N,N-Dimethyldopamine

Last updated
N,N-Dimethyldopamine
Dimethyldopamine.svg
Names
Preferred IUPAC name
4-[2-(Dimethylamino)ethyl]benzene-1,2-diol
Identifiers
3D model (JSmol)
ChemSpider
PubChem CID
UNII
  • InChI=1S/C10H15NO2/c1-11(2)6-5-8-3-4-9(12)10(13)7-8/h3-4,7,12-13H,5-6H2,1-2H3
    Key: XJTVXBWTYZCUJX-UHFFFAOYSA-N
  • CN(C)CCC1=CC(=C(C=C1)O)O
Properties
C10H15NO2
Molar mass 181.235 g·mol−1
Appearancecolorless
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

N,N-Dimethyldopamine (DMDA) is an organic compound belonging to the phenethylamine family. It is related structurally to the alkaloid epinine (N-methyldopamine) and to the major neurotransmitter dopamine (of which it is the N,N-dimethylated analog). Because of its structural relationship to dopamine, DMDA has been the subject of a number of pharmacological investigations. DMDA has been detected in Acacia rigidula .

Contents

Occurrence

DMDA has been reported from the plant Acacia rigidula Benth. (Fabaceae), in which it has been detected at levels of ~ 11-45 ppm. [1]

Chemistry

Since N,N-dimethyldopamine is chemically an amine, it is basic (a weak base, technically), but it is also a catechol (a 1,2-dihydroxybenzene), which gives it weakly acidic properties, so that the compound is amphoteric.

Preparation

Several different methods have been reported for the preparation of DMDA. An early synthesis by Buck and co-workers began with 3,4-dimethoxybenzaldehyde (veratraldehyde), which was condensed with hippuric acid to give the azlactone; this was hydrolyzed with NaOH to the corresponding pyruvic acid, which was then converted to its oxime; treatment of the oxime with acetic anhydride gave 3,4-dimethoxyphenylacetonitrile, which was catalytically reduced (H2/Pd) in the presence of excess dimethylamine to N,N-dimethyl-3,4-dimethoxyphenethylamine; finally, the methoxy-groups were cleaved with HCl to give DMDA as its hydrochloride salt. [2]

A more recent method starts with 3,4-dimethoxyphenylacetic acid, which is converted to its acid chloride with thionyl chloride; this is reacted with dimethylamine to give the dimethylamide, which is then reduced using diborane to N,N-dimethyl 3,4-dimethoxyphenethylamine; the methoxy- groups are finally cleaved with hydriodic acid to give DMDA. [3]

The shortest method is that of Borgman et al., who converted 3,4-dimethoxyphenethylamine into N,N-dimethyl 3,4-dimethoxyphenethylamine by catalytic reduction (H2/Pd) in the presence of formaldehyde; the methoxy-groups were then cleaved with hydrobromic acid. [4]

Pharmacology

One of the earliest pharmacological studies of DMDA was that of Daly and his co-workers, who studied the ability of a large number of substituted phenethylamines to release norepinephrine (NE) from the mouse heart. In this assay, a subcutaneous dose of 10 mg/kg of DMDA hydrochloride (referred to as "3,4-dihydroxy-N,N-dimethylphenethylamine HCl") failed to produce a significant change in the NE content of the heart. In comparison, a dose of 5 mg/kg, s.c., of N-methyldopamine ("3,4-dihydroxy-N-methylphenethylamine HCl") caused a 45% reduction in the NE content, while dopamine HCl itself caused a 50% decrease at a dose of 5 mg/kg, s.c. [5]

Another early pharmacological investigation of DMDA was carried out by Goldberg and co-workers, who examined the effects of a range of phenethylamine analogs in an assay based on the vasodilation produced by injection of the test drug into the renal artery of the dog. In this assay, a drug was classed as "dopamine-like" if the vasoldilation it produced was not prevented by β-blocking drugs, and did not occur if the drug was injected into the femoral artery. Although DMDA, at a dose of 0.5 mg, caused a marked bradycardia, a dose of ~ 0.75 mg did not increase renal blood flow (i.e. cause vasodilation) after administration of atropine to abolish the bradycardia. [6]

In cats pretreated with atropine and hexamethonium, DMDA is a strong vasopressor: a parenteral dose of 10 μg/kg produced a rise in blood pressure more than twice that produced by the same dose of dopamine. In an assay based on the increase in heart rate (positive chronotropic response) produced by electrical stimulation of the post-ganglionic fibers of cat cardioaccelerator nerve, an i.v. dose of ~ 15 μg/kg DMDA caused a 50% reduction of the response, compared to an approximately 10% decrease produced by the same dose of dopamine. From these and other related observations, the researchers concluded that DMDA was a potent inhibitor of the adrenergic system via stimulation of inhibitory putative (at that time) dopamine receptors on adrenergic nerve terminals. [7]

In the dog, an i.v. dose of 16 μg/kg caused an ~ 80% decrease in heart rate in the same cardioaccelerator nerve assay, compared to an ~ 8% decrease produced by dopamine. DMDA caused vasoconstriction in several isolated vascular preparations from the rabbit. The pressor activity of DMDA was partially inhibited by the α-antagonist phentolamine. From these and other observations, the investigators concluded that there were significant species-related differences between the responses to DMDA of dogs and cats, with adrenergic effects being predominant in dogs. [8]

Ginos et al. tested DMDA for effects in unilaterally-caudectomized mice (dose ≤ 120 mg/kg, i.p.), nigral-lesioned rats (dose = 10 mg/kg, i.p.), and on adenylate cyclase activity in homogenized mouse caudate nuclei (concentration = 10μM/L). DMDA showed no effects in any of these assay systems. By comparison, N-methyldopamine also had no effect in caudectomized mice at ≤ 150 mg/kg, and only a weak effect in nigral-lesioned rats at 25 mg/kg, although it was as effective as dopamine in stimulating cAMP in the adenylate cyclase assay. [3]

Borgman and co-workers reported in 1973 that at 100 mg/kg, given i.p. to mice, DMDA failed to antagonize the tremor and reduction in locomotor activity produced by pre-administration of oxotremorine. In another assay, 6 mg/kg of DMDA (i.p. in mice) only slightly antagonized the reduced locomotor activity resulting from pre-treatment with reserpine. A dose of 1 mg/kg, iv., of DMDA did not produce any hypothermia in mice. [4]

It has been stated that dopamine is behaviorally inactive due to its rapid peripheral metabolism and inability to cross the blood–brain barrier. [9] When dopamine or N-methyldopamine were injected directly into the nucleus accumbens of mice, however, doses of 12.5-50 μg produced marked hyperactivity, with the latter being somewhat more potent. In contrast, DMDA did not produce any hyperactivity in doses up to 100 μg. [10]

In a 1981 paper, Costall and co-workers reported that DMDA, in doses of 0.5–8 mg/kg given i.p. to mice, produced a dose-dependent reduction in spontaneous motor activity (occurring within a 20 minute period after drug administration). They also observed piloerection at 2 mg/kg, and prostration accompanying the 8 mg/kg or higher doses. The effects of DMDA were not altered by the administration of spiroperidol. [11]

Receptor binding studies, in competition with [3H]-spiperone, using receptors from pig anterior pituitary, have revealed the following affinities for D2 receptors exhibited by DMDA: Kahigh = 20 nM; Kalow = 10200 nM. In comparison, the corresponding affinities for N-methyldopamine are: 10.4 nM (high) and 3430 nM (low), while for dopamine they are 7.5 nM (high) and 4300 nM (low affinity state). [12]

Similar receptor binding results were obtained when DMDA and DA were assayed using a receptor preparation from rat striatum: competition against [3H]-spiperone gave affinity constants of ~ 25 nM (high affinity state) and ~ 724 nM (low) for DMDA, compared to ~ 10 nM (high) and ~ 354 nM (low) for dopamine. Both drugs were also tested for their ability to inhibit the [3H]-ACh release from mouse striatal slices evoked by K+. In this assay, the ED50 for DMDA was ~ 0.06 μM, and for dopamine it was ~ 1.9 μM. [13]

Toxicity

The LD50 for N,N-dimethyldopamine·HCl is reported as 240 mg/kg (mouse, i.p.).; [3] under the same experimental conditions, the LD50 for N-methyldopamine.HBr (epinine hydrobromide) is 212 mg/kg (mouse, i.p.), and the LD50 for dopamine·HCl is 1978 mg/kg (mouse, i.p.). [3]

See also

Related Research Articles

<span class="mw-page-title-main">Salvinorin A</span> Chemical compound

Salvinorin A is the main active psychotropic molecule in Salvia divinorum. Salvinorin A is considered a dissociative hallucinogen.

<span class="mw-page-title-main">2C-B</span> Chemical compound

2C-B (4-Bromo-2,5-dimethoxyphenethylamine) is a psychedelic drug of the 2C family. It was first synthesized by Alexander Shulgin in 1974. In Shulgin's book PiHKAL, the dosage range is listed as 12–24 mg. As a recreational drug, 2C-B is sold as a white powder sometimes pressed in tablets or gel caps. It is also referred to by a number of street names. The drug is usually taken orally, but can also be insufflated or vaporized. While being primarily a psychedelic it is also a mild entactogen.

<span class="mw-page-title-main">Tetraethylammonium</span> Polyatomic ion (N(C₂H₅)₄, charge +1)

Tetraethylammonium (TEA) is a quaternary ammonium cation with the chemical formula [Et4N]+, consisting of four ethyl groups attached to a central nitrogen atom. It is a counterion used in the research laboratory to prepare lipophilic salts of inorganic anions. It is used similarly to tetrabutylammonium, the difference being that its salts are less lipophilic, more easily crystallized and more toxic.

<span class="mw-page-title-main">Synephrine</span> Chemical compound

Synephrine, or, more specifically, p-synephrine, is an alkaloid, occurring naturally in some plants and animals, and also in approved drugs products as its m-substituted analog known as neo-synephrine. p-Synephrine and m-synephrine are known for their longer acting adrenergic effects compared to epinephrine and norepinephrine. This substance is present at very low concentrations in common foodstuffs such as orange juice and other orange products, both of the "sweet" and "bitter" variety. The preparations used in traditional Chinese medicine (TCM), also known as Zhi Shi (枳实), are the immature and dried whole oranges from Citrus aurantium. Extracts of the same material or purified synephrine are also marketed in the US, sometimes in combination with caffeine, as a weight-loss-promoting dietary supplement for oral consumption. While the traditional preparations have been in use for millennia as a component of TCM-formulas, synephrine itself is not an approved over the counter drug. As a pharmaceutical, m-synephrine (phenylephrine) is still used as a sympathomimetic, mostly by injection for the treatment of emergencies such as shock, and rarely orally for the treatment of bronchial problems associated with asthma and hay-fever.

<span class="mw-page-title-main">Chlorprothixene</span> Chemical compound

Chlorprothixene, sold under the brand name Truxal among others, is a typical antipsychotic of the thioxanthene group.

<span class="mw-page-title-main">Lysergic acid hydroxyethylamide</span> Chemical compound

D-Lysergic acid α-hydroxyethylamide, also known as D-lysergic acid methyl carbinolamide, is an alkaloid of the ergoline family, believed to be present in various species in the Convolvulaceae, as well as some species of fungi. Morning glory heavenly blue and Hawaiian baby woodrose especially contain high amounts of LSH, with content varying between species and by how fresh.

<span class="mw-page-title-main">Hordenine</span> Chemical compound

Hordenine is an alkaloid of the phenethylamine class that occurs naturally in a variety of plants, taking its name from one of the most common, barley. Chemically, hordenine is the N-methyl derivative of N-methyltyramine, and the N,N-dimethyl derivative of the well-known biogenic amine tyramine, from which it is biosynthetically derived and with which it shares some pharmacological properties. As of September 2012, hordenine is widely sold as an ingredient of nutritional supplements, with the claims that it is a stimulant of the central nervous system, and has the ability to promote weight loss by enhancing metabolism. In experimental animals, given sufficiently large doses parenterally, hordenine does produce an increase in blood pressure, as well as other disturbances of the cardiovascular, respiratory, and nervous systems. These effects are generally not reproduced by oral administration of the drug in test animals, and virtually no scientific reports of the effects of hordenine in human beings have been published.

<span class="mw-page-title-main">Adrenomedullin</span>

Adrenomedullin is a vasodilator peptide hormone of uncertain significance in human health and disease. It was initially isolated in 1993 from a pheochromocytoma, a tumor of the adrenal medulla: hence the name.

<span class="mw-page-title-main">MME (psychedelic)</span> Chemical compound

MME (2,4-dimethoxy-5-ethoxyamphetamine) is a lesser-known psychedelic drug. It is a dimethoxy-ethoxy analog of TMA-2. MME was first synthesized by Alexander Shulgin from ethylvanillin. In his book PiHKAL, the minimum dosage is listed as 40 mg and above, and the duration listed as 6–10 hours. Shulgin gives MME a ++ on the Shulgin Rating Scale.

<i>N</i>-Methylphenethylamine Chemical compound

N-Methylphenethylamine (NMPEA) is a naturally occurring trace amine neuromodulator in humans that is derived from the trace amine, phenethylamine (PEA). It has been detected in human urine and is produced by phenylethanolamine N-methyltransferase with phenethylamine as a substrate, which significantly increases PEA's effects. PEA breaks down into Phenylacetaldehyde which is further broken down into Phenylacetic acid by Monoamine oxidase. When this is inhibited by Monoamine oxidase inhibitors, it allows more of the PEA to be metabolized into nymphetamine (NMPEA) and not wasted on the weaker inactive metabolites.

<span class="mw-page-title-main">Alpha-2C adrenergic receptor</span> Protein-coding gene in the species Homo sapiens

The alpha-2C adrenergic receptor, also known as ADRA2C, is an alpha-2 adrenergic receptor, and also denotes the human gene encoding it.

<span class="mw-page-title-main">Deoxyepinephrine</span> Chemical compound

Deoxyepinephrine, also known by the common names N-methyldopamine and epinine, is an organic compound and natural product that is structurally related to the important neurotransmitters dopamine and epinephrine. All three of these compounds also belong to the catecholamine family. The pharmacology of epinine largely resembles that of its "parent", dopamine. Epinine has been found in plants, insects and animals. It is also of significance as the active metabolic breakdown product of the prodrug ibopamine, which has been used to treat congestive heart failure.

<span class="mw-page-title-main">ICI-118,551</span> Chemical compound

ICI-118,551 is a selective β2 adrenergic receptor (adrenoreceptor) antagonist or beta blocker. ICI binds to the β2 subtype with at least 100 times greater affinity than β1 or β3, the two other known subtypes of the beta adrenoceptor. The compound was developed by Imperial Chemical Industries, which was acquired by AkzoNobel in 2008.

<span class="mw-page-title-main">Phenylethanolamine</span> Chemical compound

Phenylethanolamine, or β-hydroxyphenethylamine, is a trace amine with a structure similar to those of other trace phenethylamines as well as the catecholamine neurotransmitters dopamine, norepinephrine, and epinephrine. As an organic compound, phenylethanolamine is a β-hydroxylated phenethylamine that is also structurally related to a number of synthetic drugs in the substituted phenethylamine class. In common with these compounds, phenylethanolamine has strong cardiovascular activity and, under the name Apophedrin, has been used as a drug to produce topical vasoconstriction.

<span class="mw-page-title-main">Monoamine releasing agent</span> Class of compounds

A monoamine releasing agent (MRA), or simply monoamine releaser, is a drug that induces the release of a monoamine neurotransmitter from the presynaptic neuron into the synapse, leading to an increase in the extracellular concentrations of the neurotransmitter. Many drugs induce their effects in the body and/or brain via the release of monoamine neurotransmitters, e.g., trace amines, many substituted amphetamines, and related compounds.

DO<em>x</em> Class of chemical compounds

4-Substituted-2,5-dimethoxyamphetamines (DOx) is a chemical class of substituted amphetamine derivatives featuring methoxy groups at the 2- and 5- positions of the phenyl ring, and a substituent such as alkyl or halogen at the 4- position of the phenyl ring. Most compounds of this class are potent and long-lasting psychedelic drugs, and act as highly selective 5-HT2A, 5-HT2B, and 5-HT2C receptor partial agonists. A few bulkier derivatives such as DOAM have similarly high binding affinity for 5-HT2 receptors but instead act as antagonists, and so do not produce psychedelic effects though they retain amphetamine-like stimulant effects.

<span class="mw-page-title-main">Osemozotan</span> Pharmaceutical drug

Osemozotan (MKC-242) is a selective 5-HT1A receptor agonist with some functional selectivity, acting as a full agonist at presynaptic and a partial agonist at postsynaptic 5-HT1A receptors. 5-HT1A receptor stimulation influences the release of various neurotransmitters including serotonin, dopamine, norepinephrine, and acetylcholine. 5-HT1A receptors are inhibitory G protein-coupled receptor. Osemozotan has antidepressant, anxiolytic, antiobsessional, serenic, and analgesic effects in animal studies, and is used to investigate the role of 5-HT1A receptors in modulating the release of dopamine and serotonin in the brain, and their involvement in addiction to abused stimulants such as cocaine and methamphetamine.

<i>N</i>-Methyltyramine Chemical compound

N-Methyltyramine (NMT), also known as 4-hydroxy-N-methylphenethylamine, is a human trace amine and natural phenethylamine alkaloid found in a variety of plants. As the name implies, it is the N-methyl analog of tyramine, which is a well-known biogenic trace amine with which NMT shares many pharmacological properties. Biosynthetically, NMT is produced by the N-methylation of tyramine via the action of the enzyme phenylethanolamine N-methyltransferase in humans and tyramine N-methyltransferase in plants.

<span class="mw-page-title-main">Candicine</span> Chemical compound

Candicine is a naturally occurring organic compound that is a quaternary ammonium salt with a phenethylamine skeleton. It is the N,N,N-trimethyl derivative of the well-known biogenic amine tyramine, and, being a natural product with a positively charged nitrogen atom in its molecular structure, it is classed as an alkaloid. Although it is found in a variety of plants, including barley, its properties have not been extensively studied with modern techniques. Candicine is toxic after parenteral administration, producing symptoms of neuromuscular blockade; further details are given in the "Pharmacology" section below.

<span class="mw-page-title-main">Halostachine</span> Alkaloid

Halostachine is a natural product, an alkaloid first isolated from the Asian shrub Halostachys caspica, and structurally a β-hydroxy-phenethylamine related to its better-known "parent" biogenic amine, phenylethanolamine, to the adrenergic drug synephrine, and to the alkaloid ephedrine. The pharmacological properties of halostachine have some similarity to those of these structurally-related compounds, and Halostachys caspica extracts have been included as a constituent of certain OTC dietary supplements, but halostachine has never been developed as a prescription drug. Although it is found in nature as a single stereoisomer, halostachine is more commonly available as a synthetic product in the form of its racemate. In appearance it is a colorless solid.

References

  1. B. A. Clement, C. M. Goff, and T.D. A. Forbes (1998). "Toxic amines and alkaloids from Acacia rigidula." Phytochemistry49 1377-1380.
  2. J. S. Buck, R. Baltzly and W. Ide (1938). "β-Phenethylamine derivatives. Tertiary and quaternary salts." J. Am. Chem. Soc.60 1789-1796.
  3. 1 2 3 4 J. Z. Ginos et al. (1975). "Cholinergic effects of molecular segments of apomorphine and dopaminergic effects of N,N-dialkylated dopamines." J. Med. Chem.18 1194-1200.
  4. 1 2 R. J. Borgman, J. J. McPhillips, R. E. Stitzel, and I. J. Goodman (1973). "Synthesis and pharmacology of centrally acting dopamine derivatives and analogs in relation to Parkinson's disease." J. Med. Chem.16 630-633.
  5. J. W. Daly, C. R. Creveling, and B. Witkop (1966). "The chemorelease of norepinephrine from mouse hearts. Structure-activity relationships. I. Sympathomimetic and related amines." J. Med. Chem.9 273-280.
  6. L. I. Goldberg, P. F. Sonneville, and J. L. McNay (1968). "An investigation of the structural requirements for dopamine-like renal vasodilation: phenethylamines and apomorphine." J. Pharmacol. Exp. Ther.163 188-197.
  7. M. Ilhan, J. P. Long and J. G. Cannon (1975). "Bulbocapnine's ability to antagonize the adrenergic inhibitory action of dopamine and analogs." Eur. J. Pharmacol.33 13-18.
  8. J. M. Kitzen, M. Ilhan, J. G. Cannon and J. P. Long (1976). "α-Adrenergic activity of N,N-dimethyldopamine (DMDA)." Eur. J. Pharmacol.38 365-372.
  9. J. G. Cannon, F.-L. Hsu, J. P. Long, J. R. Flynn, B. Costall, and R. J. Naylor (1978). "Preparation and biological actions of some symmetrically N,N-disubstituted dopamines." J. Med. Chem.21 248–253.
  10. B. Costall, R. J. Naylor and R. M. Pinder (1976). "Characterisation of the mechanisms for hyperactivity induction from the nucleus accumbens by phenethylamine derivatives." Psychopharmacol.48 225-231.
  11. B. Costall, S. K. Lim, and R. J. Naylor (1981). "Characterisation of the mechanisms by which purported dopamine agonists reduce spontaneous locomotor activity of mice." Eur. J. Pharmacol.73 175-188.
  12. P. Seeman, M. Watanabe, D. Grigoriadis, J. L. Tedesco, S. R. George, U. Svensson, J. L. Nilsson, and J. L. Neumeyer (1985). "Dopamine D2 receptor binding sites for agonists. A tetrahedral model." Mol. Pharmacol.28 391-399.
  13. R. A. Wallace, T. Farooqui, L. Wallace, J. Ares, Y.A. Chang, D. Miller, and N. Uretsky (1987). "Interaction of permanently charged analogs of dopamine with the D-2 dopaminergic receptor." Biochem. Pharmacol.36 3903 – 3910.