key: cord-0773924-l0n9rr6s authors: Lin, Simon X.; Curtis, Maurice A.; Sperry, Jonathan title: Pyridine Alkaloids with Activity in the Central Nervous System date: 2020-10-16 journal: Bioorg Med Chem DOI: 10.1016/j.bmc.2020.115820 sha: 25f9e502449bc93b630462f57dd4f190824db622 doc_id: 773924 cord_uid: l0n9rr6s This review discusses all pyridine alkaloids with CNS activity, their therapeutic potential, and the interesting array of sources whence they originate. Central Nervous System (CNS) disease and disorders encompass a vast range of pathologies that includes neurodegenerative disease (e.g. Alzheimer's and Parkinson's), psychiatric conditions (e.g. anxiety, depression and psychosis), epilepsy, multiple sclerosis, neuropathic pain, autism and many more. [1] [2] [3] [4] [5] [6] [7] [8] [9] The disease burden from CNS disorders is enormous. Studies have revealed that neurological disorders were the leading cause of disability-adjusted life years (DALYs; ~280 million) and the second leading cause of deaths (~9.0 million) globally . 10 The absolute number of deaths and DALYs from all CNS-related diseases between 1990 and 2016 have increased by 39% and 15%, respectively. 10 Dementia is one of the largest contributors to neurological DALYs (~10.4%), with at least 50 million people believed to be living with a form of dementia. 10 In 2017, it was estimated that ~792 million people worldwide lived with a form of mental and/or behavioural illness. [11] [12] [13] As the global population surges, the prevalence of CNS-related disease will inevitably increase and as a result, there is a pressing need to develop more effective treatment strategies. [10] [11] [12] [13] Modern medicine has famously relied on natural product-based therapeutics to treat CNS disorders due to the intimate relationship between natural products and the human brain. A recent report estimates that ~84% of approved drugs for the treatment of CNS diseases are natural products or natural product inspired, and 400 clinically approved CNS drugs can be traced back to 20 natural product scaffolds. 14 Pyridines are privileged scaffolds in medicinal chemistry 15 and the nitrogen atom in pyridine plays a crucial role in the pharmacological profile of many drugs that contain this heterocycle. 16 In this account, all pyridine alkaloids that are active in the CNS are detailed, including the array of terrestrial and marine sources from whence they originate, their bioactivity and in some cases, their use as clinically approved therapies. All pyridines, pyridones and pyridiniums are presented, but their benzo-fused (e.g. quinolines) and saturated variants (e.g. piperidines) are not covered herein. Pyridine alkaloids with CNS activity have been isolated from plants, fungi, bacteria, amphibian and marine sources, and some are present in a wide variety of life forms. This review has been structured along these lines accordingly. Tobacco is the dried leaves of Nicotiana tabacum, a plant belonging to the Solanaceae (nightshade) family. [17] [18] [19] [20] The use of N. tabacum by indigenous American Indians dates back ~8000 years, where the plant was smoked in pipe ceremonies for therapeutic and ritualistic purposes. 20 The use of Nicotiana plants was shown to English explorers in 1565 and they began growing the plants commercially in 1612 in what is now Virginia (USA). 20 Tobacco consumption was first introduced to Europe in the late 16 th century for both recreational and medical use, including the treatment of fatigue, abscesses, external wounds, nasal blockages and syphilis. [17] [18] [19] [20] In 1828, the major component responsible for the psychopharmacological response to tobacco use, nicotine (Figure 1 ), was isolated from dried N. tabacum leaves by Posselt and Reimann. 21 The chemical structure of nicotine was later established by Pictet and Crépieux through total synthesis in 1895. 22 Nicotine is also present (albeit in lower amounts) in other genera of the Solanaceae family, such as potatoes, tomatoes and green peppers. 19, 20 Today, N. tabacum is cultivated in over 120 countries worldwide, where it is used to make cigarettes and as the source of nicotine for replacement therapy (NRT). 20 Many reviews on the biological activities of nicotine in animals and a variety of cell systems are available. [17] [18] [19] [20] N N H Me Figure 1 . Nicotine. Upon smoking tobacco, nicotine is carried to the lungs, where it quickly enters the bloodstream and into the brain. 19 The effects of nicotine include heightened arousal, reduced stress and anxiety, energy increase and enhanced pain thresholds. [17] [18] [19] [20] Nicotine has also been shown to improve learning, problem-solving ability, reaction time, selective attention and vigilance in those performing repetitive tasks. 19, 20 These positive reinforcing effects induced from acute nicotine administration are critical factors in tobacco addiction. [17] [18] [19] [20] Nicotine binds to nicotinic acetylcholine receptors (nAChRs), a group of cationic ligand-gated ion channels found in both the peripheral nervous system (PNS) and CNS. [17] [18] [19] [20] There are three nAChR subtypes in the mammalian brain (α4β2-, α3β4-and α7-nAChRs); the most predominant subtype in humans, α4β2-nAChR, is where nicotine displays the highest binding affinity (K i < 1 nM), and is a full agonist at this site. [17] [18] [19] [20] The binding affinity for nicotine at the α3β4-nAChR (K i = 530 nM) and the α7-nAChR (K i = 6290 nM) are much weaker than at the α4β2-nAChR. 20 Nicotine binds as a full agonist, opening the ion channels and stimulating cation influx (e.g. sodium, potassium and calcium) to induce the release of multiple neurotransmitters including dopamine, serotonin (5-HT), norepinephrine, acetylcholine (ACh), γ-aminobutyric acid (GABA), β-endorphins and glutamate into the mesolimbic area, the corpus striatum and the frontal cortex. [17] [18] [19] [20] In particular, the release of dopamine in the mesolimbic system leads to the rewarding effects associated with nicotine use. 19 , 20 Picciotto and Zoli have demonstrated that knocking out the α4β2-subunit gene in rats eliminated the effects of nicotine and the release of dopamine. 20, 23 In related studies, the α3β4-nAChR is implicated in the cardiovascular effects of nicotine 20, 24 and the α7-nAChR is involved in learning, memory and sensory gating. 20, 25 Systematic nicotine administration increases the levels of opioid peptides in the nucleus accumbens, leading to analgesic effects. 20, 26 In vivo experiments have demonstrated that the reinforcing effects of nicotine in mice were eliminated upon co-administration with naltrexone (a non-selective opioid receptor antagonist); mice with their µ-opioid receptor gene removed exhibited reduced analgesic effects upon nicotine administration, suggesting endogenous opioids are involved in the rewarding properties of nicotine. 26 In more recent studies, it was proposed that nicotine also binds to N-methyl-D-aspartate (NMDA) receptors, causing a release of dopamine in the nucleus accumbens. 20, 27 Upon acute nicotine administration in rats, the levels of glutamate were increased in the ventral tegmental area due to the activation of nAChRs located pre-synaptically on the glutamatergic nerve terminals. 27 The increase of glutamate induces firing of dopaminergic neurons and thus elevates dopamine levels. 20, 27 However, when dizocilpine (a non-competitive NMDA receptor antagonist) was co-administered, the effects of nicotine were attenuated, inferring that the nicotine-NMDA receptor interaction is indeed important for the addictive properties of nicotine. 27 Tobacco addiction is a huge global health concern, causing more than 7 million smokingrelated deaths worldwide in 2017. 28 The World Health Organization (WHO) has reported that more than 1.3 billion people smoke tobacco daily, and the cost for the treatment of nicotine related health conditions in the USA is ~US$155 billion per annum. 20 Pure nicotine is used in NRT as a smoking cessation agent to help abate tobacco withdrawal. [17] [18] [19] [20] 29 Nicorette ® was approved as an NRT by the US Food and Drug Administration (FDA) in 1984 as a patch, gum, tablet and spray that enables controlled levels of nicotine to be administered during smoking cessation. 20, 29 There is some evidence that nicotine consumption reduces the risk of developing neurodegenerative diseases (e.g. Parkinson's disease) 30 and mood disorders (e.g. anxiety and depression). 31 Recently, preliminary investigations have reported lower rates of SARS-CoV-2 (COVID-19) infection among smokers. [32] [33] [34] Several natural products structurally related to nicotine have also been isolated from a variety of sources; many reviews on their biological activities are available, thus only key points relevant to the subject of this account are included herein. Nornicotine ( Figure 2 ) is a minor peripheral metabolite of nicotine in various mammal species (e.g. humans, monkeys and rodents). 35, 36 This pyridine alkaloid possesses a demethylated pyrrolidine ring and was extracted from N. glutinosa by Ehrenstein in 1931. 37 Subsequent phytochemical studies revealed that nornicotine is also present in many Nicotiana species and is one of the three most abundant minor alkaloids produced in N. tabacum, alongside anabasine ( Figure 3 ) and anatabine ( Figure 4 ). [38] [39] [40] Kisaki Nornicotine displays significant agonist properties at nAChR subtypes in the CNS. 35, 42 Dwoskin and co-workers have demonstrated that nornicotine evokes the release of dopamine by stimulating nAChRs from the dopaminergic presynaptic terminals in a concentration dependent manner. 42 Both mecamylamine and dihydro-β-erythroidine (nonselective nAChR antagonists) inhibited the [ 3 H]-overflow effect of nornicotine (< 100 µM) on rat striatal slices with preloaded [ 3 H]-dopamine. 42 However, the inhibitory activities of both nAChR antagonists were not observed when the nornicotine concentration was increased (> 100 µM), suggesting a nAChR-mediated mechanism was involved. 42 The effect of (-)-nornicotine on dopamine release was greater than the (+)-enantiomer at concentrations 1, 10 and 100 µM, indicating the mediated nAChR subtype is more sensitive to the (-)-enantiomer. 42 Subsequently, Papke and co-workers demonstrated that nornicotine is more potent at the rat α7-nAChR (EC 50 = 17.4 µmol/L) than at the α4β2-(EC 50 = 375 µmol/L) and α3β4-(EC 50 = 614 µmol/L) subtype receptors. 35 These findings imply that nornicotine contributes to the neuropharmacological effects of tobacco smoking via a nicotinic receptor stimulation. 35, 42 Another of the three abundant minor alkaloids found in Nicotiana plants, anabasine ( Figure 3 ), was first isolated from the toxic Asian plant Anabasis aphylla by Orechoff and Stereochemical integrity is an important pharmacological factor in determining specific biological activities of chiral natural products, and enantiomers often exhibit significant differences in their biological properties. 46 The binding affinities (K i ) and the agonist potencies (EC 50 ) of the two anabasine enantiomers at rat α4β2-and α7-nAChRs were examined. 46 however, (+)-anabasine binds more selectively at the α4β2-nAChR (K i = 0.91 µM) than at the α7-receptor (K i = 3.7 µM). 46 Subsequent toxicity analysis showed that (+)-anabasine (LD 50 = 11 mg/kg) is more toxic than (-)-anabasine (LD 50 = 16 mg/kg), implying that the stereochemistry affects both pharmacological activities and lethality. 46 Anatabine ( Figure 4 ) is the second most abundant alkaloid (~4%) present in Nicotiana species. 36, 48 This alkaloid was first isolated from the leaves of N. tabacum by Spath and Kesztler in 1937, who also established the chemical structure through total synthesis. 49 Anatabine is a pyridine-dihydropyridine structurally related to anabasine. 48 The binding affinities (K i ) of (-)-and (+)-anatabine were evaluated at rat α4β2-nAChR by displacement of [ 3 H]-cytisine radioligand binding using anabasine and nicotine as comparisons. 48 Kem and co-workers demonstrated that (+)-anatabine (K i = 119 nM) exhibited a higher binding affinity than (-)-anatabine (K i = 249 nM), ~8-and 4-fold more potent than anabasine (K i = 910 nM) respectively; 48 the nicotine K i value was found to be ~2 nM. 48 In the same study, the agonist potency (EC 50 ) and ACh stimulation efficacies (I max ) of both anatabine enantiomers at human α4β2-and α7-nAChRs were also examined. 48 At the α4β2-subtype receptor, (-)-anatabine (EC 50 = 2.65 µM; I max = 43.2%) was ~2-fold more efficacious than (+)-anatabine (EC 50 = 0.74 µM, I max = 25.0%); much higher efficacies were observed at the α7-receptors for both (-)-anatabine (EC 50 = 69.7 µM; I max = 113%) and (+)anatabine (EC 50 = 51.8 µM; I max = 105%). 48 These findings suggest that anatabine is a selective and potent ligand at α7-nAChRs. 48 Recent publications have reported that anatabine also lowers the production of β-amyloid in human brain cells 48, 51 and enhances memory and attention dysfunction in rats, 48, 52 suggesting that anatabine may represent a potential therapeutic candidate for dementia disorders such as Alzheimer's disease (AD). 48,51,52 An N-methylated anatabine isomer, N-methylanatabine ( Figure 5 ), was isolated from the leaves of N. tabacum by Spath and Kesztler in 1937; 53 its chemical structure was affirmed through total synthesis in the same study. 53 Although structurally similar to nicotine and anatabine, studies on the biological activities of N-methylanatabine are scarce in the current literature database. The interactions of N-methylanatabine and monoamine oxidase (MAO) A and B were evaluated by Castagnoli and co-workers; 54 the group established that Nmethylanatabine did not produce any significant changes of MAO-A and B activities in rat brain upon administration (data not shown). 54 In 2020, McHugh and co-workers re-examined the therapeutic potential of N-methylanatabine through an electrophysiological characterization test using Xenopus oocytes expressing the human α4β2-nAChRs. 55 The agonist potency (EC 50 ) and the maximal receptor response (I max ) of N-methylanatabine (EC 50 = 6.2 µM; I max = 26%) at the human α4β2-receptor were ~8-and 6-fold less active than nicotine (EC 50 = 0.8 µM; I max = 159%) respectively. 55 N N Me H Figure 5 . N-methylanatabine. Cotinine ( Figure 6 ), often used as a biomarker for tobacco exposure, is the major peripheral metabolite of nicotine with a half-life of 19 -24 hours in many animal species (including humans). [56] [57] [58] [59] Between 70% to 80% of the nicotine consumed by humans is oxidized to cotinine by cytochrome P450 2A6 (CYP2A6) and cytoplasmic aldehyde oxidase. 57 tabacum) 61 and Duboisia hopwoodii. 62 Several reviews and studies on the pharmacological properties of cotinine are available. 56, 58, 59, 63, 64 N N Me O H Figure 6 . Cotinine. Cotinine crosses the blood brain barrier and acts as a low affinity nAChR agonist at brain receptors. 56, 65 Vainio and co-workers demonstrated that cotinine displays weak binding affinity (K i ) and agonist potency (EC 50 ) at rat brain nAChRs labelled with [ 3 H]-epibatidine when compared with nicotine. 65 Cotinine (K i = 3.0 µM; EC 50 = 21 µM) was ~270-fold less active than nicotine (K i = 11 nM; EC 50 = 77 nM) at competing for nAChRs binding sites with [ 3 H]-epibatidine (250 pM) from rat frontal cortex; 65 a similar pattern was observed in rat hippocampus cells (data not shown). 65 These findings indicated that cotinine exhibits weak binding potency to nAChRs in the CNS and mediates its various pharmacological effects upon voluntary nicotine administration. 65 In a subsequent study, the specific receptor subtype that cotinine primarily acts on was examined by Terry Jr and co-workers. 63 More than seventy neurotransmitter receptors, transporters and enzymes (including dopamine D 1-4 , adrenergic α1-2, GABA A-B , glutamate, histamine H 1-3 , muscarinic acetylcholine receptor M 1-5 , opioid, acetylcholine 1-7 , Ca/K/Na channels, nitric oxide, bradykinin, neurokinin and acetylcholinesterase) were screened; it was found that cotinine was relatively inactive (< 50% inhibition at 10 µM) across a wide range of pharmacological targets. 63 However, cotinine (1 µM) significantly enhanced the responses evoked by low concentrations of ACh (< 40 µM) in Xenopus oocytes expressing the human α7-nAChRs, inferring an interaction of cotinine with the α7-receptor. 56, 63 Subsequent behavioural studies also showed that cotinine (1.0 -10 mg/kg) increased the exploration time in rats when it was co-administered (intraperitoneal injection) with donepezil (0.5 mg/kg). 63 Although completely inactive if given alone, cotinine could be considered an adjunctive therapeutic agent to improve the effective dose of cholinergic medications (e.g. donepezil) commonly used for AD and other memory disorders. 56, 58, [63] [64] [65] Echeverria and co-workers demonstrated that cotinine (5 mg/kg) enhances extinction of a contextual fear memory upon acute administration in rats by at least 20%, suggesting the potential therapeutic value for memory improvement and post-traumatic stress disorder (PTSD). 58 Working memory performance and depressive behaviours were improved when cotinine (0.03 -10 mg/kg) was administered in normal and MK801-(an NMDA receptor antagonist used to mimic psychotic symptoms) impaired animal models (e.g. rats and monkeys), 66,67 thus implicating cotinine as a potential therapeutic agent for attention deficit hyperactivity disorder (ADHD) and neuropsychiatric disorders (e.g. anxiety, depression and psychosis). 58, 66, 67 It was also shown that cotinine is efficacious in treating dementia-related memory impairments; 68 cotinine (0.1 µM) reduces β-amyloid (Aβ) neurotoxicity in primary cortical neurons and prevents working memory loss by decreasing the Aβ aggregation and plaque deposition in memory impaired rats. 68 These findings indicate that the neuroprotectivity and the absence of toxicity exhibited by cotinine is due to its agonist property at the α7-nAChRs. 56, 58, [63] [64] [65] [66] [67] [68] Cotinine (1 µM -3 mM) has also been shown to evoke the release of dopamine by stimulating the α7-nAChRs in a calcium-dependent manner in rat striatum; 69 the levels of serotonin and noradrenaline in rat brains increased when cotinine (2 mg/kg) was given in repeated doses, 70 suggesting potential use as an antidepressant agent through activation of α7-nAChRs. 58 Taken together, cotinine displays a safer therapeutic profile than nicotine due to its much longer half-life and a lower risk of abuse. 56, 58, 59, 64, [66] [67] [68] Cotinine induces positive changes in synaptic plasticity which warrant further investigations. In a phytochemical study attempting to discover new minor tobacco alkaloids in Nicotiana plants, a nicotine derivative consisting two 1-methyl-2-pyrrolidinyl moieties attached to a central pyridine scaffold was isolated from dried N. tabacum roots by Crooks and coworkers; 71 structural elucidation was affirmed by total synthesis and this 3,5-dinicotine alkaloid was consequently named DINIC ( Figure 7 ) due to the presence of the two Nmethylpyrrolidine rings. 71 The pharmacological activities of DINIC was investigated by evaluating its ability to displace the binding of Metanicotine (Figure 8 ), also known as Rivanicline, TC-2403 and RJR-2403, is a nicotine alkaloid examined as a potential therapeutic candidate for the treatment of neurodegenerative disorders, including AD. 72, 73 The chemical structure of metanicotine was first assigned by Pinner in 1895 through the degradation of nicotine. 74 In 1953, Wahl reported the natural occurrence of metanicotine as the biological degradation of nicotine upon its isolation from fermented tobacco. 75 Subsequent phytochemical studies revealed that metanicotine is also present in Solanaceae plants (e.g. N. tabacum and D. hopwoodii) 62 The pharmacological properties of metanicotine in the CNS were characterized by Bencherif and co-workers in 1996. 72 (EC 50 = 1.1 µM; E max = 91%), a neuroprotective anxiolytic agent used in the treatment of AD and ADHD; but ~10-fold less potent than nicotine (EC 50 = 100 nM; E max = 113%). 72, 78 In the subsequent in vivo study, metanicotine (3.6 µmol/kg; subcutaneous administration) significantly increased the levels of ACh, dopamine, norepinephrine and serotonin in rat cortex by 190%, 150%, 150% and 170% respectively. 72, 78 The agonist properties of metanicotine were evaluated in Xenopus oocytes expressing the rat α4 and β2 subunits using nicotine and ACh for comparison. At concentrations of 10 and 100 µM, metanicotine produced a higher peak activation (492%) than both nicotine (333%) and ACh (242%), suggesting that metanicotine is a highly potent agonist at the α4β2-nAChR. 72, 78 The physiological and behavioural profiles of metanicotine administration were investigated using the passive avoidance test, water-navigation performance and radial arm maze performance in rats. 72, 78 Metanicotine (0.6 µmol/kg; subcutaneous injection) significantly reversed the amnesic effects induced by scopolamine (0.5 µmol/kg; subcutaneous injection) in rats by ~50%; oral administration of metanicotine (0.3 -3.0 µmol/kg) decreased the mecamylamineinduced amnesia by 40 -50%; both long-(reference) and short-term (working) memory of rats whose forebrain cholinergic projection system impaired by ibotenic acid (10 mg/mL) were improved upon metanicotine administration (0.36, 0.72 and 1.4 µmol/kg) by ~2-to 8fold. 72, 78 Acute toxicity studies indicated that metanicotine is much less toxic than nicotine after single or repeated doses in rats and dogs (LD 50 = 1.8 mol/kg). 72, 78 These preclinical studies suggested that metanicotine is a potent and selective α4β2-nAChR agonist; its safer physiological and more desired behavioural profiles prompted further investigation. 72, 78 Metanicotine (as RJR-2403) was originally developed as an orally available medication for the treatment of neurodegenerative disorders (e.g. AD) by RJ Reynolds Tobacco Co. (USA). 79, 80 However, the development was discontinued in the preclinical phase in 2001 due to undesired adverse effects. 79 Subsequently, metanicotine (as TC-2403) was examined for ulcerative colitis (a chronic inflammatory bowel disease) due to its ability to inhibit the production of Interleukin-8. 80 Clinical trials of metanicotine were advanced into phase II as an enema formulation in 2003; a phase II placebo-controlled trial with 200 ulcerative colitis patients was carried out but unsatisfactory primary efficacy resulted in the discontinuation of the trial in 2005. 79, 80 No published reports on the efficacy or pharmacokinetics of metanicotine in humans is currently available. 80 Alangium chinense (Lour.) is a Chinese deciduous shrub commonly used in traditional Chinese medicine. 81 An alkaloid named anabasamine ( Figure 10 ), possessing a 2,3′-bipyridyl scaffold bonded to a piperidine ring, was isolated from the seeds of Anabasis aphylla (Central Asian shrub) by Mukhamedzhanov and co-workers in 1967. 82 In a subsequent phytochemical study investigating the inhibition of cholinesterases, anabasamine was found to exhibit weak but selective anti-acetylcholinesterase properties. 83 The binding affinity (K i ) of anabasamine at human blood erythrocyte acetylcholinesterase was ~8.6-fold more effective than at horse blood serum butyrylcholinesterase (51 µM vs 440 µM). 83 N N N Me Figure 10 . Anabasamine. Plants belonging to the Leguminosae family, such as Cytisus, Laburnum and Sophora, have been used in traditional medicine for hundreds of years. [84] [85] [86] [87] [88] [89] American Indians are known to have consumed the seeds of L. anagyroides (also known as Cytisus laburnum) for their purgative and emetic effects during rituals; traditional European medicine used alcoholic extracts of Cytisus plants for constipation, migraine and insomnia; the leaves of L. anagyroides were used as tobacco substitute during World War II. [84] [85] [86] [87] [88] [89] In several phytochemical studies examining the biological active secondary metabolite of L. anagyroides, 90,91 the aqueous extract of the seeds was found to contain cytisine ( Figure 11 ), a quinolizidone alkaloid fused to a bispidine ring with absolute configuration later assigned as 1R,5S through stereoselective total synthesis. 92 Subsequent isolation studies have demonstrated that cytisine is present in multiple genera of the Leguminosae family, and is most abundant in the seeds of these plants (between 59% to 80%). [93] [94] [95] Pure cytisine has been used as a respiratory analeptic, diuretic and an insecticide in Europe. [84] [85] [86] [87] [88] [89] Several detailed reviews on the biological properties of cytisine and its therapeutic applications are available. 84 . 96 Cytisine displays a million-fold binding specificity for nAChRs (K i = 0.16 nM) over muscarinic acetylcholine receptors (K i > 400 µM). 84, 96 Cytisine binds with high density in the thalamus of both rat and human brain, where the α4β2-subtype is the predominant nAChRs. 96 Cytisine was subsequently shown to selectively bind at the α4β2-nAChR (K i = 0.17 nM) in rat brain cells, with ~6-fold greater specificity than nicotine (K i = 0.95 nM); 96,97 at the α3β4-and α7-subtype receptors, the binding affinity of cytisine were 840 nM and 4200 nM respectively. 97, 98 However, at 10 µM concentration, the agonist potency of cytisine at α4β2-subtype receptor was only 56% relative to nicotine, inferring that cytisine is a partial α4β2-nAChR agonist. 98 Coe and co-workers 98 reported that the agonist potency of nicotine was reduced by 30% upon co-administration with cytisine, indicating that cytisine partially antagonizes the agonist effect of nicotine. Taken together, these findings established that cytisine is a selective, low-efficacy partial α4β2-nAChR agonist. [84] [85] [86] [87] [88] [89] The effect of cytisine on dopamine release in rat striatum has also been studied. 99 Dopaminergic toxicity caused by oxidopamine (6 µg; a selective neurotoxin that destroys dopaminergic neurons in the brain) was significantly attenuated when the rats were pre-administered with cytisine (2 mg/kg), whilst the amount of dopamine expression in substantia nigra was increased from 30% to 60%. 99 The neuroprotective properties displayed by cytisine may be beneficial in neurodegenerative disorders. [84] [85] [86] [87] [88] [89] 99, 100 In a related study, Picciotto and co-workers observed antidepressant-like properties of cytisine in mouse models. 101 Upon cytisine treatment (1.5 mg/kg), mice showed similar acute antidepressant-like responses in the tail suspension and the forced swim tests when compared with mecamylamine (a nonselective nAChR antagonist that has shown antidepressant effects), inferring that the antidepressant-like efficacy of cytisine is a result of its partial agonist property in which it competitively inhibits ACh signalling through α4β2-nAChRs. 84, 101 Given that cytisine is a selective α4β2-nAChR partial agonist that induces dopamine release and displays antidepressant-like effects, it became a therapeutic candidate as a smoking cessation agent. [84] [85] [86] [87] [88] [89] 100 Cytisine acts as a competitive antagonist in the presence of nicotine, attenuating nicotine's effect at α4β2-nAChR by shielding nicotine-induced dopaminergic activation and therefore limiting the rewarding effect from tobacco consumption; in the absence of nicotine during smoking cessation, cytisine behaves as a partial agonist and increases dopamine levels in the brain, dampening nicotine withdrawal symptoms that include irritability, depression, insomnia and fatigue. [85] [86] [87] [88] [89] 100 Unsystematic clinical trials during the 1960s and 1970s in Central and Eastern Europe suggested that cytisine maintains smoking abstinence superior to placebo [84] [85] [86] 88 and as a result, cytisine was marketed in pill form (Tabex ® ) in 1964 to treat smoking dependence in Bulgaria, East Germany, Poland and Russia. [85] [86] [87] [88] [89] However, cytisine displays low efficacy due to limited crossing of the bloodbrain barrier; [84] [85] [86] [87] [88] [89] 100 it was also reported to cause adverse effects, including nausea, vomiting and sleep disorders. [85] [86] [87] 89 Therefore, the use of cytisine as a smoking cessation aid in humans is not approved by the FDA or the European Union (EU), but is still available in some European countries. [84] [85] [86] [87] [88] [89] 100 Cytisine served as the lead compound for the development of a more efficacious α4β2-nAChR partial agonist for smoking cessation, which led to the development of Varenicline ( Figure 11 ) by Pfizer, approved by the FDA in 2006 for the treatment of nicotine addiction and smoking cessation. [84] [85] [86] [87] [88] [89] 98, 100 Varenicline displays a selective binding affinity (K i = 0.06 nM) and potent partial agonist activity (EC 50 = 3.1 µM) at the α4β2-nAChR in rats; dopamine release in rat brain was reduced by 45% upon coadministration of nicotine and varenicline. 98 Full scale clinical trials indicated that varenicline possesses a similar partial agonist profile at the α4β2-nAChR as cytisine in humans. 84, 85, 98, 100 Nausea and insomnia are the two most common side-effects in the first month of treatment, occurring in ~30% and ~26% of patients respectively, but these mostly subside upon extended administration and/or dose titration. 100 Varenicline displays a greater efficacy than bupropion (an atypical antidepressant and nicotinic receptor antagonist) and NRTs in sustaining abstinence from smoking. 84, 100 It is worth noting that early postlaunch surveillance and meta-analysis advised potential neuropsychiatric and cardiovascular conditions associated with varenicline use, but subsequent clinical studies revealed that the probability of these adverse events is low in patients that do not have a pre-existing psychiatric condition, such as depression, anxiety or schizophrenia. 100 3-Hydroxy-11-norcytisine ( Figure 12 ) is a 5-membered ring skeletal congener of cytisine isolated from the Leguminosae family. 102, 103 It was extracted from the seeds of L. anagyroides in 1989 by Hayman and Gray, 102 but has received little attention in the pharmacology library despite its obvious structural and potential biosynthetic relationship with cytisine. 103 Almost two decades later, Yohannes and Bhatti examined the biological activities of 3-hydroxy-11-norcytisine at rat α4β2-and α7-nAChRs. 103 The binding affinity (K i ) of 3-hydroxy-11-norcytisine was ~35000-and ~153-fold less effective than cytisine at The Ormosia genus of the Leguminosae contains pyridine alkaloids structurally related to cytisine. 104 The roots of O. hosiei contain hosieines A and B, whilst the stems contain hosieines C and D (Figure 13 ). 104 These cytisine-type alkaloids comprise a structurally Abundant throughout China, Huperzia serrata is a plant renowned for its diverse therapeutic applications in traditional Chinese medicine, including the treatment of contusions, swellings, pain and schizophrenia. [105] [106] [107] [108] [109] In a phytochemical study examining the bioactive secondary metabolites of H. serrata, the aqueous extract of dried H. serrata was found to contain huperzine A (Figure 14) , an alkaloid comprising an unusual bicyclo[3.3.1] ring system fused with an ethylidene group and a 2-pyridone moiety. 110 Much effort has focused on the extraction of huperzine A from other plants due to the limited quantity available from H. serrata. [105] [106] [107] [108] [109] Huperzine A has also been isolated from Lycopodiaceae and Selaginellaceae families, but also in poor yield; Phlegmariurus carinatus and P. mingcheensis of the Huperziaceae family have been reported to produce the highest yields of huperzine A. 111, 112 Several detailed reviews on the therapeutic potential of huperzine A are available. 105 The cholinesterases (ChE) are a family of enzymes present in the CNS that break down choline-based esters. 113, 114 Two types of ChE have been characterized; acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). 113 AChE specifically hydrolyzes ACh into choline and acetic acid to avoid over-stimulation in post-synaptic nerves; BuChE (also known as pseudocholinesterase) is a nonspecific ChE that breaks down different choline-based esters. 113, 114 ACh is a neurotransmitter found predominantly in the human brain and has an important role in arousal, attention, memory and motivation. 114 AD patients have lower ACh levels due to age related degeneration of their cholinergic system and/or brain injuries. 113 The cholinergic hypothesis of AD suggests a strategy to treat neurodegeneration is to restore ACh deficiency. 114 Therefore, AChE inhibitors are served as cognition enhancing agents to treat patients with mild to moderate AD, including tacrine, donepezil, physostigmine and rivastigmine. 113, 114 The inhibitory activity of huperzine A has been examined against both AChE and BuChE in vitro. 115 The inhibitory activity on AChE induced by huperzine A (IC 50 = 0.082 µM) was slightly more potent than tacrine (a nonselective ChE inhibitor; IC 50 = 0.093 µM), but ~8-fold weaker than the drug donepezil (a selective AChE inhibitor; IC 50 = 0.010 µM). 115 The inhibitory activity on BuChE induced by huperzine A (IC 50 = 74.43 µM) was ~15 and ~1000fold less potent than donepezil (IC 50 = 5.01 µM) and tacrine (IC 50 = 0.074 µM) respectively. 115 These findings indicated that huperzine A displays high selectivity for AChE over BuChE. [105] [106] [107] [108] [109] 115 Oral administration of huperzine A to rats led to significant inhibition of AChE (16% inhibition at 1 µmol/kg), ~15 and 140-fold more potent than donepezil (9% inhibition at 8 µmol/kg) and tacrine (7% inhibition at 60 µmol/kg), respectively. 115 However, upon intracerebroventricular (ICV) injection, the anti-AChE activity of huperzine A (21% inhibition at 0.066 µmol/kg) was ~3-fold less potent than donepezil (35% inhibition at 0.038 µmol/kg) but ~2-fold stronger than tacrine (11% inhibition at 0.068 µmol/kg), a pattern similar to the in vitro results. 115 The different administration routes clearly affect the bioavailability of huperzine A, with the ICV route facilitating access to the brain. [105] [106] [107] 115 In a subsequent in vivo experiment, the level of ACh in the whole rat brain upon huperzine A administration was also measured. 115 Huperzine A displayed the most prolonged increase in ACh level when compared with donepezil and tacrine, lasting for at least 6 hours after administration. 115 Moreover, the activity of choline acetyltransferase and the level of choline did not change, indicating that the increase of ACh level was not a result of an increase in ACh synthesis. 108, 109, 115 A clear inverse relationship was observed between AChE activity and ACh level, further confirming that the increase was mediated through the AChE inhibition induced by huperzine A. 115 Given that the lack of ACh in the brain is a common symptom in AD patients, the high binding specificity to AChE of huperzine A suggests therapeutic potential. [105] [106] [107] [108] [109] The effect of huperzine A on glutamate-induced neuron toxicity was investigated by Ved and co-workers. 116 Neurons derived from rat embryonic forebrain were treated for 45 minutes with either 100 µM glutamate, 100 nM huperzine A or a mixture of 100 nM huperzine A and 100 µM glutamate. Neuronal cell death caused by glutamate-induced toxicity was found to be ~55% upon treatment with glutamate alone, which was ~50% greater than the group treated with huperzine A alone. Treatment with both huperzine A and glutamate resulted in a ~30% neuron death, suggesting that huperzine A had partially suppressed the glutamate-induced toxicity in neurons. [105] [106] [107] [108] [109] 116 In the same study, the effect of huperzine A on calcium mobilization was also investigated. 116 Neurons derived from embryonic forebrain were exposed to 10 µM of either glutamate or Bay-K8644 (a potent calcium channel agonist), followed by huperzine A (100 nM) and the level of evation was measured. Upon huperzine A treatment, the glutamate-induced calcium mobilization was reduced from 811 to 668 nM, but a minimal effect was observed in the neurons that were exposed to Bay-K8644 (calcium elevation: 505 nM vs 521 nM). 116 These results inferred that huperzine A acts on glutamate receptors to exert neuroprotective properties on glutamate-induced toxicity. [105] [106] [107] [108] [109] 116 Studies examining the effect of huperzine A on the NMDA receptor, an ionotropic glutamate receptor that controls synaptic plasticity and memory function, have also been performed. 117 Huperzine A (100 µM antagonists that bind to the NMDA ion channel) were displaced by huperzine A at K i = 5.6 and 9.5 µM respectively, indicating that huperzine A interacts with NMDA receptor by binding to the ion channel via a non-competitive inhibition mechanism. [105] [106] [107] [108] [109] 117 The NMDAinduced neuronal toxicity was prevented as the survival rate of cell culture increased from 35% to 85% upon pre-treatment with huperzine A. 117 Thus, huperzine A is a potent noncompetitive NMDA ion channel antagonist and exerts neuroprotective properties by blocking NMDA-induced toxicity in neuronal cells. [105] [106] [107] [108] [109] 117 The effect of huperzine A on cognitive enhancement has been examined in several animal models (e.g. rats, chicks and monkeys). [105] [106] [107] [108] [109] 118 Tang and co-workers demonstrated that both working and reference memory deficits induced by scopolamine (a mAChR antagonist commonly used to induce cognitive deficits) were significantly improved upon huperzine A administration. 106, 108, 118 The improvements were more pronounced on working memory than on reference memory. 118 Upon oral administration to aged rats, huperzine A was ~3.5 and 9fold more potent at ameliorating memory impairments than donepezil and tacrine respectively, in agreement with the results reported by Tang and co-workers. 115 In a subsequent study, the cognitive enhancement induced by huperzine A was investigated on memory deficits induced by both scopolamine and GABA in chicks. 119 The results revealed that huperzine A was able to reverse the memory disruptions caused by scopolamine and GABA, suggesting huperzine A improved memory formation processes through both AChE inhibition and GABA receptor antagonism. 108, 119 Cai and co-workers also established that huperzine A (0.01 mg/kg; intramuscularly) significantly improved the spatial working memory impairments induced by catecholamine depleting agent reserpine (0.1 mg/kg; intramuscular) in monkeys. 120 The results revealed an inverted U-shaped dose-response pattern, inferring that huperzine A putatively improves the working memory deficits through an adrenergic mechanism. 108, 120 The LD 50 of huperzine A lies between 2 -4 mg/kg in female rats and > 4 mg/kg in male rats. [105] [106] [107] [108] [109] Huperzine A is less toxic and induces less adverse effects than classic AChE inhibitors that are currently used to treat AD (e.g. donepezil and tacrine). 106 Senna multijuga is a popular ornamental plant in many Brazilian regions because of its brightly yellow coloured flowers. 129 This species belongs to the Senna genus which is renowned for their diverse biological and pharmaceutical properties. 129, 130 The seeds of S. multijuga have been used in Brazilian traditional medicine to treat ophthalmic and skin infections, whilst the leaves are used as a sedative during rituals by indigenous South American tribes. 130 In a phytochemical study, two unusual 2-methyl-3-hydroxy-6-alkyl pyridine alkaloids, 7'-multijuguinone 1 and 12'-hydroxy-7'-multijuguinone 2 were isolated from the leaves of S. multijuga. 129 In a subsequent study, 130 The AChE inhibitory activity of 1 -7 were investigated in both isolation reports by standard bioautography (TLC assay) and microplate tests. 129, 130 Using physostigmine (a reversible AChE inhibitor) as the positive control, the preliminary bioautography studies revealed that all the pyridine alkaloids 1 -7 shown in Figure 15 were ~3-to 120-fold less active than physostigmine. 129, 130 A microplate test confirmed the bioactivity of the natural products as the assay data indicated weak anti-AChE activity (13% -52% inhibition at 350 mM) when compared with physostigmine (87% inhibition at 350 mM). 129, 130 The data suggests that the constitution of the alkyl chain at C6 is vital and the presence of the C7'-OH appears to be important for higher levels of inhibition. 129,130 Euphorbia is a genus of flowering plants in the Euphorbiaceae family renowned for their use in traditional Chinese medicine, such as the treatment of skin diseases, gonorrhoea, migraine and intestinal parasites. 131, 132 In a phytochemical study on E. prolifera, 133 Nitric oxide (NO) is a membrane-permeable gas that acts as a neuromodulator at the synaptic junctions. 135 High levels of NO will result in oxidative stress and hence neuronal inflammation in the CNS, a condition thought to play a role in neurodegenerative diseases (e.g. AD and Parkinson's disease). 135 The neuroprotective properties of euphorbialoids A -J and analogues 1 -2 were investigated by evaluating their inhibitory activity on lipopolysaccharide (LPS)-induced NO production in murine microglial BV-2 cells. 133 Euphorbialoids A -J and their unnamed congeners 1 and 2 inhibited the production of NO to varying degrees in microglial BV-2 cells when compared with 2-methyl-2-thiopseudourea sulfate (positive control; 13.2 µM), 133 suggesting that these alkaloids represent therapeutic leads for the treatment of neurodegenerative diseases. Ceropegia of the Apocynaceae family is a genus of plants that are native to Africa, Southern Asia and Australia. 136 The phytoconstituents of Ceropegia species have been routinely used in Ayurvedic medicine to treat gastric disorders, dysentery, hepatic disease, urinary tract diseases and diarrhoea. 136 In one phytochemical study, cerpegin ( Figure 17 ), a pyridine alkaloid consisting a 2-pyridone fused with a 2-furanone ring was isolated from the fleshy stem of C. juncea. 137 Cerpegin has been shown to possess many diverse biological and pharmacological activities, such as analgesic, tranquilizing, anti-inflammatory, anti-ulcer and anti-cancer properties. 136 Relevant to the subject of this review, cerpegin was found to exhibit dose-related analgesic properties against acetic acid-induced writhing in mice. 136, 138 No automatic or behavioural changes were observed up to a dose of 20 mg/kg; however, excitation, respiratory paralysis and later convulsions was produced by cerpegin with dosing greater than 400 mg/kg. 138 Tranquilizers act on the CNS to moderate brain activities and relieve hyperactive nerves to treat patients with anxiety, sleeping disorders and psychoses (e.g. schizophrenia). 136 Cerpegin displays tranquilizing properties through an unknown mechanism, but it has been suggested that this furanopyridone alkaloid competitively antagonizes both dopamine (D 2 ) and serotonin (5-HT) receptors. 136 Figure 17 . Cerpegin. Many pharmacologically active compounds have been detected from Brazilian Strychnos, a genus of flowering plants belonging to the Loganiaceae family. 139 In a methodology study attempting to modify the isolation procedure of biologically active alkaloids from Strychnos species, a monoterpene tri-substituted cyclopentapyridine alkaloid named cantleyine ( Figure 18 ) was isolated from the aqueous extract of S. trinervis roots. 139 The effect of cantleyine against CaCl 2 on voltage dependent calcium channels was investigated. 139 It was reported that when the concentration of cantleyine was increased from 120 to 490 µM, CaCl 2 -induced maximal smooth muscle contraction decreased from 90% to 45% in guinea pig ileum. These results suggested that cantleyine induces a reversible but nonselective spasmolytic action on the vascular and visceral smooth muscles due to the inhibition of Ca 2+ influx through voltagegated Ca 2+ channels, similar to that exhibited by common calcium channel inhibitors such as verapamil and nifedipine. 139 The furopyridine alkaloid haplophyllidine ( Figure 19 ) was isolated from the seeds of Haplophyllum perforatum by Shakirov and co-workers. 140 Haplophyllidine is a potent CNS depressant and synergizes the effects of narcotic/hypnotic drugs in mice, rats and rabbits. 140, 142 Simultaneous subcutaneous injection (s.c.) of luminal (< 72.5 mg/kg) and haplophyllidine (< 130 mg/kg) produced strong neurological deficits; upon co-administration with hexanal (40 mg/kg) or chloral hydrate (230 mg/kg), haplophyllidine (2 -40 mg/kg) prolonged sleep duration by 50% and 70% respectively. 142 In a subsequent study, the antagonism properties of haplophyllidine against analeptic agents, including corazol, camphor, strychnine and caffeine were studied in mice. 143 At 25 mg/kg, haplophyllidine inhibited 50% of corazol-induced convulsions and completely eliminated the convulsion effects at 75 mg/kg; co-administration of camphor (525 mg/kg) and haplophyllidine (125 -150 mg/kg) lowered the convulsion effects by 50% and lethality by 100%; haplophyllidine (100 mg/kg) produced complete mortality protection against strychnine (1.44 mg/kg), whilst at 250 mg/kg reduced death by 80% against caffeine (210 mg/kg). 143 These studies suggested that haplophyllidine exhibits pronounced sedative and anti-analeptic properties. 140, 142, 143 Gentianine (Figure 20) , an alkaloid comprising a 3-vinylpyridine fused with pyranone, was isolated from Gentiana kirilowii and its chemical structure was established upon total synthesis. 144, 145 Gentianine has also been extracted from many other Gentiana and Swertia Initial biological observations of gentianine reported that the alkaloid acts as a CNS stimulant in low doses, but becomes paralytic in higher amounts. 146, 147 The antipsychotic profile of gentianine was demonstrated by Bhattacharya and co-workers in 1974. 146 Gentianine (10 -20 mg/kg; i.p.) diminished spontaneous motility and produced sedation/ptosis in rats; when dosage was increased (50 -100 mg/kg; i.p.), hind-limb paralysis and catalepsy was observed, inferring that gentianine exhibits an antipsychotic activity. 146 Subsequent pharmacological studies evaluated the effects of gentianine on hexobarbital-induced hypnosis, amphetamine toxicity and lysergide-induced symptoms in rats. 146 Gentianine significantly potentiated the sleeping time induced by hexobarbital (100 mg/kg; i.p.) by 80%; amphetamine (20 mg/kg; i.p.) toxicity was reduced by 88% and amphetamine-induced (10 mg/kg; s.c.) stereotypic behaviours (including continuous sniffing, biting and compulsive gnawing) were blocked; gentianine completely inhibited lysergide-induced symptoms such as piloerection and tremors in mice. 146 The effects of gentianine on mice in a rotarod performance test, conditioned avoidance response and induced aggressive behaviour were examined. 146 Gentianine inhibited the ability of trained mice to remain on a rotating rod by 80%; it selectively blocked the avoidance response to the conditioned stimulus (buzzer) without affecting the escape response to the unconditioned stimulus (electric shock); aggressive behaviours induced by foot-shocks were inhibited by 60%. 146 The effects of gentianine on morphine analgesia, the anticonvulsant action of diphenylhydantoin and pentylenetetrazol-induced convulsions have also been examined in rats. 146 Upon gentianine administration, the analgesic activity of morphine (2 mg/kg; i.p.) was increased by 150%; the anticonvulsant activity of diphenylhydantoin (2.5 mg/kg; i.p.) was potentiated by 60%; pentylenetetrazol (70 mg/kg; s.c.)-induced convulsions were inhibited by 70%. 146 The LD 50 of gentianine was found to be 276 mg/kg; the alkaloid is thought to possess only a moderate to low order of toxicity due to the lack of obvious toxicity present upon prolonged i.p. administration (20 mg/kg; q.d.; 21 days) in rats. 146 Gentianine exhibits a broad range of interesting neuropharmacological properties that warrant further investigation with modern biological testing. 146, 147 3. Amphibian-derived Native to Central and South America, the poison dart frog Epipedobates anthonyi is renowned for its brightly coloured body and high toxicity. [148] [149] [150] Indigenous South American Indians apply the skin secretions to poison the tips of blow-dart weaponry for hunting and self-defence against large predators. 149 In a study examining the chemical constituents of the skin secretions, the methanolic extract of 750 E. anthonyi frog skins was concentrated and dried in vacuo to give a crude alkaloid fraction that upon purification gave (+)-epibatidine ( Figure 21) , a structurally unprecedented alkaloid comprising a 2-chloropyridine bonded to an exo-7-aza-bicycloheptane. 150 The absolute configuration of (+)-epibatidine was assigned to be 1R,2R,4S through stereoselective total synthesis. 151, 152 A detailed review on the bioactivity of epibatidine and its therapeutic potential has recently been published, 149 thus only key points will be included herein. Amphibians produce biologically active alkaloids that have aided the development of lead compounds for the treatment of various pathologies. 149 Therefore, upon the isolation of (+)epibatidine, efforts to decipher its biological mechanism were forthcoming. (+)-Epibatidine was examined in both the Straub-tail response (generally used as a positive indicator of morphine-like mechanism) and heat nociception activity. 150 In these experiments, (+)-epibatidine was reported to be a potent anti-nociceptive, ~200 times more effective than morphine (Straub-tail response ED 50 : 0.020 vs 10 mg/kg; hot plate analgesia ED 50 : 0.005 vs 1 mg/kg, respectively). 150 Epibatidine was shown to have a non-opioid mode of action due to the binding at opioid receptors being ~9000-fold less potent than morphine (IC 50 = 8800 nM vs 1 nM, respectively). Moreover, the anti-nociceptive activity of epibatidine was almost unaffected when the nonselective opioid antagonist naloxone was pre-administered to rats, proving that epibatidine does not exert its analgesic properties through the opioid receptors. [148] [149] [150] However, subsequent studies inferred that synthetic (±)-epibatidine did not generate a Straub tail response in rats, suggesting previous results were linked to high-dose syndrome or alkaloid contamination. [153] [154] [155] Regardless, epibatidine clearly displayed a nonopioid mode of action, and more biological investigations were subsequently performed to reevaluate the biological properties of this compound. 149 In 1993, Qian and co-workers reported that epibatidine is a potent nAChR agonist. 153 A group of mice was divided in half and administered with either (-)-nicotine (5 mg/kg; positive control) or (+)-epibatidine (20 µg/kg). 153 Both groups had a rapid anti-nociceptive response; the control group reached maximum response at 2 min and lasted for 10 min, whilst the epibatidine group took 5 min to reach a maximum response that lasted for 20 min. 153 In the same study, if mice were preadministered the nAChR antagonist mecamylamine (1 mg/kg) and then treated with (+)epibatidine, the anti-nociceptive dose was 289.2 µg/kg, ~22-fold greater than the group that did not receive mecamylamine (13.6 µg/kg), 153 providing further evidence that epibatidine is exerting its effects via the nAChRs. Based on these results, a radioligand binding assay was performed to investigate the binding affinity of epibatidine at nAChRs and a range of other neurotransmitter receptors. The study showed that the IC 50 153 However, at 10 µM, epibatidine failed to displace any specific ligands at a range of other neuronal receptors (i.e. GABA A , benzodiazepine, dopaminergic, serotonergic, adrenergic, glutamate/aspartate, neurokinin, bradykinin, cholecystokinin and calcium gene-related peptide). 153 These detailed studies provide very strong evidence that epibatidine exerts its anti-nociceptive effects through nAChR agonism. 148, 149, 153 There are seventeen nAChR subtypes in vertebrates and sixteen in humans, with three different receptor sites abundant in the mammalian brain (α4β2-, α3β4-and α7-nAChR). 148, 149 A series of experiments were conducted to investigate the binding selectivity of epibatidine at these different nAChR subtypes. 155 Using (-)-nicotine as the control, Gopalakrishnan and coworkers re-evaluated the binding affinity (K i ) and the agonist potency (ED 50 ) of (+)epibatidine at four different nAChR subtypes (α4β2-, α3β4-, α7-and α1β1δγ-subunits) to establish receptor binding specificity (Table 2) . 155 The α1β1δγ-nAChR was also selected as part of the investigation as it is a commonly expressed nAChR at neuromuscular junctions. These studies showed that while (+)-epibatidine and (-)-nicotine are both α4β2-nAChR agonists, fundamental differences were apparent. The binding affinity and the agonist potency of nicotine are higher at the α4β2-nAChR than the other receptor subtypes, whilst epibatidine displays strong binding affinity and agonist activity at all four nAChRs, with some slight specificity at the α4β2 and α3β4 subtypes. 154, 155 Overall, epibatidine is a much stronger nAChR agonist than nicotine. In a separate study, Rupniak and co-workers reported that both (+)-and (-)-epibatidine have very similar binding affinity at the α4β2-nAChR subtype (K i = 0.04 vs 0.06 nM) and near identical anti-nociceptive properties (IC 50 = 0.10 vs 0.24 nM) in rats. 154 These interesting results indicated that the absolute stereochemistry of epibatidine has a negligible effect on binding with the α4β2-nAChR, nor its pharmacological profile. 154 The same group also discovered that epibatidine binds at the muscarinic acetylcholine receptor (mAChR) M 1 -subtype at high doses. 148, 154 There are five mAChR subtypes (M 1 -M 5 ) in humans, but only the M 1 -subtype has been found in the brain. 156 Rupniak and co-workers investigated the binding affinity of epibatidine at M 1 -mAChR and nAChRs using a radioligand binding assay. 154 inhibition at 10 µM, respectively). 154 The Relative Affinity Ratio was also calculated for both (+)-and (-)-epibatidine to predict their antagonist/agonist efficacy at the M 1 -mAChR. 157 The ability of (+)-epibatidine to displace the mAChR antagonist [ 3 H]-N-methylscopolamine (NMS) and the mAChR agonist [ 3 H]-oxotremorine-M (oxo-M) was measured as the Apparent Affinity Constant (K app ) using the radioligand binding assay; K app (NMS) was then divided by the K app (oxo-M) to provide a measurement of antagonist/agonist efficacy. 154 The experiment was also repeated for (-)-epibatidine and the results were compared to those of carbachol (a potent nonselective mAChR agonist) and atropine (a potent nonselective mAChR antagonist). These results are summarised in Table 3 . Table 3 . M 1 -mAChR binding profile of (+)-and (-)-epibatidine vs carbachol and atropine. The Relative Affinity Ratios of both (+)-and (-)-epibatidine are significantly lower than that of the mAChR agonist carbachol, indicating that both epibatidine enantiomers are not agonists at the M 1 -mAChR. 149, 157 The affinity profile of (+)-epibatidine suggested that it has a similar affinity as the classic mAChR antagonist atropine (Ratio = 4.2 vs 2.1), whereas (-)epibatidine resembles a partial mAChR antagonist. 154, 157 Given that (+)-and (-)-epibatidine is a potent nAChR agonist and a moderate M 1 -mAChR antagonist, this natural product was initially considered a promising lead for the non-opioid treatment of pain. 148, 149 However, no in vivo experiments have been performed in non-rodents due to its low therapeutic index in rats (LD 50 < 125 nmol/kg; intravenously). 158 The toxicity of epibatidine is caused by its potent, non-selective binding at the nAChRs. 148, 149 Because these nicotinic receptors are widely distributed within the human body (e.g. brain, heart and smooth muscle) and are involved in many neurological and physiological conditions (e.g. schizophrenia, Parkinson's disease, AD, muscular paralysis, hypertension and seizures), epibatidine binding would lead to many off-target effects at important districts. [148] [149] [150] As a result, epibatidine itself is no longer investigated for therapeutic development, but its unique scaffold provides a platform for the development of safer therapeutic agents through medicinal chemistry studies. 148,149,154 Phantasmidine ( Figure 22 ) is an epibatidine congener isolated from Anthony's poison arrow frog (E. anthonyi) by Fitch and co-workers. 159 Due to the small quantity obtained (20 µg), the chemical structure was tentatively inferred from the limited spectroscopic data (MS, IR and NMR) and analogy to epibatidine. 159 The absolute configuration of phantasmidine was later established through total synthesis and it was revealed that the compound exists as a 4:1 scalemic mixture of (2aR,4aS,9aS) and (2aS,4aR,9aR) enantiomers. 160 Initial investigations by Fitch and co-workers showed that phantasmidine displayed a specific agonist activity at nAChRs expressing the β4-subunits (data not shown), suggesting that the compound might possess a different nAChR subtype specificity to epibatidine. 159 The group later conducted a more detailed pharmacological investigation on phantasmidine to elucidate its binding selectivity and agonist activity at the nAChRs. 160 The binding affinities and agonist potency of (2aR, 4aS, 9aS)-phantasmidine, (2aS, 4aR, 9aR)-phantasmidine, racemic samples of phantasmidine and epibatidine (positive control) to nAChRs (α4β2-, α3β4-and α7-subtypes) are shown in Table 4 . The results revealed that the binding affinity and the agonist activity of (±)-phantasmidine at nAChRs were ~2-to 50-fold weaker than (±)-epibatidine, whilst the binding affinity and the agonist activity of the (2aR,4aS,9aS)-enantiomer were ~2-to 45-fold greater than the (2aS,4aR,9aR)-enantiomer. 160 Interestingly, these data indicate that phantasmidine is selective for the α4β2-subtype, contradicting the initial results reported by Fitch and co-workers. 159 Toxicity investigations showed that the LD 50 values of (±)-phantasmidine and the (2aR,4aS,9aS)-enantiomer were 270 and 72 µg/kg respectively, at least 10-and 3-fold less toxic than (±)-epibatidine (LD 50 < 26 µg/kg), whilst the (2aS,4aR,9aR)-isomer was much less toxic, producing similar effects at LD 50 > 10 mg/kg. 160 It is clear that the stereochemistry of phantasmidine plays an important role in nAChR binding and toxicity. 160 Similar to epibatidine, phantasmidine itself is also no longer considered as a potential therapeutic candidate due to its low therapeutic index, however, its agonist selectivity at the neuronal α4β2-nAChR makes it a useful pharmacologic tool for the investigation of specific nAChR subtypes. 159,160 Noranabasamine ( Figure 23 ) is a des-N-methyl analogue of anabasamine that was isolated from the skins of Columbian poison dart frog Phyllobates terribilis. 163 Due to its structural similarity with nicotine and anabasine (Nicotiana alkaloids), it has been suggested that noranabasamine might highly also possess agonist activity at nAChRs; 162 however, no biological evaluation on noranabasamine is currently available. Entomogenous deuteromycetes are a taxonomically diverse group of imperfect fungi known to produce biologically active secondary metabolites due to their complex association with insect hosts. 164, 165 In a study investigating the CNS-related secondary metabolites of entomogenous fungi, militarinone A ( Figure 24 ) was isolated from the mycelial extract of Paecilomyces militaris strain RCEF0095. 164 Militarinone A is a 1,4-dihydroxy-2-pyridone alkaloid comprising a cis-1,4-dihydroxycyclohexane moiety and a polyene side chain. In subsequent studies, two structurally related 4-hydroxy-2-pyridone alkaloids, (+)-Ndeoxymilitarinone A 165 and farinosone A 166 (Figure 24 ), were isolated from P. farinosus strains RCEF0097 and RCEF0101 respectively. The neurotrophic properties of militarinone A, (+)-N-deoxymilitarinone A and farinosone A were investigated by examining their potential to stimulate neuronal differentiation in PC-12 cell lines. [164] [165] [166] The cell viability data showed that all three compounds exhibited potent neuritogenic activities when compared with an endogenous glycoprotein, the nerve growth factor (positive control; induces 80% neurite outgrowth at 50 ng/mL). Militarinone A produced 80%, 70% and 30% neurite outgrowth at 33, 10 and 3.3 µM respectively; 164 (+)-N-deoxymilitarinone A displayed a weaker neurotrophic activity than militarinone A, inducing a neurite outgrowth of 51% and 12% at 100 and 33 µM respectively, which inferred that the hydroxy group at N1 is essential for neuronal differentiation and survival; 165 farinosone A exhibited 70% and 40% neurite outgrowth at 50 and 20 µM respectively. 166 These findings showed that militarinone A, (+)- interesting therapeutic candidates for the prevention of neuronal decline. [164] [165] [166] Arthpyrone C Biological investigations on the secondary metabolites extracted from sponge-derived fungi led to the discovery of arthpyrone A and C ( Figure 24 ) from Arthrinium arundinis strain ZSDS1-F3 derived from Phakellia fusca. 167 These alkaloids comprise a 1,4-dihydroxy-2pyridone bonded to a cyclohexanol moiety and a decalin ring system. One previously discovered 1,4-dihydroxy-2-pyridone alkaloid, N-hydroxyapiosporamide (Figure 24 ), was also isolated. 168 Arthpyrone C displayed pronounced anti-AChE activity (IC 50 = 0.81 µM) when compared with classic AChE inhibitor tacrine (positive control; IC 50 = 0.48 µM), whilst arthpyrone A and N-hydroxyapiosporamide both exhibited moderate AChE inhibition (IC 50 = 47 and 39 µM, respectively). 167 These results suggest that arthpyrone C is an interesting lead compound for the development of treatments for neurodegenerative conditions. The 4-hydroxy-2-pyridone alkaloid paecilomide (Figure 24 ) was isolated from P. lilacinus by Takahashi and co-workers in 2013. 169 Two possible rotational forms, paecilomide A and B, were observed in the NOESY contour map between both H6 and H5' as well as between H7 and H4', due to the possibility of free rotation of the C7-C4' sigma bond. 169 Paecilomide displayed potent AChE inhibitory activity (57.5% inhibition at 25 µL) when compared with physostigmine (98% inhibition at 10 mg/mL), suggesting therapeutic potential. 169 Acromelic acids A and B ( Figure 25 ) are neuroexcitatory amino acids first isolated from Clitocybe acromelalga by Konno, Shirahama and Matsumoto in 1983. 170 C. acromelalga is a toxic Japanese mushroom that elicits symptoms similar to acromelalgia and erythromelalgia upon ingestion. [170] [171] [172] [173] The fresh fruiting bodies of C. acromelalga contains acromelic acids A and B; 170 stereoselective total synthesis of acromelic acid A affirmed the proposed structures and assigned the absolute configuration of both natural products in the process. 172 Acromelic acids A and B are classified as kainoids, analogues of kainic acid (Figure 25 ), a natural product found in Digenea simplex that is related to the excitatory neurotransmitter glutamate. 170, 172, 174 Kainic acid acts as a potent agonist at glutamate receptors and is used as a neurodegenerative agent in neuroscience research to mimic glutamate excitotoxicity in neurodegenerative models (e.g. AD and epilepsy). 174 Therefore, the neuroexcitatory properties of acromelic acids A and B were investigated. 171, 173, 175, 176 Electrophysiological tests were performed to examine the neurological potency of acromelic acid A at the crayfish neuromuscular junction and mice spinal cord. 173, 175 These studies showed that acromelic acid A is a powerful glutamate receptor agonist that exhibits significant depolarizing action in central neurons and ionotropic neuroexcitatory activity in both muscle fibre and brain, with a potency ~100-fold greater than kainic acid. [171] [172] [173] [174] Similar results were obtained on the frog spinal cord (data not shown). 176 In 1990, Nozoe and co-workers reported the isolation of acromelic acid C ( Figure 25 ) from C. acromelalga. 177 The chemical structure and absolute configuration of acromelic acid C was established by detailed NMR spectroscopy and by analogy to acromelic acids A and B. 177 The neurotoxicity of acromelic acid C was investigated via intraperitoneal injection into a mouse; the median lethal dose was 10 mg/kg, slightly higher than acromelic acids A and B (7 and 8 mg/kg respectively). 177 Two neuroexcitatory amino acids, acromelobic acid and an acromelobic acid analogue 1 ( Figure 25) Figure 25 . Acromelic acids A -C, acromelobic acids and kainic acid. Fusarium is a genus of filamentous fungi that produce diverse biologically active secondary metabolites, including the mycotoxin fusaric acid (Figure 26 ). [180] [181] [182] [183] [184] [185] Fusaric acid (5butylpicolinic acid) was first isolated from F. heterosporium in 1934 by Yabuta 181 and was found to exhibit weak antimicrobial properties. [180] [181] [182] [183] More than three decades later, fusaric acid was found to be a potent inhibitor of dopamine β-hydroxylase (DBH), 183 In vitro studies suggested that fusaric acid is a very potent inhibitor of DBH, ~10-fold more active than picolinic acid. 184, 185 The percentage inhibition of DBH induced by fusaric acid at concentration of 0.005, 0.05, 0.5 and 1 µM was 20%, 58%, 89% and 92%, respectively; whilst picolinic acid at 0.5 and 5 µM inhibited DBH by 55% and 83% respectively, inferring that the butyl side chain at C5 increases the DBH inhibitory effects. 184 Inhibition of DBH by fusaric acid was found to be uncompetitive and completely reversible, indicating that this compound affects the enzyme-substrate complex. 184 The binding specificity of fusaric acid was studied by examining its inhibitory activity on other oxidoreductases. 184 At 10 µM, fusaric acid did not inhibit MAO, tyrosine hydroxylase or aldehyde hydrogenase, indicating that the compound has a high degree of binding specificity for DBH. 184 A study examining the dopamine and norepinephrine levels in rat brains after a single dose administration of fusaric acid (100 mg/kg; intraperitoneal injection) have been performed. 182, 184 The norepinephrine expression in rat brains decreased significantly from 0.22 to 0.10 µg/g three hours after fusaric acid administration, whilst the level of dopamine remained constant (from 0.42 to 0.40 µg/g). The significant decrease in the level of norepinephrine without a corresponding decrease in dopamine expression in rat brains implies that fusaric acid inhibits DBH and thus reduces the production of norepinephrine. 184 These investigations showed that fusaric acid is a potent DBH inhibitor both in vivo and in vitro, 182, 184 which led to clinical trials of fusaric acid in patients with mania and depression being conducted in 1974. 185 The levels of 3-methoxy-4-hydroxyphenylglycol (the major metabolite of norepinephrine) in the cerebrospinal fluid was reduced by ~25% when patients were administered fusaric acid compared to placebo, whilst the mean concentration of homovanillic acid (the major metabolite of dopamine) almost doubled, indicating an accumulation of dopamine in the brain. 185 However, adverse behavioural changes were observed in patients with stage III mania and/or other severe pre-existing psychotic features; a few patients with mild hypomanic symptoms showed no change or slight improvements, suggesting that the effects of fusaric acid relate to the pre-existing clinical state of the patients. 182, 185 The results from these clinical trials indicated that a reduction in norepinephrine by DBH inhibition did not improve manic symptoms and therefore fusaric acid was not approved for therapeutic use. 185 Subsequent in vivo investigations have demonstrated other neurochemical effects of fusaric acid in the brain. 182, 186 In addition to inhibiting the biosynthesis of norepinephrine, fusaric acid was also found to alter the levels of melatonin, serotonin, tyrosine, tryptophan and luteinizing hormone. 186 However, inconsistent results (data not shown) were obtained from these different studies which inferred that the neurochemical effects of this mycotoxin vary with species (i.e. rodents, rabbits and swine). 186 Although fusaric acid has been shown to cause behavioural changes in test subjects, it is primarily used as a research tool as its mode of action in the brain is still not fully understood. 182, 186 Another potent DBH inhibitor named phenopicolinic acid ( Figure 26 ) was isolated from Paecilomyces sp. strain AF2562 by Nakamura and co-workers in 1975. 187 The DBH inhibitory activity of phenopicolinic acid was reported to be nearly double that of fusaric acid (IC 50 = 0.039 vs 0.05 µM). 187 In vivo experiments demonstrated that phenopicolinic acid (50 mg/kg; oral administration) reduced the blood pressure of hypertensive rats by 21%, 16%, and 23% in 1, 3 and 5 hours respectively; the LD 50 of phenopicolinic acid was ~350 mg/kg upon intraperitoneal injection. 187 Filter feeding marine species, such as sponges, host microorganisms that produce biologically active secondary metabolites with therapeutic potential. 188, 189 The aqueous extract of an Dung beetles roll faecal matter into balls for ease of transport and subsequent storage as a food source. These dung balls contain unique microorganisms that have been studied as a source of structurally unique secondary metabolites with biological active properties. 191 In one such study, Oh and co-workers isolated coprismycins A and B ( Figure 28 ) from a SNA015 strain of a Streptomyces species found in the dung balls of indigenous Korean dung beetle Copris triparititus. 191 Coprismycins A and B are densely substituted 2-aryl-3thiopyridine alkaloids that differ in their aldoxime geometry. Coprismycins A and B are structurally related to 1-methyl-4-phenyl-1,2,3,6tetrahydropyridine (MPTP), a metabolic precursor to the neurotoxin 1-methyl-4phenylpyridinium (MPP + ). 191 MPP + is known to induce neuropathological changes by killing dopamine-producing neurons in the pars compacta of the substantia nigra. Neurodegenerative diseases (e.g. Parkinson's disease) are often associated with low levels of functional dopaminergic neurons in the brain. Due to their structural similarity to MPTP, the coprismycins were evaluated against human-derived SH-SY5Y cells that express dopaminergic neuron markers. 191 SH-SY5Y cells were treated with three concentrations (1.0, 2.5 and 5.0 µM) of either coprismycin A or B for 24 hours. The assay revealed 100% cell viability, suggesting that coprismycins A and B are not toxic to dopaminergic neurons. 191 In a subsequent experiment, SH-SY5Y cells were pre-treated with different concentrations of either coprismycin A or B, followed by MPP + (800 µM). 191 The cell viability after MPP + exposure was improved from 63.0% to 69.7%, 74.2% and 80.3% upon pre-treatment of coprismycin A at concentration of 1.0, 2.5 or 5.0 µM respectively, suggesting that the MPP +induced neurotoxicity was suppressed. 191 Coprismycin B exerted similar neuroprotective properties, producing cell viability of 76.4% and 88.4% at 1 and 2.5 µM respectively; however, at 5 µM the cell viability dropped to 69.8%, indicating neurotoxicity of coprismycin B at higher concentrations. 191 Nonetheless, these findings have provided strong evidence to show that both coprismycins A and B exhibit pronounced neuroprotective properties that warrant further investigation. Two 2,2'-bipyridyl alkaloids, SF2738 D 1 and SF2738 F 2 ( Figure 28 ) were discovered in 1994 from a culture of Streptomyces sp. in Japan (Yokohama, Kanagawa). 192 Some years later, these natural products were also isolated from the bacterial strain SNA015 of Streptomyces sp. found in C. triparititus. 191 The structural similarity of SF2738 D and SF2738 F with coprismycins A and B led to an investigation of their neuroprotective properties. 191 Upon treating SF2738 D and SF2738 F to SH-SY5Y cells, there was no loss in cell viability. Moreover, the cell viability of SH-SY5Y cells increased (data not shown) when they were treated with SF2738 D and SF2738 F prior to exposure to MPP + , suggesting they have a similar neuroprotective effect to the coprismycins and are themselves also promising therapeutic leads for treating neurodegenerative disorders. 191 A potent neurotoxin was initially discovered from the marine worm Rhynchocoela by Bacq 193 in 1936 and was found to exhibit significant toxicity when injected into crabs, causing convulsions, flaccid paralysis and eventually death. 194 The structure of this compound remained unknown for many years as attempts at crystallization using standard alkaloidal precipitants were unsuccessful. Three and a half decades later, Kem and co-workers isolated the same compound from the hoplonemertine Paranemertes peregrine and elucidated its chemical structure by both total synthesis and comparison with previously published literature data. 194 Named anabaseine (Figure 29 ), this nemertine alkaloid is a double bond isomer of anatabine, possessing a tetrahydropyridyl ring with an internal imine double conjugated with the pyridine moiety. 46, 194, 195 Subsequent studies reported that anabaseine is the primary compound found in the poison glands of Messor and Aphenaenogaster ants; 46, 195 anabaseine has not yet been detected in plants. 46, 195 Anabaseine has been shown to stimulate the release of ACh and norepinephrine from rat brains upon injection; 195 several synthetic anabaseine-related analogues also displayed significant cognitive enhancement and avoidance behaviours. 46, 195 As a result, the pharmacological properties of anabaseine were examined Kem and co-workers on rat α4β2and α7-receptors, the two predominant nAChRs in mammalian CNS. 195 The binding affinity (K i ) and the agonist potency (EC 50 ) of anabaseine (K i = 0.032 µM; EC 50 = 4.2 µM) at the α4β2-nAChR were ~8-fold less active and ~3-fold more potent than nicotine (K i = 0.0041 µM; EC 50 = 14 µM) respectively; 195 at the α7-nAChR, anabaseine (K i = 0.058 µM; EC 50 = 6.7 µM) was ~7-fold more potent than nicotine (K i = 0.40 µM; EC 50 = 47 µM). 195 These findings indicated that anabaseine exhibits different binding selectivity and agonist potency at the nAChRs, inferring the significance of structural conformation with nicotinic receptor recognition sites. Further studies are required to fully understand the impact of the subtle structural differences between this neurotoxin and the tobacco alkaloids. 195 A Convenient Racemic Synthesis of Two Isomeric Tetrahydropyridyl Alkaloids: Isoanatabine and Anatabi Isoanatabine ( Figure 30 ) was isolated from the hoplonemertine Amphiporus anagulatus by Kem and co-workers in 2009. 196 This alkaloid is an anatabine isomer possessing a carboncarbon double bond in the 3,4-position of the piperidine scaffold. 48, 196 The naturally occurring enantiomeric form of isoanatabine has not yet been reported. Due to the structural similarity with nicotine, the pharmacological properties of both (-)-and (+)-isoanatabine were examined by Kem and co-workers at rat and human α4β2-nAChRs. 196 The binding affinity of (-)-isoanatabine (K i = 108 nM) at rat α4β2-nAChR was slightly stronger than the (+)enantiomer (K i = 136 nM); however, at the human α4β2-nAChR, (-)-isoanatabine was less efficacious (EC 50 = 1.01 µM; I max = 78.7%) than the (+)-enantiomer (EC 50 = 0.31 µM; I max = 102%). 196 These findings indicate that isoanatabine is a more potent ACh stimulant at the Platisidines A -C ( Figure 32 ) were isolated from an Okinawan marine sponge in the genus of Plakortis (sp. SS-11) by Koboyashi and co-workers. 198 These nicotinic acid derivatives comprise an N-methylated pyridinium-β-carboxylate with a hexadecanoyl side chain. mAChRs play important roles in various physiological functions in the human body, including memory and learning. 156 Six structurally unprecedented macrocyclic bis-1,3disubstituted pyridiniums, cyclostellettamines A -F (Figure 33 ), were isolated from a Japanese marine sponge Stelletta maxima. 199 Agelas, a genus of marine sponges commonly found on the Caribbean and Indo-Pacific coral reefs, is a rich source of pharmacologically active bromo-alkaloids. 200, 201 In a study examining the biologically active secondary metabolites of A. longissimi, the methanolic extract of the sponge was partitioned and purified to give agelongine (Figure 34) , an alkaloid comprising a pyridinium-β-carboxylate moiety bonded to a 4-bromopyrrole-2-carboxylic unit through an aliphatic chain. 200 In a subsequent study, the structurally related pyridinium alkaloid daminin (Figure 34 Agelongine exhibited a competitive, reversible antagonism at serotonergic receptors subfamily 1 (5-HT 1 ) in vitro. 200 The agonist properties of 5-hydroxytryptamine at 5-HT 1 receptors were inhibited when agelongine was introduced (IC 50 = 80 µM). 200 Moreover, agelongine (100 µM) did not affect the concentration-response of histamine, ACh and prostaglandin E 2 , suggesting that agelongine is selective for 5-HT 1 receptors; however, the specific 5-HT 1 receptor subtype used in these preliminary pharmacological tests was not stated. 200 5-HT 1 receptor subtypes, including 5-HT 1A , 5-HT 1B , 5-HT 1D , 5-HT 1E and 5-HT 1F have different distribution and function in the human body. 202 For example, 5-HT 1A , the most common 5-HT 1 receptor subtype and highly saturated in the hippocampus, is involved in the emotional mechanism; the classic anxiolytic agent buspirone is an agonist at this site. 202 In contrast, both the 5-HT 1B and 5-HT 1D receptors are widely distributed in the basal ganglia and act as autoreceptors to decrease the transmission of the neurotransmitter glutamate in the neuronal terminals. 202 The triptans are 5-HT 1B/1D receptor agonists used to treat migraine attacks in adults with or without aura. 202 Further investigations are required to fully understand how agelongine binds across the serotonergic system and therefore determine its therapeutic potential as a neuropsychiatric lead compound. Calcium ions regulate many critically important functions in the CNS, including the release of neurotransmitters and intracellular signal transductions. 203 The neuroprotective properties of daminin were determined by measuring its effect on the changes of [Ca 2+ ] i levels in neuronal cells, using glutamic acid and NMDA (neuroexcitatory agonists) as positive controls. 201 Upon treatment with either glutamic acid (200 µM) Trigonella foenum-graecum L. (fenugreek) of the Leguminosae family is a herbal plant that has been used for centuries to treat a wide range of ailments including diabetes, fever, memory loss, epilepsy and migraine. 204 The vitamin B 6 derivative trigonelline ( Figure 35 ) is a N-methylnicotinic acid that has been isolated from the seeds of fenugreek, a legume crop used as a spice and medicines in East Asia and Northern Africa. 204, 205 Subsequent studies revealed that trigonelline is also widely distributed in plants within the dicotyledonae subclass, as well as in various animal species, such as arthropods, marine poriferans and mammals. 204 Trigonelline occurs in raw coffee beans and is converted into nicotinic acid upon roasting at ~230 ºC. 204, 206 Trigonelline is a water-soluble secondary metabolite formed from nicotinate ( Figure 35 ) and is responsible for the bitterness in coffee. 204 Extension of dendrites and axons in neurons can compensate neural loss and repair damaged neuronal network in people with dementia. 207, 208 Komatsu and co-workers demonstrated that trigonelline extracted from coffee beans exhibits functional neurite outgrowth activity by inducing axonal extension in human neuroblastoma SK-N-SH cells. 207 Trigonelline (30 µM) significantly increased the percentage of human SK-N-SH cells with neurites larger than 50 µm by 15% after three days of treatment. 207 In a subsequent study, the same group also revealed that upon oral administration of trigonelline (500 mg/kg; q.d.; 15 days), male ddY mice pre-treated with Aβ (5 nmol) were able to complete more successful crossings over a previous platform position in the water maze test, indicating an improvement in memory retention. 207 Trigonelline also displays other CNS-related properties, including protection against cerebral ischemia by decreasing neuronal spike frequency from single action potential to multiple firing (0.1 mM), 209 stimulation of dopamine release (136% at 4.977 µM), 210 competitive inhibition of GABA A receptors (K i = 13 nM), 211 and weak inhibition of AChE (IC 50 = 233 µM). 212 Taken together, these findings indicate that trigonelline exhibits pronounced neuroprotective properties that warrant further investigation. In summary, pyridine alkaloids possess a diverse array of properties in the CNS that validate this class of natural products as a source of potential therapeutic leads for CNS disorders. Many pyridine alkaloids structurally related to nicotine are themselves nAChR agonists, including cytisine and epibatidine, with the former providing the basis for the development of antagonists that inhibit the activation of neuronal potentials in the nervous system and are therefore potential therapeutic leads for various CNS-related disorders, including Parkinson's and AD. Many of the pyridine alkaloids described herein have pronounced effects in the CNS, but their exact mode of action is not well understood. For example, haplophyllidine possesses sedative and anti-analeptic properties, while gentianine is a CNS stimulant with a promising antipsychotic profile. We hope that this review has provided thought-provoking insight into the therapeutic utility of pyridine alkaloids for the treatment for CNS disorders. Targeted Drug Delivery to Central Nervous System (CNS) for the Treatment of Neurodegenerative Disorders: Trends and Advances Drug Delivery to the Central Nervous System Potential Medicinal Plants for CNS Disorders: An Overview Central Nervous System Diseases and Inflammation Neurogenesis in the Adult Brain: New Strategies for Central Nervous System Diseases Drug Delivery to the Central Nervous System: A Review Crossing the Blood-Brain Barrier: A Review on Drug Delivery Strategies for Treatment of the Central Nervous System Diseases Drug Development for CNS Disorders: Strategies for Balancing Risk and Reducing Attrition CNS Disorders-Current Treatment Options and the Prospects for Advanced Therapies Global, Regional, and National Burden of Alzheimer's Disease and Other Dementias, 1990-2016: A Systematic Analysis for the Global Burden of Disease Study The Prevalence of Mental Disorders among the Homeless in Western Countries: Systematic Review and Meta-Regression Analysis The Economic Burden of Mental Disorders in China, 2005-2013: Implications for Health Policy Accessed 21 Why Are the Majority of Active Compounds in the CNS Domain Natural Products? A Critical Analysis Analysis of the Structural Diversity, Substitution Patterns, and Frequency of Nitrogen Heterocycles among Us FDA Approved Pharmaceuticals: Miniperspective The Necessary Nitrogen Atom: A Versatile High-Impact Design Element for Multiparameter Optimization Nicotine Addiction Pharmacology of Nicotine: Addiction, Smoking-Induced Disease, and Therapeutics Its Metabolism and an Overview of Its Biological Effects And The Brain Chemical Investigation of Tobacco and the Preparation of the Characteristic Active Principle of This Plant. Magazin f Ueber Phenyl-und Pyridylpyrrole Und Die Constitution Des Nicotins Acetylcholine Receptors Containing the Β2 Subunit Are Involved in the Reinforcing Properties of Nicotine Cardiovascular Responses to Microinjections of Nicotine into the Caudal Ventrolateral Medulla of the Rat Oct-3-Yl]-4-Chlorobenzamide Hydrochloride] Enhances GABAergic Synaptic Activity in Brain Slices and Restores Auditory Gating Deficits in Anesthetized Rats Nicotine Effects and the Endogenous Opioid System The Role of NMDA Receptor Antagonists in Nicotine Tolerance, Sensitization, and Physical Dependence: A Preclinical Review World Health Organization Monitoring Tobacco Use and Prevention Policies Nicotine Replacement Therapy for Smoking Cessation Effects of (−)-Nicotine and (−)-Cotinine on 6-Hydroxydopamine-Induced Oxidative Stress and Neurotoxicity: Relevance for Parkinson's Disease Altered Anxiety-Related Responses in Mutant Mice Lacking the Β4 Subunit of the Nicotinic Receptor Beyond Smoking Cessation: Investigating Medicinal Nicotine to Prevent and Treat COVID-19 Systematic Review of the Prevalence of Current Smoking among Hospitalized COVID-19 Patients in China: Could Nicotine Be a Therapeutic Option? Nicotine and SARS-CoV-2: COVID-19 May Be a Disease of the Nicotinic Cholinergic System The Pharmacological Activity of Nicotine and Nornicotine on NAChRs Subtypes: Relevance to Nicotine Dependence and Drug Discovery Zur Kenntnis Der Alkaloide Des Tabaks The Alkaloid Contents of Sixty Nicotiana Species Gas Chromatographic-Mass Spectrometric Method for Determination of Anabasine, Anatabine and Other Tobacco Alkaloids in Urine of Smokers and Smokeless Tobacco Users Enantiomeric Composition of Nornicotine, Anatabine, and Anabasine in Tobacco Phytochemical Studies on the Tobacco Alkaloids. I. Optical Rotatory Power of Nornicotine Nicotinic-Receptor Mediation of S (-) Nornicotine-Evoked [3H] Overflow from Rat Striatal Slices Preloaded with [3H] Dopamine Handbook of Pesticide Toxicology Chapter 3-Pest Control Agents from Natural Products Über Die Alkaloide von Anabasis Aphylla L.(I. Mitteil Neonicotine Recently Found as an Alkaloid in Anabasis Aphylla L.(Scientific Note Relative Toxicities and Neuromuscular Nicotinic Receptor Agonistic Potencies of Anabasine Enantiomers and Anabaseine Absolute Configuration of Anabasine from Messor and Aphaenogaster Ants A Pharmacological Comparison of Two Isomeric Nicotinic Receptor Agonists: The Marine Toxin Isoanatabine and the Tobacco Alkaloid Anatabine Ein Neues Tabakalkaloid (XI. Mitteil. Über Tabak-Basen) Determination of Tobacco Alkaloid Enantiomers Using Reversed Phase UPLC/MS/MS. Heliyon Anatabine Lowers Alzheimer's Aβ Production in Vitro and in Vivo Effects of Tobacco Smoke Constituents, Anabasine and Anatabine, on Memory and Attention in Female Rats Über Neue Basen Des Tabaks (XIII. Mitteil. Über Tabak-Alkaloide) Studies on the Interactions of Tobacco Leaf and Tobacco Smoke Constituents and Monoamine Oxidase An Electrophysiological Characterization of Naturally Occurring Tobacco Alkaloids and Their Action on Human Α4β2 and Α7 Nicotinic Acetylcholine Receptors The Nicotine Metabolite, Cotinine, Alters the Assembly and Trafficking of a Subset of Nicotinic Acetylcholine Receptors Neuropathology Of Drug Addictions And Substance Misuse New Insights into the Mechanisms of Action of Cotinine and Its Distinctive Effects from Nicotine Beyond That Expected, More than a Biomarker of Tobacco Consumption Die Constitution Des Alkaloids The Chemistry of Tobacco Fermentation. I. Conversion of the Alkaloids. D. Identification of Cotinine in Fermented Leaves Alkaloids of Duboisia Hopwoodii −) Isomers of Cotinine Augment Cholinergic Responses in Vitro and in Vivo Neuroactive Metabolite of Nicotine: Potential for Treating Disorders of Impaired Cognition Cotinine Binding to Nicotinic Acetylcholine Receptors in Bovine Chromaffin Cell and Rat Brain Membranes The Nicotine Metabolite, Cotinine, Attenuates Glutamate (NMDA) Antagonist-Related Effects on the Performance of the Five Choice Serial Reaction Time Task (5C-SRTT) in Rats Cotinine Reduces Depressivelike Behavior, Working Memory Deficits, and Synaptic Loss Associated with Chronic Stress in Mice Evaluation of Nicotine and Cotinine Analogs as Potential Neuroprotective Agents for Alzheimer's Disease S)-(−)-Cotinine, the Major Brain Metabolite of Nicotine, Stimulates Nicotinic Receptors to Evoke [3H] Dopamine Release from Rat Striatal Slices in a Calcium-Dependent Manner On the Action of Nicotine and Cotinine on Central 5-Hydroxytryptamine Neurons Identification and Synthesis of Novel Alkaloids from the Root System of Nicotiana Tabacum: Affinity for Neuronal Nicotinic Acetylcholine Receptors Pharmacological Characterization of RJR-2403: A Nicotinic Agonist with Potential Therapeutic Benefit in the Treatment of Alzheimer's Disease Nicotine-like Actions of Cis-Metanicotine and Trans-Metanicotine Über Das Vorkommen von Metanikotin in Natürlich Nikotinfreien Tabaksorten Effect of Gibberellic Acid Applications to Leaves of Nicotiana on Nornicotine, Anabasine, Metanicotine, Oxynicotine, and Nicotinic Acid Content Composition Studies on Tobacco. XX.-Bases of Cigarette Smoke Metanicotine: A Nicotinic Agonist with Central Nervous System Selectivity-in Vitro and in Vivo Characterization Nicotinic Acetylcholine Receptors: From Basic Science to Therapeutics The Comprehensive Pharmacology Reference Sesquiterpenes and Alkaloids from the Roots of Alangium Chinense Alkaloids Of Anabasis Aphylla and Their Cholinergic Activities Cytisine and Derivatives: Synthesis, Reactivity, and Applications Cytisine-from the Past to the Future Cytisine-from Ethomedical Use to the Development as a Natural Alternative for Smoking Cessation Cytisine: A Natural Product Lead for the Development of Drugs Acting at Nicotinic Acetylcholine Receptors Cytisine for the Treatment of Nicotine Addiction: From a Molecule to Therapeutic Efficacy Cytisine for Nicotine Addiction Treatment: A Review of Pharmacology, Therapeutics and an Update of Clinical Trial Evidence for Smoking Cessation Vorläufige Mittheilung Über Cytisin Und Laburnin, Zwei Neue Pflanzenbasen in Cytisus Laburnum Zur Frage Der Identität von Cytisin Und Ulexin Absolute Configurations Of (-)-Cytisine And Of Related Alkaloids Quinolizidine Alkaloids from Argyrolobium Uniflorum Alkaloids of Templetonia Incana Quinolizidine Alkaloids in Petteria Ramentacea Nicotinic Receptor Binding of [3H] Cytisine,[3H] Nicotine and [3H] Methylcarbamylcholine in Rat Brain Characterization of [3H] Cytisine Binding to Human Brain Membrane Preparations Varenicline: An Α4β2 Nicotinic Receptor Partial Agonist for Smoking Cessation In Vivo Modulation of Dopaminergic Nigrostriatal Pathways by Cytisine Derivatives: Implications for Parkinson's Disease Discovery and Development of Varenicline for Smoking Cessation a Partial Agonist of High-Affinity Nicotinic Acetylcholine Receptors, Has Antidepressant-like Properties in Male C57BL/6J Mice Quinolizidone Alkaloid from Laburnum Anagyroides First Total Synthesis of (±)-3-Hydroxy-11-Norcytisine: Structure Confirmation and Biological Characterization Cytisine-like Alkaloids from Ormosia Hosiei Hemsl. & EH Wilson A Mini-Review of Biological Characteristics, Natural Sources, Synthetic Origins, and Future Prospects Huperzine A as Potential Treatment of Alzheimer's Disease: An Assessment on Chemistry, Pharmacology, and Clinical Studies Development of Huperzine A and B for Treatment of Alzheimer's Disease Medicinal Chemistry of Bioactive Natural Products Pharmacological Profile of Huperzine A, a Novel Acetylcholinesterase Inhibitor from Chinese Herb The Structures of Huperzine A and B, Two New Alkaloids Exhibiting Marked Anticholinesterase Activity Cholinesterase Inhibitors: A New Class of Psychotropic Compounds The Role of Cholinesterase Inhibitors in Alzheimer's Disease Effect of Huperzine A, a New Cholinesterase Inhibitor, on the Central Cholinergic System of the Rat Huperzine A, a Potential Therapeutic Agent for Dementia, Reduces Neuronal Cell Death Caused by Glutamate The NMDA Receptor Ion Channel: A Site for Binding of Huperzine A Reversal of Scopolamine-Induced Deficits in Radial Maze Performance by (−)-Huperzine A: Comparison with E2020 and Tacrine Huperzine A Reverses Scopolamine-and Muscimol-Induced Memory Deficits in Chick Effect of Huperzine A on Working Memory in Reserpine-or Yohimbine-Treated Monkeys Lycodine-Type Alkaloids from Lycopodiastrum Casuarinoides and Their Acetylcholinesterase Inhibitory Activity The Alkaloids Huperzines C and D and Huperzinine From Lycopodiastrum Casuarinoides Three New Lycopodium Alkaloids from Huperzia Carinata and Huperzia Squarrosa Lycodine-Type Alkaloids from Lycopodiastrum Casuarinoides Alkaloids from Lycopodium Casuarinoides Acetylcholinesterase Inhibitory Pyridine Alkaloids of the Leaves of Senna Multijuga Pyridine Alkaloids from Senna Multijuga as Acetylcholinesterase Inhibitors Chemical and Pharmacological Research of the Plants in Genus Euphorbia New Myrsinol Diterpenes from Euphorbia Prolifera New Myrsinol Diterpenes from Euphorbia Prolifera and Their Inhibitory Activities on LPS-Induced NO Production Diterpenes from Euphorbia Seguieriana Inhibition of Nitric Oxide Production in BV2 Microglial Cells by Triterpenes from Tetrapanax Papyriferus Cerpegin Alkaloid and Its Analogues: Chemical Synthesis and Pharmacological Profiles A Pyridine Alkaloid from Ceropegia Juncea Pharmacological Actions of Cerpegin, a Novel Pyridine Alkaloid from Ceropegia Juncea. Fitoterapia (Milano) The Monoterpene Alkaloid Cantleyine from Strychnos Trinervis Root and Its Spasmolytic Properties Other than Those of Cinchona The Technology of the Isolation of the Alkaloids Dubinidine and Haplophylidine Synergy of the Alkaloid Haplophyllidin with Sleep-Inducing and Narcotic Drugs Structure of Gentianine Synthesis of Gentianine Chemical Constituents of Gentianaceae XI: Antipsychotic Activity of Gentianine Plant Secondary Metabolism Alkaloids of Terpenoid Origin (excepting indole alkaloids and ergot alkaloids Epibatidine and Pain Epibatidine: A Promising Natural Alkaloid in Health Epibatidine: A Novel (Chloropyridyl) Azabicycloheptane with Potent Analgesic Activity from an Ecuadoran Poison Frog The Synthesis of (+)-and (-)-Epibatidine Resolution of Synthetic (+)-and (-)-Epibatidine by Chiral High Performance Liquid Chromatography and Identification of the Natural Enantiomer Epibatidine Is a Nicotinic Analgesic Antinociceptive and Toxic Effects of (+)-epibatidine Oxalate Attributable to Nicotinic Agonist Activity Stable Expression, Pharmacologic Properties and Regulation of the Human Neuronal Nicotinic Acetylcholine Alpha 4 Beta 2 Receptor Acetylcholine Receptor in Alzheimer's Disease Relative Affinities of Drugs Acting at Cholinoceptors in Displacing Agonist and Antagonist Radioligands: The NMS/Oxo-M Ratio as an Index of Efficacy at Cortical Muscarinic Receptors Cholinergic Agent Attack (Nicotine, Epibatidine, and Anatoxin-a) Phantasmidine: An Epibatidine Congener from the Ecuadorian Poison Frog Epipedobates Anthonyi Absolute Configuration and Pharmacology of the Poison Frog Alkaloid Phantasmidine Synthesis of Phantasmidine Enantioselective Syntheses of Both Enantiomers of Noranabasamine Indole Alkaloids" (Calycanthine and Chimonanthine) and a Piperidinyldipyridin Militarinone A, a Neurotrophic Pyridone Alkaloid from Paecilomyces m Ilitaris +)-N-Deoxymilitarinone A, a Neuritogenic Pyridone Alkaloid from the Insect Pathogenic Fungus Paecilomyces f Arinosus Neurotrophic Alkaloidal Metabolites from the Entomogenous Deuteromycete Paecilomyces f Arinosus Pyridone Alkaloids from a Sponge-Derived Fungus Arthrinium Arundinis ZSDS1-F3 Ester Transfer Protein Inhibitor from an Insect-Associated Fungus Isolation and Structure of Aromelic Acid A and B. New Kainoids of Clitocybe Acromelalga Acromelic Acids A and B. Potent Neuroexcitatory Amino Acids Isolated from Clitocybe Acromelalga Synthesis of Acromelic Acid A, a Toxic Principle of Clitocybe Acromelalga Acromelic Acid, a Novel Excitatory Amino Acid from a Poisonous Mushroom: Effects on the Crayfish Neuromuscular Junction Kainic Acid-Induced Neurodegenerative Model: Potentials and Limitations Acromelic Acid Is a Much More Potent Excitant than Kainic Acid or Domoic Acid in the Isolated Rat Spinal Cord Effects of Acromelic Acid A on the Binding of [3H] Glutamic Acid and [3H] Kainic Acid to Synaptic Membranes and on the Depolarization at the Frog Spinal Cord Acromelic Acid C. A New Toxic Constituent of Clitocybe Acromelalga: An Efficient Isolation of Acromelic Acids Novel Neuroexcitatory Amino Acid from Clitocybe Acromelalga New Amino Acids from Clitocybe Acromelalga. Possible Intermediates in the Biogenesis of Mushroom Toxins, Acromelic Acids Production of Fusaric Acid by Fusarium Species Biochemistry of Fusarium Heterosporium in Rice Plant, Part 1: Newly Found Product of Fusaric Acid Pharmacological Activities of Fusaric Acid (5-Butylpicolinic Acid) Fusaric Acid, a Hypotensive Agent Produced by Fungi Inhibition of Dopamine β-Hydroxylase by Fusaric Acid (5-Butylpicolinic Acid) in Vitro and in Vivo Behavioral Effects of a New Dopamine-β-Hydroxylase Inhibitor (Fusaric Acid) in Man Fusaric Acid Alters Akt and Ampk Signalling in C57bl/6 Mice Brain Tissue Phenopicolinic Acid, a New Microbial Product Inhibiting Dopamine β-Hydroxylase Marine Natural Products New Natural Products from the Sponge-Derived Fungus Aspergillus Niger New Natural Products from the Sponge-Derived Fungus Aspergillus Niger Coprismycins A and B, Neuroprotective Phenylpyridines from the Dung Beetle-Associated Bacterium Novel Antibiotics SF2738A, B and C, and Their Analogs Produced by Streptomyces Sp Les Poisons Des Nemertiens Isolation and Structure of a Hoplonemertine Toxin Anabaseine Is a Potent Agonist on Muscle and NeuronalAlpha-Bungarotoxin-Sensitive Nicotinic Receptors Isoanatabine, a Naturally Occurring Α4β2 Nicotinic Receptor Agonist Hoplonemertine Worms-a New Source of Pyridine Neurotoxins Alkaloids from an Okinawan Marine Sponge of Plakortis Species Pyridine Alkaloids Which Inhibit Binding of Methyl Quinuclidinyl Benzilate (QNB) to Muscarinic Acetylcholine Receptors, from the Marine Sponge, Stelletta Maxima A Novel Bromopyrrole Alkaloid from the Sponge Agelas Longissima with Antiserotonergic Activity a Bioactive Pyrrole Alkaloid from the Mediterranean Sponge Axinella Damicornis International Union of Pharmacology Classification of Receptors for 5-Hydroxytryptamine (Serotonin) Calcium Dynamics in the Central Nervous System Trigonelline: A Plant Alkaloid with Therapeutic Potential for Diabetes and Central Nervous System Disease Medicinal Plant; The National Science Council Of Sri Lanka Trigonelline Is a Novel Phytoestrogen in Coffee Beans Trigonelline-Induced Neurite Outgrowth in Human Neuroblastoma SK-N-SH Cells Search for Natural Products Related to Regeneration of the Neuronal Network A Pyridinium Derivative from Red Sea Soft Corals Inhibited Voltage-Activated Potassium Conductances and Increased Excitability of Rat Cultured Sensory Neurones Identification of Coffee Components That Stimulate Dopamine Release from Pheochromocytoma Cells (PC-12) Effects of Coffee Components on the Response of GABAA Receptors Expressed in Xenopus Oocytes Acetylcholinesterase Enzyme Inhibitory Potential of Standardized Extract of Trigonella Foenum Graecum L and Its Constituents ☒ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.☒The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Simon Lin