Open access peer-reviewed chapter

Snake Venom and Therapeutic Potential

Written By

Mamdouh Ibrahim Nassar

Submitted: 11 October 2021 Reviewed: 27 October 2021 Published: 14 April 2022

DOI: 10.5772/intechopen.101421

From the Edited Volume

Snake Venom and Ecology

Edited by Mohammad Manjur Shah, Umar Sharif, Tijjani Rufai Buhari and Tijjani Sabiu Imam

Chapter metrics overview

793 Chapter Downloads

View Full Metrics

Abstract

Many active secretions produced by animals have been employed in the development of new drugs to treat diseases such as hypertension and cancer. Snake venom toxins contributed significantly to the treatment of many medical conditions. Snake venoms are the secretion of venomous snakes, which are synthesized and stored in specific venom glands. Many toxins from snake venom are investigated and formulated into drugs for the treatment of conditions such as cancer, hypertension, and thrombosis. Most of the venoms are complex mixture of a number of proteins, peptides, enzymes, toxins and non-protein inclusions. Cytotoxic effects of snake venom have potential to degrade and destroy tumor cells. Different species have different types of venom, which depends upon its species, geographical location, its habitat, climate and age. The purpose of this chapter is to review focusing on the therapeutic potential of snake venoms and to establish a scientific basis for diseases treatment particular antitumor.

Keywords

  • snake venom
  • cancer therapy
  • diseases treatment

1. Introduction

Snake venoms are the secretion of venomous snakes, which are synthesized and stored in special glands. The venom were synthesized and stored into the base of channeled or tubular fangs through which it is ejected. Most of the venoms are complex mixture of a number of proteins, peptides, enzymes, toxins and non-protein inclusions [1]. Some of snake venom possess biological effects on various functions, such as blood coagulation and pressure, regulation, and transmission of nerve impulses. These venoms have been studied and developed by researchers for use as pharmacological or diagnostic tools, and even drugs. Snake venom is a therapeutic agent for various diseases due to its physiologically active components [2]. More specifically, cobra venom has been used historically in Ayurveda in the treatment of arthritis and other chronic diseases [3].

Chinese physicians are implementing the use of snake venom products to treat stroke patients, and research has been conducted surrounding its analgesic, anti- cancerous and anti-inflammatory effects [2]. Cytotoxic effects of snake venom have potential to degrade and destroy tumor cells [4]. There are basically three types of snake venom according to its effects [5, 6]. (a) Hemotoxic venoms, which affects cardiovascular system and blood functions, (b) cytotoxic venoms targets specific cellular sites or muscles and (c) neurotoxic venoms harms nervous system of human body. The families, Elapidae and Viperidae, are large majority of the research done surrounding the medical application of snake venom involves species within these groups. Both elapids and vipers are front fanged snakes that belong to the superfamily Colubroidea. Notable species of the elapid family are cobras of the genus Naja, and a well-researched species in the viper family is Crotalus durissus terrificus.

Snake venom components caused retardation of growth of cancerous cells due to its therapeutic activity, potency for many diseases and disorders [7]. Many excellent publications characterized use of venoms for the treatment of various therapeutic conditions such as human diseases, cancer and inflammation [8, 9].

Advertisement

2. Components of snake venom

Snake venoms are complex mixtures; mainly it has proteins, which have enzymatic activities, inorganic cations, calcium, potassium, magnesium, zinc, nickel, cobalt, iron, and manganese. Zinc is necessary for anti-cholinesterase activity; calcium is required for activation of enzyme like phospholipase. Some snake venoms also contain carbohydrate, lipid, biogenic amines, and free amino acids [10].

Advertisement

3. Snake enzymes

Proteins found in snake venom include toxins, neurotoxins, nontoxic proteins, and many enzymes, especially hydrolytic ones. Enzymes are protein in nature including digestive hydrolases, L-amino-acid oxidase, phospholipases, thrombin-like pro-coagulant, and kallikrein-like serine proteases and metalloproteinases (hemorrhagins), which damage vascular endothelium.

Phosphodiesterases enzyme interfere with the prey’s cardiac system, mainly to lower the blood pressure. Phospholipase A2 causes hemolysis by lysing the phospholipid cell membranes of red blood cells [2]. Amino acid oxidases and proteases are used for digestion. Also amino acid oxidase triggers some other enzymes and is responsible for the yellow color of the venom. Hyaluronidase enzymes increases tissue permeability to accelerate the absorption of other enzymes into tissues Table 1.

TypeNameOrigin species
OxidoreductasesDehydrogenase LactateElapidae
L-amino-acid oxidaseAll species
CatalaseAll species
TransferasesAlanine amino transferase
HydrolasesPhospholipase A2All species
LysophospholipaseElapidae, Viperidae
AcetylcholinesteraseElapidae
Alkaline phosphataseBothrops atrox
Acid phosphataseDeinagkistrodon acutus
5′-nucleotidaseAll species
PhosphodiesteraseAll species
DeoxyribonucleaseAll species
Ribonuclease 1All species
Adenosine triphosphataseAll species
AmylaseAll species
HyaluronidaseAll species
NAD-nucleotidaseAll species
KininogenaseViperidae
Factor X activatorViperidae, Crotalinae
HeparinaseCrotalinae
α-FibrinogenaseViperidae, Crotalinae
β-FibrinogenaseViperidae, Crotalinae
α-β-FibrinogenaseBitis gabonica
Fibrinolytic enzymeCrotalinae
Prothrombin activatorCrotalinae
CollagenaseViperidae
ElastaseViperidae
LyasesGlucosaminate ammonia-lyase

Table 1.

Main enzymes of snake venom [1].

Advertisement

4. Polypeptide toxins

Pllypeptides include cytotoxins, cardiotoxins, and postsynaptic neurotoxins (such as α-bungarotoxin and α-Cobratoxin), which bind to acetylcholine receptors at neuromuscular junctions. Also polypeptides contains metals, peptides, lipids, nucleosides, carbohydrates, amines, and oligopeptides. Chemical composition variations of snake venom due to geographical and Ontogenic of the different species [3].

4.1 Proteolytic enzymes

These enzymes catalyze the breakdown of tissue proteins and peptides. They are also known as peptide hydrolases, protease, endopeptidases and proteinases. Some metal ions of the proteolytic enzymes help in catalysis involved in the activity of certain venom proteases and phospholipases [10].

4.2 Arginine ester hydrolase

Non-cholinesterase enzymes, it causes hydrolysis of the ester or peptide linkage, to which an arginine residue contributes the carboxyl group. This activity was found in snake, crotalid, viperid and some sea snake venoms [10]. Several arginine ester hydrolases have been isolated from the venoms of different snake species. These enzymes eventually showed fibrinogenolytic, caseinolytic, bradykinin releasing or edema-inducing activities. Most of them are serine proteases [11].

4.3 Thrombin-like enzymes

Snake venom thrombin-like enzymes (SVTLEs) constitute the major portion (10–24%) of snake venom and these are the second most abundant enzymes present in the crude venom. These enzymes are glycoprotein in nature, and act as defibrinating anticoagulants in vivo, whereas in vitro they clot plasma, heparinised plasma and purified fibrinogen. It used as therapeutic agent for the treatment of various diseases such as congestive heart failure, ischemic stroke, thrombotic disorders. Thrombin like enzymes such as crotalase, agkistrodon, ancrod and batroxobin can be purified from different snake venoms [12].

4.4 Collagenase

Collagenase enzymes are proteinase in nature that digests collagen and mesenteric collagen fibers [13]. Collagenase are also compounds of snake venoms, may induce disruption of retinal veins that, in turn, result in retinal hemorrhage. Collagenase could as drug leading to the development of new treatments due to its proteolytic properties in their pathophysiology.

4.5 Hyaluronidase

hyaluronidase beyond its role as a spreading factor venom it deserves to be explored as a therapeutic target for inhibiting the systemic distribution of venom bite. It acts upon connective tissues and decreases their viscosity, catalyzes the cleavage of internal glycoside bonds. Hyaluronidase enzyme has been found to be ubiquitously distributed in snake venoms. Hyaluronidase enzyme by itself is non-toxic and has long been known as ‘spreading factor’. The breakdown in the hyaluronic barrier allows some other fractions of venom to penetrate the organ tissues [2].

4.6 Phospholipase

Phospholipases are enzymes that hydrolyse glycerophospholipids. It catalyzes the calcium dependent hydrolysis of the 2-acyl ester bond thereby producing free fatty acids and lysophospho lipid. Neurotoxic phospholipases A2 (PLA2s) very large superfamily of enzymes composed of 16 groups within six major types. PLA2s can bind to and hydrolyse membrane phospholipids of the motor nerve terminal to cause degeneration of the nerve terminal and skeletal muscle. PLA2 can also cause hydrolysis of membrane phospholipids, and liberation of some bioactive products [14]. The biotechnological effectively of PLA2 inhibitors may support the therapeutic potential with antiophidian activity.

4.7 Phosphodiestsrase

Snake poisonous venom phosphodiesterase is a zinc metalloenzyme that share a number of mechanistic features with the nucleotidyl transferases. Zinc of this enzyme is activated by magnesium, and catalyze α-β phosphoryl bond cleavage. Phosphodiesterase releases 5-mononucleotide chain act as an exonucleotidase, thereby affecting DNA and RNA functions [15].

4.8 Acetylcholinesterase

Snake acetylcholinesterase in general is found in cobra and sea snake but absent in viperid and crotalid venoms. It plays a role in cholinergic transmission which located at the neuro-muscular junction of vertebrates.

Advertisement

5. Pharmaceutical assessment of snake venom

Some of snake venom components which have spurred the development of novel pharmaceutical compounds. Snake venom are investigated for the treatment of many diseases as cancer, hypertension, and thrombosis. Venoms of rattlesnakes and other crotalids produce alterations in resistance of blood vessels, changes in blood cells and coagulation and changes in cardiac and pulmonary dynamics. Also it may cause alterations in nervous system and respiratory system [16, 17, 18, 19, 20]. The potency of venom and its effect on human depend on the type and amount of venom injected and the site where it is deposited. Different other parameters and therapeutic derived such as hypotension and nerve shock and fall in blood pressure and varying degree of shock followed by a decrease in heamatocrit values are associated with snake venom [21, 22, 23].

Advertisement

6. Snake venom in medicine

Snake venoms are a cocktail of potent compounds which specifically and avidly target numerous essential molecules with high efficacy. The individual effects of all venom toxins integrate into lethal dysfunctions of almost any organ system. Such toxin mimetic may help in influencing a specific body function pharmaceutically for the sake of man’s health. Such snake toxin-derived mimetic are in clinical use, trials, or consideration for further pharmaceutical exploitation, especially in the fields of hemostasis, thrombosis, coagulation, and metastasis. Snake venom has great potential use as a medicine, because of all the compounds it contains, and their specific actions. Two analgesics derive from cobra venom; Cobroxin is used like morphine to block nerve transmission, and Nyloxin reduces severe arthritis pain [24]. Arvin compound from Malayan pitviper is an effective anticoagulant. Venom components allow researchers to develop novel drugs for treatment many diseases such as, nerve epilepsy, multiple sclerosis, myasthenia gravis, Parkinson’s disease, and poliomyelitis, musculoskeletal disease [24].

Advertisement

7. Snake venom and diseases treatment

Given that snake venom contains many biologically active ingredients, some may be useful to treat disease [25].

Phospholipases type A2 (PLA2s) from the Tunisian vipers Cerastes cerastes and Macrovipera lebetina have been found to have antitumor activity [26, 27]. PLA2s hydrolyze phospholipids, thus could act on bacterial cell surfaces, providing novel antimicrobial activities [28]. The analgesic activity of many snake venom proteins has been long known [29, 30] and the main challenge is how to deliver protein to the nerve cells.

Advertisement

8. Serotherapy of snake venom

Serotherapy using antivenom is a common current treatment, both adaptive immunity and serotherapy are specific to the type of snake; venom with identical physiological action do not cross-neutralize [31, 32].

Advertisement

9. Snake venom therapy of hepatocellular carcinoma

Hepatocellular carcinoma (HCC) represents up to 90% of all liver malignancies. Recently, the World Health Organization (WHO) reported that HCC is the fifth most common tumor worldwide, and the second most common cause of cancer-associated deaths. For the majority of advanced HCC cases, curative treatments are not possible, and the prognosis is dismal because of underlying cirrhosis as well as poor tumor response to standard chemotherapy. For patients with advanced HCC, the only approved molecular targeted therapy is sorafenib (SOR), the first orally active multi-kinase inhibitor. It provides only temporary therapeutic efficacy by increasing the survival rate by approximately 3 months [33]. Besides, a great inter-individual variation in the pharmacokinetics of SOR, due to systemic overexposure, has contributed to its toxicity [34, 35]. Therapeutic approaches to identify and develop novel compounds such as snake venom components are urgent to have potential ability for cancer treatment [36]. Moreover, better finding alternative natural safe, and better ways to treat cancer with less toxicity and deteriorated effect on normal cells is highly desirable [37].

The combining snake venoms (SVs) could synergistically enhance the antiproliferative effects at low doses on liver cancer cells (HepG2). In such Research the gene expression for apoptotic, inflammatory, antioxidant and cell cycle regulator was determined [38].

Varies compounds from venomous animals such as spiders, scorpions, snakes, caterpillars, centipedes, wasp, bees, toads, ants, and frogs have largely shown biotechnological and pharmacological applications against many diseases including cancer [39, 40, 41, 42]. Venoms obtained from snakes were reported to exhibit a cytotoxic effect against tumor cells [26]. This potency is due to inhibiting cell proliferation, promoting cell death through activating the apoptotic mechanisms [43, 44]. Meanwhile snake venom increased cytochrome-c production, modulating the expression levels of proteins that controlling the cell cycle, and treat triggering damages in the cell membranes [45, 46, 47].

The complex mixtures of snake venom, L-amino acid oxidase (LAAO) are a effect as anticancer therapeutic activity and through the induction of oxidative stress in cancer cells [48]. L-amino acid oxidase (LAAO) has been reported to exhibit a potent anti-tumor activity to different cancer cell lines including [49]. LAAO can selectively bind to the cancer cell surface at specific phospholipid compositions to deliver the hydrogen peroxide [47, 48, 49, 50]. LAAO mediates its cytotoxicity to the cell surface and produces H2O2 [49, 51, 52]. Moreover studies are confirmed this safer effect on animal models [38]. In terms of cytotoxicity, combined administration of LAAO with SOR has reduced the cell death on normal liver cells THLE-2 as compared to a single administration [38]. On the other hand the administration of LAAO and SV alone or in combination with SOR has significantly induced cell death and apoptosis in HepG2 cells as compared to control untreated cells [53]. Additionally, [54] showed that the LAAO isolated from Ophiophagus hannah venom selectively kills cancer cells via the apoptotic pathway by regulating the caspase 3, 7 activity but is non-toxic to normal cells. One of the consequences of the excessive damage caused by the reactive oxygen species (ROS) is changes in mitochondrial membrane permeability causing Ca+2 overload that result in cytochrome c release and apoptotic death [55].

Advertisement

10. Therapeutic effects of snake venom on rheumatoid

Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease in which the immune system primarily attacks healthy tissue of synovial joints (NIH). The disease affects between 0.5–1.0% of the developed world population, and is a significant cause of disability [56]. The primary characteristic of RA is the progressive destruction and inflammation of synovial joints, most commonly in metacarpophalangeal, proximal interphalangeal, metatarsophalangeal, wrist, and knee joints. Articular manifestations include symmetric joint swelling, tenderness, stiffness, and motion impairment, and general symptoms such as fevers, fatigue, weight loss, and discomfort are also common [57].

Snake venom has been used for treatment of rheumatoid arthritis and pain management. Venom from the families Elapidae and Viperidae have been shown to have anti-inflammatory and analgesic effects. Snake venom has anti-inflammatory effects by reducing levels of pro- inflammatory cytokines and increasing levels of anti-inflammatory cytokines [58]. Additionally, snake venom can reduce structural damage from prolonged inflammation by acting as a (tumor necrosis factor alpha), TNF-alpha blocker, and by inhibiting the proliferation of fibroblast-like synoviocytes. The mechanisms of snake venom pain modulation seen in murine pain models follow the cholinergic and opioidergic systems. Analgesic findings involving the cholinergic system concluded not only that the effects of snake venom have similar effects to morphine, but also that no withdrawal symptoms were observed after administration of venom stopped. These results show incredible promise for a non-addictive analgesic that could be used for pain management in rheumatoid arthritis patients [58].

A study found that while the general health status of RA patients in Norway improved between the years of 1994 and 2001, alleviation of pain remained the highest priority in both cohorts [59]. In another study, 88% of participants selected pain as their top priority for improvement during a year of treatment [60]. Pain scores are also disproportionately greater in women, minorities, and those with lesser levels of education, and pain is a top contributor to emotional health in RA patients [61, 62].

One of the main treatments for pain in RA patients is the administration of disease modifying antirheumatic drugs (DMARDs), which act peripherally to reduce the inflammatory response and the pain associated with it. Additionally, non-steroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen and naproxen are often suggested to patients to manage their pain. These medications can be coupled with over the counter medications such as acetaminophen to further alleviate pain. When the combination of NSAID and acetaminophen administration has failed to provide relief, weak opioids are considered [63]. Therapies for RA have generally shifted focus from symptom management to the treatment of underlying inflammation that causes the symptoms [64]. Biologic disease modifying drugs are act to reduce immune responses in the body such as TNF inhibitors are used to block tumor necrosis factor (a proinflammatory cytokine) activity. Similarly, Abatacept prevents the overactivity of T cells, and Tocilluzumab inhibits the activity of another proinflammatory protein, IL-6 [65, 66].

Mechanical and thermal hyperalgesia have been found to be suppressed in several murine models with the administration of snake venom. Inflammation can also affect central pain processing, so a decrease in inflammation with snake venom could positively affect central pain and sensitization as well. The effects of snake venom from elapids and vipers on cholinergic and opioidergic mechanisms of pain are arguably the most promising relevant to treating rheumatoid arthritis. In one study, snake venom acting on cholinergic receptors to produce analgesia was found to be just as effective as morphine, with a longer lasting effect [67]. A handful of studies have utilized venom from elapids, particularly the species Naja kaouthia and Naja naja, in murine arthritis models to study the anti-inflammatory and anti-arthritic properties of the venom or its specific components [68]. Observed the effects of NN-32, a cytotoxic protein from N. naja venom, on arthritic rats. It was found that while arthritic rats showed significantly increased levels of inflammatory cytokines TNF-α, IL-17, and cytokine-induced neutrophil chemoattractant 1 (CINC-1, a rat cytokine (homolog of IL-8) with hyperalgesic properties) compared to non-arthritic control rats, NN-32 treatment significantly decreased levels of these cytokines. Another study by the same researchers found that IL-10 levels were decreased in adjuvant induced arthritic rats, but the levels were significantly restored when treated by N. kaouthia venom [69].

Produced similar results using cobratoxin, a neurotoxin from a Naja cobra, on complete Freund’s adjuvant (CFA) induced arthritis rats [70]. The arthritic rats showed increased serum levels of (tumor necrosis factor) TNF-α, IL-1, and IL-2, and decreased levels of IL-10. With the cobratoxin treatment, the rats exhibited lower proinflammatory cytokine levels, and a reversal of the CFA induced IL-10 decrease [69]. Found similar results with neurotoxin-NNA, another peptide from N. naja atra: Treatment with the peptide exhibited a dose dependent decrease in TNF-α and IL-1β levels in rat models of inflammation. These studies add to the evidence that cobra venom could modulate the production of inflammatory cytokines in RA and subsequently reduce inflammatory pain.

Compared the effects of cobratoxin from N. naja atra to dexamethasone, a corticosteroid that relieves inflammation. This revealed the dexamethasone administered to arthritic rats showed greater effects on acute inflammation than the cobrotoxin, but inhibition of the long-term inflammatory process (observed by a decrease of cytokines IL-6, TNF-α, and IL- 1β) was strong in both. The maintenance of the levels suggests that orally administered CTX has anti-inflammatory properties by decreasing pro-inflammatory cytokine levels and maintaining pro-inflammatory cytokine levels. Rats treated with CTX showed slightly greater anti-inflammatory and analgesic effects, suggesting the potential for components of venom to function as NSAIDs [69].

11. Snake venom therapy of joint destruction

The use of tumor necrosis factor (TNF) blockers, a more recent therapeutic option for RA, provides a correlation between the cytokine TNF-α and bone erosion. Several studies have found that the five TNF blockers that are currently in use have all been correlated with continued inhibition of bone erosion [71]. The positive effect of TNF inhibitors provides evidence that a decrease in the cytokine TNF-α could have beneficial effects on reducing not only initial inflammatory pain but also pain induced by bone erosion and other structural changes. Additionally, the anti arthritic and anti inflammatory activity of NN-32, a cytotoxic protein from Indian spectacle cobra snake (Naja naja) venom showed significant decrease in physical and urinary parameters, serum enzymes, serum cytokines levels as compared to arthritic control group of rats. NN-32 treatment recovered carrageenan induced inflammation [72]; Cobratoxin (CTX), the long-chain α-neurotoxin from Thailand cobra venom, has been demonstrated to have analgesic action in rodent pain models [73]. Structural changes of bone and cartilage are a hallmark of inflammatory joint diseases such as rheumatoid arthritis (RA), psoriatic arthritis (PsA), and ankylosing spondylitis (AS) [74, 75] found that cobrotoxin from N. naja atra venom inhibited the activation of nuclear factor kappa B (NF-κB). NF-κB is a transcriptional factor that plays a role in inflammation by expressing pro-inflammatory cytokines, including TNF-α, and inhibition of NF-κB has been shown to delay progression of joint destruction in animal arthritis models. Another study also found that cobrotoxin has an inhibitory effect on NF-κB activation, which led to decreased levels of TNF-α [76]. These studies indicate that cobra venom can decrease proinflammatory cytokine levels, affecting as anti-inflammatory properties pain associated with physical destruction of the joint. These properties could reduce both peripheral and central inflammation, and potentially prevent further joint damage and sensitization of nerves [77].

12. Therapeutic potential of snake venom on cancer

The anti-cancer potential of snake venom depend on its protein peptides and enzymes which bind to cancer cell membranes, affecting the migration and proliferation of these cells [78].

Cancer is characterized by uncontrolled cell division, cell transformation, and escape of apoptosis, invasion, angiogenesis and metastasis. Induction of apoptosis is the most important mechanism of many anticancer agents. Snake integrins are important in cell adhesion, cell migration, tissue organization, cell growth, hemostasis and inflammatory responses, so they are in the study for the development of drugs for the treatment of cancer [53]. The induction of the apoptosis manifests the control on the tumor size and number of tumor cells hence establishing the application of apoptosis inducers as vital components in the treatment of cancer [55].

Isolation and purification of L-amino acid oxidases (LAAOs) from Bothrops leucurus (Bl-LAAO) and cobra was effected on platelet function and cytotoxicity [79, 80]. The mechanism of this enzyme action may be related to the inhibition of thymidine incorporation and an interaction with DNA [81]. Also different tumor cell lines were found to susceptible from lytic action and from synthetic peptide. Also NN-32 showed cytotoxicity on EAC cells, increased survival time of inoculated EAC mice, reduced solid tumor volume and weight. NN-32 increased proapoptotic protein [82]. Pharmacokinetics effect of cytotoxin from Chinese cobra (N. naja atra) venom was studied on rabbits [49]. Plasma levels of the cytotoxin were analyzed by a biotinavidin enzyme-linked immunosorbent assay.

The extraction of specific protein Okinawa Habu apoxin protein-1 (OHAP-1) from Okinawa Habu venom studied for its toxic effects [83]. In this study, OHAP-1 could induce apoptosis in some glioma cell. Also the apoptotic effect of OHAP-1 on malignant glioma cells could be through the generation of intracellular ROS and p53 protein expression. Antitumor activity using snake venom (Lapemis curtus) caused decreasing of Hep2 tumor volume and considered as an important indicator of reduction of tumor burden [84]. Cardiotoxin III (CTX III), was isolated from N. naja atra venom, and reported its anticancer activity [85]. The anti-tumor potential as well as its cytotoxicity and hemolysis activity was occurred as a galactoside-binding lectin which isolated from B. leucurus venom [86]. Purification of BjcuL, a lectin from Bothrops jararacussu venom was observed its cytotoxic effects to gastric carcinoma cells. This confirmed cytotoxicity of BJcuL on tumor cells mainly by altering cell adhesion and through induction of apoptosis [87].

13. Anti-microbial potency of snake venom

Snakes venoms were assayed in order to investigate their antimicrobial activities giving promising results [88]. Since 1930s, cobra venom has been used to treat various diseases like asthma, polio, multiple sclerosis, rheumatism, severe pain and trigeminal neuralgia. Among antimicrobial components that have been isolated from snake venom are (i) L-amino acid oxidase (LAAO), and (ii) phospholipase A2 (PLA2) [89]. The LAAO antibacterial action appears to result from hydrogen peroxide generated by the oxidative action of the enzymes, as the effect is abolished in the presence of hydrogen peroxide scavengers such as catalase [10, 90, 91, 92, 93]. Also antimicrobial peptides including cathelicidins, nerve growth factor and omwaprin have been isolated from various venomous snake species [94, 95, 96]. The antibacterial effects of cobra venom LAAO were affected against strains including S. aureus, S. epidermidis, P. aeruginosa, Klebsiella pneumoniae, E. coli, gram-positive and negative bacteria [92, 97]. Purified L-amino acid oxidase from Bothrops pauloensis snake venom had bactericidal activities [98, 99].

Electron microscopic assessments of both Gram-positive and Gram-negative bacterial strains suggested that the H2O2 produced by LAO induced bacterial membrane rupture and consequently loss of cytoplasmatic content [100, 101]. Akbu-LAAO an L-amino acid oxidase isolated from the venom of Agkistrodon blomhoffii ussurensis snake exhibited a strong bacteriostasis effect on S. aureus [102].

The most mode of action involved in the bactericidal activity of LAAOs is that H2O2 causes oxidative stress in the target cell, triggering disorganization of the plasma membrane and cytoplasm and consequent cell death Table 2 [103, 104].

Snake speciesAntibacterial componentEffective against
Bothrops mattogrosensisBmLAAOGram positive and negative bacteria
Ophiophagus HannahKing cobra L-amino acid oxidase (Oh-LAAO)Gram positive and negative bacteria
B. alternatusBalt-LAAO-IE. coli and S. aureus
Daboia russellii siamensisDRS-LAAOS. aureus (ATCC 25923),
P. aeruginosa (ATCC 27853) and E. coli (ATCC 25922).
King cobra venomLAAOS. aureus, S. epidermidis,
P. aeruginosa, K. pneumoniae, and E. coli
B. pauloensisBp-LAAONot specific
Bothriechis schlegeliiBsLAAOS. aureus and Acinetobacter baumannii
Naja naja oxianaLAAOB. subtilis and E. coli
Crotalus durissus cascavellaCasca LAAO(Xanthomonas axonopodis pv passiflorae) and S. mutans
Crotalus durissus cumanensisCdcLAAOS. aureus and A. baumannii
Vipera lebetinaLAAOGram-negative and Gram-positive bacteria
Agkistrodon blomhoffiiussurensisAkbu-LAAOS. aureus
Trimeresurus mucrosquamatusTM-LAOE. coli, S. aurues and
B. dysenteriae
Trimeresurus jerdoniiTJ-LAOE. coli, S. aureus, P. aeruginosa, and Bacillus megaterium.
Bothrops marajoensisBmarLAAOS. aureus, and P. aeruginosa
Bothrops jararacaLAAOS. aureus
Agkistrodon haly PallasLAAOE coli K12D31
B. leucurusBleuLAAOS. aureus

Table 2.

Anti-bacterial profile of various snake venom LAAOs [88].

13.1 Anti-microbial activity of phospholipase A2 (PLA2)

Phospholipase has antimicrobial activity against E. coli and S. aureus as well as the Gram-positive bactericidal activity of sPLA(2)-I [105]. Also Phospholipases A2 (PLA2S) isolated from C. durissus terrificus venom showed antimicrobial activity against Xanthomonas axonopodis pv. Passiflorae Table 3 [106].

Snake speciesAntibacterial componentEffective against
Bungarus fasciatusBFPAE. coli and S. aureus
Agkistrodon sppAgkTx-IIS. aureus, P. vulgaris and Burkholderia pseudomallei
Echis carinatusEcTx-IEnterobacter aerogenes, E. coli, P. vulgaris, P. mirabilis, P. aeruginosa and S. aureus
Vipera berus berusVBBPLA2B. subtilis
Bothrops asperPLA2 myotoxinsS. typhimurium and S. aureus
Porthidium nasutumPnPLA2S. aureus

Table 3.

Antibacterial profile of various snake venom Phospholipae A2s [88].

13.2 Antimicrobial activity of peptides

Peptides are have a critical defense against all kinds of microorganisms, bacteria, fungi, and viruses. Peptides play an important role in the bactericidal effect. Antimicrobial peptides can be divided into four structural groups known as α-helical, β-sheet, α-hairpin, and extended peptides [107].

13.3 Cathelicidin

Cathelicidin-BF found in the venom of the snake Bungarus fasciatus in treating Salmonella typhimurium infection. Cathelicidins are a family of antimicrobial peptides acting as multifunctional effectors molecule in innate immunity. Cathelicidin-BF had been purified from the snake venoms of B. fasciatus (BF) and it was the first identified cathelicidin antimicrobial peptide in reptiles [88]. S. epidermidis, was also effectively killed by Cathelicidin-BF [108, 109].

Cathelicidin-BF is active against Salmonella infected-mice and it showed strong antibacterial activity against various bacteria [110]. Cathelicidin from the venom of B. fasciatus has antibacterial activity against drug-resistant E. coli, P. aeruginosa, and S. aureus. Also cathelicidin BF-30 had stronger antimicrobial activities against a broad spectrum of microorganisms [111].

14. Conclusions

Snake venoms are the complex mixtures of several biologically active proteins, peptides, enzymes, and organic and inorganic compounds.

Snake venoms are very important agents for many types of diseases as well as antimicrobial, anti-inflammation, anti-rheumatoid and cancer therapy. Snake venoms acts by inhibiting cell proliferation and promoting cell death by different means: induction of apoptosis in cancer cell, increasing Ca2+ influx; inducing cytochrome C release; decreasing or increasing the expression of proteins that control cell cycle; leading to damage of cell membranes. Snake venoms contain many components that act on the peripheral nervous system for killing or immobilizing prey. All the above mentioned attracted our attention to develop of a new drugs from snake venoms will be useful as therapeutic agents of many diseases.

References

  1. 1. Leon G, Sanchez L, Hernandez A, Villalta M, Herrera M, Segura A, et al. Immune response towards snake venoms. Inflammation & Allergy Drug Targets. 2011;10:381-398
  2. 2. Sudhakar KA, Dumantraj AR, Sonali C. A review on Snake venom: An unrevealed medicine for human ailments: Great scope for pharmaceutical research. International Journal of Research in Ayurveda and Pharmacy. 2017;8(2):35-41. DOI: 10.7897/2277-4343.08280
  3. 3. Gomes A, Bhattacharya S, Chakraborty M, Bhattacharjee P, Mishra R, Gomes A. Anti-arthritic activity of Indian monocellate cobra (Naja kaouthia) venom on adjuvant induced arthritis. Toxicon. 2010;55(2-3):670-673. DOI: 10.1016/j.toxicon.2009.10.007
  4. 4. Marsh N, Williams V. Practical applications of snake venom toxins in haemostasis. Toxicon. 2005;45:1171-1181
  5. 5. Jin H, Varner J. Integrins: Roles in cancer development and as treatment targets. British Journal of Cancer. 2004;90:561-565
  6. 6. Debatin KM, Krammerr P. Death receptors in chemotherapy and cancer. Oncogene. 2004;23:2950-2966
  7. 7. Kalam Y, Isbister GK, Mirtschin P, Hodgson WC, Konstantakopoulos N. Validation of a cell-based assay to differentiate between the cytotoxic effects of elapid snake venoms. Journal of Pharmacological and Toxicological Methods. 2011;63:137-142
  8. 8. Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian Journal of Experimental Biology. 2010;48:93-103
  9. 9. Santos MMDV, Santana CD, Giglio JR, Da Silva RJ, Sampaio SV, Soares AM, et al. Antitumoural effect of an L-amino acid oxidase isolated from Bothrops jararaca snake venom. Basic & Clinical Pharmacology & Toxicology. 2008;102:533-542
  10. 10. Kitchens CS, Eskin TA. Fatality in a case of envenomation by Crotalus adamanteus initially successfully treated with polyvalent ovine antivenom followed by recurrence of defibrinogenation syndrome. Journal of Medical Toxicology. 2008;4:180-183
  11. 11. Pmdrio-Escarso SH, Soares AM, Rodrigues VM, Mancin AC, Reis ML, Ballejo G, et al. Isolation and characterization of an arginine ester hydrolase from Bothrops jararacussu venom which induces contractions of the isolated rat uterus. Biochemistry and Molecular Biology International. 1999;47(4):699-706
  12. 12. Ullah A, Masood R, Ali I, Ullah K, Ali H, Akbar H, et al. Thrombin-like enzymes from snake venom: Structural characterization and mechanism of action. International Journal of Biological Macromolecules. 2018;114(788):811. DOI: 10.1016/j.ijbiomac.2018.03.164
  13. 13. Chu CW, Tsai TS, Tsai IH, Lin YS, Tu MC. Prey envenomation does not improve digestive performance in Taiwanese pit vipers (Trimeresurus gracilis and T. stejnegeri stejnegeri). Comparative Biochemistry & Physiology. 2009;152A:579-585
  14. 14. Wuster W, Peppin L, Pook CE, Walker DE. A nesting of vipers: Phylogeny, historical biogeography and patterns of diversification of the Viperidae (Squamata: Serpentes). Molecular Phylogenetics and Evolution. 2008;49:445-459
  15. 15. Tsai CH, Yang SH, Chien CM, Lu MC, Lo CS, Lin YH, et al. Mechanisms of cardiotoxin III-induced apoptosis in human colorectal cancer colo205 cells. Clinical and Experimental Pharmacology & Physiology. 2006;33:177-182
  16. 16. Gallacci M, Cavalcante WLG. Understanding the in vitro neuromuscular activity of snake venom Lys49 phospholipase A2 homologues. Toxicon. 2010;55:1-11
  17. 17. Marcussi S, Sant'Ana CD, Oliveira CZ, Rueda AQ, Menaldo DL, Beleboni RO, et al. Snake venom phospholipase A2 inhibitors: Medicinal chemistry and therapeutic potential. Current Topics in Medicinal Chemistry. 2007;7:743-756
  18. 18. Gutierrez JM, Alexandra R, Escalante T, Díaz C. Hemorrhage induced by snake venom metalloproteinases: Biochemical and biophysical mechanisms involved in microvessel damage. Toxicon. 2005;45:997-1011
  19. 19. Marshall DM. Enzyme activities and biological functions of snake venoms. Applied Herpetology. 2005;2:109-123
  20. 20. Aguilar I, Guerrero B, Salazar AM, Giron ME, Perez JC, Sanchez EE, et al. Individual venom variability in the south American rattlesnake Crotalus durissus cumanensis. Toxicon. 2007;50:214-224
  21. 21. Gutierrez JM, Ownby CL. Skeletal muscle degeneration induced by venom phospholipases A2: Insights into the mechanisms of local and systemic myotoxicity. Toxicon. 2003;42:915-931
  22. 22. Yamazaki Y, Takani K, Atoda H, Morita T. Snake venom vascular endothelial growth factors (VEGFs) exhibit potent activity through their specific recognition of KDR (VEGF receptor 2). The Journal of Biological Chemistry. 2003;278:51985-51988
  23. 23. Chippaux JP, Williams V, White J. Snake venom variability: Methods of study, results and interpretation. Toxicon. 1991;29:1279-1303
  24. 24. Estevão-Costa M-I, Sanz-Soler R, Johanningmeier B, Eble JA. Snake venom components in medicine: From the symbolic rod of Asclepius to tangible medical research and application. International Journal of Biochemistry & Cell Biology. 2018;104:94-113. DOI: 10.1016/j.biocel.2018.09.011
  25. 25. McCleary RJ, Kini RM. Non-enzymatic proteins from snake venoms: A gold mine of pharmacological tools and drug leads. Toxicon. 2013;62:56-74. DOI: 10.1016/j.toxicon.2012.09.008
  26. 26. Vyas VK, Brahmbhatt K, Bhatt H, Parmar U. Therapeutic potential of snake venom in cancer therapy: Current perspectives. Asian Pacific Journal of Tropical Biomedicine. 2013;3(2):156-162. DOI: 10.1016/S2221-1691(13)60042-8
  27. 27. Jain D, Kumar S. Snake venom: A potent anticancer agent. Asian Pacific Journal of Cancer Prevention. 2012;13(10):4855-4860. DOI: 10.7314/apjcp.2012.13.10.4855
  28. 28. de Oliveira Junior NG, e Silva Cardoso MH, Franco OL. Snake venoms: Attractive antimicrobial proteinaceous compounds for therapeutic purposes. Cellular and Molecular Life Sciences. 2013;70(24):4645-4658. DOI: 10.1007/s00018-013-1345-x
  29. 29. Woolf CJ. Pain: Morphine, metabolites, mambas, and mutations. The Lancet. Neurology. 2013;12(1):18-20. DOI: 10.1016/S1474-4422(12)70287-9
  30. 30. Osipov A, Utkin Y. Effects of snake venom polypeptides on central nervous system. Central Nervous System Agents in Medicinal Chemistry. 2012;12(4):315-328. DOI: 10.2174/187152412803760618
  31. 31. Reptile Venom Research. Australian Reptile Park. Archived: February 2, 2010. Retrieved: December 21, 2010
  32. 32. Boulenger GA. The Snakes of Europe, Publisher. London: Methuen; 1913
  33. 33. Siegel AB, Zhu AX. Metabolic syndrome and hepatocellular carcinoma: Two growing epidemics with a potential link. Cancer. 2009;115:5651-5661
  34. 34. Gao J, Xie L, Yang WS, Zhang W, Gao S, Wang J, et al. Risk factors of hepatocellular carcinoma–current status and perspectives. Asian Pacific Journal of Cancer Prevention. 2012;13:743-752
  35. 35. Boudou-Rouquette P, Narjoz C, Golmard JL, Thomas-Schoemann A, Mir O, Taieb F, et al. Early sorafenib-induced toxicity is associated with drug exposure and UGTIA9 genetic polymorphism in patients with solid tumors: A preliminary study. PLoS One. 2012;7:e42875
  36. 36. El-Magd MA, Mohamed Y, El-Shetry ES, Elsayed SA, Abo-Gazia M, Abdel-Aleem GA, et al. Melatonin maximizes the therapeutic potential of non-preconditioned MSCs in a DEN-induced rat model of HCC. Biomedicine & Pharmacotherapy. 2019;114:108732
  37. 37. Shaban AM, Hammouda O, Abou Ghazala L, Raslan M, El-Magd MA. Ethyl acetate fraction of garlic (Allium sativum) inhibits the viability of MCF7 and HepG2 through induction of apoptosis and G2/M phase cell cycle arrest. Journal of Applied Pharmaceutical Science. 2018;8:142-150
  38. 38. Mansour GH, El-Magd MA, Mahfouz DH, Abdelhamid IA, Mahamed MF, Ibrahim NS, et al. Bee venom and its active component Melittin synergistically potentiate the anticancer effect of Sorafenib against HepG2 cells. Bioorganic Chemistry, November 2021;116:105329
  39. 39. Nassar MI, Elzayat EM. Anti-cancer and anti-microbial and neurological effects using wasp venom peptides agents: Review. Acta Scientific Microbiology. 2020;3(1):97-102
  40. 40. Nassar MI. Wasps venom new trend for treatment of cancer, microbial and pathogenic diseases. Journal of Pharmaceutical Microbiology. 2020;6(2):30-31
  41. 41. Ibrahim NM, Abd El-Monem DH, Youssef M, Ibrahim SM, Mohamed SM, Abd-Aldayem MS, et al. Bee venom drug potentiality on the macromolecules damage of the larval gut of Hermetia Illucens (L.), (Diptera: Stratiomyidae). Journal of the Egyptian Society of Parasitology. 2020;50(3):488-493
  42. 42. Mansour GH, El-Magd MA, Mahfouz DH, Abdelhamid IA, Mohamed MF, Ibrahim NS, et al. Bee venom and its active component Melittin synergistically potentiate the anticancer effect of Sorafenib against HepG2 cells. Bioorganic Chemistry. 2021;3(116):105329. DOI: 10.1016/j.bioorg.2021.105329
  43. 43. Estevão-Costa M-I, Sanz-Soler R, Johanningmeier B. Johannes A Eble. Snake venom components in medicine: From the symbolic rod of Asclepius to tangible medical research and application. The International Journal of Biochemistry & Cell Biology. 2018;09:011. DOI: 10.1016/j.biocel.2018.09.011
  44. 44. Mackessy SP. Handbook of Venoms and Toxins of Reptiles. Boca Raton, FL: CRC Press/Taylor & Francis Group; 2009
  45. 45. Bauchot R. Snakes: A Natural History. New York City, NY, USA: Sterling Publishing Co., Inc.; 1994. pp. 194-209. ISBN 978-1-4027-3181-5
  46. 46. Condrea E, Devries A, Mager J. Hemolysis and splitting of human erythrocyte phospholipids by snake venoms. Biochimica et Biophysica Acta (BBA)—Specialized Section on Lipids and Related Subjects. 1964;84(1):60-73. DOI: 10.1016/0926-6542(64)90101-5
  47. 47. Tan KK, Bay BH, Gopalakrishnakone P. L-amino acid oxidase from snake venom and its anticancer potential. Toxicon. 2018;144:7-13
  48. 48. Ullah A. Structure-function studies and mechanism of action of snake venom L-amino acid oxidases. Frontiers in Pharmacology. 2020;11:110-110
  49. 49. Guo C, Liu S, Dong P, Zhao D, Wang C, Tao Z, et al. Akbu-LAAO exhibits potent anti-tumor activity to HepG2 cells partially through produced H2O2 via TGF-β signal pathway. Scientific Reports. 2015;5:18215
  50. 50. Abdelkafi-Koubaa Z, Aissa I, Morjen M, Kharrat N, El Ayeb M, Gargouri Y, et al. Interaction of a snake venom L-amino acid oxidase with different cell types membrane. International Journal of Biological Macromolecules. 2016;82:757-764
  51. 51. Ande SR, Kommoju PR, Draxl S, Murkovic M, Macheroux P, Ghisla S, et al. Mechanisms of cell death induction by L-amino acid oxidase, a major component of ophidian venom. Apoptosis: An International Journal on Programmed Cell Death. 2006;11:1439-1451
  52. 52. Burin SM, Berzoti-Coelho MG, Cominal JG, Ambrosio L, Torqueti MR, Sampaio SV, et al. The L-amino acid oxidase from Calloselasma rhodostoma snake venom modulates apoptomiRs expression in Bcr-Abl-positive cell lines. Toxicon. 2016;120:9-14
  53. 53. Zhang H, Teng M, Niu L, Wang Y, Wang Y, Liu Q, et al. Purification, partial characterization, crystallization and structural determination of AHP-LAAO, a novel L-amino-acid oxidase with cell apoptosis-inducing activity from Agkistrodon halys pallas venom. Acta Crystallographica Section D: Structural Biology—Acta Crystallographica. 2004;60:974-977
  54. 54. Lee ML, Fung SY, Chung I, Pailoor J, Cheah SH, Tan NH. King cobra (Ophiophagus hannah) venom L-amino acid oxidase induces apoptosis in PC-3 cells and suppresses PC-3 solid tumor growth in a tumor xenograft mouse model. International Journal of Medical Sciences. 2014;11:593-601
  55. 55. Torii S, Yamane K, Mashima T, Haga N, Yamamoto K, Fox JW, et al. Molecular cloning and functional analysis of apoxin I, a snake venom-derived apoptosis-inducing factor with L-amino acid oxidase activity. Biochemistry. 2000;39:3197-3205
  56. 56. Boonen A, Severens JL. The burden of illness of rheumatoid arthritis. Clinical Rheumatology. 2011;30(1):3-8. DOI: 10.1007/s10067-010-1634-9
  57. 57. Grassi W, De Angelis R, Lamanna G, Cervini C. The clinical features of rheumatoid arthritis. European Journal of Radiology. 1998;27:S18-S24. DOI: 10.1016/S0720-048X(98)00038-2
  58. 58. Maggie M. Potential Therapeutic Effects of Snake Venom Components on Pain Management in Rheumatoid Arthritis Patients. University Honors Theses. Paper 1075; 2021. Available from: https://doi.org/10.15760/honors.1101
  59. 59. Heiberg T, Finset A, Uhlig T, Kvien TK. Seven year changes in health status and priorities for improvement of health in patients with rheumatoid arthritis. Annals of the Rheumatic Diseases. 2005;64(2):191-195. DOI: 10.1136/ard.2004.022699
  60. 60. Klooster PM, Veehof MM, Taal E, Riel Piet LCM, van de Laar MAFJ. Changes in priorities for improvement in patients with rheumatoid arthritis during 1 year of anti-tumour necrosis factor treatment. Annals of the Rheumatic Diseases. 2007;66(11):1485-1490. DOI: 10.1136/ard.2007.069765
  61. 61. Wolfe F, Michaud K. Assessment of pain in rheumatoid arthritis: Minimal clinically significant difference, predictors, and the effect of anti-tumor necrosis factor therapy. The Journal of Rheumatology. 2007;34(8):1674-1683
  62. 62. Lee YC. Effect and treatment of chronic pain in inflammatory arthritis. Current Rheumatology Reports. 2013;15(1):300
  63. 63. Lee YC. Arthritis Research Introductory: Cell-Cell Adhesion Trafficking and Angiogenesis. Ann Arbor, MI: Internal Medicine—Rheumatology, University of Michigan Medical School; 2012
  64. 64. Colmegna I, Ohata BR, Menard HA. Current understanding of rheumatoid arthritis therapy. Clinical Pharmacology & Therapeutics. 2012;91(4):607-620
  65. 65. Johns Hopkins Arthritis Center. Rheumatoid Arthritis Treatment. 2018. Available from: https://www.hopkinsarthritis.org/arthritis-info/rheumatoid-arthritis/ra-treatment/
  66. 66. Vardeh D, Naranjo JF. Peripheral and central sensitization. In: Pain Medicine. Cham: Springer; 2017. pp. 15-17. DOI: 10.1007/978-3-319-43133-8_4
  67. 67. Cheng B, Zhou X, Zhu Q, Gong S, Qin Z, Reid P, et al. Cobratoxin inhibits pain-evoked discharge of neurons in thalamic parafascicular nucleus in rats: Involvement of cholinergic and serotonergic systems. Toxicon: Official Journal of the International Society on Toxinology. 2009;54:224-232. DOI: 10.1016/j.toxicon.2009.04.007
  68. 68. Gomes A. Snake venom—An anti arthritis natural product. Al Ameen Journal of Medical Sciences. 2010;3(3):176
  69. 69. Ruan Y, Yao L, Zhang B, Zhang S, Guo J. Anti-inflammatory effects of neurotoxin-Nna, a peptide separated from the venom of Naja naja atra. BMC Complementary and Alternative Medicine. 2013;13(1):1-5. DOI: 10.1186/1472-6882-13-86
  70. 70. Zhu KZ, Liu YL, Gu JH, Qin ZH. Antinociceptive and anti-inflammatory effects of orally administrated denatured naja naja atra venom on murine rheumatoid arthritis models. Evidence-based Complementary and Alternative Medicine. 2013;2013(4):1-10. DOI: 10.1155/2013/616241
  71. 71. Chen CX, Chen JY, Kou JQ, Xu YL, Wang SZ, Zhu Q, et al. Suppression of inflammation and arthritis by orally administered cardiotoxin from Naja naja atra. Evidence-based Complementary and Alternative Medicine. 2015;2015:1-12. DOI: 10.1155/2015/387094
  72. 72. Gomes A, Datta P, Das T, Biswas AK, Gomes A. Anti arthritic and anti inflammatory activity of a cytotoxic protein NN-32 from Indian spectacle cobra (Naja naja) venom in male albino rats. Toxicon. 2014;90:106-110. DOI: 10.1016/j.toxicon.2014.07.002
  73. 73. Liu YL, Lin HM, Zou R, Wu JC, Han R, Raymond LN, et al. Suppression of complete Freund's adjuvant-induced adjuvant arthritis by cobratoxin. Acta Pharmacologica Sinica. 2009;30(2):219-227. DOI: 10.1038/aps.2008.20
  74. 74. Schett G, Coates LC, Ash ZR, Finzel S, Conaghan PG. Structural damage in rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis: Traditional views, novel insights gained from TNF blockade, and concepts for the future. Arthritis Research & Therapy. 2011;13(Suppl 1):S4. DOI: 10.1186/1478-6354-13-S1-S4
  75. 75. Zhu Q, Huang J, Wang SZ, Qin ZH, Lin F. Cobrotoxin extracted from Naja atra venom relieves arthritis symptoms through anti-inflammation and immunosuppression effects in rat arthritis models. Journal of Ethnopharmacology. 2016;194:1087-1095. DOI: 10.1016/j.jep.2016.11.009
  76. 76. Park MH, Song HS, Kim KH, Son DJ, Lee SH, Yoon DY, et al. Cobrotoxin inhibits NF-κB activation and target gene expression through reaction with NF-κB signal molecules. Biochemistry. 2005;44(23):8326-8336. DOI: 10.1021/bi050156h
  77. 77. Metzger, Maggie, Potential therapeutic effects of snake venom components on pain management in rheumatoid arthritis patients [University Honors Theses. Paper 1075. 2021]. DOI: 10.15760/honors.1101
  78. 78. Vyas VK, Brahmbhatt K, Bhatt H, Parmar U. Therapeutic potential of snake venom in cancer therapy: Current perspectives. Asian Pacific Journal of Tropical Biomedicine. 2013;3(2):156-162. DOI: 10.1016/S2221-1691(13)60042-8
  79. 79. Naumann GB, Silva LF, Silva L, Faria G, Richardson M, Evangelista K. Cytotoxicity and inhibition of platelet aggregation caused by an L-amino acid oxidase from Bothrops leucurus venom. Biochimica et Biophysica Acta. 2011;1810:683-694
  80. 80. Ahn MY, Lee BM, Kim YS. Characterization and cytotoxicity of L-amino acid oxidase from the venom of king cobra (Ophiophagus hannah). The International Journal of Biochemistry & Cell Biology. 1997;29:911-919
  81. 81. Gebrim LC, Marcussi S, Menaldo DL, De Menezes CSR, Nomizo A, Hamaguchi A. Antitumor effects of snake venom chemically modified Lys49 phospholipase A2-like BthTX-I and a synthetic peptide derived from its C-terminal region. Biologicals. 2009;37:222-229
  82. 82. Das T, Bhattacharya S, Halder B, Biswas A, Gupta SD, Gomes A. Cytotoxic and antioxidant property of a purified fraction (NN-32) of Indian Naja naja venom on Ehrlich ascites carcinoma in BALB/c mice. Toxicon. 2011;57:1065-1072
  83. 83. Sun LK, Yoshii Y, Hyodo A, Tsurushima H, Saito A, Harakuni T, et al. Apoptotic effect in the glioma cells induced by specific protein extracted from Okinawa habu (Trimeresurus flavoviridis) venom in relation to oxidative stress. Toxicology In Vitro. 2003;17:169-177
  84. 84. Karthikeyan R, Karthigayan S, Sri Balasubashini M, Somasundaram ST, Balasubramanian T. Inhibition of Hep2 and HeLa cell proliferation in vitro and EAC tumor growth in vivo by Lapemis curtus (Shaw 1802) venom. Toxicon. 2008;51(157-161):50
  85. 85. Lin KL, Su JC, Chien CM, Chuang PW, Chang LS, Lin SR. Down-regulation of the JAK2/PI3K-mediated signaling activation is involved in Taiwan cobra cardiotoxin III-induced apoptosis of human breast MDA-MB-231 cancer cells. Toxicon. 2010;55:1263-1273
  86. 86. Nunes ES, Souza MA, Vaz AF, Silva TG, Aguiar JS, Batista AM. Cytotoxic effect and apoptosis induction by Bothrops leucurus venom lectin on tumor cell lines. Toxicon. 2012;59:667-671
  87. 87. Nolte S, De Castro DD, Barea AC, Gomes J, Magalhães A, Mello Zischler LF. A lectin purified from Bothrops jararacussu venom, induces apoptosis in human gastric carcinoma cells accompanied by inhibition of cell adhesion and actin cytoskeleton disassembly. Toxicon. 2012;59:81-85
  88. 88. Iqbal Alam M, Ojha R, Alam MA, Quasimi H. Therapeutic potential of snake venoms as antimicrobial agents. Frontiers in Drug, Chemistry and Clinical Research, Oat. 2019;2:3-9. DOI: 10.15761/FDCCR.1000136
  89. 89. Reid PF. Alpha-cobratoxin as a possible therapy for multiple sclerosis: A review of the literature leading to its development for this application. Critical Reviews in Immunology. 2007;27:291-302
  90. 90. Ferreira BL, Santos DO, Dos Santos AL, Rodrigues CR, de Freitas CC, et al. Comparative analysis of viperidae venoms antibacterial profile: A short communication for proteomics. Evidence-based Complementary and Alternative Medicine. 2011;2011:1-4
  91. 91. San TM, Vejayan J, Shanmugan K, Ibrahim H. Screening antimicrobial activity of venoms from snakes commonly found in Malaysia. Journal of Applied Sciences. 2010;10:2328-2332
  92. 92. Lee ML, Tan NH, Fung SY, Sekaran SD. Antibacterial action of a heat-stable form of L-amino acid oxidase isolated from king cobra (Ophiophagus hannah) venom. Comparative Biochemistry and Physiology. Toxicology & Pharmacology. 2011;153:237-242
  93. 93. Samy PR, Gopalakrishnakone P, Ho B, Chow VT. Purification, characterization and bactericidal activities of basic phospholipase A2 from the venom of Agkistrodon halys (Chinese pallas). Biochimie. 2008;90:1372-1388
  94. 94. Xie JP, Yue J, Xiong YL, Wang WY, Yu SQ. In vitro activities of small peptides from snake venom against clinical isolates of drug-resistant mycobacterium tuberculosis. International Journal of Antimicrobial Agents. 2003;22:172-174
  95. 95. Nair DG, Fry BG, Alewood P, Kumar PP, Kini RM. Antimicrobial activity of omwaprin, a new member of the waprin family of snake venom proteins. The Biochemical Journal. 2007;402:93-104
  96. 96. Wang Y, Hong J, Liu X, Yang H, Liu R. Snake cathelicidin from Bungarus fasciatus is a potent peptide antibiotics. PLoS One. 2008;3:e3217
  97. 97. Ciscotto P, Machado de Avila RA, Coelho EA, Oliveira J, Diniz CG, et al. Antigenic, microbicidal and antiparasitic properties of an l-amino acid oxidase isolated from Bothrops jararaca snake venom. Toxicon. 2009;53:330-341
  98. 98. Rodrigues RS, da Silva JF, Boldrini França J, Fonseca FP, Otaviano AR, et al. Structural and functional properties of Bp-LAAO, a new L-amino acid oxidase isolated from Bothrops pauloensis snake venom. Biochimie. 2009;91:490-501
  99. 99. Samel M, Vija H, Kurvet I, Künnis-Beres K, Trummal K, et al. Interactions of PLA2-s from Vipera lebetina, Vipera berus berus and Naja naja oxiana venom with platelets, bacterial and cancer cells. Toxins (Basel). 2013;5:203-223
  100. 100. Toyama MH, Toyama Dde O, Passero LF, Laurenti MD, Corbett CE, et al. Isolation of a new L-amino acid oxidase from Crotalus durissus cascavella venom. Toxicon. 2006;47:47-57
  101. 101. Tõnismägi K, Samel M, Trummal K, Rönnholm G, Siigur J, et al. L-amino acid oxidase from Vipera lebetina venom: Isolation, characterization, effects on platelets and bacteria. Toxicon. 2006;48:227-237
  102. 102. Sun MZ. Biochemical, functional and structural characterization of Akbu-LAAO: A novel snake venom L-amino acid oxidase from Agkistrodon blomhoffii ussurensis. Biochimie. 2010;92:343-349
  103. 103. Kitani Y, Kikuchi N, Zhang G, Ishizaki S, Shimakura K, et al. Antibacterial action of L-amino acid oxidase from the skin mucus of rockfish Sebastes schlegelii. Comparative Biochemistry and Physiology. Part B, Biochemistry & Molecular Biology. 2008;149:394-400
  104. 104. Zhang H, Teng M, Niu L, Wang Y, Wang Y, et al. Purification, partial characterization, crystallization and structural determination of AHP-LAAO, a novel L-amino-acid oxidase with cell apoptosis-inducing activity from Agkistrodon halys pallas venom. Acta Crystallographica. Section D, Biological Crystallography. 2004;60:974-977
  105. 105. Xu C, Ma D, Yu H, Li Z, Liang J, et al. A bactericidal homodimeric phospholipases A2 from Bungarus fasciatus venom. Peptides. 2007;28:969-973
  106. 106. Oliveira DG. Structural and functional characterization of basic PLA2 isolated from Crotalus durissus terrificus venom. Journal of Protein Chemistry. 2002;21:161-168
  107. 107. Bhattacharjya S, Ramamoorthy A. Multifunctional host defense peptides: Functional and mechanistic insights from NMR structures of potent antimicrobial peptides. The FEBS Journal. 2009;276:6465-6473
  108. 108. Wang Y, Zhang Z, Chen L, Guang H, Li Z, et al. Cathelicidin-BF, a snake cathelicidin-derived antimicrobial peptide, could be an excellent therapeutic agent for acne vulgaris. PLoS One. 2011;6:e22120
  109. 109. Xia X, Zhang L, Wang Y. The antimicrobial peptide cathelicidin-BF could be a potential therapeutic for salmonella typhimurium infection. Microbiological Research. 2015;171:45-51
  110. 110. Zhao H, Gan TX, Liu XD, Jin Y, Lee WH, et al. Identification and characterization of novel reptile cathelicidins from elapid snakes. Peptides. 2008;29:1685-1691
  111. 111. Zhou H, Dou J, Wang J, Chen L, Wang H, et al. The antibacterial activity of BF-30 in vitro and in infected burned rats is through interference with cytoplasmic membrane integrity. Peptides. 2011;32:1131-1138

Written By

Mamdouh Ibrahim Nassar

Submitted: 11 October 2021 Reviewed: 27 October 2021 Published: 14 April 2022