VERATRIDINE

VERATRIDINE

* Please be kindly noted products are not for therapeutic use. We do not sell to patients.

VERATRIDINE
Category Mycotoxins
Catalog number BBF-04109
CAS 71-62-5
Molecular Weight 673.79
Molecular Formula C36H51NO11
Purity ≥90% by HPLC

Online Inquiry

Description

Veratridine, a pentacyclic triterpene acid compound, has been found to be a steroid-derived alkaloid neurotoxin and could be used as a voltage-gated Na+ channel opener.

Specification

Synonyms Veratrin (Amorphous); (3β,4α,16β)-4,12,14,16,17,20-Hexahydroxy-4,9-epoxycevan-3-yl 3,4-dimethoxybenzoate
Storage Store at -20°C (dark)
IUPAC Name [(1R,2S,6S,9S,10R,11S,12S,14R,15S,18S,19S,22S,23S,25R)-1,10,11,12,14,23-hexahydroxy-6,10,19-trimethyl-24-oxa-4-azaheptacyclo[12.12.0.02,11.04,9.015,25.018,23.019,25]hexacosan-22-yl] 3,4-dimethoxybenzoate
Canonical SMILES CC1CCC2C(C3(C(CC4(C5CCC6C7(C5(CC4(C3CN2C1)O)OC6(C(CC7)OC(=O)C8=CC(=C(C=C8)OC)OC)O)C)O)O)O)(C)O
InChI InChI=1S/C36H51NO11/c1-19-6-11-26-31(3,40)35(43)25(17-37(26)16-19)33(42)18-34-24(32(33,41)15-27(35)38)10-9-23-30(34,2)13-12-28(36(23,44)48-34)47-29(39)20-7-8-21(45-4)22(14-20)46-5/h7-8,14,19,23-28,38,40-44H,6,9-13,15-18H2,1-5H3/t19-,23-,24-,25-,26-,27-,28-,30-,31+,32+,33+,34+,35-,36-/m0/s1
InChI Key FVECELJHCSPHKY-YFUMOZOISA-N
Source Veratridine is a steroid-derived alkaloid from plants in the Liliaceae family.

Properties

Appearance White Powder
Boiling Point 814.5°C at 760 mmHg
Melting Point 180°C
Density 1.45 g/cm3
Solubility Soluble in ethanol (50 mg/mL)

Toxicity

Carcinogenicity No indication of carcinogenicity to humans (not listed by IARC).
Mechanism Of Toxicity Veratridine is a steroid-derived alkaloid from plants in the Liliaceae family that functions as a neurotoxin by abolishing inactivation of sodium ion channels. It binds to intramembrane receptor site 2 and increases intracellular Ca2+ concentration. It acts by preferentially binding to activated Na+ channels causing persistent activation that leads to increased nerve excitability. The activation of the action potential Na+ ionophore by veratridine is time- and concentrationdependent and completely reversible.
Toxicity LD50 in mice (mg/kg): 1.35 i.p. (Swiss, Bauer); 0.42 i.v. (Krayer); 6.3 s.c. (Tanaka)

Reference Reading

1. Effects of veratridine on sodium currents and fluxes
W Ulbricht Rev Physiol Biochem Pharmacol . 1998;133:1-54. doi: 10.1007/BFb0000612.
Veratridine causes Na+ channels to stay open during a sustained membrane depolarization by abolishing inactivation. The consequential Na+ influx, either by itself or by causing a maintained depolarization, leads to many secondary effects such as increasing pump activity, Ca2+ influx, and in turn exocytosis. If the membrane is voltage clamped in the presence of the alkaloid, a lasting depolarizing impulse induces, following the "normal" transient current, another much more slowly developing Na+ current that reaches a constant level after a few seconds. Repolarization then is followed by an inward tail current that slowly subsides. Development of these slow currents is enhanced by additional treatment with agents that inhibit inactivation. Most of these phenomena can be satisfactorily explained by assuming that Na+ channels must open before veratridine binds to them, and that the slow current changes reflect the kinetics of binding and unbinding. It is unclear, however, where the alkaloid stays when it is not bound. Although the effect sets in promptly, once this pool is filled, access to it from outside must be impeded since in most preparations veratridine can only partially be washed out. Cooling acts as if the available concentration is reduced, but this reversible "reduction" takes much longer to develop than the cold-induced changes in kinetics. Several authors assume that the binding site, site 2, is accessed from the lipid phase of the membrane. Considerations of this kind are often based on experiments with batrachotoxin, the widely used site-2 ligand which has a much higher affinity and acts as a full agonist in contrast to the partial agonist veratridine. Batrachotoxin thus lends itself to binding studies using radiolabeled derivatives. Such experiments may eventually lead to the characterization of neurotoxin site 2; the first promising steps have been taken. Modern techniques of molecular biology will almost certainly be successful, and one hopes for point-mutated channels with distinctly different reactions also to veratridine. A considerable amount of research is still required to clarify the structural basis for the numerous allosteric interactions with other sites, the mechanism of the very large potential shift of activation, the reduced single-channel conductance and selectivity, and the chemical nature of the different affinities of the site-2 toxins. Note Added in Proof. A report on point mutations with effects on neurotoxin site 2 (see Sect. 8) has just appeared: Wang S-Y, Wang GK (1988) Point mutations in segment I-S6 render voltage-gated Na+ channels resistant to batrachotoxin. Proc Natl Acad USA 95:2653-2658. In microliter muscle Na+ channels expressed in mammalian cells, mutation Asn434Lys leads to complete, Asn434Ala to partial insensitivity to 5 mM batrachotoxin. (Asn434 corresponds to Asn419 of Trainer et al. 1996). The mutant channel displays almost normal current kinetics and in the presence of veratridine little, if any, slow tail current. However, veratridine inhibits peak Na+ currents in the mutant which may point to a complex structure of site 2.
2. Veratridine Can Bind to a Site at the Mouth of the Channel Pore at Human Cardiac Sodium Channel NaV1.5
Jens Meiler, Tiffany Shields, Andrew M Glazer, Alican Gulsevin, Brett M Kroncke, Dan M Roden Int J Mol Sci . 2022 Feb 17;23(4):2225. doi: 10.3390/ijms23042225.
The cardiac sodium ion channel (NaV1.5) is a protein with four domains (DI-DIV), each with six transmembrane segments. Its opening and subsequent inactivation results in the brief rapid influx of Na+ions resulting in the depolarization of cardiomyocytes. The neurotoxin veratridine (VTD) inhibits NaV1.5 inactivation resulting in longer channel opening times, and potentially fatal action potential prolongation. VTD is predicted to bind at the channel pore, but alternative binding sites have not been ruled out. To determine the binding site of VTD on NaV1.5, we perform docking calculations and high-throughput electrophysiology experiments in the present study. The docking calculations identified two distinct binding regions. The first site was in the pore, close to the binding site of NaV1.4 and NaV1.5 blocking drugs in experimental structures. The second site was at the "mouth" of the pore at the cytosolic side, partly solvent-exposed. Mutations at this site (L409, E417, and I1466) had large effects on VTD binding, while residues deeper in the pore had no effect, consistent with VTD binding at the mouth site. Overall, our results suggest a VTD binding site close to the cytoplasmic mouth of the channel pore. Binding at this alternative site might indicate an allosteric inactivation mechanism for VTD at NaV1.5.
3. Renal effects of veratridine
K U N CHAUDHRI Br J Pharmacol Chemother . 1959 Mar;14(1):74-82. doi: 10.1111/j.1476-5381.1959.tb00931.x.
Veratridine hydrochloride injected subcutaneously into unanaesthetized rats inhibited water diuresis. A linear relationship between log dose and antidiuretic effect could be established over the dose range 50 to 200 mug./100 g. of body weight. When veratridine hydrochloride was injected intravenously in doses from 10 to 30 mug./100 g., this relationship was also linear. In terms of its antidiuretic action, the alkaloid was approximately five times as effective when given intravenously. Rats anaesthetized with urethane responded to an intravenous injection with a more pronounced inhibition than unanaesthetized animals. Protoveratrine injected intravenously into unanaesthetized rats showed no clear relationship between dose and magnitude of antidiuretic effect. Veratridine hydrochloride injected intravenously had a pronounced hypotensive effect in both anaesthetized and unanaesthetized rats. Treatment with atropine did not affect this hypotensive action significantly. Atropine given subcutaneously 30 min. before an intravenous injection of veratridine hydrochloride abolished or diminished the inhibitory effect of veratridine on water diuresis. Veratridine hydrochloride injected intravenously into unanaesthetized rats caused a marked depression of the clearance of inulin and p-aminohippurate. In unanaesthetized rats with an osmotic diuresis, veratridine hydrochloride produced its usual antidiuretic effect. The urine of rats injected with veratridine hydrochloride produced an antidiuretic effect when injected intravenously into other animals. The antidiuretic potency of such urines was not affected by treatment with thioglycollate. Animals injected with veratridine excreted small amounts of a veratridine-like substance in the urine. These results do not suggest that veratridine in antidiuretic and hypotensive doses stimulated the neurohypophysis in the rat.

Spectrum

Predicted LC-MS/MS Spectrum - 10V, Positive

Experimental Conditions

Ionization Mode: Positive
Collision Energy: 10 eV
Instrument Type: QTOF (generic), spectrum predicted by CFM-ID
Mass Resolution: 0.0001 Da
Molecular Formula: C36H51NO11
Molecular Weight (Monoisotopic Mass): 673.3462 Da
Molecular Weight (Avergae Mass): 673.7902 Da

Recommended Products

BBF-01825 Loganin Inquiry
BBF-03754 CASTANOSPERMINE Inquiry
BBF-00764 Cerebroside C Inquiry
BBF-03755 Actinomycin D Inquiry
BBF-05862 Epirubicin Inquiry
BBF-02614 Nystatin Inquiry

Bio Calculators

Stock concentration: *
Desired final volume: *
Desired concentration: *

L

* Our calculator is based on the following equation:
Concentration (start) x Volume (start) = Concentration (final) x Volume (final)
It is commonly abbreviated as: C1V1 = C2V2

* Total Molecular Weight:
g/mol
Tip: Chemical formula is case sensitive. C22H30N4O c22h30n40
g/mol
g

Recently viewed products

Online Inquiry

Verification code
cartIcon
Inquiry Basket