Journal of Clinical Toxicology

Journal of Clinical Toxicology
Open Access

ISSN: 2161-0495

+44 1478 350008

Review Article - (2014) Volume 4, Issue 2

Pathophysiological and Pharmacological Effects of Snake Venom Components: Molecular Targets

Laraba-Djebari Fatima* and Chérifi Fatah
USTHB, Laboratory of Cellular and Molecular Biology, Algeria
*Corresponding Author: Laraba-Djebari Fatima, USTHB, Faculty of Biological Sciences, Laboratory Of Cellular And Molecular Biology; BP 32 El-Alia, Bab Ezzouar, Algiers, Algeria, Tel: 213-21-24-7950, Fax: +21321336077 Email:

Abstract

Snake venoms are a mixture of hydrolases which produce complex pathogenesis such as bleeding, dermo/myonecrosis, inflammation and coagulation disorders. The toxicity of venoms cannot be attributed to only one component. It is well known that venom components present antagonist activities, while some of them work synergistically. Binding to their intra- and extra-cellular or molecular targets, leads these components to generate severe disturbances which might concern several systems through complex mechanisms. Some of these mechanisms are still not yet elucidated. Thus, some of these components can act at different steps of blood coagulation by activating or inhibiting several molecular or cellular targets thereby inducing blood disorders. Despite their effects, it is well established that some of components from snake venoms present beneficial effects when acting alone as purified entity. Appropriate treatments of snakebite victims need a complete understanding of the pharmacological roles of the different venom components. Thus, this review emphasizes the toxicological relevance of snake venoms mainly those of Viperidae and their components as pharmacological bioactive tools.

Keywords: Snake venoms; Toxicology; Bleeding; Myotoxicity; Hemostasis; Biomolecules; Immunotherapy

Introduction

Snakes are the most feared venomous animals in the world due to their induced morbidity and mortality worldwide which represent 5,400,000 bites over 2,500,000 fatalities followed by about 125,000 deaths [1]. However, some retrospective studies reported that the incidence, mortality and long term disability due to snakebites were shown to be much higher [2-4]. They are poikilothermic and carnivorous reptiles, they are very abundant in hot regions of the world [5]. Snakes belong to the phylum of vertebrates and class of reptiles; they form with the Saurians, the order of Squamates which are divided into four families: Elapidae located in southeast of Asia, in central and southern America and Australia. Hydrophidae distributed in Asia [6]. Crotalidae are found in North America as well as in South Asia. Viperidae are more abundant in Europe, Southeast of Asia and very common in Africa. This family was represented by more than 250 species which are adapted to environmental conditions; it appeared 25 million years before. Snakes represent the most venomous animals, their venoms are complex mixtures of molecules that induce diverse effects on the human systems (hemorrhage, edema, myonecrosis and bleeding disorders) (Figure 1) [7,8].

clinical-toxicology-Pathophysiological-effects

Figure 1: Pathophysiological effects induced by snake venoms.

Due to the richness, heterogeneity and synergistic or antagonistic action of different components, the mechanisms of all these effects are not yet all fully understood. Envenoming induced by snakebite is characterized by local tissue damage involving hemorrhage, blistering, myonecrosis and inflammation. The inflammatory response has relevance in the evolution of tissue damage; it is associated with edema, pain, leukocyte infiltration and release of several mediators. Pathogenesis induced by snake venoms is multi-factorial and complex; it is characterized by local and systemic alterations.

The induced symptoms after bites vary in humans, depending on the amount of inoculated venom, bite site, age, weight and response of each bitten patient. Several studies reported clinical symptoms in relationship with the biochemical variability of venom composition which leads to tissue damage causing failure of various vital organs. Death can occur few days or several weeks after snake bite.

Snake Venoms and their Relevant Components

Venoms

Snake venoms are rich bio-resource of biologically active compounds, but only one percent of these molecules have been characterized; therefore, many of the bioactive components remain to be explored. These biomolecules present diverse activities which could also be used as tools mainly for medical research or diagnosis [9-11]. Identification and characterization of toxic compounds present in snake venoms are the main step not only to understand the pathophysiological changes observed after bites, but they can also be useful to improve the treatment after snake bites.

Components

Secreted proteins represent the major components of snake venoms, they are encoded by poly-adenylated mRNA of venomous glands (12S and 20S) [12]. These proteins have diverse biological activities, some of them are hydrolytic enzymes which help snake in its digestion, and some others are able to induce metabolic dysfunctions of the prey and/or to kill it [13]. In addition to secreted proteins, other components are also found in the venoms, such as lipids, polysaccharides, nucleotides, nucleosides, free amino acids, riboflavin, serotonin and histamine. Pharmacological active substances of the venoms are enzymes and low molecular weight peptides. Some of these enzymes may contribute to the toxic activity of the venoms [14]. Main targets of isolated enzymes from snake venoms are cell membranes, vascular wall and blood coagulation cascade [15,16]. Snake venoms and mainly those of Viperidae contain also molecules that act on the four interconnected blood systems as producers of inflammatory mediators, (i) coagulation system, (ii) fibrinolysis, (iii) complement and (iiii) kinin system (Figure 2) [17].

clinical-toxicology-Involvement-SVSPs

Figure 2: Involvement of SVSPs and SVMPs in hemostatic disturbances [9-11,20,21,24,25].

Proteinases

Knowledge of the combined effects of different components of venom leads to the better understanding of the observed symptoms in snake envenomation [18]. Proteolytic enzymes are particularly involved in the pathogenesis of tissue necrosis, hemorrhage and bleeding disorders. Proteinases isolated from viper venoms represent a heterogeneous group of enzymatic proteins of 15 and 100 kDa [19]. Some of these proteinases act on blood coagulation factors and can be pro-coagulant or anti-coagulant since they may exert activating or inhibiting effects of plasma factors. They are also endowed with fibrino(geno) lytic activities as well as thrombin and plasmin. Venom proteases are divided into two broad classes of enzymes: Snake Venom Serine Proteases (SVSPs) and Snake Venom Metalloproteinases (SVMPs) [20-23]. The structure of these enzymes is stabilized by disulfide bridges [20-22,24]. These enzymes are able to hydrolyze natural substrates such as casein, hemoglobin and fibrinogen as well as synthetic substrates [25].

Snake Venom Serine proteases (SVSPs):

Serine proteases are abundant in snake venoms, they have been identified in venoms mainly from the subfamily of Crotalinae (Agkistrodon, Crotalus, Lachesis, Trimeresurus), Viperinae (Cerastes cerastes, Cerastesvipera and Bitisgabonica) and Colubrinae (Dipholidustypus) [25]. According to their effects on the hemostatic system, they can be classified as kallikrein-like which lead to the release of bradykinin or as fibrinogenases [26,27]. Most of these proteases affect several targets of hemostasis, platelets, plasma coagulation and fibrinolytic system [25]. They are called "Thrombinic enzymes from snake venom" or Snake Venom Thrombin-Like Enzymes (SVTLEs). SVSPs (20 to 100 kDa) present preserved a common domain which consists in a catalytic triad of three basic amino acid histidine (His 57), serine (Ser 195), hence the name "serine protease" and aspartic acid (Asp 102), each of these amino acids play a key role in the catalytic activity. SVSPs target mainly the coagulation cascade and act as potent platelet aggregating molecules and/or as exogenous plasma factors (Figure 2) [28]. Several proteolytic enzymes are purified from snake venoms; among them those purified from Cerastescerastes venom as α, β-fibrinogenases based on their ability to hydrolyze the fibrinogen such as the procoagulant proteinases and fibrinogenases (RP34, afaâcytin and CC3-SPase). Afaâcytin, RP34 and CC3-SPase displayed, respectively, α,ß-fibrinogenase and α-fibrinogenase activity [26,29].

Snake Venom Metalloproteinases (SVMPs):

Several SVMPs (22 to 100 kDa) isolated from snake venoms have been characterized as Zn2+- metalloproteinases which have components of the basement membrane of the endothelial cells as main targets [30,31]. Hemostatic system disorders are the most studied biological effect induced by SVMPs [14,32]. In addition to their procoagulant and anti-coagulant activities, SVMPs are also involved in the pathogenesis of edema, inflammation, myonecrosis, skin damage and the development of cardiovascular shock [32-37]. They are able to inhibit platelet aggregation which potentiates the effect of bleeding [38]. SVMPs are also able to degrade the extracellular matrix components (collagen, proteoglycans, laminin and fibronectin) inducing massive extravasation of blood [36,39]. These metalloproteinases are responsible of the induced local and systemic bleeding after bites; affecting various organs (heart, liver, lungs, intestines and brain), so they are called hemorragins. They can also cause swelling, blisters and necrosis. These enzymes are, therefore, widely involved in the pathogenesis of tissue necrosis [40]. SVMPs are synthesized in vivo as inactive zymogens, organized into domains containing a signal peptide, a pro-domain and a conserved catalytic domain with a binding site for zinc, disintegrin-type or lectin-domain can be associated at these domains [15,32,41-43]. SVMPs, Zn2+- or both Zn2+/Ca2+-dependent, are characterized by a required sequence (HEXXHXXGXXH) for the Zn2+ binding, which is essential for their proteolytic activity [44-47]. Histidyl residues are involved in zinc binding, while the glutamate residue allows the polarization of water molecule by zinc. Based on their structures and molecular masses, SVMPs were previously classified into four classes [47-50]. However, further investigations on SVMPs structure updated this classification into only three classes. The mature P-I class contains only a metalloproteinase domain. This domain is followed by disintegrin domain in class P-II while in the P-III class these two domains are linked to disintegrin-like and cysteine-rich domains [51-53]. Isolated metalloproteinases from snake venoms are able also to disrupt hemostatic system since they exhibit α or β-fibrinogenase activities or inhibit platelet aggregation enhancing bleeding effect [15,32,38].

Disintegrins and C-type Lectins

Protein components of snake venoms belong to families of disintegrins and C-type lectins are, increasingly, used in biomedical research, in diagnostic or/and therapeutic purposes. Due to their effects on various platelet receptors (GPIb, GPIIb/IIIa, GPVI, α2β1.), disintegrins and C-type lectin domains have been considered as modulators of the platelet aggregation (Figure 3).

clinical-toxicology-modulator-platelets

Figure 3: Agonists of platelet receptors involved in adhesion and aggregation, the relationship with C-type lectin proteins (CLPs) as anticoagulant, procoagulant and modulator of platelets.

By binding to αIIbβ3 integrin on activated platelets, and thus preventing its interaction with fibrinogen, RGD disintegrins inhibit platelet aggregation induced by a wide range of agonists, e.g. ADP, α-thrombin, collagen and arachidonic acid. Based on their ability to inhibit adhesion, migration, proliferation and invasion of different cancer cell lines some of these disintegrins and C-type lectins have been described as anti-tumoral potential effect. C-type lectins have also in vivo and in vitro an anti-angiogenic powerful by interacting with integrins of endothelial cells [54,55]. Several disintegrins have been identified in vitro and in vivo as potent inhibitors of platelet aggregation, their structures served also as a model in drug design. Some of them are clinically used as anti-platelets for coronary artery diseases, such as Eptifibatide (Integrilin) and Tirofiban. Eptifibatide is a cycle peptide derivative of the disintegrin barbourin isolated from dusky pigmy rattlesnake (Sistrususbarbouri), while, Tirofiban, is a synthetic molecule mimicking the disintegrin Echistatin from Echiscarinatus[56,57]. The possibility to identify lectins using new receptors (other than GPIb, GPVI and α2β1) could be for help to develop and provide new opportunities in the diagnosis and treatment of hemostasis. Meanwhile, new anti-tumor and anti-angiogenic activities of some lectins (lebecetin, lebectin and BJcuL) open new therapeutic perspectives in the field of cancer treatment.

Phospholipases A2 (PLA2s)

Snake venoms are one of the richest sources of PLA2s. They often contain a large number of iso-enzymes such as in Naja naja, Vipera russelli, Trimeresurus flavoviridis venoms [58]. Snake venom PLA2s are divided into two groups according to their primary sequence and the position of their disulfide bridges [59,60]. PLA2s of Elapidae and Hydrophidae venoms belong to the Group I, while those of Viperinae and Crotalinae belong to the Group II. The structure of these two groups is very similar, it consists of 120-125 amino acid residues, stabilized by seven disulfide bridges [59,61,62]. SV-PLA2s induced various biological effects such as neurotoxic, myotoxic, cytolytic, edematic, cardiotoxic and anticoagulant effects [63-67].

Myotoxic PLA2s and myotoxic peptides:

PLA2s are the most abundant myotoxic components of snake venoms; they induce similar degenerative events in muscle cells. The myotoxic PLA2s may be endowed with a phospholipase activity or not [68-70]. The active site of myotoxic PLA2s is highly conserved (His 48, Asp 49, Tyr 52 and Asp 99). The residue Asp 49 binds to calcium, which is essential to the PLA2 activity [66]. Myotoxins devoid in phospholipase activity, have the same active site residues (His 48, Tyr 52 and Asp 99). The residue Asp 49 could be substituted by Lysine or Serine. Otherwise, the substitution of aspartate 49 could prevent the binding of calcium, resulting in a loss of phospholipase activity. This type of molecule nevertheless retains myotoxicity [66,71]. The region (115-129 in C-terminus) of the Lys49 PLA2s is responsible for their ability to alter the integrity of the bilayer membrane. The onset of muscle damage even in the absence of phospholipase activity showed that the catalytic domain of PLA2s differ of their biological activities, including their myotoxic activity (Figure 4).

clinical-toxicology-Probable-mechanism

Figure 4: Probable mechanism of myotoxin.

However, Ser49 or Lys49 PLA2s altered muscle tissue by an independent mechanism of enzymatic hydrolysis of membrane phospholipids [72-74].

Myotoxins without PLA2 activity are peptides of 42 to 52 amino acid residues, stabilized by three disulfide bonds. This activity is related to their two successive sequences at their C-terminus (Figure 4). The first cationic sequence is rich in positive charged residues, while the second sequence is rich in hydrophobic residues; both are bound by three to four disulfide bridges.

Toxicological Relevance of Snake Venoms: Envenomation And Therapy

Envenomation

Biodistribution of snake venoms and their components:

Many studies on biodistribution of snake venoms were performed using sandwich ELISA, the biodistribution could explain their induced clinical symptoms [75]. These studies showed that snake venoms are rapidly absorbed from the injection site and diffused to the tissue and the vascular compartment; however, their elimination is very slow. Conducted study on Vipera aspis venom radio-labeled with Iodine125 and injected by i.v. route (260 mg/kg of rabbit weight) showed a rapid distribution with an estimated time of 15 ± 3.6 minutes determined by radio-labeling and 31 ± 8.4 minutes determined by sandwich ELISA [75]. Further study on Walterinnesia aegyptia venom showed a rapid absorption with 20 ± 2.1 minutes and a wide distribution of venom in vascular and tissular compartments of rabbit [76]. This biodistribution was correlated to that of Vipera aspis venom injected by i.v. route which [76]. Parallaly, after envenomation with Vipera aspis, observed symptoms appeared slowly and sustainably develop [76]. Furthermore, the toxicokinetic study of the venom injected subcutaneous is widely distributed throughout the body, indicating that the components of venom widely diffuse out of the vascular compartment; this would result in very long half-life time elimination. Vipera aspis venom injected intramuscularly revealed large volume of distribution of the venom (Vd = 2 L.kg) and low amount of venom (less than 4%) was excreted through the kidneys [77]. In another hand, biodistribution of Cerastes cerastes and Vipera lebetina venoms carried out by sandwich ELISA in the different compartments of rats revealed that rapid diffusion of venoms from the serum to the tissues. Maximum serum concentration is reached after 3 hours of envenomation. The kidneys, liver, lungs, heart and pancreas, are the main target organs for Cerastes cerastes and Vipera lebetina venoms. The absence of the venoms in the brain is attributed to the presence of the blood- lymphatic barrier that prevents the passage of toxins in the venom of the brain [76].

Induced pathophysiology by venom components and involved mechanisms II.1.2.1. Inflammatory response:

Involvement of inflammatory process in the pathogenesis of snake envenomation was reported since 90s [50,77,78]. It is well known that snake venoms contain various activities able to activate several pathways (Figure 5).

clinical-toxicology-Inflammatory-response

Figure 5: Inflammatory response induced by snake venom components (Fg: Fibrinogen, vWF: Von Willbrand Factor, GP: Glycoprotein).

Increase of capillary permeability was reported after snake envenomation leading to the release of several mediators. Many components of snake venoms (PLA2s, bioamines and proteinases) contribute to the induced inflammatory response which is initiated by an increase of vascular permeability followed by cell infiltration [79]. Several studies reported that SVMPs such as BaP1 from Bothrops asper and Jararhagin from Bothrops jararaca, are involved in the inflammatory pathogenesis leading to an increase of pro-inflammatory cytokine production [80,81]. The induced inflammatory response by snake venoms particularly those ofViperidae, is amplified by the presence of metalloproteinases, serine proteases, phospholipases A2 and other non-enzymatic proteins such as disintegrins and C-type lectins, which alter the vessel walls and trigger tissue damage. SVMPs play a relevant role in the complex multifactorial inflammatory response induced by snakebite. High levels of IL-6 and IL-1β were observed in muscular tissue of envenomed animals by Bothropsaspervenom [82]. However, a rapid increase of IL-6 levels versus a late onset of IL-1 and TNF-α was observed after injection of Bothropsaspervenom[78]. It was reported that Promutoxin (R49sPLA2) isolated from the venom of Probothropsmuscrosquamatus, induced a release of IL-12, TNF-α, IL-6 and IL-1β from a cell culture of human monocytes and IL-2 cytokines, TNF and IL-6 from human T lymphocytes. Some SV-PLA2s hydrolyze membrane phospholipids of platelets, leading to the release of agonists, mainly arachidonic acid a precursor of several inflammatory substances such as prostaglandins and leukotrienes [9,11]. Activation of the complement system results in the formation of many additional degradation products that serve as important mediators of inflammation. Snake venoms stimulate the activation of mast cells which lead to histamine release, inducing vascular permeability and vasodilatation leading to extravasation. Furthermore, the kinin system can also be activated directly by the proteinases of snake venoms that activate the release of bradykinin [83,84]. This system is initiated by activation of Hageman factor (FXII) following tissue injury. This plasmatic factor activates in turn, the pre-kalikrein into kalikrein, in presence of kininogen, leading to vasoactive peptides causing fever and pain. Bradykinin is a nano-peptide responsible for the increased vascular permeability due to its binding with specific receptors on sensory neurons; it, therefore, activates the alternative complement pathway which amplifies the inflammatory response. SVMPs such as fibrolase isolated from Agkistrodon contortrix contortrix venom, is described to be involved in the biosynthesis/degradation pathways of bradykinin [85].

Blood disorders

Coagulation and fibrinolysis:

Proteolytic enzymes isolated from snake venoms were identified as α, β or γ fibrinogenases depending on their ability to hydrolyze the fibrinogen in vitro [10].

Several thrombin-like molecules isolated from snake venoms induce the release of either fibrinopeptides A or B or both of them. These fibrinopeptides act as mediators of inflammation and induce vascular permeability and neutrophil chemotaxis [15,11]. The formed thrombus is considered as a potent chemotactic agent for neutrophils and in vascular permeability acting on the kinin system. Viperidae and Crotalidae venoms are also able to induce fibrinolysis. Purification and characterization of three procoagulant proteinases (RP34, Afaâcytin and CC3-SPase proteinase) from Cerastes cerastes venom, showed fibrinogenolytic activities when analyzed by SDS-PAGE, afaâcytin and RP34 displayed, respectively, α,ß-fibrinogenase and α-fibrinogenase activity [9,10,25]. Like afaâcytin, CC3-SPase is also characterized as an α,β-fibrinogenase due to the release of both A and B fibrinopeptides. TSV-PA, a serine proteinase purified from the venom of Trimeresurusstejnegeri, plays a role of plasminogen activator. It shares about 70% of homology with other serine proteases and has significant structural similarities with the t-PA, it converts plasminogen to plasmin in the same way that the latter by the cleavage of bond Arg561-Val562 [86,87].

Hemorrhage:

Spontaneous systemic bleeding is caused by hemorrhagins which damage vascular endothelium. Additional effects caused by snake envenomations (coagulopathies, hemorrhage, impaired and few platelets, and vessel wall damage) can result in severe bleeding, a common cause of death after bites by Viperidae, Elapidae and Colubridae. SVMPs through their disintegrin domain may have multiple roles; it would allow to the amplification of hemorragin action affecting platelet aggregation [9,50]. Disintegrins interact and block platelets integrins via their integrin-binding tri-peptide motifs; RGD (Arginine-Glycine-Aspartate) is the most common motif between many disintegrins, while other variants of motifs can also be found in some disintegrins e.g., KGD, WGD, MGD, KTS, RTS and MLD [88]. These motifs can act and inhibit platelet aggregation through their selective binding to integrins such as GPIIb/IIIa or αIIBβ3. SVMPs degrade also platelet membrane glycoproteins and their ligands such as collagen, GPIb and Von Willbrand factor [89,90].

Myotoxicity

Snake envenomation induces prominent local tissue damage that often results in permanent disability and systemic alterations associated with haemorrhage, coagulopathies, cardiovascular shock and renal failure. Clinical reports indicate that, in humans, the main invalidating effect is the irreversible disruption of muscle tissue [91]. Tissue necrosis is a relevant local effect caused after snakebites, it is considered as a serious consequence in severe cases of envenomation. When myonecrosis appears tissues are altered leading to the gangrene and infections. This type of complication can be the cause of amputation. Indeed, myotoxins of snake venoms affect mainly the plasma membrane of muscle cells to which they bind through their cationic sequence [73,92]. Molecular mechanism by which they caused the muscle tissue damage is not yet fully elucidated. Myonecrosis is due to the myotoxins that induce irreversible damage of skeletal muscle fibers. These molecules bind to the plasma membrane of muscle cells and alter its permeability and integrity (Figure 4). The induced muscle tissue damage could be due to the penetration of myotoxins into muscle cells by endocytosis, probably through membrane receptors onto the surface of muscle cells or following hydrolysis of phospholipids causing membrane disruption. These molecules enter into the cytosol, reach and alter the membrane of mitochondria and sarcoplasmic reticulum of muscle cells. The intracellular effect of these toxins occurs only after their initial action on the plasma membrane, which marks the onset of degenerative events [93,94].

Treatment

The complexity of the snake venoms and their induced effects after envenomation makes difficult their treatment. However, more attention was given to loco-regional disorders that sometimes lead to amputations and permanent disabilities. Human suffering attributable to snake bites remains a public health problem in many countries of the world, several people over the world are known to be envenomed and some them are killed or maimed by snakes every year. Preventive efforts should be aimed towards education of regions at-risk to reduce contact with snakes and to understand snakes’ behavior [88]. Whatever the therapy used, it should include not only the neutralization of toxicity but also the other effects induced by venoms (hemorrhage and necrosis…). To treat snake envenoming, the production and clinical use of antivenom must be improved. Although antivenom was effective in the neutralization of systemic complications. It has limited effectiveness against the development of local damage, metabolic dysfunctions and tissue damage caused by venom components that are responsible for its health hazards due to their fast distribution and effects such as hemorrhage, myotoxicity and edema-forming [93]. Collaboration between physicians, epidemiologists and toxinologists should enhance the understanding and treatment of envenoming [88].

Beneficial Effects of Isolated Components from Snake Venoms

Most of venom compounds acquire interesting properties increasingly used in biomedical research and as tools in diagnosis and/or therapies. Indeed, the specific nature of coagulant or anti-coagulant properties of venoms makes them useful to better understand the haemostatic mechanisms [10,11]. Some of these components act synergistically at different stages of the coagulation cascade [95]. SVSPs and SVMPs present an interest as biomedicines and may be used as diagnostic tools; they act on this system as pro-coagulants, anticoagulants, and on platelet aggregation as pro- or anti-platelet [10]. Some of these molecules are used in the diagnosis and treatment of thrombotic and heart diseases. Components of snake venoms contain two categories of components that act antagonistically through activation or inhibition of coagulation factors and platelet aggregation. These compounds, able to hydrolyze the coagulation factors with high specificity, are divided into serine proteinases and metalloproteinases. Plasma defibrinogenation induced by snake venom components is of interest. Indeed, Arvin “ancrod”, isolated from Agkistrodonrhodostomavenom, is one of the molecules used in patients treated with anti-coagulants. It is also used to lower the levels of fibrinogen in the treatment of peripheral vascular disorders. Botroxobin, another molecule isolated from the venom of Bothropsatrox and Bothropsmoojeni, converts fibrinogen into fibrin by releasing only fibrinopeptide A. It is used for its defibrinogenating effect in the treatment of thrombotic diseases. The coagulating properties of Afaâcytin (Bothropsasper), insensitive to specific plasma thrombin inhibitors could be useful as hemostatic agent in some cases of bleeding and thrombocytopenia such as observed in post-operative situations [4,28]. The fibrinogenase RP34 could serve as defibrinogenating agent in the case of certain diseases [24,28,96]. All these defibrinogenating biomolecules share their properties, may be used as tools in clinical applications or in basic research. More in-depth studies in pharmacology, toxicology of these biomolecules are to be undertaken to determine their mode of action in vivo.

Conclusion

Snake venoms, considered to be one of the most important bio-resources, include pharmacologically active molecules. To better understand the diversity of biological actions of snake venoms and propose new treatment for some pathologies, many studies focused on purification and characterization of new bioactive compounds from venoms. Currently, biomolecules of snake venoms are of great fundamental diagnostic and therapeutic interest. Characterization, biological properties establishment of these bioactive molecules and investigation on their mechanisms, may lead to their eventual use for therapeutic purposes. Therapeutically, proteinases, disintegrins and C-type lectins from snake venoms are widely used as anticoagulants or/and anti-platelets. Furthermore, they are valuable tools for understanding the different mechanisms of hemostasis and are also used in the diagnosis of dysfunctions related to coagulation factors such as enzyme activity in thrombin-like venoms that are used for the fibrinogenopathy screening. Venom compounds are also used for diagnostic analysis of various coagulation factors (factors V, VII, X, platelet factor III, protein C and factor of Willbrand).

References

  1. Chippaux JP (1998) Snake-bites: appraisal of the global situation. Bull World Health Organ 76: 515-524.
  2. Hati AK, Mandal M, De MK, Mukherjee H, Hati RN (1992) Epidemiology of snake bite in the district of Burdwan, West Bengal. J Indian Med Assoc 90: 145-147.
  3. White J (2004) Viperid snakes. In: Dart R (Eds), Medical Toxicology. Lippincott, Williams and Wilkins, pp. 1579-1591.
  4. Audebert F, Urtizberea M, Sabouraud A, Scherrmann JM, Bon C (1994) Pharmacokinetics of Viperaaspis venom after experimental envenomation in rabbits. J PharmacolExpTher 268: 1512-1517.
  5. Marsh N, Whaler B (1978) The effects of snake venoms on the cardiovascular and haemostatic mechanisms. Int J Biochem 9: 217-220.
  6. Oshaka A (1979) An approach to the physiological mechanism involved in haemorrhagic principles as a useful analytical tool. Animal, plant and microbial toxins 123: 17-28.
  7. Chérifi F, Rousselle JC, Namane A, Laraba-Djebari F (2010) CCSV-MPase, a Novel Procoagulant Metalloproteinase from Cerastescerastes Venom: Purification, Biochemical Characterization and Protein Identification. Protein J 29: 466-474.
  8. Chérifi F, Laraba-Djebari F (2013) Isolated biomolecules of pharmacological interest in hemostasis from Cerastescerastes venom. J Venom Anim Toxins Incl Trop Dis 19: 11.
  9. Chérifi F, Namane A, Laraba-Djebari F (2014) Isolation, functional characterization and proteomic identification of CC2-PLA2 from Cerastescerastes venom: a basic platelet-aggregation-inhibiting factor. Protein Journal 33:66-77.
  10. Qiu Y, Choo YM, Yoon HJ, Jin BR (2012) Molecular cloning and fibrin(ogen)olytic activity of a bumblebee (Bombushypocritasapporoensis) venom serine protease. Journal of Asia-Pacific Entomology 15: 79-82.
  11. Bon C, Arocas V, Braud S, Francischcti I, Leduc M (2000) Snake venom in thombosis and haemostatis. XII th word congress of the International Society of Toxinologie, Paris, September: 18-22.
  12. Menaldo DL, Bernardes CP, Santos-Filho NA, Moura LA, Fuly AL et al. (2012) Biochemical characterization and comparative analysis of two distinct serine proteases from Bothropspirajai snake venom. Biochimie 94: 2545-2558.
  13. Marrakchi N, Barbouche R, Guermazi S, Bon C, el Ayeb M (1997) Procoagulant and platelet-aggregating properties of cerastocytin from Cerastescerastes venom. Toxicon 35: 261-272.
  14. Valeriano-Zapana JA, Segovia-Cruz FS, Rojas-Hualpa JM, Martins-de-Souza D, Ponce-Soto LA, et al. (2012) Functional and structural characterization of a new serine protease with thrombin-like activity TLBan from Bothropsandianus(Andean lancehead) snake venom. Toxicon 59: 231-240.
  15. Laing GD, Moura-da-Silva AM (2005) Jararhagin and its multiple effects on hemostasis. Toxicon 45: 987-996.
  16. Djebari FL, Martin-Eauclaire MF (1990) Purification and characterization of a phospholipase A2 from Cerastescerastes (horn viper) snake venom. Toxicon 28: 637-646.
  17. Ramírez GA, Fletcher PL Jr, Possani LD (1990) Characterization of the venom from CrotalusmolossusnigrescensGloyd (black tail rattlesnake): isolation of two proteases. Toxicon 28: 285-297.
  18. Maruyama M, Sugiki M, Yoshida E, Shimaya K, Mihara H (1992) Broad substrate specificity of snake venom fibrinolytic enzymes: possible role in haemorrhage. Toxicon 30: 1387-1397.
  19. Jin Y, Lu QM, Wei JF, Li DS, Wang WY, et al. (2001) Purification and characterization of jerdofibrase, a serine protease from the venom of Trimeresurusjerdonii snake. Toxicon 39: 1203-1210.
  20. Bortoleto RK, Murakami MT, Watanabe L, Soares AM, Arni RK (2002) Purification, characterization and crystallization of Jararacussin-I, a fibrinogen-clotting enzyme isolated from the venom of Bothropsjararacussu. Toxicon 40: 1307-1312.
  21. Samel M, Subbi J, Siigur J, Siigur E (2002) Biochemical characterization of fibrinogenolytic serine proteinases from Viperalebetina snake venom. Toxicon 40: 51-54.
  22. Cho SY, Hahn BS, Yang KY, Kim YS (2001) Purification and characterization of calobin II, a second type of thrombin-like enzyme from Agkistrodoncaliginosus (Korean viper). Toxicon 39: 499-506.
  23. Laraba-Djebari F, Martin-Eauclaire MF, Marchot P (1992) A fibrinogen-clotting serine proteinase from Cerastescerastes (horned viper) venom with arginine-esterase and amidase activities: purification, characterization and kinetic parameter determination. Toxicon 30: 1399-1410.
  24. Braud S, Parry MA, Maroun R, Bon C, Wisner A (2000) The contribution of residues 192 and 193 to the specificity of snake venom serine proteinases. J BiolChem 275: 1823-1828.
  25. Serrano SM, Mentele R, Sampaio CA, Fink E (1995) Purification, characterization, and amino acid sequence of a serine proteinase, PA-BJ, with platelet-aggregating activity from the venom of Bothropsjararaca. Biochemistry 34: 7186-7193.
  26. Fox JW, Serrano SM (2009) Timeline of key events in snake venom metalloproteinase research. J Proteomics 72: 200-209.
  27. Laraba-Djebari F, Martin-Eauclaire MF, Mauco G, Marchot P (1995) Afaâcytin, an aß-fibrinogenase from Cerastescerastes (horned viper) venom, activates purified factor X and induces serotonin release from human blood platelets. Eur J Biochem 233: 756-765.
  28. Gutiérrez JM, Rucavado A (2000) Snake venom metalloproteinases: their role in the pathogenesis of local tissue damage. Biochimie 82: 841-850.
  29. Mebs D, Pohlmann S, Von Tenspolde W (1988) Snake venom hemorrhagins: neutralization by commercial antivenoms. Toxicon 26: 453-458.
  30. Markland FS (1998) Snake venoms and the hemostatic system. Toxicon 36: 1749-1800.
  31. Marsh NA (1994) Snake venoms affecting the haemostatic mechanism: a consideration of their mechanism, practical applications and biological significance. Blood CoagulFibrinolysis 5: 399-410.
  32. Mackessy SP (1996) Characterization of the major metalloprotease isolated from the venom of the northern pacific rattlesnake, Crotalusviridisoreganus. Toxicon 34: 1277-1285.
  33. Rucavado A, Núñez J, Gutiérrez JM (1998) Blister formation and skin damage induced by BaP1, a haemorrhagic metalloproteinase from the venom of the snake Bothropsasper. Int J ExpPathol 79: 245-254.
  34. Gutiérrez JM, Romero M, Díaz C, Borkow G, Ovadia M (1995) Isolation and characterization of a metalloproteinase with weak hemorrhagic activity from the venom of the snake Bothropsasper (terciopelo). Toxicon 33: 19-29.
  35. Norris RL, Dery R, Johnson C, Williams S, Rose K, et al. (2003) Regional vs systemic antivenom administration in the treatment of snake venom poisoning in a rabbit model: a pilot study. Wilderness Environ Med 14: 231-235.
  36. Lomonte B, Gutiérrez JM, Borkow G, Ovadia M, Tarkowski A, et al. (1994) Activity of hemorrhagic metalloproteinase BaH-1 and myotoxin II from Bothropsasper snake venom on capillary endothelial cells in vitro. Toxicon 32: 505-510.
  37. Mebs D (1986) Myotoxic activity of phospholipases A2 isolated from cobra venoms: neutralization by polyvalent antivenoms. Toxicon 24: 1001-1008.
  38. Rucavado A, Flores-Sánchez E, Franceschi A, Magalhaes A, Gutiérrez JM (1999) Characterization of the local tissue damage induced by LHF-II, a metalloproteinase with weak hemorrhagic activity isolated from Lachesismutamuta snake venom. Toxicon 37: 1297-1312.
  39. Paine MJ, Desmond HP, Theakston RDG, Crampton JM (1992) Purification, cloning and molecular characterization of a high molecular weight hemorrhagic metalloprotease, jararhagin, from Bothropsjararaca venom. Insights into the disintegrin gene family. J BiolChem 267: 22869-22876.
  40. Takeya H, Nishida S, Nishino N, Makinose Y, Omori-Satoh T, et al. (1993) Primary structures of platelet aggregation inhibitors (disintegrins) autoproteolytically released from snake venom hemorrhagic metalloproteinases and new fluoregenic peptide substrates for these enzymes. J Biochem 113: 473-783.
  41. Matsui T, Fujimura Y, Titani K (2000) Snake venom proteases affecting hemostasis and thrombosis. BiochimBiophysActa 1477: 146-156.
  42. Tsai IH, Wang YM, Chiang TY, Chen YL, Huang RJ (2000) Purification, cloning and sequence analyses for pro-metalloprotease-disintegrin variants from Deinagkistrodonacutus venom and subclassification of the small venom metalloproteases. Eur J Biochem 267: 1359-1367.
  43. Jia LG, Shimokawa K, Bjarnason JB, Fox JW (1996) Snake venom metalloproteinases: structure, function and relationship to the ADAMs family of proteins. Toxicon 34: 1269-1276.
  44. Gong W, Zhu X, Liu S, Teng M, Niu L (1998) Crystal structures of acutolysin A, a three-disulfide hemorrhagic zinc metalloproteinase from the snake venom of Agkistrodonacutus. J MolBiol 283: 657-668.
  45. Hati R, Mitra P, Sarker S, Bhattacharyya KK (1999) Snake venom hemorrhagins. Crit Rev Toxicol 29: 1-19.
  46. Hati R, Mitra P, Sarker S, Bhattacharyya KK (1999) Snake venom hemorrhagins. Crit Rev Toxicol 29: 1-19.
  47. Zhang D, Botos I, Gomis-Rüth FX, Doll R, Blood C, et al. (1994) Structural interaction of natural and synthetic inhibitors with the venom metalloproteinase, atrolysin C (form d). ProcNatlAcadSci U S A 91: 8447-8451.
  48. Bjarnason JB, Fox JW (1994) Hemorrhagic metalloproteinases from snake venoms. PharmacolTher 62: 325-372.
  49. Franceschi A, Rucavado A, Mora N, Gutiérrez JM (2000) Purification and characterization of BaH4, a hemorrhagic metalloproteinase from the venom of the snake Bothropsasper. Toxicon 38: 63-77.
  50. Menezes MC,Paes Leme AF, Melo RL, Silva CA, Della Casa M, et al. (2008) Activation of leukocyte rolling by the cysteine-rich domain and the hyper-variable region of HF3, a snake venom hemorrhagic metalloproteinase. FEBS Lett. 28:3915-3921.
  51. Wagstaff SC, Harrison RF (2006) Venom gland EST analysis of the saw-scaled viper, Echisocellatus, reveals novel alpha9beta1 integrin-binding motifs in venom metalloproteinases and a new group of putative toxins, renin-like aspartic proteases. Gene 377:21-32.
  52. Calvete JJ, Marcinkiewicz C, Monleón D, Esteve V, Celda B, et al. (2005) Snake venom disintegrins: evolution of structure and function. Toxicon 45: 1063-1074.
  53. Pereira-Bittencourt M, Carvalho DD, Gagliardi AR, Collins DC (1999) The effect of a lectin from the venom of the snake, Bothropsjararacussu, on tumor cell proliferation. Anticancer Res 19: 4023-4025.
  54. Wang WJ, Huang TF (2002) Purification and characterization of a novel metalloproteinase, acurhagin, from Agkistrodonacutus venom. ThrombHaemost 87: 641-650.
  55. Liu S, Sun MZ, Greenaway FT (2006) A novel plasminogen activator from AgkistrodonblomhoffiiUssurensis venom (ABUSV-PA): purification and characterization. BiochemBiophys Res Commun 348: 1279-1287.
  56. Morita T (2005) Structures and functions of snake venom CLPs (C-type lectin-like proteins) with anticoagulant-, procoagulant-, and platelet-modulating activities. Toxicon 45: 1099-1114.
  57. Kini RM, Iwanaga S (1986) Structure-function relationships of phospholipases. I: Prediction of presynaptic neurotoxicity. Toxicon 24: 527-541.
  58. Davidson FF, Dennis EA (1990) Evolutionary relationships and implications for the regulation of phospholipase A2 from snake venom to human secreted forms. J MolEvol 31: 228-238.
  59. Lomonte B, Moreno E, Tarkowski A, Hanson LA, Maccarana M (1994) Neutralizing interaction between heparins and myotoxin II, a lysine 49 phospholipase A2 from Bothropsasper snake venom. Identification of a heparin-binding and cytolytic toxin region by the use of synthetic peptides and molecular modeling. J BiolChem 269: 29867-29873.
  60. Valentin E, Lambeau G (2000) What can venom phospholipases A(2) tell us about the functional diversity of mammalian secreted phospholipases A(2)? Biochimie 82: 815-831.
  61. Nicolas JP, Lambeau G, Lazdunski M (1995) Identification of the binding domain for secretory phospholipases A2 on their M-type 180-kDa membrane receptor. J BiolChem 270: 28869-28873.
  62. Kini RM, Iwanaga S (1986) Structure-function relationships phospholipases II: charge density distribution and the myotoxicity of presynaptically neurotoxic phospholipases. Toxicon 24: 895-905.
  63. Lambeau G, Ancian P, Nicolas JP, Cupillard L, Zvaritch E, et al. (1996) [A family of receptors for secretory phospholipases A2]. C R SeancesSocBiolFil 190: 425-435.
  64. Arni RK, Ward RJ (1996) Phospholipase A2--a structural review. Toxicon 34: 827-841.
  65. Ward RJ, Alves AR, Neto JR, Arni RK, Casari G (1998) A sequence space analysis of lys49 phospholipases A2: clues towards identification of residues involved in a novel mechanism of membrane damage and in myotoxicity. Protein Engineering 11: 285-294.
  66. Escarso SHA, Soares AM, Rodriguez VM, Angulo Y, Diaz C, et al. (2000) Myotoxic phospholipase A2 in Bothrops snake venoms: effect of chemical modifications on the enzymatic and pharmacological properties of Bothrops toxin from Bothropsjararacussu. Biochimie 82: 755-763.
  67. Kini RM, Evans HJ (1992) Structural domains in venom proteins: evidence that metalloproteinases and non-enzymatic platelet aggregation inhibitors (disintegrins) from snake venoms are derived by proteolysis from a common precursor. Toxicon 30: 265-293.
  68. Fuly AL, Calil-Elias S, Zingali RB, Guimarães JA, Melo PA (2000) Myotoxic activity of an acidic phospholipase A2 isolated from Lachesismuta (Bushmaster) snake venom. Toxicon 38: 961-972.
  69. Gutiérrez JM, Romero M, Díaz C, Borkow G, Ovadia M (1995) Isolation and characterization of a metalloproteinase with weak hemorrhagic activity from the venom of the snake Bothropsasper (terciopelo). Toxicon 33: 19-29.
  70. Lomonte B, Gutiérrez JM, Furtado MF, Otero R, Rosso JP, et al. (1990) Isolation of basic myotoxins from Bothropsmoojeni and Bothropsatrox snake venoms. Toxicon 28: 1137-1146.
  71. Francis B, Gutierrez JM, Lomonte B, Kaiser II (1991) Myotoxin II from Bothropsasper (Terciopelo) venom is a lysine-49 phospholipase A2. Arch BiochemBiophys 284: 352-359.
  72. Lomonte B, Pizarro-Cerdá J, Angulo Y, Gorvel JP, Moreno E (1999) Tyr-->Trp-substituted peptide 115-129 of a Lys49 phospholipase A(2) expresses enhanced membrane-damaging activities and reproduces its in vivo myotoxic effect. BiochimBiophysActa 1461: 19-26.
  73. Zuliani JP, Gutiérrez JM, Casais e Silva LL, CoccuzzoSampaio S, Lomonte B, et al. (2005) Activation of cellular functions in macrophages by venom secretory Asp-49 and Lys-49 phospholipases A(2). Toxicon 46: 523-532.
  74. Selistre de Araujo HS, White SP, Ownby CL (1996) cDNA cloning and sequence analysis of a lysine-49 phospholipase A2 myotoxin from Agkistrodoncontortrixlaticinctus snake venom. Arch BiochemBiophys 326: 21-30.
  75. Audebert F, Sorkine M, Bon C (1992) Envenoming by viper bites in France: clinical gradation and biological quantification by ELISA. Toxicon 30: 599-609.
  76. RivièreG, Choumet V, Saliou B, Debray M, Bon C (1998) Absorption and elimination of viper venom after antivenom administration. J PharmacolExpTher 285: 490-495.
  77. Lomonte B, Tarkowski A, Hanson LA (1993) Host response to Bothropsasper snake venom. Analysis of edema formation, inflammatory cells, and cytokine release in a mouse model. Inflammation 17: 93-105.
  78. Sebia-Amrane F, Laraba-Djebari F (2013) Pharmaco-modulations of induced edema and vascular permeability changes by Viperalebetina venom: inflammatory mechanisms. Inflammation 36: 434-443.
  79. Rucavado A, Lomonte B, Ovadia M, Gutiérrez JM (1995) Local tissue damage induced by BaP1, a metalloproteinase isolated from Bothropsasper (Terciopelo) snake venom. ExpMolPathol 63: 186-199.
  80. Clissa PB, do Nascimento N, Rogero JR (1999) Toxicity and immunogenicity of Crotalusdurissusterrificus venom treated with different doses of gamma rays. Toxicon 37: 1131-1141.
  81. Chaves F, León G, Alvarado VH, Gutiérrez JM (1998) Pharmacological modulation of edema induced by Lys-49 and Asp-49 myotoxic phospholipases A2 isolated from the venom of the snake Bothropsasper (terciopelo). Toxicon 36: 1861-1869.
  82. Wei JF, Wei XL, Chen QY, Huang T, Qiao LY, et al. (2006) N49 phospholipase A2, a unique subgroup of snake venom group II phospholipase A2. BiochimBiophysActa 1760: 462-471.
  83. León G, Sánchez L, Hernández A, Villalta M, Herrera M, et al. (2011) Immune response towards snake venoms. Inflamm Allergy Drug Targets 10: 381-398.
  84. Clemetson KJ (2010) Snaclecs (snake C-type lectins) that inhibit or activate platelets by binding to receptors. Toxicon 56: 1236-1246.
  85. Koh YS, Chung KH, Kim DS (2001) Biochemical characterization of a thrombin-like enzyme and a fibrinolytic serine protease from snake (Agkistrodonsaxatilis) venom. Toxicon 39: 555-560.
  86. Serrano SM, Mentele R, Sampaio CA, Fink E (1995) Purification, characterization, and amino acid sequence of a serine proteinase, PA-BJ, with platelet-aggregating activity from the venom of Bothropsjararaca. Biochemistry 34: 7186-7193.
  87. Warrell DA, Gutiérrez JM, Calvete JJ, Williams D (2013) New approaches & technologies of venomics to meet the challenge of human envenoming by snakebites in India. Indian J Med Res 138: 38-59.
  88. Jangprasert P, Rojnuckarin P2 (2014) Molecular cloning, expression and characterization of albolamin: a type P-IIa snake venom metalloproteinase from green pit viper (Cryptelytropsalbolabris). Toxicon 79: 19-27.
  89. Marcinkiewicz C (2013) Applications of snake venom components to modulate integrin activities in cell-matrix interactions. Int J Biochem Cell Biol 45: 1974-1986.
  90. Gopalakrishnakone P, Dempster DW, Hawgood BJ, Elder HY (1984) Cellular and mitochondrial changes induced in the structure of murine skeletal muscle by crotoxin, a neurotoxic phospholipase A2 complex. Toxicon 22: 85-98.
  91. Falconi M, Desideri A, Rufini S (2000) Membrane-perturbing activity of Viperidaemyotoxins: an electrostatic surface potential approach to a puzzling problem. J MolRecognit 13: 14-19.
  92. Montecucco C, Gutiérrez JM, Lomonte B (2008) Cellular pathology induced by snake venom phospholipase A2 myotoxins and neurotoxins: common aspects of their mechanisms of action. Cell Mol Life Sci 65: 2897-2912.
  93. Hamza L, Girardi T, Castelli S, Gargioli C, Cannata S, et al. (2010) Isolation and characterization of a myotoxin from the venom of Macroviperalebetinatransmediterranea. Toxicon 56: 381-390.
  94. Hamza L, Gargioli C, Castelli S, Rufini S, Laraba-Djebari F (2010) Purification and characterization of a fibrinogenolytic and hemorrhagic metalloproteinase isolated from Viperalebetina venom. Biochimie 92: 797-805.
  95. Vyas VK, Brahmbhatt K, Bhatt H, Parmar U, Patidar R (2013) Therapeutic potential of snake venom in cancer therapy: current perspectives. Asian Pac J Trop Biomed 3: 156-162.
Citation: Fatima L, Fatah C (2014) Pathophysiological and Pharmacological Effects of Snake Venom Components: Molecular Targets. J Clin Toxicol 4:190.

Copyright: © 2014 Fatima L, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top