Iranian Journal of Medical Sciences

Document Type : Review Article

Authors

1 Research Center for Health Sciences, Institute of Health, Department of Medical Entomology and Vector Control, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran

2 Li Ka Shing Center for Health Research and Innovation, University of Alberta, Edmonton, AB, Canada

3 Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran

Abstract

Venomous arthropods such as scorpions and bees form one of the important groups with an essential role in medical entomology. Their venom possesses a mixture of diverse compounds, such as peptides, some of which have toxic effects, and enzymatic peptide Phospholipase A2 (PLA2) with a pharmacological potential in the treatment of a wide range of diseases. Bee and scorpion venom PLA2 group III has been used in immunotherapy, the treatment of neurodegenerative and inflammatory diseases. They were assessed for antinociceptive, wound healing, anti-cancer, anti-viral, anti-bacterial, anti-parasitic, and anti-angiogenesis effects. PLA2 has been identified in different species of scorpions and bees. The anti-leishmania, anti-bacterial, anti-viral, and anti-malarial activities of scorpion PLA2 still need further investigation. Many pieces of research have been stopped in the laboratory stage, and several studies need vast investigation in the clinical phase to show the pharmacological potential of PLA2. In this review, the medical significance of PLA2 from the venom of two arthropods, namely bees and scorpions, is discussed.

Keywords

What’s Known

The venom of arthropods such as scorpions and bees is a complex mixture of polypeptides, bioactive proteins, and enzymes. Phospholipase A2 (PLA2) is one of the important characterized enzymes in the venom, with different therapeutic potentials in medicine.

What’s New

Newly discovered mechanism of phospholipase A2 (PLA2) in disease treatment Considering the results of the latest research on bee and scorpion venom PLA2 in the treatment of different diseases New results related to the anti-viral activity and wound healing effects of bee venom PLA2

Introduction

Arthropods, as the largest phylum of animals, include the insect, arachnid, crustacean, and myriapod classes, which constitute approximately 80% of the known species on earth. 1 - 3 Bees and scorpions are two species in this phylum reported to be under investigation, with a focus on their venom based on its pharmacological, medical, and industrial significance. 1 - 4 Scorpions belong to the Arachnida class and order with a variety of about 2200 species. 5 , 6 They utilize an apparatus called the venom gland and telson in caching their pray, defending against predators, and some intraspecific communications. 7 The venom contains a wide range of peptides, proteins, mucoproteins, enzymes such as L-amino acid oxidase, a serine protease, hyaluronidase, and metalloproteinase, nucleotides, salt, biogenic amines, and phospholipase A2 (PLA2). 8 - 10 The peptides from scorpion venom were shown to have anti-cancer, anti-bacterial, anti-viral, anti-parasitic, and anti-epileptic properties, and can potentiate bradykinin activities as well. 11 - 18

Honey bees have an important role in pollinating and honey production and have a venom gland in their abdominal cavity as a weapon to protect themselves against predators. 19 The venom consisted of a mixture of a variety of compounds, including peptides (apamin, adolapin, melittin), bioactive amines, several non-peptide compounds (free amino acids and lipids), and enzymes (hyaluronidase and PLA2). 20 - 23 Several studies demonstrated that bee venom has radio-protective, anti-nociceptive, anti-inflammatory, and anti-mutagenic activity. 24 - 30 The PLA2 in the venom of arthropods such as bees and scorpions belongs to the PLA2 group III and has a wide range of pharmacological properties. 31 PLA2 hydrolyzes glycerophospholipids at the sn-2 position and results in the release of fatty acids and lysophospholipids. 32 In mammals, secretory phospholipase A2 (sPLA2) has an important role in proliferation, maturation, and inflammation, 33 - 35 while bee venom sPLA2 of group III is also identical to the mammalian sPLA2s. 36 This review investigated the bee and scorpion venom PLA2 and its medical therapeutic potential.

Pharmacological Function of Bee Venom PLA2

Specific Immunotherapy (SIT)

Specific immunotherapy is defined as an increase in the allergen doses via the subcutaneous route. 37 Nowadays, bee venom injection is used to treat bee venom allergy in 95% of sensitive patients with local and systemic anaphylactic reactions. 38 - 41 Bee venom immunotherapy has an important role in specific immunotherapy, providing natural immunity against venom allergens, but it sometimes leads to allergic side effects among 20-40% of the patients. 40 , 42 In these patients, PLA2 is responsible for allergens and immunogenic particles up to 80%. 42 - 44 The PLA2 in bee venom consisted of 12% whole venom proteins. 44 In PLA2, three T-cell epitopes have been identified, including PLA45-62 (PI), PLA82-92 (PII), and PLA113-124 (PIII). 42

In venom therapy, T helper type 1 (Th1) (Interleukin-2 [IL-2] and Interferon-gamma [IFN-γ]) and T helper type 2 (Th2) (IL-4, IL-5, and IL-13) cells shift to IL-10 secreting Type 1 regulatory T-cells (Tr1 cell), and this process leads to a decrease in T-cell responsiveness. So, Th1- and Th2-type cytokine secretions and T-cell proliferation are suppressed. 40 Regulatory T-cells (Tregs) have an important role in allergic reactions by retaining immune homeostasis and regulatory functions. 45 Additionally, Tregs suppress immunoglobulin E (IgE) antibody production and increase immunoglobulin G4 (IgG4) production as non-inflammatory isotypes. 40 , 46 Some studies have shown that venom immunotherapy reduces the PLA-IgE antibody but increases immunoglobulin G2a (IgG2a), which can inhibit anaphylactic reactions. 47

Cancer Therapy

Bee venom can induce apoptosis in cancer cells and inhibit their growth by increasing intracellular Ca2+ and reactive oxygen species (ROS). 48 - 50 The two important compounds in bee venom are melittin (an amphiphilic peptide) and PLA2, which induce necrosis, cytolysis, and apoptosis through the enhancement of Ca2+ entry and calpain activity, as well as death receptor signaling activation. 1 , 51 - 53 Actually, in the apoptosis process, caspase-3 is activated by bee venom in the synovial fibroblasts. 54 Several studies have revealed that cancer cells are destroyed by the activation of matrix metalloproteinase (MMP) and caspase, a mechanism that is related to bee venom anti-tumor activity. 48 , 55 - 57

Several cancer cells, such as leukemic cells and breast, prostate, lung, liver, renal, mammary, and bladder cancer cells, can be treated by melittin and PLA2 of bee venom. 29 , 48 , 49 , 58 The melittin in bee venom is a PLA2 activator. 59 - 62 The activation of PLA2 via melittin, as an antiapoptotic factor, indicates the anti-tumor activity of bee venom, 48 which can disrupt the cell membrane by moving the anions and increasing their cytotoxic effect. 63 Moreover, PLA2 can also influence the proliferation of tumor cells. 48 In renal cancer, it was shown that bee venom PLA2 with the combined effect of phosphatidylinositol-(3, 4)-bisphosphate (PtdIns (3, 4) P2) has a synergistic effect in inducing cell death. 64 , 65

Neurodegenerative Disease Therapy

The potential mechanism in group III sPLA2, such as bee venom PLA2, is a therapeutic strategy for prion diseases, as a family of neurodegenerative diseases, which is characterized by gliosis and neuronal vacuolation. 66 Prion diseases are disorders that occur with the misfolding of the isoform or protease-resistant prion protein (PrPcs) of the cellular prion protein (PrPc). The overexpression of PrPc by prion protein (PrP) peptide 106-126 induces neurotoxicity. 67 PLA2, which is expressed in the peripheral and central nervous system (CNS), 35 , 68 activates the protein kinase B (AKT) in mammals and regulates neuronal survival and outgrowth. 36 In neuritogenesis, sPLA2 modulates phosphatidylinositol 3- kinase (PI3K)/ protein kinase B (AKT) signaling, so that PrP (106-126)-mediated neurotoxicity is blocked. 35 , 69

The potential mechanism in group III sPLA2, such as bee venom PLA2, is a therapeutic strategy for prion disease characterized by gliosis and neuronal vacuolation. 66 In fact, bee venom PLA2 is a protective agent against PrP(106-126)-mediated neurotoxicity in prion diseases. 69 Alzheimer’s disease is another degenerative disease of the CNS that is relevant to neurotoxic microglial activation. 70 , 71 Microglia activation has an important role in the accumulation of amyloid-beta (Aβ), which is associated with the progression of this disease via the secretion of tumor necrosis factor-alpha (TNF-α), IL-6, IL-1, and free radicals as inflammatory molecules. 72 , 73 A study has indicated that bee venom PLA2 decreases the activation of neurotoxic microglial in triple-transgenic (3xTg) mouse models. 70 On the other hand, bee venom PLA2 alters the apoptotic signal pathway and induces Treg expansion. In fact, bvPLA2 increases CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) and PD-1 (programmed cell death protein 1) as anti-apoptotic molecules. 74

Parkinson’s disease (PD) is another age-related neurodegenerative disease that is extremely common after Alzheimer’s disease and is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). 75 - 77 PD is associated with the accumulation of post-translationally modified alpha-synuclein (α-syn), which results in the activation of microglial, interleukin-1β (IL-1β), TNFα, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase enzyme, and ROS, affecting the CNS and speeding up PD pathogenesis. 78 Several studies have illustrated that bee venom PLA2 can inhibit the apoptosis of dopaminergic neurons and protect glutamate-induced neurotoxicity. 76 , 79 - 83 Technically, bee venom phospholipase A2 (bvPLA2) promotes the survival of dopaminergic neurons through Treg overexpansion. 84 On the other hand, in transgenic mice, it was shown that bee venom PLA2 can decrease the activation of microglia in A53T transgenic mice, so that α-syn, which is accumulated in the spinal cord, is reduced. This can be considered as a novel strategy for the treatment of PD. 83

Anti-inflammatory Effect

Bee venom is used as anti-inflammatory medication in the treatment of chronic inflammatory diseases such as multiple sclerosis and rheumatoid arthritis. 85 - 90 Recent investigations have shown that bee venom PLA2 is responsible for the anti-inflammation effects by reducing the aggregation of immune cells. 88 , 91 - 93

Shine and others found that bee venom PLA2 has therapeutic effects via the depletion of Tregs and is a good candidate for the treatment of fibrosis and radiation pneumonitis. 88 These Tregs are influenced by bee venom PLA2 in the airways, which leads to the prevention of the chronic obstructive disease of allergic asthmatic symptoms, influencing the release of IL-4, IL-5, and IL-13, the production of allergen-specific IgE, and the aggregation of eosinophils and basophils in the airways. 94 - 97

Bee venom PLA2 is capable of neuroprotection and suppressing the microglial activation, which is activated in degenerative neurons such as PD. 84 Bee venom PLA2 can induce Treg differentiation and suppress the secretion of prostaglandin E2 (PGE2) by a cluster of differentiation 26 (CD206)+dendritic cells (DCs). PLA2-stimulated DCs release PGE2 which binds to a cluster of differentiation 4 (CD4)+T cells on the prostaglandin E2 (PGE2) receptor 2 subtype (EP2) and regulates the expression of forkhead box P3 (Foxp3). 84 So, bee venom PLA2 can be a pharmacological candidate for treating neuro-inflammatory diseases such as PD.

It has been demonstrated that in the treatment of PD, bee venom PLA2 (bvPLA2) has anti-inflammatory effects and neuro-protective activity by the inhibition of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). 98 It has also been noted that bee venom PLA2 has an important role in anti-inflammatory effects and Alzheimer’s disease amelioration through the modulation of Tregs and production of IL-10. 99 , 100 Bee venom PLA2 can ameliorate memory impairment by the inhibition of nuclear factor kappa B (NF-κB), a key transcription factor, 101 and inhibit the phosphorylation of signal transducer and activator of transcription 3 (STAT3), leading to the anti-inflammatory effects. 101

It was shown that bee venom PLA2 can modulate the Treg cell population and prevent Cis-diamminedichloroplatinum (cisplatin)-induced renal inflammation and nephrotoxicity. 91 , 102 In 2014, Kim and others found that bee venom PLA2 has hepatoprotective effects by modulating the Treg and inducing IL-10 in acetaminophen-induced acute toxicity in liver and kidneys. 103 The investigation showed that bee venom PLA2 decreases lipid accumulation in the aorta and foam cells formation by increasing Treg cells. Technically, increasing the Treg cells leads to an increase in high-density lipoprotein cholesterol (HDL-C) and a decrease in low-density lipoprotein cholesterol (LDL-C). Therefore, bee venom PLA2 can be a potential therapeutic agent for atherosclerosis disease. 104

Antinociceptive Effect

Bee venom is used to treat diseases such as tendonitis, wounds, bursitis, shingles, and even burns. 105 In addition, it is also used as an antinociceptive in patients. 20 , 77 , 86 , 105 , 106 Studies have indicated that pre-treatment by bee venom PLA2 has three crucial roles in patients using oxaliplatin, which is widely prescribed for lung, ovarian, and breast cancers. It can prevent oxaliplatin-induced neuropathic pains by inhibiting the development of cold and mechanical allodynia, inhibiting macrophage infiltration, and decreasing IL-1β in the lumbar dorsal root ganglia. 107 - 110

Wound-healing Effects

Bee venom can promote wound healing by enhancing the responses of toll-like receptor 3 in keratinocytes. In other words, it increases the uptake of polyinosinic: polycytidylic acid, and affects the production of IL-8 in keratinocytes, leading to the therapeutic potential of bee venom PLA2 for healing skin wounds. 111

Atopic dermatitis, also known as atopic eczema, is a biphasic inflammatory skin disorder. 112 It was shown that bee venom PLA2 reduces atopic skin lesion inflammation in n Balb/c mouse models, induced by 2,4-dinitrochlorobenzene (DNCB), and house dust mite, Dermatophagoides farinae extract (DFE). 113 Bee venom PLA2 inhibited the cytokine levels, serum IgE, the infiltration of mast cells (MCs), and epidermal thickness in an atopic dermatitis model, induced by the interaction of DNCB and DFE with CD206 mannose receptor. 114

Anti-virus Activity

Hewawaduge and others indicated that bee venom PLA2 blocks the receptors on the cell surface involved in virus attachment and inhibits virus replication, revealing the in vitro antiviral activity of bee venom PLA2 against coxsackievirus (H3), adenovirus (AdV), enterovirus-71 (EV-71), vesicular stomatitis virus (VSV), and herpes simplex virus (HSV). 115 It was demonstrated that the p3bv peptide (amino acids 21 to 35 of bee venom PLA2) inhibits human immunodeficiency virus 1 (HIV-1) replication by binding to CXC-chemokine receptor 4 (CXCR4). 116 Therefore, the pharmacological potential of bee venom PLA2 against viruses was suggested.

Anti-parasitic and Anti-bacterial Activities

Bee venom PLA2 was shown to have a specific role in killing or inhibiting gram-negative bacteria (Enterobacter cloacae, Citrobacter freundii, Escherichia coli) and Trypanosoma brucei brucei, which causes sleep sickness in tropical counties. 117 , 118 The antibacterial activity of bee venom PLA2 was demonstrated against gram-positive bacteria, such as Staphylococcus aureus, and gram-negative bacteria, including Escherichia coli, Pseudomonas aeruginosa, and Klebsiella aerogenes. 119 It was noted that the expression of bee venom PLA2 is decreased in transgenic mosquitos (Anopheles stephensi) by blocking the oocyst formation of Plasmodium berghei and interfering with the transmission of the parasite. 120 Additionally, bee venom PLA2 can induce stage-specific growth arrest in intraerythrocytic Plasmodium falciparum by modifying serum lipoproteins. 121

All information related to the pharmacological function of bee venom PLA2 is gathered in table 1.

References Source Pharmacological function Disease Mechanism
Akdis CA et al. 46 von Garnier C et al. 47 Hori S et al. 35 von Ozdemir C et al. 40 Bee venom Specific immunotherapy (SIT) Allergy 1. Conversion Th1 and Th2 cells to IL-10, suppressing the T cell proliferation and cytokine secretions
2. Suppressing IgE antibody production by Tregs and increasing IgG4 production
3. Reducing PLA-IgE antibody but increasing IgG2a for anaphylactic reaction inhibition
Lad PJ et al. 63 Jang M-H et al. 49 Oršolić N et al. 29 Moon D-O et al. 55 Rahman KW et al. 57 Putz T et al. 64 Li B et al. 58 Putz T et al. 65 Cho H-J et al. 56 Ozdemir C et al. 40 Oršolić N et al. 48 Nabiuni M et al. 51 Gajski G et al. 53 Chaisakul J et al. 50 Kong G-M et al. 52 Cancer therapy Leukemic cells 1. Increasing intracellular Ca2+, reactive oxygen species (ROS), enhancing calpain activity, and death receptor signaling activation
Breast cancer
Prostate cancer
Lung cancer
Liver cancer 2. Activation of matrix metalloproteinase (MMP), and caspase
Renal cancer 3. disrupting the cell membrane
Mammary cancer 4. Inducing cell death with the cooperation of phosphatidylinositol-(3, 4)-bisphosphate (PtdIns (3, 4) P2)
Bladder cancer
Brandner S et al. 66 Jeong J-K et al. 69 Neurodegenerative disease therapy Prion Blocking PrP(106-126)-mediated
Ye M et al. 70 Baek H et al. 74 Alzheimer 1. Decreasing the activation of neurotoxic microglia
2. Increases CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) and PD-1 (Programmed cell death protein 1)
Doo A-R et al. 80 Doo A-R et al. 79 Lee SM et al. 81 Chung ES et al. 84 Ye M et al. 83 Awad K et al. 76 Parkinson 1. Inhibiting the apoptosis of dopaminergic neurons and protecting glutamate-induced neurotoxicity
2. Promote the survival of dopaminergic neurons through Treg overexpression
3. Reducing the accumulation of α-syn in the spinal cord
Lee J-D et al. 27 Castro HJ et al. 87 Liu X et al. 85 Lee G et al. 93 Anti-inflammatory effect Multiple sclerosis Rheumatoid arthritis Reducing the aggregation of immune cells
Shin D et al. 88 Fibrosis radiation pneumonitis Depletion of Tregs
Park et al. 97 Allergic asthmatic Production of allergen-specific IgE and aggregation of eosinophils and basophils
Hirsch E et al. 122 Block ML et al. 71 Chung ES et al. 84 Parkinson 1. Suppressing the secretion of prostaglandin E2 (PGE2) PGE2 by CD206 (cluster of differentiation 26) CD206 + dendritic cells (DCs)
2. Neuroprotective activity by inhibition of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)
Chen C-H et al. 100 Baek H et al. 74 Alzheimer Modulation of Tregs and production of IL-10
Kang G-H et al. 104 Atherosclerosis Increasing the Treg cells, increasing high-density lipoprotein cholesterol (HDL-C), and decreasing low-density lipoprotein cholesterol (LDL-C)
Arany I et al. 102 Kim H et al. 103 Liver injury Modulation of the Treg cell Treg and inducing IL-10 in acetaminophen-induced acute toxicity
Arany I et al. 102 Kim H et al. 91 Kidney injury Modulate Treg cells population and prevent Cis-diamminedichloroplatinum (cisplatin)-induced renal inflammation and nephrotoxicity
Muggia FM et al. 108 Petit T et al. 109 Alcindor T et al. 107 Li D et al. 110 Antinociceptive effect - Inhibition of the development of cold and mechanical allodynia and the inhibition of macrophage infiltration and decreasing the IL-1β
Nakashima A et al. 111 Wound healing effects Skin wounds Increasing the polyinosinic: polycytidylic acid (poly(I: C)) uptake and production of IL-8 in keratinocyte
Jung KH et al. 113 Shin D et al. 114 Atopic dermatitis Interaction with CD206 mannose receptor
Fenard D et al. 116 Hewawaduge C et al. 115 Anti-virus activity Coxsackievirus (H3), Adenovirus (AdV), Enterovirus-71 (EV-71), Vesicular stomatitis virus (VSV), Herpes simplex virus (HSV), and Human immunodeficiency virus 1 (HIV-1) Blocking cell surface receptors
Boulanger N et al. 117 Perumal Samy et al. 119 Boutrin MCF at al. 118 Antiparasite and anti-bacterial activities Sleep sickness (Trypanosoma brucei brucei) Killing or growth-inhibiting
Gram-negative bacteria (Enterobacter cloacae, Citrobacter freundii, Escherichia coli, Pseudomonas aeruginosa, and Klebsiella aerogenes)
Gram-positive bacteria (Staphylococcus aureus)
Deregnaucourt C et al. 121 Moreira LA et al. 120 Malaria (Plasmodium berghei and Plasmodium falciparum) 1. Blocking oocyst formation of Plasmodium berghei in Anopheles stephensi mosquito
2. Growth arrest of the intraerythrocytic stage in Plasmodium falciparum
Table 1.Pharmacological function of bee venom Phospholipase A2

Pharmacological Function of Scorpion Venom PLA2

Anti-angiogenesis and Anti-tumoral Activity

So far, several PLA2s have been identified in the venoms of different scorpion species, including enterotoxin from Heterometrus laoticus, 123 hemilipin and hemilipin2 from the Iranian scorpion Hemiscorpius lepturus, 124 , 125 phospholipin and imperatoxin from Pandinus imperator, 126 , 127 phaiodactylipin from Anuroctonus phaiodactylus, 128 and PLA2 from Scorpio maurus, 129 and Heterometrus fulvipes. 130 Angiogenesis is defined as the growth of new vessels from existing vessels. 131 Hemilipin is a heterodimeric protein, sPLA2 enzyme, which has been isolated from the venom of the Iranian scorpion Hemiscorpius lepturus. This enzyme reduces the expression levels of three important proangiogenic factors, including the vascular endothelial growth factor-A, -C, and -D (VEGF-A, VEGF-C, VEGF-D), the vascular endothelial growth factor receptor-1 and -2 (VEGFR-1 and VEGFR-2), hepatocyte growth factor (HGF), and endoglin expression (CD105). It also induces an anti-angiogenesis effect in vivo and in vitro, both on human umbilical vein endothelial cells (HUVECs) and human pulmonary artery endothelial cells (HPAECs), as well as the chick embryo chorioallantoic membrane (CAM). 124

Hemilipin2, a new heterodimeric PLA2 that has been identified in the Iranian scorpion Hemiscorpius lepturus, was shown to have anti-angiogenesis activity. A small subunit of Hemilipin2 was demonstrated in vitro and in vivo to have anti-angiogenesis activity in HUVECs, HPAECs, and CAM models, without any apoptotic or cytotoxic effect. 125 The recombinant protein of heterodimeric PLA2 found in Scorpio Maurus has shown anti-angiogenesis effects on HUVECs through the inhibition of migration, invasion, and adhesion activities, 132 while angiogenesis is a key process in metastasis and tumor growth, 133 which lends to its important role in cancer therapy. Native and recombinant proteins of PLA2 derived from Scorpio Maurus venom have demonstrated anti-tumor activities through interference with the function of α5β1 and αvβ3 integrin receptors in human vascular endothelial cells (HMEC-1). 134

All information related to the pharmacological function of scorpion venom PLA2 is presented in table 2.

References Source Species of scorpion Enzymes name Pharmacological function Mechanism
Jridi et al. 124 Scorpion venom Hemiscorpius lepturus Hemilipin Anti-angiogenesis Reducing the expression of VEGF-A, VEGF-C, VEGF-D, VEGFR-1, VEGFR-2, HGF, and CD105
Jridi et al. 125 Hemiscorpius lepturus Hemilipin2 Anti-angiogenesis -
Krayem N et al. 134 and Krayem N et al. 132 Scorpio Maurus PLA2 Anti-angiogenesis and anti-tumor Interference with α5β1 and αvβ3 integrin receptors function
Table 2.Pharmacological function of scorpion venom Phospholipase A2

Discussion

Venomous arthropods such as scorpions and bees are among the important groups with an essential significance in medical entomology. 135 Their venom possesses a mixture of diverse compounds, such as peptides, some of which have toxic effects, and enzymatic peptide PLA2, with pharmacological potential in the treatment of a wide range of diseases. 9 Therefore, studying the effectiveness of venom peptides is an extremely important task, which can lead to the introduction of new drug candidates in the treatment of various diseases. Bee and scorpion venom PLA2 group III has been used in immunotherapy and the treatment of neurodegenerative and inflammatory diseases. They were assessed for antinociceptive, wound-healing, anti-cancer, anti-viral, antibacterial, anti-parasitic, and anti-angiogenesis effects.

Conclusion

PLA2 group III has been identified in different species of scorpions and bees. The anti-leishmania, anti-bacterial, anti-viral, and anti-malarial activities of scorpion PLA2 still require further investigation. Many researches in this area have been stopped in the laboratory stage, and several studies need vast investigation in the clinical phase to show the pharmacological potential of PLA2.

Authors’ Contribution

P.S.A and K.A contributed to Study Design and Study Implementation, P.S.A, K.A, H.A, and D.M contributed to Analysis and Interpretation of Data, Major Contribution to Writing. All authors have read and approved the final manuscript and agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Conflict of Interest: None declared.

References

  1. Jo M, Park MH, Kollipara PS, An BJ, Song HS, Han SB, et al. 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. 2012; 258:72-81. DOI | PubMed
  2. Mamelak AN, Jacoby DB. Targeted delivery of antitumoral therapy to glioma and other malignancies with synthetic chlorotoxin (TM-601). Expert Opin Drug Deliv. 2007; 4:175-86. DOI | PubMed
  3. Remijsen Q, Verdonck F, Willems J. Parabutoporin, a cationic amphipathic peptide from scorpion venom: much more than an antibiotic. Toxicon. 2010; 55:180-5. DOI | PubMed
  4. Hider RC. Honeybee venom: a rich source of pharmacologically active peptides. Endeavour. 1988; 12:60-5. DOI | PubMed
  5. Najafian M, Ghorbani A, Zargar M, Baradaran M, Baradaran N. Scorpion stings in pregnancy: an analysis of outcomes in 66 envenomed pregnant patients in Iran. J Venom Anim Toxins Incl Trop Dis. 2020; 26:e20190039. Publisher Full Text | DOI | PubMed
  6. Lourenco WR. Scorpions and life-history strategies: from evolutionary dynamics toward the scorpionism problem. J Venom Anim Toxins Incl Trop Dis. 2018; 24:19. Publisher Full Text | DOI | PubMed
  7. Morgenstern D, Rohde BH, King GF, Tal T, Sher D, Zlotkin E. The tale of a resting gland: transcriptome of a replete venom gland from the scorpion Hottentotta judaicus. Toxicon. 2011; 57:695-703. DOI | PubMed
  8. Al-Asmari AK, Islam M, Al-Zahrani AM. In vitro analysis of the anticancer properties of scorpion venom in colorectal and breast cancer cell lines. Oncol Lett. 2016; 11:1256-62. Publisher Full Text | DOI | PubMed
  9. Ding J, Chua PJ, Bay BH, Gopalakrishnakone P. Scorpion venoms as a potential source of novel cancer therapeutic compounds. Exp Biol Med (Maywood). 2014; 239:387-93. DOI | PubMed
  10. Mander L, Liu HW. Comprehensive natural products II: chemistry and biology. Elsevier: Amsterdam; 2010.
  11. Salem ML, Shoukry NM, Teleb WK, Abdel-Daim MM, Abdel-Rahman MA. In vitro and in vivo antitumor effects of the Egyptian scorpion Androctonus amoreuxi venom in an Ehrlich ascites tumor model. Springerplus. 2016; 5:570. Publisher Full Text | DOI | PubMed
  12. Tong-ngam P, Roytrakul S, Sritanaudomchai H. BmKn-2 scorpion venom peptide for killing oral cancer cells by apoptosis. Asian Pac J Cancer Prev. 2015; 16:2807-11. DOI | PubMed
  13. de la Salud Bea R, Petraglia AF, Ascuitto MR, Buck QM. Antibacterial Activity and Toxicity of Analogs of Scorpion Venom IsCT Peptides. Antibiotics (Basel). 2017; 6Publisher Full Text | DOI | PubMed
  14. Hong W, Li T, Song Y, Zhang R, Zeng Z, Han S, et al. Inhibitory activity and mechanism of two scorpion venom peptides against herpes simplex virus type 1. Antiviral Res. 2014; 102:1-10. Publisher Full Text | DOI | PubMed
  15. Gao B, Xu J, Rodriguez Mdel C, Lanz-Mendoza H, Hernandez-Rivas R, Du W, et al. Characterization of two linear cationic antimalarial peptides in the scorpion Mesobuthus eupeus. Biochimie. 2010; 92:350-9. DOI | PubMed
  16. Flores-Solis D, Toledano Y, Rodriguez-Lima O, Cano-Sanchez P, Ramirez-Cordero BE, Landa A, et al. Solution structure and antiparasitic activity of scorpine-like peptides from Hoffmannihadrurus gertschi. FEBS Lett. 2016; 590:2286-96. DOI | PubMed
  17. Dehong MFWSG. Antiepileptic effect of scorpion venom on SD rats with Epilepsy induced by kainic acid [J]. Heilongjiang Medicine and Pharmacy. 2002; 6
  18. Araujo RL, Gomez MV. Potentiation of bradykinin action on smooth muscle by a scorpion venom extract. Gen Pharmacol. 1976; 7:123-6. DOI | PubMed
  19. Hora ZA, Altaye SZ, Wubie AJ, Li J. Proteomics Improves the New Understanding of Honeybee Biology. J Agric Food Chem. 2018; 66:3605-15. DOI | PubMed
  20. Son DJ, Lee JW, Lee YH, Song HS, Lee CK, Hong JT. Therapeutic application of anti-arthritis, pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol Ther. 2007; 115:246-70. DOI | PubMed
  21. Rady I, Siddiqui IA, Rady M, Mukhtar H. Melittin, a major peptide component of bee venom, and its conjugates in cancer therapy. Cancer Lett. 2017; 402:16-31. Publisher Full Text | DOI | PubMed
  22. Al-Ani I, Zimmermann S, Reichling J, Wink M. Pharmacological synergism of bee venom and melittin with antibiotics and plant secondary metabolites against multi-drug resistant microbial pathogens. Phytomedicine. 2015; 22:245-55. DOI | PubMed
  23. Zhou J, Zhao J, Zhang S, Shen J, Qi Y, Xue X, et al. Quantification of melittin and apamin in bee venom lyophilized powder from Apis mellifera by liquid chromatography-diode array detector-tandem mass spectrometry. Anal Biochem. 2010; 404:171-8. DOI | PubMed
  24. Lee G, Bae H. Bee Venom Phospholipase A2: Yesterday’s Enemy Becomes Today’s Friend. Toxins (Basel). 2016; 8:48. Publisher Full Text | DOI | PubMed
  25. Varanda EA, Monti R, Tavares DC. Inhibitory effect of propolis and bee venom on the mutagenicity of some direct- and indirect-acting mutagens. Teratog Carcinog Mutagen. 1999; 19:403-13. PubMed
  26. 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. DOI | PubMed
  27. Lee JY, Kang SS, Kim JH, Bae CS, Choi SH. Inhibitory effect of whole bee venom in adjuvant-induced arthritis. In Vivo. 2005; 19:801-5. PubMed
  28. Baek YH, Huh JE, Lee JD, Choi DY, Park DS. 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; 1073-1074:305-10. DOI | PubMed
  29. 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; 41:861-70. DOI | PubMed
  30. Orsolic N, Knezevic A, Sver L, Terzic S, Hackenberger BK, Basic I. Influence of honey bee products on transplantable murine tumours. Vet Comp Oncol. 2003; 1:216-26. DOI | PubMed
  31. Li JH, Zhang CX, Shen LR, Tang ZH, Cheng JA. Expression and regulation of phospholipase A2 in venom gland of the chinese honeybee, Apis cerana cerana. Arch Insect Biochem Physiol. 2005; 60:1-12. DOI | PubMed
  32. Kini RM. Excitement ahead: structure, function and mechanism of snake venom phospholipase A2 enzymes. Toxicon. 2003; 42:827-40. DOI | PubMed
  33. Sato H, Taketomi Y, Isogai Y, Miki Y, Yamamoto K, Masuda S, et al. Group III secreted phospholipase A2 regulates epididymal sperm maturation and fertility in mice. J Clin Invest. 2010; 120:1400-14. Publisher Full Text | DOI | PubMed
  34. Sato H, Kato R, Isogai Y, Saka G, Ohtsuki M, Taketomi Y, et al. Analyses of group III secreted phospholipase A2 transgenic mice reveal potential participation of this enzyme in plasma lipoprotein modification, macrophage foam cell formation, and atherosclerosis. J Biol Chem. 2008; 283:33483-97. Publisher Full Text | DOI | PubMed
  35. Masuda S, Yamamoto K, Hirabayashi T, Ishikawa Y, Ishii T, Kudo I, et al. Human group III secreted phospholipase A2 promotes neuronal outgrowth and survival. Biochem J. 2008; 409:429-38. DOI | PubMed
  36. Valentin E, Ghomashchi F, Gelb MH, Lazdunski M, Lambeau G. Novel human secreted phospholipase A(2) with homology to the group III bee venom enzyme. J Biol Chem. 2000; 275:7492-6. DOI | PubMed
  37. Guerin V, Dubarryr M, Robic D, Brachet F, Rautureau M, Andre C, et al. Microsphere entrapped bee-venom phospholipase A2 retains specific IgE binding capacity: a possible use for oral specific immunotherapy. J Microencapsul. 2002; 19:761-5. DOI | PubMed
  38. Urbanek R, Kemeny DM, Richards D. Sub-class of IgG anti-bee venom antibody produced during bee venom immunotherapy and its relationship to long-term protection from bee stings and following termination of venom immunotherapy. Clin Allergy. 1986; 16:317-22. DOI | PubMed
  39. Lichtenstein LM, Valentine MD, Sobotka AK. A case for venom treatment in anaphylactic sensitivity to hymenoptera sting. N Engl J Med. 1974; 290:1223-7. DOI | PubMed
  40. Ozdemir C, Kucuksezer UC, Akdis M, Akdis CA. Mechanisms of immunotherapy to wasp and bee venom. Clin Exp Allergy. 2011; 41:1226-34. DOI | PubMed
  41. Hunt KJ, Valentine MD, Sobotka AK, Benton AW, Amodio FJ, Lichtenstein LM. A controlled trial of immunotherapy in insect hypersensitivity. N Engl J Med. 1978; 299:157-61. DOI | PubMed
  42. Muller U, Akdis CA, Fricker M, Akdis M, Blesken T, Bettens F, et al. Successful immunotherapy with T-cell epitope peptides of bee venom phospholipase A2 induces specific T-cell anergy in patients allergic to bee venom. J Allergy Clin Immunol. 1998; 101:747-54. DOI | PubMed
  43. Akdis CA, Akdis M, Blesken T, Wymann D, Alkan SS, Muller U, et al. Epitope-specific T cell tolerance to phospholipase A2 in bee venom immunotherapy and recovery by IL-2 and IL-15 in vitro. J Clin Invest. 1996; 98:1676-83. Publisher Full Text | DOI | PubMed
  44. Irsch J, Konig C, Lohndorf A, Tesch H, Krieg T, Merk H, et al. The frequency of phospholipase A2 binding of basophilic granulocytes does not decrease during bee-venom-specific immunotherapy. Allergy. 1999; 54:742-7. DOI | PubMed
  45. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003; 299:1057-61. DOI | PubMed
  46. Akdis CA, Blesken T, Akdis M, Wuthrich B, Blaser K. Role of interleukin 10 in specific immunotherapy. J Clin Invest. 1998; 102:98-106. Publisher Full Text | DOI | PubMed
  47. von Garnier C, Astori M, Kettner A, Dufour N, Heusser C, Corradin G, et al. Allergen-derived long peptide immunotherapy down-regulates specific IgE response and protects from anaphylaxis. Eur J Immunol. 2000; 30:1638-45. DOI | PubMed
  48. Orsolic N. Bee venom in cancer therapy. Cancer Metastasis Rev. 2012; 31:173-94. DOI | PubMed
  49. Jang MH, Shin MC, Lim S, Han SM, Park HJ, Shin I, et al. 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. DOI | PubMed
  50. Chaisakul J, Hodgson WC, Kuruppu S, Prasongsook N. Effects of Animal Venoms and Toxins on Hallmarks of Cancer. J Cancer. 2016; 7:1571-8. Publisher Full Text | DOI | PubMed
  51. Nabiuni M, Safaeinejad Z, Parivar K, Divsalar A, Nazari Z. Antineoplastic effects of honey bee venom. Zahedan J Res Med Sci. 2013; 15:1-5.
  52. Kong GM, Tao WH, Diao YL, Fang PH, Wang JJ, Bo P, et al. Melittin induces human gastric cancer cell apoptosis via activation of mitochondrial pathway. World J Gastroenterol. 2016; 22:3186-95. Publisher Full Text | DOI | PubMed
  53. Gajski G, Garaj-Vrhovac V. Melittin: a lytic peptide with anticancer properties. Environ Toxicol Pharmacol. 2013; 36:697-705. DOI | PubMed
  54. Hong SJ, Rim GS, Yang HI, Yin CS, Koh HG, Jang MH, et al. Bee venom induces apoptosis through caspase-3 activation in synovial fibroblasts of patients with rheumatoid arthritis. Toxicon. 2005; 46:39-45. DOI | PubMed
  55. Moon DO, Park SY, Heo MS, Kim KC, Park C, Ko WS, et al. 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; 6:1796-807. DOI | PubMed
  56. Cho HJ, Jeong YJ, Park KK, Park YY, Chung IK, Lee KG, et al. Bee venom suppresses PMA-mediated MMP-9 gene activation via JNK/p38 and NF-kappaB-dependent mechanisms. J Ethnopharmacol. 2010; 127:662-8. DOI | PubMed
  57. Rahman KM, Sarkar FH, Banerjee S, Wang Z, Liao DJ, Hong X, et al. Therapeutic intervention of experimental breast cancer bone metastasis by indole-3-carbinol in SCID-human mouse model. Mol Cancer Ther. 2006; 5:2747-56. DOI | PubMed
  58. Li B, Gu W, Zhang C, Huang XQ, Han KQ, Ling CQ. Growth arrest and apoptosis of the human hepatocellular carcinoma cell line BEL-7402 induced by melittin. Onkologie. 2006; 29:367-71. DOI | PubMed
  59. Fletcher JE, Jiang MS. Possible mechanisms of action of cobra snake venom cardiotoxins and bee venom melittin. Toxicon. 1993; 31:669-95. DOI | PubMed
  60. Shier WT. Activation of high levels of endogenous phospholipase A2 in cultured cells. Proc Natl Acad Sci U S A. 1979; 76:195-9. Publisher Full Text | DOI | PubMed
  61. Altinbas B, Topuz BB, Yilmaz MS, Aydin C, Savci V, Jochem J, et al. The mediation of the central histaminergic system in the pressor effect of intracerebroventricularly injected melittin, a phospholipase A2 activator, in normotensive rats. Prostaglandins Leukot Essent Fatty Acids. 2012; 87:153-8. DOI | PubMed
  62. Grandison L. Stimulation of anterior pituitary prolactin release by melittin, an activator of phospholipase A2. Endocrinology. 1984; 114:1-7. DOI | PubMed
  63. Lad PJ, Shier WT. Activation of microsomal guanylate cyclase by a cytotoxic polypeptide: melittin. Biochem Biophys Res Commun. 1979; 89:315-21. DOI | PubMed
  64. Putz T, Ramoner R, Gander H, Rahm A, Bartsch G, Thurnher M. Antitumor action and immune activation through cooperation of bee venom secretory phospholipase A2 and phosphatidylinositol-(3,4)-bisphosphate. Cancer Immunol Immunother. 2006; 55:1374-83. DOI | PubMed
  65. Putz T, Ramoner R, Gander H, Rahm A, Bartsch G, Bernardo K, et al. 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. 2007; 56:627-40. DOI | PubMed
  66. Brandner S, Klein MA, Frigg R, Pekarik V, Parizek P, Raeber A, et al. Neuroinvasion of prions: insights from mouse models. Exp Physiol. 2000; 85:705-12. DOI | PubMed
  67. Sim VL. Prion disease: chemotherapeutic strategies. Infect Disord Drug Targets. 2012; 12:144-60. DOI | PubMed
  68. Kolko M, DeCoster MA, de Turco EB, Bazan NG. Synergy by secretory phospholipase A2 and glutamate on inducing cell death and sustained arachidonic acid metabolic changes in primary cortical neuronal cultures. J Biol Chem. 1996; 271:32722-8. DOI | PubMed
  69. Jeong JK, Moon MH, Bae BC, Lee YJ, Seol JW, Park SY. Bee venom phospholipase A2 prevents prion peptide induced-cell death in neuronal cells. Int J Mol Med. 2011; 28:867-73. DOI | PubMed
  70. Ye M, Chung HS, Lee C, Yoon MS, Yu AR, Kim JS, et al. Neuroprotective effects of bee venom phospholipase A2 in the 3xTg AD mouse model of Alzheimer’s disease. J Neuroinflammation. 2016; 13:10. Publisher Full Text | DOI | PubMed
  71. Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol. 2005; 76:77-98. DOI | PubMed
  72. Jana M, Palencia CA, Pahan K. Fibrillar amyloid-beta peptides activate microglia via TLR2: implications for Alzheimer’s disease. J Immunol. 2008; 181:7254-62. Publisher Full Text | DOI | PubMed
  73. Muehlhauser F, Liebl U, Kuehl S, Walter S, Bertsch T, Fassbender K. Aggregation-Dependent interaction of the Alzheimer’s beta-amyloid and microglia. Clin Chem Lab Med. 2001; 39:313-6. DOI | PubMed
  74. Baek H, Park SY, Ku SJ, Ryu K, Kim Y, Bae H, et al. Bee Venom Phospholipase A2 Induces Regulatory T Cell Populations by Suppressing Apoptotic Signaling Pathway. Toxins (Basel). 2020; 12Publisher Full Text | DOI | PubMed
  75. Hirsch EC, Hunot S. Neuroinflammation in Parkinson’s disease: a target for neuroprotection?. Lancet Neurol. 2009; 8:382-97. DOI | PubMed
  76. Awad K, Abushouk AI, AbdelKarim AH, Mohammed M, Negida A, Shalash AS. Bee venom for the treatment of Parkinson’s disease: How far is it possible?. Biomed Pharmacother. 2017; 91:295-302. DOI | PubMed
  77. Kim HW, Kwon YB, Han HJ, Yang IS, Beitz AJ, Lee JH. Antinociceptive mechanisms associated with diluted bee venom acupuncture (apipuncture) in the rat formalin test: involvement of descending adrenergic and serotonergic pathways. Pharmacol Res. 2005; 51:183-8. DOI | PubMed
  78. Olson KE, Gendelman HE. Immunomodulation as a neuroprotective and therapeutic strategy for Parkinson’s disease. Curr Opin Pharmacol. 2016; 26:87-95. Publisher Full Text | DOI | PubMed
  79. Doo AR, Kim SN, Kim ST, Park JY, Chung SH, Choe BY, et al. Bee venom protects SH-SY5Y human neuroblastoma cells from 1-methyl-4-phenylpyridinium-induced apoptotic cell death. Brain Res. 2012; 1429:106-15. DOI | PubMed
  80. Doo AR, Kim ST, Kim SN, Moon W, Yin CS, Chae Y, et al. Neuroprotective effects of bee venom pharmaceutical acupuncture in acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse model of Parkinson’s disease. Neurol Res. 2010; 32:88-91. DOI | PubMed
  81. Lee SM, Yang EJ, Choi SM, Kim SH, Baek MG, Jiang JH. Effects of bee venom on glutamate-induced toxicity in neuronal and glial cells. Evid Based Complement Alternat Med. 2012; 2012:368196. Publisher Full Text | DOI | PubMed
  82. Kim KH, Lee SY, Shin J, Hwang JT, Jeon HN, Bae H. Dose-Dependent Neuroprotective Effect of Standardized Bee Venom Phospholipase A2 Against MPTP-Induced Parkinson’s Disease in Mice. Front Aging Neurosci. 2019; 11:80. Publisher Full Text | DOI | PubMed
  83. Ye M, Chung HS, Lee C, Hyun Song J, Shim I, Kim YS, et al. Bee venom phospholipase A2 ameliorates motor dysfunction and modulates microglia activation in Parkinson’s disease alpha-synuclein transgenic mice. Exp Mol Med. 2016; 48:e244. Publisher Full Text | DOI | PubMed
  84. Chung ES, Lee G, Lee C, Ye M, Chung HS, Kim H, et al. Bee Venom Phospholipase A2, a Novel Foxp3+ Regulatory T Cell Inducer, Protects Dopaminergic Neurons by Modulating Neuroinflammatory Responses in a Mouse Model of Parkinson’s Disease. J Immunol. 2015; 195:4853-60. DOI | PubMed
  85. Liu XD, Zhang JL, Zheng HG, Liu FY, Chen Y. Clinical randomized study of bee-sting therapy for rheumatoid arthritis. Zhen Ci Yan Jiu. 2008; 33:197-200. PubMed
  86. Lee JD, Park HJ, Chae Y, Lim S. An Overview of Bee Venom Acupuncture in the Treatment of Arthritis. Evid Based Complement Alternat Med. 2005; 2:79-84. Publisher Full Text | DOI | PubMed
  87. Castro HJ, Mendez-Lnocencio JI, Omidvar B, Omidvar J, Santilli J, Nielsen HS, et al. A phase I study of the safety of honeybee venom extract as a possible treatment for patients with progressive forms of multiple sclerosis. Allergy Asthma Proc. 2005; 26:470-6. PubMed
  88. Shin D, Lee G, Sohn SH, Park S, Jung KH, Lee JM, et al. Regulatory T Cells Contribute to the Inhibition of Radiation-Induced Acute Lung Inflammation via Bee Venom Phospholipase A(2) in Mice. Toxins (Basel). 2016; 8Publisher Full Text | DOI | PubMed
  89. Billingham ME, Morley J, Hanson JM, Shipolini RA, Vernon CA. Letter: An anti-inflammatory peptide from bee venom. Nature. 1973; 245:163-4. DOI | PubMed
  90. Im EJ, Kim SJ, Hong SB, Park JK, Rhee MH. Anti-Inflammatory Activity of Bee Venom in BV2 Microglial Cells: Mediation of MyD88-Dependent NF-kappaB Signaling Pathway. Evid Based Complement Alternat Med. 2016; 2016:3704764. Publisher Full Text | DOI | PubMed
  91. Kim H, Lee H, Lee G, Jang H, Kim SS, Yoon H, et al. Phospholipase A2 inhibits cisplatin-induced acute kidney injury by modulating regulatory T cells by the CD206 mannose receptor. Kidney Int. 2015; 88:550-9. DOI | PubMed
  92. Hossen MS, Shapla UM, Gan SH, Khalil MI. Impact of Bee Venom Enzymes on Diseases and Immune Responses. Molecules. 2016; 22Publisher Full Text | DOI | PubMed
  93. Lee G, Kang G-H, Bae H. Bee venom phospholipase A2 suppression of experimental autoimmune encephalomyelitis is dependent on its enzymatic activity. Molecular & Cellular Toxicology. 2019; 15:307-13. DOI
  94. Geha RS. Regulation of IgE synthesis in humans. J Allergy Clin Immunol. 1992; 90:143-50. DOI | PubMed
  95. Chung KF, Barnes PJ. Cytokines in asthma. Thorax. 1999; 54:825-57. Publisher Full Text | DOI | PubMed
  96. Mucida DS, de Castro Keller A, Fernvik EC, Russo M. Unconventional strategies for the suppression of allergic asthma. Curr Drug Targets Inflamm Allergy. 2003; 2:187-95. DOI | PubMed
  97. Park S, Baek H, Jung KH, Lee G, Lee H, Kang GH, et al. Bee venom phospholipase A2 suppresses allergic airway inflammation in an ovalbumin-induced asthma model through the induction of regulatory T cells. Immun Inflamm Dis. 2015; 3:386-97. Publisher Full Text | DOI | PubMed
  98. Kim KH, Kim M, Lee J, Jeon HN, Kim SH, Bae H. Comparison of the Protective Effects of Bee Venom Extracts with Varying PLA2 Compositions in a Mouse Model of Parkinson’s Disease. Toxins (Basel). 2019; 11Publisher Full Text | DOI | PubMed
  99. Baek H, Lee CJ, Choi DB, Kim NS, Kim YS, Ye YJ, et al. Bee venom phospholipase A2 ameliorates Alzheimer’s disease pathology in Abeta vaccination treatment without inducing neuro-inflammation in a 3xTg-AD mouse model. Sci Rep. 2018; 8:17369. Publisher Full Text | DOI | PubMed
  100. Chen CH, Zhou W, Liu S, Deng Y, Cai F, Tone M, et al. Increased NF-kappaB signalling up-regulates BACE1 expression and its therapeutic potential in Alzheimer’s disease. Int J Neuropsychopharmacol. 2012; 15:77-90. DOI | PubMed
  101. Ham HJ, Han SB, Yun J, Yeo IJ, Ham YW, Kim SH, et al. Bee venom phospholipase A2 ameliorates amyloidogenesis and neuroinflammation through inhibition of signal transducer and activator of transcription-3 pathway in Tg2576 mice. Transl Neurodegener. 2019; 8:26. Publisher Full Text | DOI | PubMed
  102. Arany I, Safirstein RL. Cisplatin nephrotoxicity. Semin Nephrol. 2003; 23:460-4.
  103. Kim H, Keum DJ, Kwak J, Chung HS, Bae H. Bee venom phospholipase A2 protects against acetaminophen-induced acute liver injury by modulating regulatory T cells and IL-10 in mice. PLoS One. 2014; 9:e114726. Publisher Full Text | DOI | PubMed
  104. Kang GH, Lee S, Choi DB, Shin D, Kim J, Yang H, et al. Bee Venom Phospholipase A2 Ameliorates Atherosclerosis by Modulating Regulatory T Cells. Toxins (Basel). 2020; 12Publisher Full Text | DOI | PubMed
  105. Chen J, Lariviere WR. The nociceptive and anti-nociceptive effects of bee venom injection and therapy: a double-edged sword. Prog Neurobiol. 2010; 92:151-83. Publisher Full Text | DOI | PubMed
  106. Lee JH, Kwon YB, Han HJ, Mar WC, Lee HJ, Yang IS, et al. Bee venom pretreatment has both an antinociceptive and anti-inflammatory effect on carrageenan-induced inflammation. J Vet Med Sci. 2001; 63:251-9. DOI | PubMed
  107. Alcindor T, Beauger N. Oxaliplatin: a review in the era of molecularly targeted therapy. Curr Oncol. 2011; 18:18-25. Publisher Full Text | DOI | PubMed
  108. Muggia FM. Recent updates in the clinical use of platinum compounds for the treatment of gynecologic cancers. Semin Oncol. 2004; 31:17-24. DOI | PubMed
  109. Petit T, Benider A, Yovine A, Bougnoux P, Spaeth D, Maindrault-Goebel F, et al. Phase II study of an oxaliplatin/vinorelbine combination in patients with anthracycline- and taxane-pre-treated metastatic breast cancer. Anticancer Drugs. 2006; 17:337-43. DOI | PubMed
  110. Li D, Kim W, Shin D, Jung Y, Bae H, Kim SK. Preventive Effects of Bee Venom Derived Phospholipase A(2) on Oxaliplatin-Induced Neuropathic Pain in Mice. Toxins (Basel). 2016; 8Publisher Full Text | DOI | PubMed
  111. Nakashima A, Tomono S, Yamazaki T, Inui M, Morita N, Ichimonji I, et al. Phospholipase A2 from bee venom increases poly(I:C)-induced activation in human keratinocytes. Int Immunol. 2020; 32:371-83. DOI | PubMed
  112. Brown S, Reynolds NJ. Atopic and non-atopic eczema. BMJ. 2006; 332:584-8. Publisher Full Text | DOI | PubMed
  113. Jung KH, Baek H, Kang M, Kim N, Lee SY, Bae H. Bee Venom Phospholipase A2 Ameliorates House Dust Mite Extract Induced Atopic Dermatitis Like Skin Lesions in Mice. Toxins (Basel). 2017; 9Publisher Full Text | DOI | PubMed
  114. Shin D, Choi W, Bae H. Bee Venom Phospholipase A2 Alleviate House Dust Mite-Induced Atopic Dermatitis-Like Skin Lesions by the CD206 Mannose Receptor. Toxins (Basel). 2018; 10Publisher Full Text | DOI | PubMed
  115. Hewawaduge C, Lee B-H, Kim T-H, Uddin MB, Kim J-H, Kim CG, et al. Phospholipase A2 isolated from the venom of honey bees prevents viral attachment in mammalian cells. Journal of Biomedical and Translational Research. 2016; 17:75-8. DOI
  116. Fenard D, Lambeau G, Maurin T, Lefebvre JC, Doglio A. A peptide derived from bee venom-secreted phospholipase A2 inhibits replication of T-cell tropic HIV-1 strains via interaction with the CXCR4 chemokine receptor. Mol Pharmacol. 2001; 60:341-7. DOI | PubMed
  117. Boulanger N, Brun R, Ehret-Sabatier L, Kunz C, Bulet P. Immunopeptides in the defense reactions of Glossina morsitans to bacterial and Trypanosoma brucei brucei infections. Insect Biochem Mol Biol. 2002; 32:369-75. DOI | PubMed
  118. Boutrin MC, Foster HA, Pentreath VW. The effects of bee (Apis mellifera) venom phospholipase A2 on Trypanosoma brucei brucei and enterobacteria. Exp Parasitol. 2008; 119:246-51. DOI | PubMed
  119. Perumal Samy R, Gopalakrishnakone P, Thwin MM, Chow TK, Bow H, Yap EH, et al. Antibacterial activity of snake, scorpion and bee venoms: a comparison with purified venom phospholipase A2 enzymes. J Appl Microbiol. 2007; 102:650-9. DOI | PubMed
  120. Moreira LA, Ito J, Ghosh A, Devenport M, Zieler H, Abraham EG, et al. Bee venom phospholipase inhibits malaria parasite development in transgenic mosquitoes. J Biol Chem. 2002; 277:40839-43. DOI | PubMed
  121. Deregnaucourt C, Schrevel J. Bee venom phospholipase A2 induces stage-specific growth arrest of the intraerythrocytic Plasmodium falciparum via modifications of human serum components. J Biol Chem. 2000; 275:39973-80. DOI | PubMed
  122. Hirsch EC, Breidert T, Rousselet E, Hunot S, Hartmann A, Michel PP. The role of glial reaction and inflammation in Parkinson’s disease. Ann N Y Acad Sci. 2003; 991:214-28. DOI | PubMed
  123. Incamnoi P, Patramanon R, Thammasirirak S, Chaveerach A, Uawonggul N, Sukprasert S, et al. Heteromtoxin (HmTx), a novel heterodimeric phospholipase A(2) from Heterometrus laoticus scorpion venom. Toxicon. 2013; 61:62-71. DOI | PubMed
  124. Jridi I, Catacchio I, Majdoub H, Shahbazeddah D, El Ayeb M, Frassanito MA, et al. Hemilipin, a novel Hemiscorpius lepturus venom heterodimeric phospholipase A2, which inhibits angiogenesis in vitro and in vivo. Toxicon. 2015; 105:34-44. DOI | PubMed
  125. Jridi I, Catacchio I, Majdoub H, Shahbazzadeh D, El Ayeb M, Frassanito MA, et al. The small subunit of Hemilipin2, a new heterodimeric phospholipase A2 from Hemiscorpius lepturus scorpion venom, mediates the antiangiogenic effect of the whole protein. Toxicon. 2017; 126:38-46. DOI | PubMed
  126. Zamudio FZ, Conde R, Arevalo C, Becerril B, Martin BM, Valdivia HH, et al. The mechanism of inhibition of ryanodine receptor channels by imperatoxin I, a heterodimeric protein from the scorpion Pandinus imperator. J Biol Chem. 1997; 272:11886-94. DOI | PubMed
  127. Conde R, Zamudio FZ, Becerril B, Possani LD. Phospholipin, a novel heterodimeric phospholipase A2 from Pandinus imperator scp6pion venom. FEBS Lett. 1999; 460:447-50. DOI | PubMed
  128. Valdez-Cruz NA, Batista CV, Possani LD. Phaiodactylipin, a glycosylated heterodimeric phospholipase A from the venom of the scorpion Anuroctonus phaiodactylus. Eur J Biochem. 2004; 271:1453-64. DOI | PubMed
  129. Louati H, Krayem N, Fendri A, Aissa I, Sellami M, Bezzine S, et al. A thermoactive secreted phospholipase A(2) purified from the venom glands of Scorpio maurus: relation between the kinetic properties and the hemolytic activity. Toxicon. 2013; 72:133-42. DOI | PubMed
  130. Ramanaiah M, Parthasarathy PR, Venkaiah B. Purification and properties of phospholipase A2 from the venom of scorpion, (Heterometrus fulvipes). Biochem Int. 1990; 20:931-40. PubMed
  131. Iruela-Arispe ML, Zovein A. Fetal and Neonatal Physiology. Elsevier: Amsterdam; 2017. DOI
  132. Krayem N, Abdelkefi-Koubaa Z, Marrakchi N, Luis J, Gargouri Y. Anti-angiogenic effect of phospholipases A2 from Scorpio maurus venom glands on Human Umbilical Vein Endothelial Cells. Toxicon. 2018; 145:6-14. DOI | PubMed
  133. Lee YS, Kim YH, Shin EK, Kim DH, Lim SS, Lee JY, et al. Anti-angiogenic activity of methanol extract of Phellinus linteus and its fractions. J Ethnopharmacol. 2010; 131:56-62. DOI | PubMed
  134. Krayem N, Abdelkefi-Koubaa Z, Marrakchi N, Gargouri Y, Luis J. Native and recombinant phospholipases A2 of Scorpio maurus venom glands impair angiogenesis by targeting integrins alpha5beta1 and alphavbeta3. Int J Biol Macromol. 2018; 116:305-15. DOI | PubMed
  135. Vetter RS, Visscher PK. Bites and stings of medically important venomous arthropods. Int J Dermatol. 1998; 37:481-96. DOI | PubMed