Next Article in Journal
The Pressure of Fusarium Disease and Its Relation with Mycotoxins in The Wheat Grain and Malt
Previous Article in Journal
Detection of Five Mycotoxins in Different Food Matrices in the Malaysian Market by Using Validated Liquid Chromatography Electrospray Ionization Triple Quadrupole Mass Spectrometry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

D. russelii Venom Mediates Vasodilatation of Resistance Like Arteries via Activation of Kv and KCa Channels

by
Rahini Kakumanu
1,
Sanjaya Kuruppu
2,
Lachlan D. Rash
3,
Geoffrey K. Isbister
4,
Wayne C. Hodgson
1,* and
Barbara K. Kemp-Harper
1
1
Department of Pharmacology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton VIC 3800, Australia
2
Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton VIC 3800, Australia
3
Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, St Lucia QLD 4072, Australia
4
Clinical Toxicology Research Group, University of Newcastle, Callaghan NSW 2308, Australia
*
Author to whom correspondence should be addressed.
Toxins 2019, 11(4), 197; https://doi.org/10.3390/toxins11040197
Submission received: 8 February 2019 / Revised: 17 March 2019 / Accepted: 28 March 2019 / Published: 1 April 2019
(This article belongs to the Section Animal Venoms)

Abstract

:
Russell’s viper (Daboia russelii) venom causes a range of clinical effects in humans. Hypotension is an uncommon but severe complication of Russell’s viper envenoming. The mechanism(s) responsible for this effect are unclear. In this study, we examined the cardiovascular effects of Sri Lankan D. russelii venom in anaesthetised rats and in isolated mesenteric arteries. D. russelii venom (100 μg/kg, i.v.) caused a 45 ± 8% decrease in blood pressure within 10 min of administration in anaesthetised (100 μg/kg ketamine/xylazine 10:1 ratio, i.p.) rats. Venom (1 ng/mL–1 μg/mL) caused concentration-dependent relaxation (EC50 = 145.4 ± 63.6 ng/mL, Rmax = 92 ± 2%) in U46619 pre-contracted rat small mesenteric arteries mounted in a myograph. Vasorelaxant potency of venom was unchanged in the presence of the nitric oxide synthase inhibitor, L-NAME (100 µM), or removal of the endothelium. In the presence of high K+ (30 mM), the vasorelaxant response to venom was abolished. Similarly, blocking voltage-dependent (Kv: 4-aminopryidine; 1000 µM) and Ca2+-activated (KCa: tetraethylammonium (TEA; 1000 µM); SKCa: apamin (0.1 µM); IKCa: TRAM-34 (1 µM); BKCa; iberiotoxin (0.1 µM)) K+ channels markedly attenuated venom-induced relaxation. Responses were unchanged in the presence of the ATP-sensitive K+ channel blocker glibenclamide (10 µM), or H1 receptor antagonist, mepyramine (0.1 µM). Venom-induced vasorelaxtion was also markedly decreased in the presence of the transient receptor potential cation channel subfamily V member 4 (TRPV4) antagonist, RN-1734 (10 µM). In conclusion, D. russelii-venom-induced hypotension in rodents may be due to activation of Kv and KCa channels, leading to vasorelaxation predominantly via an endothelium-independent mechanism. Further investigation is required to identify the toxin(s) responsible for this effect.

1. Introduction

Snake bite is a globally important health issue [1,2]. Snake venom has three purposes: a defensive mechanism against predators, an aid to capture prey, and/or a tool to deter/challenge competitors [3]. Venom is a complex cocktail of toxins and enzymes that have a wide range of biological activities targeting major physiological pathways and organs [4]. Ninety to ninety-five percent of snake venom consists of proteins and peptides, many of which are toxic to humans [5]. These components often possess enzymatic activity and ligand binding abilities that, in combination and/or separately, result in the clinical envenoming symptoms in humans and other organisms [3,6]. While most components of snake venom, such as neurotoxins [7,8,9], myotoxins [10,11,12], pro-coagulants, and anticoagulant, haemolytic and local tissue necrotic factors [13,14,15] have been studied in detail, toxins targeting the cardiovascular system are less well understood.
We have previously demonstrated two distinct cardiovascular activities due to snake envenoming: “cardiovascular collapse” (defined as an irreversible rapid drop in blood pressure) [16,17] versus “prolonged hypotension” (defined as a gradual decline in blood pressure that is reversible) [18]. A number of hypotheses have been postulated in regards to the mechanism(s) of “cardiovascular collapse.” This includes the potential involvement of prothrombin activators or pro-coagulant toxins present in snake venoms [19,20]. However, we have previously demonstrated, using an in vivo animal model, that death adder (A. rugosus) venom causes collapse even though it contains no pro-coagulant toxins [16]. The release of depletable endogenous mediators has also been postulated to induce “cardiovascular collapse” [16].
Hypotension is defined as a blood pressure that is below the expected normal range for an individual in a given environment. Factors that can affect blood pressure include age, weight, medications, dehydration, or underlying medical conditions. Physiologically, hypotension can occur due to reduced systemic vascular resistance, reduced cardiac output, hypovolemia, vascular obstruction, or blood volume redistribution [21]. Known snake toxins that can affect blood pressure include natriuretic peptides, bradykinin-potentiating peptides, incretin mimetics, and sarafotoxins [22].
In this study, we examined in more detail the hypotensive effects of snake venom and pharmacologically characterized the vasodilatory effects of Sri Lankan Russell’s viper (D. russelii) venom. D. russelii is regarded as one of the most medically important venomous snakes, as it causes the highest rate of mortality and morbidity due to snake bite in Asia [23,24]. It is mainly found in South Asia and is responsible for 73% of snake envenoming cases in the Anuradhapura District, Sri Lanka [25,26]. Clinical manifestations of envenoming include neuromuscular paralysis, coagulopathy, acute kidney failure, and hypotension [27]. Since the venom consists mainly of neurotoxins and myotoxins (~80%), these toxins have been well characterized pharmacologically [11,24,28], but the cardiovascular effects of this venom are less clear.

2. Results and Discussion

D. russelii is one of the most medically important species of snakes found in South East Asia, and is responsible for the highest rate of mortality and morbidity in this region due to snake envenoming [28,29,30,31]. The clinical syndromes include coagulopathy, mild neurotoxicity, acute kidney injury, bleeding, and hypotension [11,29,32]. While the neurotoxicity [24,28], acute kidney injury, and coagulopathy [33,34,35] have been studied in great depth, little is known about the cardiovascular effects. In the current study, we investigated the prolonged hypotensive effect of D. russelii venom both in vivo and in vitro in rodents.

2.1. In Vivo Studies

D. russelii venom (100 µg/kg, i.v., n = 4) caused prolonged (30 min) hypotension (45 ± 8% decrease in mean arterial pressure) within 10 min of administration (Figure 1). In addition, the heart rate of rats decreased ~20% i.e. from 289 ± 85 b.p.m. (n = 4), just prior to venom administration, to 242 ± 71 b.p.m. (n = 4) at the time of maximum decrease in blood pressure.

2.2. Vasorelaxant Responses to D. russelii Venom

We next identified D. russelii venom as a potent dilator (EC50 = 145.4 ± 63.6 ng/mL, Rmax = 92 ± 2%; Figure 2) of isolated rat small mesenteric arteries. Interestingly, vasorelaxation was unchanged following endothelial denudation (EC50 = 137.6 ± 51.6 ng/mL, Rmax = 87 ± 7%) or inhibition of nitric oxide synthase by L-NAME (100 µM, EC50 = 130.7 ± 72.5 ng/mL, Rmax = 91 ± 5%), indicating that the venom is likely directly targeting the vascular smooth muscle to cause relaxation. Thus, we next sought to characterize the mechanism(s) via which the venom mediates endothelium-independent vasorelaxation.
In the presence of the histamine H1 receptor antagonist, mepyramine (0.1 µM), there was no change in vasorelaxation (EC50 = 159.4 ± 82.3, Rmax = 76 ± 10%) compared to venom alone, indicating that histamine does not appear to play a role in D. russelii-venom-induced vasorelaxation. In contrast, raising the extracellular concentration of K+ to 30 mM abolished the vasorelaxation effects of venom (Rmax = 5 ± 3%, p < 0.05, Table 1), suggesting that the venom also modulates relaxation of small resistance-like arteries in part via activation of K+ channels.

2.3. Contribution of Potassium Channels to D. russelii-Mediated Vasorelaxation

It is well known that potassium channels play an integral role in maintaining the membrane potential and therefore, contractile tone in smooth muscle cells [36]. The distribution and nature of potassium channels vary depending upon the size of the vessel, as well as the type of vascular bed [37,38]. There are at least four different subtypes of potassium channels present in mesenteric arterial smooth muscle cells. These include inward rectifier (KIR), voltage-gated (KV), ATP sensitive (KATP), and Ca2+-activated (KCa) potassium channels [36,39]. When K+ channels are activated, it leads to vascular smooth muscle cell hyperpolarization and relaxation, thereby leading to a decrease in blood pressure and increased blood flow [36].
Whilst there are many different venoms that are known to contain potassium channel inhibiting peptides, the current study has identified an apparent ability of D. russelii venom to activate potassium channels.
Vasorelaxation to D. russelii venom in rat small mesenteric arteries was unchanged in the presence of the ATP-sensitive K+ channel inhibitor, glibenclamide (10 µM; EC50 = 237.7 ± 62.1, Rmax = 84 ± 4%, Figure 3a). However, the voltage-gated K+ channel inhibitor 4-aminopyridine, (1000 µM) abolished venom-induced relaxation, reducing the response at 1000 ng/mL to 7 ± 6% (p < 0.01; Table 1). The non-selective KCa channel blocker TEA, (1000 µM, Rmax = 50 ± 14%), markedly attenuated venom-induced vasorelaxation. Similarly, blocking large (BKCa; iberiotoxin, 0.1 µM, Rmax = 29 ± 6%), intermediate (IKCa; TRAM-34 1 µM, Rmax = 29 ± 15%), and small (SKCa; apamin, 0.1 µM, Rmax = 11 ± 5%, Figure 3b, Table 1) Ca2+-activated K+ channels significantly inhibited venom-induced vasorelaxation.
These findings suggest the involvement of both KCa and Kv channels in D. russelii-venom-induced vasorelaxation. Given both KCa and Kv channels are sensitive to Ca2+, our observations raise the interesting possibility that the venom may modulate intracellular Ca2+ levels, thereby indirectly modulating K+ channel function. Indeed, our observation that the vasorelaxant effect of the venom was attenuated in the presence of the TRPV4 antagonist, RN-1734 (10 µM), further supports this concept. Specifically, in the presence of RN-1734, the potency of the venom was decreased approximately five-fold, and the response to 1000 ng/mL significantly reduced from 86 ± 4 to 50 ± 7% (Figure 3c). The TRPV4 agonist, GSK1016790A, also caused concentration-dependent relaxation (EC50 = 2.8 ± 0.7 µM, Rmax = 84 ± 5%) in rat small mesenteric arteries, a response which was unchanged following endothelial denudation (EC50 = 1.3 ± 0.6 µM, Rmax = 77 ± 10%). In the presence of RN-1734 (10 µM), there was an apparent two-fold decrease in the potency of GSK1016790A, yet this change failed to reach statistical significance (EC50 = 3.5 ± 0.9 µM, Figure 3d, Table 1). The greater inhibitory effect of RN-1734 against D. russelii-venom-induced vasorelaxation, as compared to GSK1016790A, may reflect a lower efficacy of the venom as a TRPV4 activator or an ability of RN-1734 to target TRPV4-independent signaling pathways potentially activated by the venom, identification of which are beyond the scope of the current study.
Whilst TRPV4 receptors are abundantly expressed on the endothelium [40], they are also present on the vascular smooth muscle [40,41]. Here, activation of TRPV4 leads to Ca2+ influx, the generation of Ca2+ sparks from the sarcoplasmic reticulum, and the subsequent activation of BKCa and vasorelaxation [42,43]. As such, D. russelii venom may cause vasorelaxation of resistance arteries in part via activation of vascular smooth muscle TRPV4 and opening of KCa. Future patch-clamp studies are required to support this hypothesis, and the potential for the venom to directly activate KCa and Kv channels remains.

3. Conclusions

D. russelii venom is known to cause an array of clinical manifestations in envenomed patients. These include coagulopathy, mild neurotoxicity, acute kidney injury, and sometimes severe hypotension. In this study, we have demonstrated that the hypotensive response may be indicative of a vasodilatory action of the venom at the level of the resistance vasculature. We have shown that D. russelii venom is a potent dilator of resistance-like arteries, mediating its response via activation of Kv and KCa channels, which may be modulated, in part, via signaling downstream of TRPV4. Future studies, including electrophysiological experiments and separation of venom components using HPLC, as well as investigating the potential contribution of other potassium channels such as KIR channels and Na+/K+-ATPase, will aid in identifying the toxin(s) responsible for relaxation and provide further insight into the underlying mechanisms.

4. Materials and Methods

4.1. In Vivo Blood Pressure Experiments

Animal experiments were approved by the Monash University Ethics Committee (MARP/2014/097). Sprague-Dawley male rats (weight 280–350g) were anaesthetized with a mixture of ketamine (100 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.). A midline incision was made and cannulae inserted into the trachea for mechanical ventilation, if required. Cannulae were also inserted into the left jugular vein for administration of venom and the right carotid artery to record arterial blood pressure. The arterial cannula was connected to a pressure transducer (PowerLab/400 system, ADInstruments Inc, Sydney, NSW, Australia). Blood pressure was then allowed to stabilize for approximately 10–15 min. Body temperature was maintained at 37 °C using an overhead lamp and heated rat table. Venom (100 µg/kg) was administrated via the jugular vein followed by flushing with saline. Responses to the venom were measured as percentage change in mean arterial pressure (MAP).

4.2. Isolation of Rat Small Mesenteric Arteries

Male Sprague-Dawley rats (200–250g) were euthanized via CO2 inhalation (95% CO2, 5% O2), followed by exsanguination. Small mesenteric arteries (second-order branch of the superior mesenteric artery) were isolated, cut into 2 mm lengths, and mounted on 40 µm wires in small vessel myographs [44]. Vessels were maintained in physiological salt solution (composed of (in mM) 119 NaCl, 4.7 KCl, 1.17 MgSO4, 25 NaHCO3, 1.8 KH2PO4, 2.5 CaCl2, 11 glucose and 0.026 EDTA) at 37 °C, and were bubbled with carbogen (95% O2, 5% CO2). In a subset of arteries, the endothelium was gently denuded via insertion of a 40 µm wire inside the lumen and rubbing the vessel walls. The mesenteric arteries were allowed to equilibrate for 30 min under zero force and then a 5 mN resting tension was applied. Changes in isometric tension were recorded using Myograph Interface Model 610 M v2.2 (DMT, Aarhus, Denmark) and PowerLab/835 (ADInstruments Inc). Data were recorded with the data acquisition program Chart (v5, ADInstruments). Following a 30 min equilibration period at 5 mN, the mesenteric arteries were contracted maximally (Fmax) using a K+ depolarizing solution (K+-containing physiological salt solution (KPSS); composed of (in mM) 123 KCl, 1.17 MgSO4, 1.18 KH2PO4, 2.5 CaCl2, 25 NaHCO3 and 11 glucose). The integrity of the endothelium was confirmed by relaxation to acetylcholine (ACh, 10 µM) in tissues pre-contracted with the thromboxane A2 mimetic, U46619 (1 µM). Vessels with a relaxation response to ACh < 20% were considered endothelium-denuded, while vessels with a relaxation response to ACh > 80% were considered endothelium-intact. Arteries were washed with physiological salt solution and the tension was allowed to return to baseline.

4.3. Vasorelaxation Experiments

Cumulative concentration–response curves to venom (1 ng/mL–1 µg/mL) were constructed in vessels pre-contracted submaximally to ~50% Fmax with titrated concentrations of U46619 (0.01–0.2 µM). Responses to venom were obtained in endothelium-intact mesenteric arteries pre-incubated for 30 min with either 30 mM K+ [45], mepyramine (0.1 µM), L-NAME (100 µM) [44], TEA (1000 µM) [46], iberiotoxin (0.1 µM), apamin (0.1 µM), TRAM-34 (1 µM), 4-aminopyridine (1000 µM) [44], glibenclamide (10 µM) [47,48] or RN-1734 (10 µM). Cumulative concentration-response curves were also constructed to venom alone (1 ng/mL–1 µg/mL) or GSK1016790A in the presence and absence of RN-1734 (10 µM). In a subset of endothelium-denuded arteries, the vasorelaxation of venom and GSK1016790A were also examined. Sodium nitroprusside (SNP; 10 µM) [49] was added at the end of each concentration response curve to ensure maximum relaxation. Only one concentration–response curve to venom was obtained in each vessel segment due to tachyphylaxis [49,50].

4.4. Data Analysis and Statistical Procedures

4.4.1. In Vivo

For the anaesthetized rat experiments, pulse pressure was defined as the difference between systolic and diastolic blood pressures. Mean arterial pressure (MAP) was calculated as diastolic blood pressure plus one-third of pulse pressure. Heart rate (HR) was determined from the blood pressure trace.

4.4.2. In Vitro

Blood vessel relaxation was expressed as a percentage reversal of the U46619 pre-contraction (i.e., cumulative relaxation responses to venom or GSK1016790A were measured as a change in tension from the stable U46619 contraction, and expressed as a percentage of this contractile response). Individual relaxation curves were fitted to a sigmoidal logistic equation and EC50 values (concentration of agonist resulting in a 50% relaxation) were calculated. Statistical comparisons between the experimental groups’ mean EC50 and maximum relaxation (Rmax) values were made using either a student’s unpaired t-test or a one-way ANOVA with Bonferroni’s post hoc comparison. Where EC50 values could not be obtained, concentration–response curves were compared by means of a two-way ANOVA (repeated measures). n = Number of artery segments from separate animals. Data represent the mean ± SEM (error bars on graph). Statistical significance was defined as p < 0.05. All data analysis was performed using GraphPad Prism v5.02 (GraphPad Software, San Diego, CA, USA) [44].

4.5. Reagents

Reagents and their sources were: U46619 (Cayman Chemical company, Ann Arbor, MI, USA), glibenclamide, TEA, 4-aminopyridine, TRAM-34, mepyramine, L-NAME, SNP, isoprenaline, ACh, RN-1734 and GSK1016790A (Sigma-Aldrich, St Louis, MO, USA), and iberiotoxin (In vitro Technologies, Melbourne, VIC, Australia). Stock solutions of U46619 (1 mM) were made up in absolute ethanol. All subsequent dilutions of stock solutions were in distilled water. All other drugs were dissolved in distilled water, and all dilutions were prepared fresh daily.

Author Contributions

Conceptualization, W.C.H., B.K.K.-H., L.D.R. and R.K.; methodology, B.K.K.-H. and R.K.; validation, B.K.K.-H. and R.K.; formal analysis, B.K.K.-H. and R.K.; investigation, R.K.; resources, G.K.I., W.C.H. and B.K.K.-H.; data curation, B.K.K.-H. and R.K.; writing—original draft preparation, R.K.; writing—review and editing, W.C.H., B.K.K.-H., G.K.I., L.D.R., S.K. and R.K.; supervision, W.C.H., B.K.K.-H., G.K.I. and S.K.; funding acquisition, G.K.I. and W.C.H.

Funding

This study was funded by an Australian National Health and Medical Research Council (NHMRC) Senior Research Fellowship (ID: 1061041) awarded to G.K.I. and a NHMRC Centres for Research Excellence Grant ID: 1110343 awarded to G.K.I. and W.C.H.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chippaux, J.P. Estimating the global burden of snakebite can help to improve management. PLoS Med. 2008, 5, 221. [Google Scholar] [CrossRef]
  2. Warrell, D.A. Snake bite. Lancet 2010, 375, 77–88. [Google Scholar] [CrossRef]
  3. Casewell, N.R.; Wüster, W.; Vonk, F.J.; Harrison, R.A.; Fry, B.G. Complex cocktails: The evolutionary novelty of venoms. Trends Ecol. Evol. 2013, 28, 219–229. [Google Scholar] [CrossRef]
  4. Fry, B.G.; Roelants, K.; Champagne, D.E.; Scheib, H.; Tyndall, J.D.; King, G.F.; Nevalainen, T.J.; Norman, J.A.; Lewis, R.J.; Norton, R.S. The toxicogenomic multiverse: Convergent recruitment of proteins into animal venoms. Ann. Rev. Genomics Hum. Genet. 2009, 10, 483–511. [Google Scholar] [CrossRef]
  5. Mackessy, S.P. Handbook of Venoms and Toxins of Reptiles; CRC Press: Boca Raton, FL, USA, 2009. [Google Scholar]
  6. Calvete, J.J.; Sanz, L.; Angulo, Y.; Lomonte, B.; Gutiérrez, J.M. Venoms, venomics, antivenomics. FEBS Lett. 2009, 583, 1736–1743. [Google Scholar] [CrossRef] [Green Version]
  7. Chaisakul, J.; Konstantakopoulos, N.; Smith, A.I.; Hodgson, W.C. Isolation and characterisation of P-EPTX-Ap1a and P-EPTX-Ar1a: Pre-synaptic neurotoxins from the venom of the northern (Acanthophis praelongus) and Irian Jayan (Acanthophis rugosus) death adders. Biochem. Pharmacol. 2010, 80, 895–902. [Google Scholar] [CrossRef]
  8. Kuruppu, S.; Smith, A.I.; Isbister, G.K.; Hodgson, W.C. Neurotoxins from Australo-Papuan elapids: A biochemical and pharmacological perspective. Crit. Rev. Toxicol. 2008, 38, 73–86. [Google Scholar] [CrossRef]
  9. Barber, C.; Isbister, G.; Hodgson, W. Alpha neurotoxins. Toxicon 2013, 66, 47–58. [Google Scholar] [CrossRef]
  10. Wickramaratna, J.C.; Fry, B.G.; Aguilar, M.I.; Kini, R.M.; Hodgson, W.C. Isolation and pharmacological characterization of a phospholipase A2 myotoxin from the venom of the Irian Jayan death adder (Acanthophis rugosus). Br. J. Pharmacol. 2003, 138, 333–342. [Google Scholar] [CrossRef]
  11. Silva, A.; Johnston, C.; Kuruppu, S.; Kneisz, D.; Maduwage, K.; Kleifeld, O.; Smith, A.I.; Siribaddana, S.; Buckley, N.A.; Hodgson, W.C. Clinical and pharmacological investigation of myotoxicity in Sri Lankan Russell’s viper (Daboia russelii) envenoming. PLoS Negl. Trop. Dis. 2016, 10, e0005172. [Google Scholar] [CrossRef]
  12. Gutiérrez, J.; Lomonte, B. Phospholipase A2 myotoxins from Bothrops snake venoms. Toxicon 1995, 33, 1405–1424. [Google Scholar] [CrossRef]
  13. Gutiérrez, J.M.; Lomonte, B. Phospholipases A2: Unveiling the secrets of a functionally versatile group of snake venom toxins. Toxicon 2013, 62, 27–39. [Google Scholar] [CrossRef]
  14. Isbister, G.K.; Brown, S.G.; MacDonald, E.; White, J.; Currie, B.J. Current use of Australian snake antivenoms and frequency of immediate-type hypersensitivity reactions and anaphylaxis. Med. J. Aust. 2008, 188, 473. [Google Scholar] [PubMed]
  15. Joseph, R.; Pahari, S.; Hodgson, W.C.; Kini, R.M. Hypotensive agents from snake venoms. Cardiovasc. Hematol. Disord. Drug Targets 2004, 4, 437–459. [Google Scholar] [CrossRef]
  16. Chaisakul, J.; Isbister, G.K.; Kuruppu, S.; Konstantakopoulos, N.; Hodgson, W.C. An examination of cardiovascular collapse induced by Eastern brown snake (Pseudonaja textilis) venom. Toxicol. Lett. 2013, 221, 205–211. [Google Scholar] [CrossRef] [PubMed]
  17. Chaisakul, J.; Isbister, G.K.; Konstantakopoulos, N.; Tare, M.; Parkington, H.C.; Hodgson, W.C. In vivo and in vitro cardiovascular effects of Papuan taipan (Oxyuranus scutellatus) venom: Exploring “sudden collapse”. Toxicol. Lett. 2012, 213, 243–248. [Google Scholar] [CrossRef]
  18. Chaisakul, J.; Isbister, G.K.; Tare, M.; Parkington, H.C.; Hodgson, W.C. Hypotensive and vascular relaxant effects of phospholipase A2 toxins from Papuan taipan (Oxyuranus scutellatus) venom. Eur. J. Pharmacol. 2014, 723, 227–233. [Google Scholar] [CrossRef]
  19. Tibballs, J. The cardiovascular, coagulation and haematological effects of Tiger snake (Notechis scutatus) venom. Anaesth. Intensive Care 1998, 26, 529. [Google Scholar] [CrossRef]
  20. Tibballs, J.; Sutherland, S.; Rivera, R.; Masci, P. The cardiovascular and haematological effects of purified prothrombin activator from the common Brown snake (Pseudonaja textilis) and their antagonism with heparin. Anaesth. Intensive Care 1992, 20, 28–32. [Google Scholar] [CrossRef] [PubMed]
  21. Klabunde, R. Cardiovascular Physiology Concepts; Lippincott Williams & Wilkins: Baltimore, MD, USA, 2011. [Google Scholar]
  22. Hodgson, W.C.; Isbister, G.K. The application of toxins and venoms to cardiovascular drug discovery. Curr. Opin. Pharmacol. 2009, 9, 173–176. [Google Scholar] [CrossRef]
  23. De Silva, A.; Ranasinghe, L. Epidemiology of snake-bite in Sri Lanka: A review. Ceylon Med. J. 1983, 28, 144. [Google Scholar]
  24. Silva, A.; Kuruppu, S.; Othman, I.; Goode, R.J.; Hodgson, W.C.; Isbister, G.K. Neurotoxicity in Sri Lankan Russell’s viper (Daboia russelii) envenoming is primarily due to U1-viperitoxin-Dr1a, a pre-synaptic neurotoxin. Neurotox. Res. 2017, 31, 11–19. [Google Scholar] [CrossRef]
  25. Alirol, E.; Sharma, S.K.; Bawaskar, H.S.; Kuch, U.; Chappuis, F. Snake bite in South Asia: A review. PLoS Negl. Trop. Dis. 2010, 4, e603. [Google Scholar] [CrossRef]
  26. Phillips, R.E.; Theakston, R.D.G.; Warrekk, D.A.; Galigedara, Y.; Abeysekera, D.; Dissanayaka, P.; Hutton, R.A.; Aloysius, D.J. Paralysis, rhabdomyolysis and haemolysis caused by bites of Russell’s viper (Vipera russelli pulchella) in Sri Lanka: Failure of Indian (haffkine) antivenom. QJM 1988, 68, 691–715. [Google Scholar]
  27. Warrell, D.A. Snake venoms in science and clinical medicine 1. Russell’s viper: Biology, venom and treatment of bites. Trans. R. Soc. Trop. Med. Hyg. 1989, 83, 732–740. [Google Scholar] [CrossRef]
  28. Silva, A.; Maduwage, K.; Sedgwick, M.; Pilapitiya, S.; Weerawansa, P.; Dahanayaka, N.J.; Buckley, N.A.; Siribaddana, S.; Isbister, G.K. Neurotoxicity in Russell’s viper (Daboia russelii) envenoming in Sri Lanka: A clinical and neurophysiological study. Clin. Toxicol. 2016, 54, 411–419. [Google Scholar] [CrossRef]
  29. Kularatne, S.A.; Silva, A.; Weerakoon, K.; Maduwage, K.; Walathara, C.; Paranagama, R.; Mendis, S. Revisiting Russell’s viper (Daboia russelii) bite in Sri Lanka: Is abdominal pain an early feature of systemic envenoming? PLoS ONE 2014, 9, e90198. [Google Scholar] [CrossRef]
  30. Kasturiratne, A.; Pathmeswaran, A.; Fonseka, M.; Lalloo, D.; Brooker, S.; De Silva, H. Estimates of disease burden due to land-snake bite in Sri Lankan hospitals. Southeast Asian J. Trop. Med. Public Health 2005, 36, 733. [Google Scholar]
  31. Kasturiratne, A.; Wickremasinghe, A.R.; de Silva, N.; Gunawardena, N.K.; Pathmeswaran, A.; Premaratna, R.; Savioli, L.; Lalloo, D.G.; de Silva, H.J. The global burden of snakebite: A literature analysis and modelling based on regional estimates of envenoming and deaths. PLoS Med. 2008, 5, e218. [Google Scholar] [CrossRef]
  32. Phillips, R.; Warrell, D. Bites by Russell’s viper (Vipera russelli siamensis) in Burma: Haemostatic, vascular, and renal disturbances and response to treatment. Lancet 1985, 326, 1259–1264. [Google Scholar]
  33. O’Leary, M.A.; Isbister, G.K. A turbidimetric assay for the measurement of clotting times of procoagulant venoms in plasma. J. Pharmacol. Toxicol. Methods 2010, 61, 27–31. [Google Scholar] [CrossRef]
  34. Maduwage, K.; Isbister, G.K. Current treatment for venom-induced consumption coagulopathy resulting from snakebite. PLoS Negl. Trop. Dis. 2014, 8, e3220. [Google Scholar] [CrossRef]
  35. Mukherjee, A.K. Characterization of a novel pro-coagulant metalloprotease (RVBCMP) possessing α-fibrinogenase and tissue haemorrhagic activity from venom of Daboia russelli russelli (Russell’s viper): Evidence of distinct coagulant and haemorrhagic sites in RVBCMP. Toxicon 2008, 51, 923–933. [Google Scholar] [CrossRef]
  36. Standen, N.; Quayle, J. K+ channel modulation in arterial smooth muscle. Acta Physiol. Scand. 1998, 164, 549–557. [Google Scholar] [CrossRef]
  37. Jackson, W.F. Potassium channels in the peripheral microcirculation. Microcirculation 2005, 12, 113–127. [Google Scholar] [CrossRef]
  38. Quayle, J.M.; Bonev, A.D.; Brayden, J.E.; Nelson, M.T. Calcitonin gene-related peptide activated ATP-sensitive K+ currents in rabbit arterial smooth muscle via protein kinase A. J. Physiol. 1994, 475, 9–13. [Google Scholar] [CrossRef]
  39. Michelakis, E.D.; Reeve, H.L.; Huang, J.M.; Tolarova, S.; Nelson, D.P.; Weir, E.K.; Archer, S.L. Potassium channel diversity in vascular smooth muscle cells. Can. J. Physiol. Pharmacol. 1997, 75, 889–897. [Google Scholar] [CrossRef]
  40. Baylie, R.; Brayden, J. TRPV channels and vascular function. Acta Physiol. 2011, 203, 99–116. [Google Scholar] [CrossRef]
  41. Inoue, R.; Jensen, L.J.; Shi, J.; Morita, H.; Nishida, M.; Honda, A.; Ito, Y. Transient receptor potential channels in cardiovascular function and disease. Circ. Res. 2006, 99, 119–131. [Google Scholar] [CrossRef]
  42. Earley, S.; Heppner, T.J.; Nelson, M.T.; Brayden, J.E. TRPV4 forms a novel Ca2+ signaling complex with ryanodine receptors and BKCa channels. Circ. Res. 2005, 97, 1270–1279. [Google Scholar] [CrossRef]
  43. Earley, S.; Pauyo, T.; Drapp, R.; Tavares, M.J.; Liedtke, W.; Brayden, J.E. TRPV4-dependent dilation of peripheral resistance arteries influences arterial pressure. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, 1096–1102. [Google Scholar] [CrossRef]
  44. Andrews, K.L.; Irvine, J.C.; Tare, M.; Apostolopoulos, J.; Favaloro, J.L.; Triggle, C.R.; Kemp-Harper, B.K. A role for nitroxyl (HNO) as an endothelium-derived relaxing and hyperpolarizing factor in resistance arteries. Br. J. Pharmacol. 2009, 157, 540–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Boussery, K.; Delaey, C.; Van de Voorde, J. The vasorelaxing effect of CGRP and natriuretic peptides in isolated bovine retinal arteries. Investig. Ophthalmol. Vis. Sci. 2005, 46, 1420–1427. [Google Scholar] [CrossRef] [PubMed]
  46. Kakumanu, R.; Hodgson, W.; Ravi, R.; Alagon, A.; Harris, R.; Brust, A.; Alewood, P.; Kemp-Harper, B.; Fry, B. Vampire venom: Vasodilatory mechanisms of vampire bat (Desmodus rotundus) blood feeding. Toxins 2019, 11, 26. [Google Scholar] [CrossRef]
  47. Lei, S.; Mulvany, M.J.; Nyborg, N.C.B. Characterization of the CGRP receptor and mechanisms of action in rat mesenteric small arteries. Pharmacol. Toxicol. 1994, 74, 130–135. [Google Scholar] [CrossRef]
  48. Isomoto, S.; Kondo, C.; Yamada, M.; Matsumoto, S.; Higashiguchi, O.; Horio, Y.; Matsuzawa, Y.; Kurachi, Y. A novel sulfonylurea receptor forms with BIR (Kir6.2) a smooth muscle type ATP-sensitive K+ channel. J. Biol. Chem. 1996, 271, 24321–24324. [Google Scholar] [CrossRef] [PubMed]
  49. Yuill, K.H.; Yarova, P.; Kemp-Harper, B.K.; Garland, C.J.; Dora, K.A. A novel role for HNO in local and spreading vasodilatation in rat mesenteric resistance arteries. Antioxid. Redox Signal. 2011, 14, 1625–1635. [Google Scholar] [CrossRef]
  50. Favaloro, J.L.; Kemp-Harper, B.K. Redox variants of NO (NO· and HNO) elicit vasorelaxation of resistance arteries via distinct mechanisms. Am. J. Physiol. Heart Circ. Physiol. 2009, 296, 1274–1280. [Google Scholar] [CrossRef]
Figure 1. Original recording showing the hypotensive response to D. russelii venom (100 g/kg, i.v.) in an anaesthetized rat. Venom (RVV; Russell’s viper venom) was added as indicated by the line on the trace.
Figure 1. Original recording showing the hypotensive response to D. russelii venom (100 g/kg, i.v.) in an anaesthetized rat. Venom (RVV; Russell’s viper venom) was added as indicated by the line on the trace.
Toxins 11 00197 g001
Figure 2. Cumulative concentration-response curves to D. russelii venom in rat small mesenteric arteries in the absence or presence of L-NAME (100 µM), 30 mM K+, mepyramine (0.1 µM), or following endothelial denudation (−E). Values are expressed as % reversal of U46619 pre-contraction and given as mean ± SEM, where n = number of animals. # p < 0.05, response at 1000 ng/mL versus control (+E) (one-way ANOVA, Bonferonni’s post-hoc test).
Figure 2. Cumulative concentration-response curves to D. russelii venom in rat small mesenteric arteries in the absence or presence of L-NAME (100 µM), 30 mM K+, mepyramine (0.1 µM), or following endothelial denudation (−E). Values are expressed as % reversal of U46619 pre-contraction and given as mean ± SEM, where n = number of animals. # p < 0.05, response at 1000 ng/mL versus control (+E) (one-way ANOVA, Bonferonni’s post-hoc test).
Toxins 11 00197 g002
Figure 3. Cumulative concentration–response curves to D. russelii venom in rat small mesenteric arteries in the absence or presence of (a) 4-aminopyridine (1000 µM) or glibenclamide (10 µM), (b) TEA (1000 µM), apamin (0.1 µM), iberiotoxin (0.1 µM) or TRAM-34 (1 µM), or (c) RN-1734 (10 µM). (d) Cumulative concentration–response curves to GSK1016790A in the absence (control +E) or presence of RN-1734 (10 µM) or in endothelium-denuded vessels (control −E). Values are expressed as % reversal of pre-contraction and given as mean ± SEM, where n = number of animals. # p < 0.05, response at 1000 ng/mL versus control (one-way ANOVA, Bonferonni’s post-hoc test), + p < 0.05, response at 1000 ng/mL versus control (student’s unpaired t-test), * p < 0.05 vs control concentration-response curve (two-way repeated measures ANOVA).
Figure 3. Cumulative concentration–response curves to D. russelii venom in rat small mesenteric arteries in the absence or presence of (a) 4-aminopyridine (1000 µM) or glibenclamide (10 µM), (b) TEA (1000 µM), apamin (0.1 µM), iberiotoxin (0.1 µM) or TRAM-34 (1 µM), or (c) RN-1734 (10 µM). (d) Cumulative concentration–response curves to GSK1016790A in the absence (control +E) or presence of RN-1734 (10 µM) or in endothelium-denuded vessels (control −E). Values are expressed as % reversal of pre-contraction and given as mean ± SEM, where n = number of animals. # p < 0.05, response at 1000 ng/mL versus control (one-way ANOVA, Bonferonni’s post-hoc test), + p < 0.05, response at 1000 ng/mL versus control (student’s unpaired t-test), * p < 0.05 vs control concentration-response curve (two-way repeated measures ANOVA).
Toxins 11 00197 g003
Table 1. Effect of treatments on D. russelii- or GSK1016790A-induced vasorelaxation in rat small mesenteric arteries.
Table 1. Effect of treatments on D. russelii- or GSK1016790A-induced vasorelaxation in rat small mesenteric arteries.
TreatmentD. russelii Venom
EC50 (ng/mL)Rmax (%)n
Control (+E)145.4 ± 63.692 ± 28
Control (−E)137.6 ± 51.687 ± 75
L-NAME130.7 ± 72.591 ± 55
Mepyramine159.4 ± 82.376 ± 104
K+ND5 ± 3 #5
Control276.5 ± 69.388 ± 215
TEAND50 ± 14 #7
ApaminND11 ± 5 #8
IberiotoxinND29 ± 6 #8
TRAM-34ND29 ± 156
Control328.7 ± 110.589 ± 29
Glibenclamide237.7 ± 62.184 ± 44
4-AminopyridineND7 ± 6 #5
Control273.6 ± 57.686 ± 46
RN-1734ND50 ± 7 *6
GSK1016790A
Control (+E)2.8 ± 0.7 µM84 ± 56
Control (−E)1.3 ± 0.6 µM77 ± 104
RN-17343.5 ± 0.9 µM79 ± 35
Values as % reversal of the level of pre-contraction; +E = endothelium intact; −E = endothelium denuded; ND = Not determined; # p < 0.05, 1-way ANOVA as compared to control * p < 0.05, student’s unpaired t-test.

Share and Cite

MDPI and ACS Style

Kakumanu, R.; Kuruppu, S.; Rash, L.D.; Isbister, G.K.; Hodgson, W.C.; Kemp-Harper, B.K. D. russelii Venom Mediates Vasodilatation of Resistance Like Arteries via Activation of Kv and KCa Channels. Toxins 2019, 11, 197. https://doi.org/10.3390/toxins11040197

AMA Style

Kakumanu R, Kuruppu S, Rash LD, Isbister GK, Hodgson WC, Kemp-Harper BK. D. russelii Venom Mediates Vasodilatation of Resistance Like Arteries via Activation of Kv and KCa Channels. Toxins. 2019; 11(4):197. https://doi.org/10.3390/toxins11040197

Chicago/Turabian Style

Kakumanu, Rahini, Sanjaya Kuruppu, Lachlan D. Rash, Geoffrey K. Isbister, Wayne C. Hodgson, and Barbara K. Kemp-Harper. 2019. "D. russelii Venom Mediates Vasodilatation of Resistance Like Arteries via Activation of Kv and KCa Channels" Toxins 11, no. 4: 197. https://doi.org/10.3390/toxins11040197

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop