax expression.
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that affects motor neurons in the brain and the spinal cord, resulting in weakness and atrophy of muscles [53]. Yang et al. showed that melittin treatment (0.1 μg/g twice a week s.c. acupuncture on bilateral point ST36) improves anti-neuroinflammatory ability of proteasome in the CNS of ALS model mice [41]. In this model, administration of melittin reduces phosphorylation of p38 and the microglial cell number in the brainstem and spinal cord; in addition, melittin-treated mice exhibit declined neuronal death in the spinal cord and improved motor control that improves motor function. Furthermore, melittin alleviates the misfolding of proteins by activation of chaperones and reducing post-transcriptional modification of α-synuclein, a major process of ALS pathogenesis. The results demonstrate the anti-neuroinflammatory effects of melittin. Although the main problem of ALS occurs in CNS, ALS also affects other organs, including the liver, spleen, and lung. Melittin treatment attenuates inflammation and stimulates the signaling for cell survival in the spleen and lung in an animal model of ALS [42]. Administration of melittin suppresses the expression of CD14 and Iba-1 (inflammatory proteins) in the lung of symptomatic ALS transgenic mice. In addition, melittin increases the expression of pERK and Bcl-2 (cell survival factors) and suppresses the expression of C14 and COX-2 in the spleen of symptomatic ALS mice.
Dantas et al. examined the pharmacological effects of melittin in mice, with particular emphasis on dopaminergic related behaviors [54]. The animals were submitted to behavioral tests, such as the apomorphine rotation test, catalepsy test, and open field test. The results showed that melittin-treated mice displayed reduced effects caused by apomorphine, though there is no alteration in motor activity or cataleptic effects with melittin treatment. The authors reported that melittin exhibits anti-psychotic properties and could be an alternative to treat psychotic disorders, cutting down the side effects of neuroleptic medicines. Melittin also decreases apomorphine-induced stereotypes. The data indicate the anti-psychotic activity of melittin in a mice model.

2.3. Application for Atherosclerosis

Atherosclerosis is an inflammatory disease in which plaque builds up inside arteries. This chronic inflammatory disorder of the arteries is a one of major causes of death in adults. Plaque is made up of cholesterol, triglycerides, remnants of dead cells, and immune cells. In the plaque, inflammatory immune cells including macrophage and helper T cells produce inflammatory cytokines, a main trigger of plaque growth [55]. Modulation of NF-κB signaling, which plays a critical role in apoptosis and cell proliferation, is regarded as a potential therapeutic target for the treatment of atherosclerosis [56,57].
In vitro studies show an effect of melittin on the proliferation and apoptosis of vascular smooth muscle cells (VSMCs). The NF-κB signal pathway was investigated as a potent mechanism for apoptosis of cultured rat aortic VSMCa. Melittin suppresses not only platelet-derived growth factor receptor (PDGFR) β-tyrosine phosphorylation, but also downstream intracellular signal transduction in rat aortic VSMCa [43] and blocks phosphorylation of AKT induced by the PDGRF signal [44]. In addition, phosphorylation of extracellular signal-regulated kinase 1/2, an upstream signal of NF-κB, is slightly suppressed by melittin treatment. Melittin treatment, besides, induces expression of pro-apoptotic proteins, including p53, Bax, and caspase-3, but decreases expression of anti-apoptotic protein Bcl-2 [44]. In addition, in vivo experiments demonstrate that melittin treatment suppresses atherosclerosis via athero-protective functions in High-Fat/LPS treated mice [45]. Melittin treatment decreases total cholesterol and triglyceride levels and recovers heart and descending aorta. In addition, melittin decreases the expression levels of TNF-α, IL-1β, VCAM-1, ICAM-1, and TGFβ-1 in atherosclerotic mice.
Cho et al. used mass spectrometry and gel electrophoresis to elucidate the anti-atherosclerotic mechanisms of melittin. They identified differentially expressed proteins by melittin treatment in human VSMCs stimulated by TNF-α [46]. In their experiments, a proteomics analysis identify 33 proteins with consistently different expression patterns after melittin treatment. The identified proteins include anti-apoptotic proteins like as stress-70 protein and annexin A1, Several target molecules of inflammation and proliferation such as prohibitin, annexin-1, and calreticulin were identified. Furthermore, intracellular signal pathway analysis shows that EGFR and NF-κB are the main signaling molecules of inflammation in TNF-α treated human VSMCs.

2.4. Application for Arthritis

The Hong group proposed target inactivation of NF-κB by directly binding to the p50 subunit as anti-arthritic mechanism of bee venom and melittin. They demonstrated that melittin inhibits LPS-induced p50 translocation into nucleus resulting in reduced transcription of inflammatory genes [20]. Melittin directly binds to p50 (affinity [Kd] = 1.2 × 10−8 M). They also investigated the anti-inflammatory effect of melittin via interaction with IKKs. In a mouse macrophage cell line and synoviocytes acquired from rheumatoid patients, melittin suppresses the TNF-α/LPS-induced production of NO and PGE2 [47]. In addition, they demonstrated that the JNK pathway is involved in the inhibition of inflammatory target gene expression and NF-κB activation by melittin [48].

2.5. Application for Liver Inflammation

Park et al. reported that melittin attenuates hepatic injury, inflammation and hepatic fibrosis. Melittin inhibits TNF-α secretion and expression of IL-1β and IL-6 in the TNF-α-treated hepatic stellate cells (HSCs). Melittin attenuates inflammation and fibrosis by inhibiting the NF-κB signaling pathway in thioacetamide-induced liver fibrosis. In addition, they suggested that the regulated inflammatory response may affect anti-fibrotic effect of melittin in the activated HSCs [49]. Subsequently, they demonstrate that melittin inhibits liver failure via blocking NF-κB signaling and apoptotic pathways in the D-galactosamine/LPS-induced mouse liver failure model [50].
Melittin decreases the high rate of lethality, alleviates hepatic pathological injury, attenuates hepatic inflammatory responses and inhibits hepatocyte apoptosis. They also elucidated the inhibitory mechanism of melittin for NF-κB transcription in TNF-α-induced hepatic damage [51]. In TNF-α-treated hepatocytes, melittin inhibits DNA binding of NF-κB as well as promoter activity of NF-κB, suggesting that melittin inhibits apoptosis of hepatocytes through suppression of NF-κB activation. Recently, they showed that administration of melittin attenuated inflammation and fibrosis of the bile duct. Their experimental results suggest that melittin may have therapeutic applications in chronic liver injury [52]. Figure 1 summarizes the major mechanisms of the anti-inflammatory action of melittin.

3. Adverse Effects of Melittin

Melittin is the major constituent of apitoxin and is known as an allergenic peptide. It is also responsible for cell lysis and death. Accumulated melittin peptides disrupt phospholipid packing in the cell membrane, resulting in cell lysis [23]. Melittin provokes the lysis of plasma membranes and intracellular membranes. Melittin and bee venom phospholipase A2 (PLA2) show synergistic action with lipid membranes, leading to cell damage [58].

3.1. Allergic Reactions

Melittin is considered as an allergen of bee venom since research in the 1970s showed that it induces an IgE response in around one-third of honeybee venom-sensitive patients [59] and melittin appears to be allergenic in several patients [60]. However, the results are possibly due to contamination with other bee venom components. Bee venom is a complex mixture of melittin, apamine, mast cell degranulating peptide, histamine, adolapin, oligopeptides, phospholipids, saccharides, phospholipase A2, hyaluconidase, acid phosphatase, etc. [26]. Purified melittin potentially contains residues of strong allergens such as hyaluronidase, phospholipase A2 and acid phosphatasen. As the purity of currently available purified melittin is not high (e.g., Sigma-Aldrich M7391: 65%–85% and Sigma-Aldrich M2272: ≥85%, (Sigma Aldrich Inc., St. Louis, MO, USA)) synthesized melittin (e.g., Sigma-Aldrich M4171: ≥97%) should be used to confirm whether melittin causes allergic reactions or not.

3.2. Hemolysis, Cytotoxic Effects

Melittin is also known for its high lytic activity on human erythrocyte cells [61,62]. It directly binds on erythrocytes and releases hemoglobin. It is reported that the maximum capacity and the apparent dissociation constant for melittin binding to human erythrocytes is 1.8 × 107 molecules/cell and 30 nM, respectively [61]. When melittin provokes hemolysis, swelling of the erythrocytes is observed after leakage of cations occurs from membrane of the cells. In its initial phase, melittin increases permeability of ions and the release of hemoglobin is followed [61,62]. Melittin is cytotoxic for human peripheral blood lymphocytes (HPBLs) in a dose- and time-dependent manner. It leads to granulation, morphologic changes, and finally lysis of cells [63].

3.3. Genotoxic Effects

Gajski et al. report that low doses (non-cytotoxic concentrations) of melittin can increase DNA damage in HPBLs [63]. In their experiments, comet and micronucleus assays show decreased proliferation of lymphocyte and increased formation of nuclear buds and micronuclei. Moreover, melittin modulates gene expressions related with apoptosis (Bax, Bcl-2, Cas-3, Cas-7), DNA damage response (TP53, CDKN1A, GADD45α, MDM), and oxidative stress (CAT, GCLC, GPX1, GSR, SOD1). The observed genotoxicity coincides with reduction of glutathione level, increased formation of reactive oxygen species, increased phospholipase C activity and lipid peroxidation, showing the induction of oxidative stress.

4. Attempts to Overcome the Adverse Effect of Melittin

Even though melittin has demonstrated significant therapeutic properties, its toxicity must be neutralized for use as an anti-inflammatory agent. The cytotoxic effect of melittin is beneficial in part for anti-tumor purposes, but it hinders therapeutic application of melittin for other purposes. Since the native form of melittin causes non-specific cell lysis and toxicity, attempts including mutation and fusion proteins to reduce the toxicity of melittin to deliver it to specific targeted lesions are ongoing. Asthana et al. showed that alanine substitution in the leucine zipper motif of melittin results in a significant reduction of its hemolytic, but not anti-bacterial activity [64]. This suggests that lesion- specific mutations can reduce the toxicity of melittin without affecting its therapeutic activity. Rayahin et al. showed that the melittin fusion protein with glutathione S-transferase exhibits anti-inflammatory properties and minimal toxicity [65]. Development of delivery techniques using nanocarriers enable safe delivery of melittin to targeted lesions without affecting non-target cells [27,66,67].

5. Conclusions

Studies over the previous decade have advanced our knowledge about bee venom-derived melittin. Herein, we summarize the inflammation-specific effects of melittin and discuss its underlying mechanisms (Table 1, Figure 1). However, parts of the underlying mechanisms still remain unclear and the toxicity of this peptide still prevents its use for anti-inflammatory activity. Melittin could possibly be used for anti-inflammatory purposes if careful provisions are taken to avoid adverse effects. Technical developments will help to modify this toxic peptide into a safe therapeutic agent. We consider that synthesized melittin and its derivatives [68,69,70] should be used to overcome the effects of contaminants from bee venom and for developing novel pharmaceutical agents. The future therapeutic application of melittin on inflammatory disorders will depend on new study protocols to validate the efficiency and safety of melittin. In addition, the anti-inflammatory effect of bee venom PLA2 was recently established [71]. As melittin and bee venom PLA2 are synergistic [58,72], it is worth studying the anti-inflammatory effect of melittin and bee venom PLA2 co-treatment.

Acknowledgments

This work was supported by the Basic Science Research Program administered by the National Research Foundation (NRF), which is funded by the Ministry of Science, ICT and Future Planning (No. 2013-068954).

Author Contributions

G. Lee and H. Bae conceived the ideas for this manuscript. G. Lee wrote the manuscript. Both authors revised and approved this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Nathan, C. Points of control in inflammation. Nature 2002, 420, 846–852. [Google Scholar] [CrossRef] [PubMed]
  2. Laveti, D.; Kumar, M.; Hemalatha, R.; Sistla, R.; Naidu, V.G.; Talla, V.; Verma, V.; Kaur, N.; Nagpal, R. Anti-inflammatory treatments for chronic diseases: A review. Inflamm. Allergy Drug Targets 2013, 12, 349–361. [Google Scholar] [CrossRef] [PubMed]
  3. Gajski, G.; Garaj-Vrhovac, V. Radioprotective effects of honeybee venom (Apis mellifera) against 915-MHz microwave radiation-induced DNA damage in Wistar rat lymphocytes: In vitro study. Int. J. Toxicol. 2009, 28, 88–98. [Google Scholar] [CrossRef] [PubMed]
  4. Varanda, E.A.; Monti, R.; Tavares, D.C. Inhibitory effect of propolis and bee venom on the mutagenicity of some direct- and indirect-acting mutagens. Teratog. Carcinog. Mutagen. 1999, 19, 403–413. [Google Scholar] [CrossRef]
  5. Yoon, H.; Kim, M.J.; Yoon, I.; Li, D.X.; Bae, H.; Kim, S.K. Nicotinic acetylcholine receptors mediate the suppressive effect of an injection of diluted bee venom into the GV3 acupoint on oxaliplatin-induced neuropathic cold allodynia in rats. Biol. Pharm. Bull. 2015, 38, 710–714. [Google Scholar] [CrossRef] [PubMed]
  6. Lim, B.S.; Moon, H.J.; Li, D.X.; Gil, M.; Min, J.K.; Lee, G.; Bae, H.; Kim, S.K.; Min, B.I. Effect of bee venom acupuncture on oxaliplatin-induced cold allodynia in rats. Evid. Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  7. Baek, Y.H.; Huh, J.E.; Lee, J.D.; Choi do, Y.; Park, D.S. Antinociceptive effect and the mechanism of bee venom acupuncture (Apipuncture) on inflammatory pain in the rat model of collagen-induced arthritis: Mediation by alpha2-adrenoceptors. Brain Res. 2006, 1073–1074, 305–310. [Google Scholar] [CrossRef] [PubMed]
  8. Huh, J.E.; Baek, Y.H.; Lee, M.H.; Choi, D.Y.; Park, D.S.; Lee, J.D. Bee venom inhibits tumor angiogenesis and metastasis by inhibiting tyrosine phosphorylation of VEGFR-2 in LLC-tumor-bearing mice. Cancer Lett. 2010, 292, 98–110. [Google Scholar] [CrossRef] [PubMed]
  9. Moon, D.O.; Park, S.Y.; Heo, M.S.; Kim, K.C.; Park, C.; Ko, W.S.; Choi, Y.H.; Kim, G.Y. Key regulators in bee venom-induced apoptosis are Bcl-2 and caspase-3 in human leukemic U937 cells through downregulation of ERK and Akt. Int. Immunopharmacol. 2006, 6, 1796–1807. [Google Scholar] [CrossRef] [PubMed]
  10. Hu, H.; Chen, D.; Li, Y.; Zhang, X. Effect of polypeptides in bee venom on growth inhibition and apoptosis induction of the human hepatoma cell line SMMC-7721 in vitro and Balb/c nude mice in vivo. J. Pharm. Pharmacol. 2006, 58, 83–89. [Google Scholar] [CrossRef] [PubMed]
  11. Orsolic, N.; Sver, L.; Verstovsek, S.; Terzic, S.; Basic, I. Inhibition of mammary carcinoma cell proliferation in vitro and tumor growth in vivo by bee venom. Toxicon 2003, 41, 861–870. [Google Scholar] [CrossRef]
  12. Orsolic, N.; Knezevic, A.; Sver, L.; Terzic, S.; Hackenberger, B.K.; Basic, I. Influence of honey bee products on transplantable murine tumours. Vet. Comp. Oncol. 2003, 1, 216–226. [Google Scholar] [CrossRef] [PubMed]
  13. Jang, M.H.; Shin, M.C.; Lim, S.; Han, S.M.; Park, H.J.; Shin, I.; Lee, J.S.; Kim, K.A.; Kim, E.H.; Kim, C.J. Bee venom induces apoptosis and inhibits expression of cyclooxygenase-2 mRNA in human lung cancer cell line NCI-H1299. J. Pharmacol. Sci. 2003, 91, 95–104. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, X.; Chen, D.; Xie, L.; Zhang, R. Effect of honey bee venom on proliferation of K1735M2 mouse melanoma cells in vitro and growth of murine B16 melanomas in vivo. J. Pharm. Pharmacol. 2002, 54, 1083–1089. [Google Scholar] [CrossRef] [PubMed]
  15. Park, Y.C.; Koh, P.S.; Seo, B.K.; Lee, J.W.; Cho, N.S.; Park, H.S.; Park, D.S.; Baek, Y.H. Long-term effectiveness of bee venom acupuncture and physiotherapy in the treatment of adhesive capsulitis: A one-year follow-up analysis of a previous randomized controlled trial. J. Altern. Complement. Med. 2014, 20, 919–924. [Google Scholar] [CrossRef] [PubMed]
  16. Kim, H.; Lee, G.; Park, S.; Chung, H.S.; Lee, H.; Kim, J.Y.; Nam, S.; Kim, S.K.; Bae, H. Bee venom mitigates cisplatin-induced nephrotoxicity by regulating CD4(+)CD25(+)Foxp3(+) regulatory t cells in mice. Evid. Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  17. Choi, M.S.; Park, S.; Choi, T.; Lee, G.; Haam, K.K.; Hong, M.C.; Min, B.I.; Bae, H. Bee venom ameliorates ovalbumin induced allergic asthma via modulating CD4+CD25+ regulatory T cells in mice. Cytokine 2012, 61, 256–265. [Google Scholar] [CrossRef] [PubMed]
  18. Lee, K.G.; Cho, H.J.; Bae, Y.S.; Park, K.K.; Choe, J.Y.; Chung, I.K.; Kim, M.; Yeo, J.H.; Park, K.H.; Lee, Y.S.; et al. Bee venom suppresses LPS-mediated NO/iNOS induction through inhibition of PKC-alpha expression. J. Ethnopharmacol. 2009, 123, 15–21. [Google Scholar] [CrossRef] [PubMed]
  19. Lee, J.Y.; Kang, S.S.; Kim, J.H.; Bae, C.S.; Choi, S.H. Inhibitory effect of whole bee venom in adjuvant-induced arthritis. In Vivo 2005, 19, 801–805. [Google Scholar] [PubMed]
  20. Park, H.J.; Lee, S.H.; Son, D.J.; Oh, K.W.; Kim, K.H.; Song, H.S.; Kim, G.J.; Oh, G.T.; Yoon, D.Y.; Hong, J.T. Antiarthritic effect of bee venom: Inhibition of inflammation mediator generation by suppression of NF-kappaB through interaction with the p50 subunit. Arthritis Rheum. 2004, 50, 3504–3515. [Google Scholar] [CrossRef] [PubMed]
  21. Habermann, E. Bee and wasp venoms. Science 1972, 177, 314–322. [Google Scholar] [CrossRef] [PubMed]
  22. Tosteson, M.T.; Tosteson, D.C. The sting. Melittin forms channels in lipid bilayers. Biophys. J. 1981, 36, 109–116. [Google Scholar] [CrossRef]
  23. Raghuraman, H.; Chattopadhyay, A. Melittin: A membrane-active peptide with diverse functions. Biosci. Rep. 2007, 27, 189–223. [Google Scholar] [CrossRef] [PubMed]
  24. Van den Bogaart, G.; Guzman, J.V.; Mika, J.T.; Poolman, B. On the mechanism of pore formation by melittin. J. Biol. Chem. 2008, 283, 33854–33857. [Google Scholar] [CrossRef] [PubMed]
  25. Gajski, G.; Garaj-Vrhovac, V. Melittin: A lytic peptide with anticancer properties. Environ. Toxicol. Pharmacol. 2013, 36, 697–705. [Google Scholar] [CrossRef] [PubMed]
  26. Son, D.J.; Lee, J.W.; Lee, Y.H.; Song, H.S.; Lee, C.K.; Hong, J.T. Therapeutic application of anti-arthritis, pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol. Ther. 2007, 115, 246–270. [Google Scholar] [CrossRef] [PubMed]
  27. Pan, H.; Soman, N.R.; Schlesinger, P.H.; Lanza, G.M.; Wickline, S.A. Cytolytic peptide nanoparticles (‘NanoBees’) for cancer therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2011, 3, 318–327. [Google Scholar] [CrossRef] [PubMed]
  28. Moreno, M.; Giralt, E. Three valuable peptides from bee and wasp venoms for therapeutic and biotechnological use: melittin, apamin and mastoparan. Toxins 2015, 7, 1126–1150. [Google Scholar] [CrossRef] [PubMed]
  29. Hou, K.K.; Pan, H.; Schlesinger, P.H.; Wickline, S.A. A role for peptides in overcoming endosomal entrapment in sirna delivery—A focus on melittin. Biotechnol. Adv. 2015, 33, 931–940. [Google Scholar] [CrossRef] [PubMed]
  30. Huang, H.W. Molecular mechanism of antimicrobial peptides: The origin of cooperativity. Biochim. Biophys. Acta 2006, 1758, 1292–1302. [Google Scholar] [CrossRef] [PubMed]
  31. Fletcher, J.E.; Jiang, M.S. Possible mechanisms of action of cobra snake venom cardiotoxins and bee venom melittin. Toxicon 1993, 31, 669–695. [Google Scholar] [CrossRef]
  32. Dempsey, C.E. The actions of melittin on membranes. Biochim. Biophys. Acta 1990, 1031, 143–161. [Google Scholar] [CrossRef]
  33. Leyden, J.J. The evolving role of Propionibacterium acnes in acne. Semin. Cutan. Med. Surg. 2001, 20, 139–143. [Google Scholar] [CrossRef] [PubMed]
  34. Vowels, B.R.; Yang, S.; Leyden, J.J. Induction of proinflammatory cytokines by a soluble factor of Propionibacterium acnes: Implications for chronic inflammatory acne. Infect. Immun. 1995, 63, 3158–3165. [Google Scholar] [PubMed]
  35. Kim, J. Review of the innate immune response in acne vulgaris: Activation of toll-like receptor 2 in acne triggers inflammatory cytokine responses. Dermatology 2005, 211, 193–198. [Google Scholar] [CrossRef] [PubMed]
  36. Aslam, I.; Fleischer, A.; Feldman, S. Emerging drugs for the treatment of acne. Expert Opin. Emerg. Drugs 2015, 20, 91–101. [Google Scholar] [CrossRef] [PubMed]
  37. Lee, W.R.; Kim, K.H.; An, H.J.; Kim, J.Y.; Chang, Y.C.; Chung, H.; Park, Y.Y.; Lee, M.L.; Park, K.K. The protective effects of melittin on Propionibacterium acnes-induced inflammatory responses in vitro and in vivo. J. Investig. Dermatol. 2014, 134, 1922–1930. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, W.R.; Kim, K.H.; An, H.J.; Kim, J.Y.; Han, S.M.; Lee, K.G.; Park, K.K. Protective effect of melittin against inflammation and apoptosis on Propionibacterium acnes-induced human THP-1 monocytic cell. Eur. J. Pharmacol. 2014, 740, 218–226. [Google Scholar] [CrossRef] [PubMed]
  39. Moon, D.O.; Park, S.Y.; Lee, K.J.; Heo, M.S.; Kim, K.C.; Kim, M.O.; Lee, J.D.; Choi, Y.H.; Kim, G.Y. Bee venom and melittin reduce proinflammatory mediators in lipopolysaccharide-stimulated BV2 microglia. Int. Immunopharmacol. 2007, 7, 1092–1101. [Google Scholar] [CrossRef] [PubMed]
  40. Han, S.M.; Kim, J.M.; Park, K.K.; Chang, Y.C.; Pak, S.C. Neuroprotective effects of melittin on hydrogen peroxide-induced apoptotic cell death in neuroblastoma SH-SY5Y cells. BMC Complement. Altern. Med. 2014, 14. [Google Scholar] [CrossRef]
  41. Yang, E.J.; Kim, S.H.; Yang, S.C.; Lee, S.M.; Choi, S.M. Melittin restores proteasome function in an animal model of ALS. J. Neuroinflamm. 2011, 8. [Google Scholar] [CrossRef] [PubMed]
  42. Lee, S.H.; Choi, S.M.; Yang, E.J. Melittin ameliorates the inflammation of organs in an amyotrophic lateral sclerosis animal model. Exp. Neurobiol. 2014, 23, 86–92. [Google Scholar] [CrossRef] [PubMed]
  43. Son, D.J.; Kang, J.; Kim, T.J.; Song, H.S.; Sung, K.J.; Yun do, Y.; Hong, J.T. Melittin, a major bioactive component of bee venom toxin, inhibits PDGF receptor beta-tyrosine phosphorylation and downstream intracellular signal transduction in rat aortic vascular smooth muscle cells. J. Toxicol. Environ. Health A 2007, 70, 1350–1355. [Google Scholar] [CrossRef] [PubMed]
  44. Son, D.J.; Ha, S.J.; Song, H.S.; Lim, Y.; Yun, Y.P.; Lee, J.W.; Moon, D.C.; Park, Y.H.; Park, B.S.; Song, M.J.; et al. Melittin inhibits vascular smooth muscle cell proliferation through induction of apoptosis via suppression of nuclear factor-kappaB and Akt activation and enhancement of apoptotic protein expression. J. Pharmacol. Exp. Ther. 2006, 317, 627–634. [Google Scholar] [CrossRef] [PubMed]
  45. Kim, S.J.; Park, J.H.; Kim, K.H.; Lee, W.R.; Kim, K.S.; Park, K.K. Melittin inhibits atherosclerosis in LPS/high-fat treated mice through atheroprotective actions. J. Atheroscler. Thromb. 2011, 18, 1117–1126. [Google Scholar] [CrossRef] [PubMed]
  46. Cho, H.J.; Kang, J.H.; Park, K.K.; Choe, J.Y.; Park, Y.Y.; Moon, Y.S.; Chung, I.K.; Chang, H.W.; Kim, C.H.; Choi, Y.H.; et al. Comparative proteome analysis of tumor necrosis factor alpha-stimulated human vascular smooth muscle cells in response to melittin. Proteome Sci. 2013, 11. [Google Scholar] [CrossRef] [PubMed]
  47. Park, H.J.; Son, D.J.; Lee, C.W.; Choi, M.S.; Lee, U.S.; Song, H.S.; Lee, J.M.; Hong, J.T. Melittin inhibits inflammatory target gene expression and mediator generation via interaction with IkappaB kinase. Biochem. Pharmacol. 2007, 73, 237–247. [Google Scholar] [CrossRef] [PubMed]
  48. Park, H.J.; Lee, H.J.; Choi, M.S.; Son, D.J.; Song, H.S.; Song, M.J.; Lee, J.M.; Han, S.B.; Kim, Y.; Hong, J.T. JNK pathway is involved in the inhibition of inflammatory target gene expression and NF-kappaB activation by melittin. J. Inflamm. 2008, 5. [Google Scholar] [CrossRef] [PubMed]
  49. Park, J.H.; Kum, Y.S.; Lee, T.I.; Kim, S.J.; Lee, W.R.; Kim, B.I.; Kim, H.S.; Kim, K.H.; Park, K.K. Melittin attenuates liver injury in thioacetamide-treated mice through modulating inflammation and fibrogenesis. Exp. Biol. Med. 2011, 236, 1306–1313. [Google Scholar] [CrossRef] [PubMed]
  50. Park, J.H.; Kim, K.H.; Lee, W.R.; Han, S.M.; Park, K.K. Protective effect of melittin on inflammation and apoptosis in acute liver failure. Apoptosis 2012, 17, 61–69. [Google Scholar] [CrossRef] [PubMed]
  51. Park, J.H.; Lee, W.R.; Kim, H.S.; Han, S.M.; Chang, Y.C.; Park, K.K. Protective effects of melittin on tumor necrosis factor-alpha induced hepatic damage through suppression of apoptotic pathway and nuclear factor-kappa B activation. Exp. Biol. Med. 2014, 239, 1705–1714. [Google Scholar] [CrossRef] [PubMed]
  52. Kim, K.H.; Sung, H.J.; Lee, W.R.; An, H.J.; Kim, J.Y.; Pak, S.C.; Han, S.M.; Park, K.K. Effects of melittin treatment in cholangitis and biliary fibrosis in a model of xenobiotic-induced cholestasis in mice. Toxins 2015, 7, 3372–3387. [Google Scholar] [CrossRef] [PubMed]
  53. Zarei, S.; Carr, K.; Reiley, L.; Diaz, K.; Guerra, O.; Altamirano, P.F.; Pagani, W.; Lodin, D.; Orozco, G.; Chinea, A. A comprehensive review of amyotrophic lateral sclerosis. Surg. Neurol. Int. 2015, 6. [Google Scholar] [CrossRef] [PubMed]
  54. Dantas, C.G.; Nunes, T.L.G.M.; Paixão, A.O.; Reis, F.P.; Júnior, W.L.; Cardoso, J.C.; Gramacho, K.P.; Gomes, M.Z. Pharmacological evaluation of bee venom and melittin. Rev. Bras. Farmacogn. 2014, 24, 67–72. [Google Scholar] [CrossRef]
  55. Ross, R. Atherosclerosis--an inflammatory disease. N. Engl. J. Med. 1999, 340, 115–126. [Google Scholar] [CrossRef]
  56. Kucharczak, J.; Simmons, M.J.; Fan, Y.; Gelinas, C. To be, or not to be: NF-kappaB is the answer—Role of Rel/NF-kappaB in the regulation of apoptosis. Oncogene 2003, 22, 8961–8982. [Google Scholar] [CrossRef] [PubMed]
  57. Pamukcu, B.; Lip, G.Y.; Shantsila, E. The nuclear factor—kappa B pathway in atherosclerosis: A potential therapeutic target for atherothrombotic vascular disease. Thromb. Res. 2011, 128, 117–123. [Google Scholar] [CrossRef] [PubMed]
  58. Damianoglou, A.; Rodger, A.; Pridmore, C.; Dafforn, T.R.; Mosely, J.A.; Sanderson, J.M.; Hicks, M.R. The synergistic action of melittin and phospholipase A2 with lipid membranes: Development of linear dichroism for membrane-insertion kinetics. Protein Pept. Lett. 2010, 17, 1351–1362. [Google Scholar] [CrossRef] [PubMed]
  59. Paull, B.R.; Yunginger, J.W.; Gleich, G.J. Melittin: An allergen of honeybee venom. J. Allergy Clin. Immunol. 1977, 59, 334–338. [Google Scholar] [CrossRef]
  60. Sobotka, A.K.; Franklin, R.M.; Adkinson, N.F., Jr.; Valentine, M.; Baer, H.; Lichtenstein, L.M. Allergy to insect stings. II. Phospholipase A: The major allergen in honeybee venom. J. Allergy Clin. Immunol. 1976, 57, 29–40. [Google Scholar] [CrossRef]
  61. Tosteson, M.T.; Holmes, S.J.; Razin, M.; Tosteson, D.C. Melittin lysis of red cells. J. Membr. Biol. 1985, 87, 35–44. [Google Scholar] [CrossRef] [PubMed]
  62. DeGrado, W.F.; Musso, G.F.; Lieber, M.; Kaiser, E.T.; Kezdy, F.J. Kinetics and mechanism of hemolysis induced by melittin and by a synthetic melittin analogue. Biophys. J. 1982, 37, 329–338. [Google Scholar] [CrossRef]
  63. Gajski, G.; Domijan, A.M.; Zegura, B.; Stern, A.; Geric, M.; Novak Jovanovic, I.; Vrhovac, I.; Madunic, J.; Breljak, D.; Filipic, M.; et al. Melittin induced cytogenetic damage, oxidative stress and changes in gene expression in human peripheral blood lymphocytes. Toxicon 2016, 110, 56–67. [Google Scholar] [CrossRef] [PubMed]
  64. Asthana, N.; Yadav, S.P.; Ghosh, J.K. Dissection of antibacterial and toxic activity of melittin: A leucine zipper motif plays a crucial role in determining its hemolytic activity but not antibacterial activity. J. Biol. Chem. 2004, 279, 55042–55050. [Google Scholar] [CrossRef] [PubMed]
  65. Rayahin, J.E.; Buhrman, J.S.; Gemeinhart, R.A. Melittin-glutathione S-transferase fusion protein exhibits anti-inflammatory properties and minimal toxicity. Eur. J. Pharm. Sci. 2014, 65, 112–121. [Google Scholar] [CrossRef] [PubMed]
  66. Soman, N.R.; Baldwin, S.L.; Hu, G.; Marsh, J.N.; Lanza, G.M.; Heuser, J.E.; Arbeit, J.M.; Wickline, S.A.; Schlesinger, P.H. Molecularly targeted nanocarriers deliver the cytolytic peptide melittin specifically to tumor cells in mice, reducing tumor growth. J. Clin. Investig. 2009, 119, 2830–2842. [Google Scholar] [CrossRef] [PubMed]
  67. Bei, C.; Bindu, T.; Remant, K.C.; Peisheng, X. Dual secured nano-melittin for the safe and effective eradication of cancer cells. J. Mater. Chem. B Mater. Biol. Med. 2015, 3, 25–29. [Google Scholar] [PubMed]
  68. Kreil, G.; Bachmayer, H. Biosynthesis of melittin, a toxic peptide from bee venom. Detection of a possible precursor. Eur. J. Biochem. 1971, 20, 344–350. [Google Scholar] [CrossRef] [PubMed]
  69. Tosteson, M.T.; Levy, J.J.; Caporale, L.H.; Rosenblatt, M.; Tosteson, D.C. Solid-phase synthesis of melittin: Purification and functional characterization. Biochemistry 1987, 26, 6627–6631. [Google Scholar] [CrossRef] [PubMed]
  70. Subbalakshmi, C.; Nagaraj, R.; Sitaram, N. Biological activities of C-terminal 15-residue synthetic fragment of melittin: Design of an analog with improved antibacterial activity. FEBS Lett. 1999, 448, 62–66. [Google Scholar] [CrossRef]
  71. Lee, G.; Bae, H. Bee venom phospholipase A2: Yesterday’s enemy becomes today’s friend. Toxins 2016, 8. [Google Scholar] [CrossRef] [PubMed]
  72. Sharma, S.V. Melittin-induced hyperactivation of phospholipase A2 activity and calcium influx in ras-transformed cells. Oncogene 1993, 8, 939–947. [Google Scholar] [PubMed]
Figure 1. Major mechanisms for the anti-inflammatory activities of melittin. Melittin suppresses signal pathways of TLR2, TLR4, CD14, NEMO, and PDGFRβ. By inhibiting these pathways melittin decreases activation of p38, ERK1/2, AKT, PLCγ1 as well as translocation of NF-κB into the nucleus. This inhibition results in reduced inflammation in skin, arota, joint, liver, and neuronal tissue. TLR, toll-like receptor; CD, cluster of differentiation; NEMO, nuclear factor kappa-B essential modulator; PDGFRβ, Platelet-derived growth factor receptor beta.
Figure 1. Major mechanisms for the anti-inflammatory activities of melittin. Melittin suppresses signal pathways of TLR2, TLR4, CD14, NEMO, and PDGFRβ. By inhibiting these pathways melittin decreases activation of p38, ERK1/2, AKT, PLCγ1 as well as translocation of NF-κB into the nucleus. This inhibition results in reduced inflammation in skin, arota, joint, liver, and neuronal tissue. TLR, toll-like receptor; CD, cluster of differentiation; NEMO, nuclear factor kappa-B essential modulator; PDGFRβ, Platelet-derived growth factor receptor beta.
Molecules 21 00616 g001
Table 1. Anti-inflammatory effects of melittin.
Table 1. Anti-inflammatory effects of melittin.
Disease ModelSpecific EffectsExperimental SystemDoseReference
Acne vulgaris
  • Reduced IKK, IκB, NF-κB and p38 phosphorylation
HaCaT cells, in vitro0.1–1 µg/mL[37]
  • Reduced swelling and granulomatous responses
mouse, in vivo, melittin and vaseline mixture applied to the surface of ear1–100 µg/ear
  • Suppressed TLR2 and CD14
  • Inhibited mRNA expression of TNF-α, IL-1β, IL-8, and IFN-γ.
  • Decreased expression of TNF-α and IL-1β by regulation of TLR2 and 4
human THP-1 monocytic cell, in vitro0.1–1 µg/mL[38]
  • Inhibited apoptosis and cleavage of caspase-3, -8, and PARP
Neuro inflammtion
  • Suppressed NO and iNOS expression
BV2 microglia, in vitro0.5–2 µg/mL[39]
  • Suppressed NF-κB activation by blocking degradation of IκBα and phosphorylation JNK and Akt
  • Suppressed expression IL-1β, IL-6, TNF-α, PGE2
  • Increased cell viability and decrease apoptosis
SH-SY5Y cells, in vitro0.5–2 µg/mL[40]
Amyotrophic lateral sclerosis
  • Decreased number of microglia and phospo-p38 in the spinal cord and brainstem
mouse, in vivo, s.c. injection at ST36 acupoint twice a week0.1 µg/g[41]
  • Improved motor function and inhibit neuronal death in the spinal cord
  • Inhibited a-synuclein misfolding
  • Suppressed expression of Iba-1 and CD14 in the lung
mouse, in vivo, s.c. injection at ST36 acupoint three times a week0.1 µg/g[42]
  • Suppressed expression of CD14 and COX-2 in spleen
  • Increased pERK and Bcl-2 in spleen
Atherosclerosis
  • Inhibited PDGR β-tyrosine phosphorylation and its intracellular signal transduction
rat aortic vascular smooth muscle cell, in vitro0.4–0.8 µg/mL[43,44]
  • Decreased total cholesterol and triglyceride but increased HDL in serum
mouse, in vivo, i.p. injection, twice a week0.1 mg/kg[45]
  • Decreased expression of TNF-α, IL-1β, VCAM-1, ICAM-1, and TGF-β1
  • Inhibited expression IL-1β, TNF-α and NF-κB activation
human monocytic cell line THP-1 derived macrophages, in vitro0.1–1 µg/mL
  • Increased prohibitin, annexin-1 expression
TNF-α stimulated human vascular smooth muscle cells, in vitro2 µg/mL[46]
  • Inhibited calreticulin expression reduced the phosphorylation of EGFR, and ERK and the expression of NF-κB in nuclear
Arthritis
  • Inhibited expression of LPS-induced COX-2, PGE2, cPLA2, NO and iNOS
raw 264.7 and synoviocytes obtained from patients with rheumatoid arthritis, in vitro5–10 µg/mL[20,47,48]
  • Inhibited JUK and NF-κB activation, release of IκB, and nuclear translocation of the p50 subunit
Liver inflammation
  • Suppressed inflammation, fibrosis, and expression of VCAM-1, IL-6 and TNF-α in the liver
mouse, in vivo, i.p. injection, twice a week for 12 weeks0.1 mg/kg[49]
  • Suppressed expression of IL-1β, IL-6 and TNF-α
rat primary hepatic stellate cells, in vitro0.1–1 µg/mL
  • Suppressed apoptosis and TNF-α, IL-1β and NF-κB signaling in GalN/LPS induced acute hepatic failure
mouse, in vivo, i.p. injection0.1 mg/kg[50]
  • Suppressed apoptotic pathway and NF-κB activation
mouse hepatocyte cell lines AML120.5–2 µg/mL[51]
  • Suppressed expressions of TNF-α, IL-6 and p-STAT3 in chronic liver injury
mouse, in vivo, i.p. injection, twice a week for 4 weeks0.1 mg/kg[52]

Share and Cite

MDPI and ACS Style

Lee, G.; Bae, H. Anti-Inflammatory Applications of Melittin, a Major Component of Bee Venom: Detailed Mechanism of Action and Adverse Effects. Molecules 2016, 21, 616. https://doi.org/10.3390/molecules21050616

AMA Style

Lee G, Bae H. Anti-Inflammatory Applications of Melittin, a Major Component of Bee Venom: Detailed Mechanism of Action and Adverse Effects. Molecules. 2016; 21(5):616. https://doi.org/10.3390/molecules21050616

Chicago/Turabian Style

Lee, Gihyun, and Hyunsu Bae. 2016. "Anti-Inflammatory Applications of Melittin, a Major Component of Bee Venom: Detailed Mechanism of Action and Adverse Effects" Molecules 21, no. 5: 616. https://doi.org/10.3390/molecules21050616

Article Metrics

Back to TopTop