Aconitine

Source: Wikipedia, the free encyclopedia.
Aconitine
Names
IUPAC name
8-(acetyloxy)-20-ethyl-3α,13,15-trihydroxy-1α,6α,16β-trimethoxy-4-(methoxymethyl)aconitan-14α-yl benzoate
Other names
Acetylbenzoylaconine
Identifiers
3D model (
JSmol
)
ChEBI
ChEMBL
ChemSpider
ECHA InfoCard
100.005.566 Edit this at Wikidata
EC Number
  • 206-121-7
IUPHAR/BPS
KEGG
UNII
  • InChI=1S/C34H47NO11/c1-7-35-15-31(16-41-3)20(37)13-21(42-4)33-19-14-32(40)28(45-30(39)18-11-9-8-10-12-18)22(19)34(46-17(2)36,27(38)29(32)44-6)23(26(33)35)24(43-5)25(31)33/h8-12,19-29,37-38,40H,7,13-16H2,1-6H3/t19-,20-,21+,22-,23+,24+,25-,26?,27+,28-,29+,31+,32-,33+,34-/m1/s1 checkY
    Key: XFSBVAOIAHNAPC-XTHSEXKGSA-N checkY
  • InChI=1/C34H47NO11/c1-7-35-15-31(16-41-3)20(37)13-21(42-4)33-19-14-32(40)28(45-30(39)18-11-9-8-10-12-18)22(19)34(46-17(2)36,27(38)29(32)44-6)23(26(33)35)24(43-5)25(31)33/h8-12,19-29,37-38,40H,7,13-16H2,1-6H3/t19-,20-,21+,22-,23+,24+,25-,26?,27+,28-,29+,31+,32-,33+,34-/m1/s1
    Key: XFSBVAOIAHNAPC-XTHSEXKGBF
  • COC[C@]12CN(C)[C@@H]3[C@H]4[C@H](OC)C1[C@@]3([C@H](C[C@H]2O)OC)[C@@H]5C[C@]6(O)[C@@H](OC)[C@H](O)[C@@]4(OC(C)=O)[C@H]5C6OC(=O)c7ccccc7
  • O=C(O[C@H]5[C@]3(O)C[C@H]4[C@@]16C2N(CC)C[C@]([C@H]1[C@@H](OC)[C@@H]2[C@@](OC(=O)C)([C@@H](O)[C@@H]3OC)[C@H]45)(COC)[C@H](O)C[C@@H]6OC)c7ccccc7
Properties
C34H47NO11
Molar mass 645.73708
Appearance solid
Melting point 203 to 204 °C (397 to 399 °F; 476 to 477 K)
H2O: 0.3 mg/mL

ethanol: 35 mg/mL

Hazards
GHS labelling:
GHS06: Toxic
Danger
H300, H330
P260, P264, P270, P271, P284, P301+P310, P304+P340, P310, P320, P321, P330, P403+P233, P405, P501
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).
☒N verify (what is checkY☒N ?)

Aconitine is an alkaloid toxin produced by various plant species belonging to the genus Aconitum (family Ranunculaceae), known also commonly by the names wolfsbane and monkshood. Monkshood is notorious for its toxic properties.

Structure and reactivity

Biologically active isolates from

organic solvents such as chloroform or diethyl ether.[3][4] Aconitine is also soluble in mixtures of alcohol
and water if the concentration of alcohol is high enough.

Like many other alkaloids, the basic

methoxy group, by heating aconitine in methanol, to produce a 8-deacetyl-8-O-methyl derivatives.[6] If aconitine is heated in its dry state, it undergoes a pyrolysis to form pyroaconitine ((1α,3α,6α,14α,16β)-20-ethyl-3,13-dihydroxy-1,6,16-trimethoxy-4-(methoxymethyl)-15-oxoaconitan-14-yl benzoate) with the chemical formula C32H43NO9.[7][8]

Mechanism of action

Aconitine can interact with the voltage-dependent sodium-ion channels, which are proteins in the cell membranes of excitable tissues, such as cardiac and skeletal muscles and neurons. These proteins are highly selective for sodium ions. They open very quickly to depolarize the cell membrane potential, causing the upstroke of an action potential. Normally, the sodium channels close very rapidly, but the depolarization of the membrane potential causes the opening (activation) of potassium channels and potassium efflux, which results in repolarization of the membrane potential.

Aconitine binds to the channel at the neurotoxin binding site 2 on the alpha subunit (the same site bound by batrachotoxin, veratridine, and grayanotoxin).[9] This binding results in a sodium-ion channel that stays open longer. Aconitine suppresses the conformational change in the sodium-ion channel from the active state to the inactive state. The membrane stays depolarized due to the constant sodium influx (which is 10–1000-fold greater than the potassium efflux). As a result, the membrane cannot be repolarized. The binding of aconitine to the channel also leads to the channel to change conformation from the inactive state to the active state at a more negative voltage.[10] In neurons, aconitine increases the permeability of the membrane for sodium ions, resulting in a huge sodium influx in the axon terminal. As a result, the membrane depolarizes rapidly. Due to the strong depolarization, the permeability of the membrane for potassium ions increases rapidly, resulting in a potassium reflux to release the positive charge out of the cell. Not only the permeability for potassium ions but also the permeability for calcium ions increases as a result of the depolarization of the membrane. A calcium influx takes place. The increase of the calcium concentration in the cell stimulates the release of the neurotransmitter acetylcholine into the synaptic cleft. Acetylcholine binds to acetylcholine receptors at the postsynaptic membrane to open the sodium-channels there, generating a new action potential.

Research with mouse nerve-hemidiaphragm muscle preparation indicate that at low concentrations (<0.1 μM) aconitine increases the electrically evoked acetylcholine release causing an induced muscle tension.[11] Action potentials are generated more often at this concentration. At higher concentration (0.3–3 μM) aconitine decreases the electrically evoked acetylcholine release, resulting in a decrease in muscle tension. At high concentration (0.3–3 μM), the sodium-ion channels are constantly activated, transmission of action potentials is suppressed, leading to non-excitable target cells or paralysis.

Biosynthesis and total synthesis of related alkaloids

Aconitine is biosynthesized by the

monkshood plant via the terpenoid biosynthesis pathway (MEP chloroplast pathway).[12] Approximately 700 naturally occurring C19-diterpenoid alkaloids have been isolated and identified, but the biosynthesis of only a few of these alkaloids are well understood.[13]

Likewise, only a few alkaloids of the aconitine family have been synthesized in the laboratory. In particular, despite over one hundred years having elapsed since its isolation, the prototypical member of its family of norditerpenoid alkaloids, aconitine itself, represents a rare example of a well-known natural product that has yet to succumb to efforts towards its total synthesis. The challenge that aconitine poses to synthetic organic chemists is due to both the intricate interlocking hexacyclic ring system that makes up its core and the elaborate collection of oxygenated functional groups at its periphery. A handful of simpler members of the aconitine alkaloids, however, have been prepared synthetically. In 1971, the Weisner group discovered the total synthesis of talatisamine (a C19-norditerpenoid).[14] In the subsequent years, they also discovered the total syntheses of other C19-norditerpenoids, such as chasmanine,[15] and 13-deoxydelphonine.[16]

Schematic for the Wiesner Syntheses of Napelline. Deoxydelphonine and Talatisamine

The total synthesis of napelline (Scheme a) begins with aldehyde 100.[14] In a 7 step process, the A-ring of napelline is formed (104). It takes another 10 steps to form the lactone ring in the pentacyclic structure of napelline (106). An additional 9 steps creates the enone-aldehyde 107. Heating in methanol with potassium hydroxide causes an aldol condensation to close the sixth and final ring in napelline (14). Oxidation then gives rise to diketone 108 which was converted to (±)-napelline (14) in 10 steps.

A similar process is demonstrated in Wiesner's synthesis of 13-desoxydelphinone (Scheme c).

ketal
114. The addition of heat, DMSO and o-xylene rearranges this ketol (115), and after 5 more steps (±)-13-desoxydelphinone (15) is formed.

Lastly, talatisamine (Scheme d) is synthesized from diene 116 and nitrile 117.

enone 120. Subsequently, this allene is added to produce photoadduct
121. This adduct group is cleaved and rearrangement gives rise to the compound 122. In 7 steps, this compound forms 123, which is then rearranged, in a similar manner to compound 114, to form the aconitine-like skeleton in 124. A racemic relay synthesis is completed to produce talatisamine (13).

More recently, the laboratory of the late David Y. Gin completed the total syntheses of the aconitine alkaloids nominine[17] and neofinaconitine.[18]

Metabolism

Aconine: an amorphous, bitter, non-poisonous alkaloid, derived from the decomposition of aconitine

Aconitine is metabolized by cytochrome P450 isozymes (CYPs). There has been research in 2011 in China to investigate in-depth the CYPs involved in aconitine metabolism in human liver microsomes.[19] It has been estimated that more than 90 percent of currently available human drug metabolism can be attributed to eight main enzymes (CYP 1A2, 2C9, 2C8, 2C19, 2D6, 2E1, 3A4, 3A5).[20] The researchers used recombinants of these eight different CYPs and incubated it with aconitine. To initiate the metabolism pathway the presence of NADPH was needed. Six CYP-mediated metabolites (M1–M6) were found by liquid chromatography, these six metabolites were characterized by mass-spectrometry. The six metabolites and the involved enzymes are summarized in the following table:

Metabolite Name Involved CYPs
M1 O-Demethyl-aconitine CYP3A4, CYP3A5, CYP2D6, CYP2C8
M2 16-O-Demethyl-aconitine CYP3A4, CYP3A5, CYP2D6, CYP2C9
M3 N-deethyl-aconitine CYP3A4, CYP3A5, CYP2D6, CYP2C9
M4 O-didemethyl-aconitine CYP3A5, CYP2D6
M5 3-Dehydrogen-aconitine CYP3A4, CYP3A5
M6 Hydroxyl-aconitine CYP3A5, CYP2D6

Selective inhibitors were used to determine the involved CYPs in the aconitine metabolism. The results indicate that aconitine was mainly metabolized by CYP3A4, 3A5 and 2D6. CYP2C8 and 2C9 had a minor role to the aconitine metabolism, whereas CYP1A2, 2E1 and 2C19 did not produce any aconitine metabolites at all. The proposed metabolic pathways of aconitine in human liver microsomes and the CYPs involved to it are summarized in the table above.

Uses

Aconitine was previously used as an antipyretic and analgesic and still has some limited application in herbal medicine, although the narrow therapeutic index makes calculating appropriate dosage difficult.[21] Aconitine is also present in Yunnan Baiyao, a proprietary traditional Chinese medicine.[22]

Toxicity

Consuming as little as 2

milligrams of pure aconitine or 1 gram of the plant itself may cause death by paralyzing respiratory or heart functions.[23] Toxicity may occur through the skin; even touching the flowers can numb finger tips.[23][citation needed
]

The toxic effects of aconitine have been tested in a variety of animals, including mammals (dog, cat, guinea pig, mouse, rat and rabbit), frogs and pigeons. Depending on the route of exposure, the observed toxic effects were

arrhythmias or death.[23][24] According to a review of different reports of aconite poisoning in humans, the following clinical features were observed:[21]

Progression of symptoms: the first symptoms of aconitine poisoning appear approximately 20 minutes to 2 hours after oral intake and include paresthesia, sweating and nausea. This leads to severe vomiting, colicky diarrhea, intense pain and then paralysis of the skeletal muscles. Following the onset of life-threatening arrhythmia, including ventricular tachycardia and ventricular fibrillation, death finally occurs as a result of respiratory paralysis or cardiac arrest.[25]

lowest published lethal dose (LDLo) for mice is 1 mg/kg orally and 0.100 mg/kg intraperitoneally. The lowest published toxic dose (TDLo) for mice is 0.0549 mg/kg subcutaneously. LD50 value for rats is 0.064 mg/kg intravenously. The LDLo for rats is 0.040 mg/kg intravenously and 0.250 mg/kg intraperitoneally. The TDLo for rats is 0.040 mg/kg parenterally. For an overview of more test animal results (LD50, LDLo and TDLo) see the following table.[24]

Species observed Type of test Route of exposure Dose data (mg/kg) Toxic effects
Human LDLo Oral 0.028 Behavioral: excitement

Gastrointestinal: hypermotility, diarrhea Gastrointestinal: other changes

Human LDLo Oral 0.029 Details of toxic effects not reported other than lethal dose value
Cat LD50 Intravenous 0.080 Behavioral: convulsions or effect on seizure threshold
Cat LDLo Subcutaneous 0.100 Details of toxic effects not reported other than lethal dose value
Guinea pig LD50 Intravenous 0.060 Behavioral: convulsions or effect on seizure threshold
Guinea pig LDLo Subcutaneous 0.050 Details of toxic effects not reported other than lethal dose value
Guinea pig LDLo Intravenous 0.025 Cardiac: arrhythmias (including changes in conduction)
Mouse LD50 Intraperitoneal 0.270 Details of toxic effects not reported other than lethal dose value
Mouse LD50 Intravenous 0.100 Sense Organs and Special Senses (Eye): lacrimation

Behavioral: convulsions or effect on seizure threshold Lungs, Thorax, or Respiration: dyspnea

Mouse LD50 Oral 1 Details of toxic effects not reported other than lethal dose value
Mouse LD50 Subcutaneous 0.270 Details of toxic effects not reported other than lethal dose value
Mouse LDLo Intraperitoneal 0.100 Details of toxic effects not reported other than lethal dose value
Mouse LDLo Oral 1 Behavioral: convulsions or effect on seizure threshold

Cardiac: arrhythmias (including changes in conduction) Gastrointestinal: hypermotility, diarrhea

Mouse TDLo Subcutaneous 0.0549 Peripheral Nerve and Sensation: local anesthetic

Behavioral: analgesia

Rabbit LDLo Subcutaneous 0.131 Details of toxic effects not reported other than lethal dose value
Rat LD50 Intravenous 0.080 Behavioral: convulsions or effect on seizure threshold
Rat LD50 Intravenous 0.064 Details of toxic effects not reported other than lethal dose value
Rat LDLo Intraperitoneal 0.250 Cardiac: other changes

Lungs, Thorax, or Respiration: dyspnea

Rat LDLo Intravenous 0.040 Cardiac: arrhythmias (including changes in conduction)
Rat TDLo Parenteral 0.040 Cardiac: arrhythmias (including changes in conduction)
Frog LDLo Subcutaneous 0.586 Details of toxic effects not reported other than lethal dose value
Pigeon LDLo Subcutaneous 0.066 Details of toxic effects not reported other than lethal dose value
  • Note that LD50 means lethal dose, 50 percent kill; LDLo means lowest published lethal dose; TDLo means lowest published toxic dose

For humans the lowest published oral lethal dose of 28 μg/kg was reported in 1969.

Diagnosis and treatment

For the analysis of the Aconitum alkaloids in biological specimens such as blood, serum and urine, several

GC-MS methods have been described. These employ a variety of extraction procedures followed by derivatisation to their trimethylsilyl derivatives. New sensitive HPLC-MS methods have been developed as well, usually preceded by SPE purification of the sample.[25] The antiarrhythmic drug lidocaine has been reported to be an effective treatment of aconitine poisoning of a patient. Considering the fact that aconitine acts as an agonist of the sodium channel receptor, antiarrhythmic agents which block the sodium channel (Vaughan-Williams' classification I) might be the first choice for the therapy of aconitine induced arrhythmias.[26] Animal experiments have shown that the mortality of aconitine is lowered by tetrodotoxin. The toxic effects of aconitine were attenuated by tetrodotoxin, probably due to their mutual antagonistic effect on excitable membranes.[27] Also paeoniflorin seems to have a detoxifying effect on the acute toxicity of aconitine in test animals. This may result from alternations of pharmacokinetic behavior of aconitine in the animals due to the pharmacokinetic interaction between aconitine and paeoniflorin.[28] In addition, in emergencies, one can wash the stomach using either tannic acid or powdered charcoal. Heart stimulants such as strong coffee or caffeine may also help until professional help is available.[29]

Famous poisonings

During the Indian Rebellion of 1857, a British detachment was the target of attempted poisoning with aconitine by the Indian regimental cooks. The plot was thwarted by John Nicholson who, having detected the plot, interrupted the British officers just as they were about to consume the poisoned meal. The chefs refused to taste their own preparation, whereupon it was force-fed to a monkey who "expired on the spot". The cooks were hanged.

Aconitine was the poison used by George Henry Lamson in 1881 to murder his brother-in-law in order to secure an inheritance. Lamson had learned about aconitine as a medical student from professor Robert Christison, who had taught that it was undetectable—but forensic science had improved since Lamson's student days.[30][31][32]

Rufus T. Bush, American industrialist and yachtsman, died on September 15, 1890, after accidentally taking a fatal dose of aconite.

In 1953 aconitine was used by a Soviet biochemist and poison developer, Grigory Mairanovsky, in experiments with prisoners in the secret NKVD laboratory in Moscow. He admitted killing around 10 people using the poison.[33]

In 2004 Canadian actor

Andre Noble
died from aconitine poisoning. He accidentally ate some monkshood while he was on a hike with his aunt in Newfoundland.

In 2009

Lakhvinder Cheema (who died as a result of the poisoning) and his current fiancée Gurjeet Choongh. Singh received a life sentence with a 23-year minimum for the murder on February 10, 2010.[34]

In 2022, twelve diners at a restaurant in York Region became acutely ill following a meal. All twelve became seriously ill and four of them were admitted to the intensive care unit after the suspected poisoning.[35]

In popular culture

Aconitine was a favorite poison in the ancient world. The poet Ovid, referring to the proverbial dislike of stepmothers for their step-children, writes:

Lurida terribiles miscent aconita novercae.[36]

Fearsome stepmothers mix lurid aconites.

Aconitine was also made famous by its use in

Twin Peaks (season 3) Part 13, aconitine is suggested to poison the main character.[37]

Monk's Hood is the name of the third Cadfael Novel written in 1980 by

Ellis Peters. The novel was made into an episode of the television series Cadfael starring Derek Jacobi
.

In the third season of the Netflix series You, two of the main characters poison each other with Aconite. One survives (due to a lower dose and an antidote), and the other is killed.

Hannah McKay (Yvonne Strahovski), a serial killer in the Showtime series Dexter uses Aconite on at least three occasions to poison her victims.

In season two episode sixteen of the series Person Of Interest aconitine is shown in a syringe stuck to the character Shaw (Sarah Shahi) nearly being injected and causing her death, until she is rescued by Reese (Jim Caviezel)

In a 2017 episode of The Doctor Blake Mysteries, fight manager Gus Jansons (Steve Adams) murdered his boxer, Mickey Ellis (Trey Coward), during a match by applying aconitine he had put in petroleum jelly and applying it to a cut over the boxer’s eye. He feared being blackmailed over a murder he helped cover up. He had made the poison from wolfsbane he had seen in a local garden.[38]

See also

References

  1. ^ Biogenetically, aconitine is not a 'true' alkaloid, as it is not ultimately derived from amino acids. Aconitine is ultimately derived from isoprene, so it is technically a terpenoid and a pseudoalkaloid.
  2. PMID 24040959
    .
  3. ^ "Aconitine". Sigma Aldrich. Retrieved 22 July 2016.
  4. ^ "Aconitine sc-202441 Material Safety Data Sheet" (PDF). Santa Cruz Biotechnology.
  5. .
  6. .
  7. .
  8. ^ "Pyroaconitine ChemSpider ID: 10211301". Chemspider.
  9. S2CID 21509335
    .
  10. .
  11. .
  12. ^ Viberti F, Raveggi E. "Aconitine: How Poisonous, How Harmful?". flipper e nuvola. Retrieved 26 April 2017.
  13. PMID 19275222
    .
  14. ^ .
  15. ^ .
  16. ^ .
  17. .
  18. .
  19. .
  20. .
  21. ^ .
  22. ^ "Yunnan Baiyao finally discloses toxic ingredient". GoKunming. 2014-04-07.
  23. ^ a b c "Aconite". Drugs.com. 9 August 2019. Retrieved 23 June 2020.
  24. ^ a b "RTECS". Oct 2011.
  25. ^
    S2CID 2490984
    .
  26. .
  27. .
  28. .
  29. .
  30. .
  31. .
  32. .
  33. ^ Лаборатория Икс [Laboratory X]. Novaya Gazeta (in Russian). 2010-05-06. Archived from the original on 2010-05-30. Retrieved 2013-04-08.
  34. ^ "Poisoning in west London in 2009". BBC TV News. 2010-02-10.
  35. ^ "12 People Poisoned at Toronto-Area Restaurant". 30 August 2022.
  36. ^ Ovid, Metamorphoses, 1.147
  37. ^ Jensen, Jeff (7 August 2017). "Twin Peaks recap: 'The Return: Part 13'". Entertainment Weekly. Meredith Corporation. Retrieved 4 May 2020. Clark offered to sell him Aconitine, a toxin with a rich literary history.
  38. ^ December Media Pty. “A Lethal Combination.” The Doctor Blake Mysteries, Season 5, Episode 1. Australian Broadcasting Corporation, 17 September 2017.

External links

  • Media related to Aconitine at Wikimedia Commons