ARTICLE DOI: 10.31480/2330-4871/158

Editorial | Volume 9 | Issue 2 Open Access

Therapeutic Potential of Salvinorin A and Its Analogues in Various Neurological Disorders

Joseph Cichon1, Renyu Liu1* and Hoang V. Le2*

1Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA

2Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, USA

*Corresponding Authors: Renyu Liu, MD, PhD, Professor, Departments of Anesthesiology and Critical Care, and Neurology, Perelman School of Medicine at the University of Pennsylvania, 336 John Morgan building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA, E-mail: RenYu.Liu@pennmedicine.upenn.edu
Hoang V. Le, PhD, Assistant Professor of Medicinal Chemistry, Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, 419 Faser Hall, University, MS 38677, USA, E-mail: hle@olemiss.edu

Editor: Jun-Ming Zhang, MD, Msc, Professor, Vice Chair for Research Anesthesia, Endowed Chair of Anesthesia Research and Education, University of Cincinnati College of Medicine, 231 Albert Sabin Way Cincinnati, OH 45267-0531, United States, E-mail: Jun-ming.zhang@uc.edu

Received: June 12, 2022 | Accepted: June 26, 2022 | Published: June 29, 2022

Citation: Cichon J, Liu R, Le HV. Therapeutic Potential of Salvinorin A and Its Analogues in Various Neurological Disorders. Transl Perioper & Pain Med 2022; 9(2):452-457


Introduction


While the hallucinogenic plant Salvia divinorum has been safely consumed by humans for centuries for religious or recreational purposes without long-term side effects, its active component, salvinorin A, and molecular mechanism of action were not known until recently [1]. Notably, salvinorin A is a potent kappa opioid receptor (KOR) agonist that is highly selective for KOR without significant interactions with any other receptors [2]. Different from classic opioid receptor ligands, which are opiate, this is the first molecule that has no nitrogen in its chemical structure but can interact with an opioid receptor with high affinity and potency (Figure 1) [1]. All classic opioid receptor agonists or antagonists have nitrogen in their chemical structures; therefore, it had been hypothesized that nitrogen was essential for a compound to have any opioid receptor activity until salvinorin A was discovered. In addition to its unique chemical structure, salvinorin A has very high selectivity towards KOR. Thus, it has been used as a great tool to study the pure pharmacological effects of KOR agonism. The C2 ester in the chemical structure quickly undergoes hydrolysis to yield the inactive metabolite salvinorin B, resulting in salvinorin A undergoing fast metabolism in the body. Despite its short duration of action, a single dose of salvinorin A, like other classic psychedelics, induces rapid and robust changes in the patterns of neural activity and connectivity in disease-related neuronal circuits. While not fully understood, these neurophysiological effects might explain salvinorin A's rapid and durable therapeutic effects in neuropsychiatric disorders that are difficult to treat. Furthermore, the unique chemical structure of salvinorin A provides a useful backbone for chemists to develop novel compounds targeting KOR and other opioid receptors for structure-activity relationships, structure-function analyses, and novel medication design purposes [3,4].

Figure 1: Chemical structures of salvinorin A and some well-known analogues of salvinorin A. Herkinorin, PR-38, salvindolin, compound 1, and compound 2 (now known as salvidenin) are dual KOR/MOR agonists. RB-64 is a G-protein-biased KOR agonist that forms a covalent bond with KOR. β-Tetrahydropyran salvinorin B is a long-acting KOR agonist. 16-Ethynyl salvinorin A is a nonbiased KOR agonist. 16-Bromo salvinorin A is a G-protein-biased KOR agonist. 1α-Hydroxysalvinorin A is a KOR antagonist. KOR, kappa opioid receptor. MOR, mu opioid receptor.

Chemical Properties of Salvinorin A


Salvinorin A is a neo-clerodane diterpenoid. It is not soluble in water or lipids, which is a significant hurdle for clinical deliverable formulation. In contrast to classic KOR ligands, salvinorin A is not an alkaloid, indicating that it cannot be rendered into a salt to improve solubility for drug development purposes. The melting point of salvinorin A is high (238-240 ℃) [5], and the powder form of salvinorin A is relatively stable. However, once taken, salvinorin A is hydrolyzed quickly by esterases due to the hydrolyzable nature of the ester functional group at C2, which is an advantage for clinical practices that need the benefits of a KOR agonist with a short-acting property and a short duration of hallucinatory or dissociative side effects.

Molecular Target of Salvinorin A


Salvinorin A (Figure 1) was first isolated from Salvia divinorum in 1982 by Ortega and coworkers [6], and it is one of the most potent, naturally occurring opioid agonists, with high selectivity and affinity towards KOR (Ki = 4 nM, EC50 in [35S]GTPγS binding assay = 2.2 nM) [7]. In 2002, Roth and coworkers discovered that salvinorin A targeted KORs expressed in both human embryonic kidney-293 cells (Ki = 16 nM) and guinea pig brain (Ki = 4.3 nM) [1]. They also discovered the nonbiased agonism nature of salvinorin A towards KOR when it was observed to activate both the G-protein signaling pathway (EC50 in cAMP assay = 4.73 nM) and β-arrestin-recruitment pathway (EC50 in the Tango assay = 10.5 nM) [8].

Analogues of Salvinorin A


Many analogues of salvinorin A have been developed to target different opioid receptors [4,7]. The most common alteration to the structure of salvinorin A is the replacement of the acetate at C2. Many different functional groups at this position, including carbonates, carbamates, alternative ester groups, ethers, amines, amides, sulfonic esters, sulfonamides, thioesters, and halides, have been incorporated and studied [4]. Data from these studies have suggested that the C2 position is critical for KOR binding and activation [7]. Notably, several C2 esters with a conjugated ring, an aromatic ring, or fused rings, such as herkinorin [9], PR-38 [10], salvindolin [11], compound 1, [12] and compound 2 (now known as salvidenin) [3] (Figure 1), displayed dual agonism on KOR and mu opioid receptor (MOR) [3]. Dual KOR/MOR agonists have been shown to retain analgesic activity while showing reduced undesirable adverse effects compared to pure KOR agonists or pure MOR agonists [3]. For example, male C57BL/6NHsd mice treated with salvidenin showed a significant increase in the latency to paw response in a hot plate test (single dose 2 mg/kg, i.p.) compared to vehicle-treated mice, which indicated antinociception, and showed a significant increase in the amount of time spent on the open arms in an elevated plus maze test (single dose 5 mg/kg, i.p.), which indicated anxiolysis [3]. Another notable C2 ester is RB-64 (Figure 1), which is the first and only known salvinorin-based agonist that forms a covalent bond with KOR [8]. RB-64 is also known to be G-protein biased. It displayed functional selectivity for G-protein over β-arrestin recruitment by a factor of 96 [8]. Therefore, RB-64, along with salvinorin A, have been widely used to study the specific effects and differences between G-protein signaling and β-arrestin recruitment signaling.

Other alternations to the structure of salvinorin A, such as the replacement of the methyl ester at C4, modifications of substituents on rings A and C, and replacement of the furan ring, have also been prepared and evaluated [4]. Unlike the acetate group at C2, the methyl ester at C4 requires more forcing conditions for hydrolysis and modification. So far, all of the modifications at the C4 position have resulted in a loss of binding affinity and potency on KOR [4]. Similarly, so far, none of the modifications of substituents on rings A and C or replacement of the furan ring have resulted in compounds with improved binding affinity and potency on KOR compared to salvinorin A [4,7]. Recently, substitution on the furan ring at C16 led to two interesting salvinorin A analogues, 16-ethynyl salvinorin A and 16-bromo salvinorin A (Figure 1) [13]. While 16-ethynyl salvinorin A displayed balanced signaling properties, 16-bromo salvinorin A showed a significant G-protein signaling bias. In acute nociceptive and inflammatory pain mouse models, 16-ethynyl salvinorin A showed significant antinociceptive effects and reduced side effects compared to salvinorin A, while 16-bromo salvinorin A displayed modest antinociceptive effects and lacked anxiogenic effects [13].

Interestingly, some small modifications to the salvinorin structure at C1 or C10 were observed to switch the functionality of the molecule from an agonist to an antagonist of KOR [14]. There are only six salvinorin-based compounds in the literature that have demonstrated antagonistic activity against any of the opioid receptors; 1α-hydroxysalvinorin A (Figure 1) is the most potent and selective antagonist for KOR [3]. KOR antagonists have been shown to alleviate depressive and anxiety-related disorders, which are the common issues related to withdrawal that can lead to drug relapse. Recently, 1α-hydroxysalvinorin A and norbinaltorphimine (nor-BNI) were studied on C57BL/6N mice for spontaneous cocaine withdrawal [15]. Administration of 1α-hydroxysalvinorin A (5 mg/kg, i.p.) was shown to reduce spontaneous cocaine-withdrawal behaviors, comparable to nor-BNI (5 mg/kg, i.p.). Notably, 1α-hydroxysalvinorin A produced anti-anxiety-like effects in the light-dark transition test that was not observed with nor-BNI (both 5 mg/kg, i.p.). Still, the mechanisms of action of KOR antagonists on withdrawal have not been known.

Salvinorin A and Neuronal Circuits Modulation


The psychoactive effects of salvinorin A have in large part hindered its development and characterization as a therapeutic agent in modern medicine. The salvinorin A-induced experience is hallmarked by drastic perceptual changes, hallucination (visions and auditory), intense feelings of depersonalization and derealization, and dissociative effects (unresponsiveness to environmental stimuli) [16]. In rodents and non-human primates, salvinorin A causes a degree of sedation and impaired locomotion [17]. These experiences have been sought out by Mazatec shamans for medical and religious purposes since neolithic times [18]. Interestingly, only until recently has there been a growing appreciation of previously known psychedelic substances, including salvinorin A, in the treatment of complex human brain disorders and chronic disease states. Increasing evidence suggests these agents induce rapid and robust therapeutic effects in select patient populations who suffer from anxiety, chronic stress, depression/mood disorders, substance disorders, and pain conditions [19]. While most of these agents target serotonin 2A receptor (psilocybin and lysergic acid diethylamide (LSD)) or N-methyl-D-aspartate (NMDA) receptor (ketamine and nitrous oxide), salvinorin A mediates its effects through KOR activation. KOR couples primarily to Gi/o proteins that regulate intracellular IP3- and cAMP3-based second messenger cascades. Activation of this signaling pathway decreases cellular excitability via an increase of the inward rectifier potassium currents [20]. Additionally, KOR down-regulates N-type calcium currents, which, via the reduction of presynaptic calcium influx, likely reduces the release of both excitatory and inhibitory neurotransmitters. Collectively, KOR activation would be inhibitory by reducing both the input signals and the postsynaptic responses. The neurophysiological effects of salvinorin A in vivo and in the treatment of various neurological disorders are not known. However, based on KOR's expression profile, including the striatum, several cortical areas (deep cortical layers (V)), limbic areas (hippocampus and amygdala), hypothalamus, spinal cord, and the claustrum, salvinorin A has been hypothesized to downregulate the neuronal activity in distinct circuits tied to the aforementioned areas [21]. Thus, salvinorin A might be well suited to acutely inhibit subsets of hyperactive neurons to resculpt abnormal patterns of neuronal activity, potentially engage various forms of synaptic plasticity, and normalize brain function (Figure 2). In ischemic stroke, for example, abnormal glutamate release drives hyperexcitability and brain injury and worsens functional outcomes. Administration of salvinorin A to patients early in stroke evolution might suppress glutamate-induced hyperactivation to minimize brain injury. This hypothesis could be carried over to treat neuropsychiatric diseases characterized by circuits with motifs of hyperactivation; for example, anxiety disorders and post-traumatic stress disorders have been linked to cortico-amygdala hyperactivation where salvinorin A might provide specific suppression of these activity patterns and the grounds for weakening such connections.

Figure 2: Schematic showing the potential of salvinorin A in modulating neuronal activity and plasticity in the brain. Certain disease states could drive hyperexcitability in distinct circuits. Delivery of salvinorin A and KOR activation can rapidly decrease neuronal activity and synaptic plasticity. These rapid and sustained effects might reshape the connectivity and patterns of neuronal activity towards normal brain function. KOR, kappa opioid receptor. K, potassium. Ca, calcium. cAMP, cyclic adenosine monophosphate. PKA, protein kinase A.

Potential Clinical Implications


Salvinorin A has therapeutic potential in various complex diseases and conditions including pain [22], addiction [23], depression [24], itching [23], and stroke (Table 1) [25]. In general, salvinorin A's short-acting activity has hindered its clinical utility since most of the aforementioned clinical situations prefer relatively long-acting compounds. Some success has been made to extend its half-life by replacing the ester at C2 with more stable functional groups, such as carbamates, ethers, and amides. A recent study showedthat β-tetrahydropyran salvinorin B (Figure 1), a C2-ether analogue of salvinorin A, had a longer duration of action and displayed analgesic and anti-inflammatory effects in mice [26]. Our group has been focusing on the application of salvinorin A in acute stroke by taking advantage of its high potency and rapid onset. The effectiveness of salvinorin A in acute stroke rescue has been demonstrated in multiple animal models, including rodents [27,28], piglets [29,30], and monkeys [25]. The short-lived dissociative effects of salvinorin A have been considered by some as a hard stop in this patient population. However, it is possible that the unwanted effects might actually be therapeutic for neuronal circuit function and brain health. Salvinorin A's rapid change in neuronal activity and potential for synaptic plasticity in the brain might set the stage for the beneficial effects that are essential for stroke recovery. Additionally, in some patients where psychoactive effects might be deemed intolerable, molecular modifications of salvinorin A to add dual MOR activation could be reasonable [3]. A summary of the potential clinical usages of salvinorin A and its analogues is presented in Table 1.

Table 1: Therapeutic potentials of salvinorin A and its analogues.

Compound

Therapeutic Potential

 

Salvinorin A

Analgesic [31], addiction therapy [32], anti-cocaine seeking therapy [33], antidepressant [32], antipruritic [34], stroke rescue [25,28,35]

Salvindolin

Antidepressant [11], analgesic [11]

PR-38

Colitis [36], irritable bowel syndrome [37], antipruritic [38]

1α-Hydroxysalvinorin A

Anxiolytic [15], cocaine withdrawal therapy [15]

RB-64

Analgesic [8]

Salvidenin

Analgesic [3], anxiolytic [3]

16-Ethynyl salvinorin A

Analgesic [13], attenuation of cocaine-induced reinstatement [39]

16-Bromo salvinorin A

Analgesic [13], attenuation of cocaine-induced reinstatement [39]

β-Tetrahydropyran salvinorin B

Analgesic [26], anti-inflammatory [26], cocaine withdrawal therapy [40], addiction therapy [40]

Herkinorin

Anti-inflammatory [41], stroke rescue [42]

Conflicts of Interest


The authors declare no conflict of interest.

Acknowledgment


Work was supported by NIH (P30GM122733 pilot project award to H.V.L.) and FAER (MRTG to J.C). Dr. Renyu Liu appreciates NIH grant support (1R01GM111421). The content is solely the responsibility of the authors and does not necessarily represent the official views of these funders. The authors appreciate the help from Dr. John Grothusen in preparing Table 1 and editing. Figure 2 was created via Biorender.com.

References


  1. Roth BL, Baner K, Westkaemper R, Siebert D, Rice KC, et al. (2002) Salvinorin A: a potent naturally occurring nonnitrogenous kappa opioid selective agonist. Proc Natl Acad Sci USA 99: 11934-11939.
  2. Yan F, Roth BL (2004) Salvinorin A: a novel and highly selective kappa-opioid receptor agonist. Life Sci 75: 2615-2619.
  3. Akins NS, Mishra N, Harris HM, Dudhipala N, Kim SJ, et al. (2022) 6,5-fused ring, C2-salvinorin ester, dual kappa and mu opioid receptor agonists as analgesics devoid of anxiogenic effects. ChemMedChem 17: e202100684.
  4. Roach JJ, Shenvi RA (2018) A review of salvinorin analogs and their kappa-opioid receptor activity. Bioorganic Med Chem Lett 28: 1436-1445.
  5. Orton E, Liu R (2014) Salvinorin A: A mini review of physical and chemical properties affecting its translation from research to clinical applications in humans. Transl Perioper & Pain Med 1: 9-11.
  6. Ortega A, Blount JF, Manchand PS (1982) Salvinorin, a new trans-neoclerodane diterpene from Salvia divinorum (Labiatae). J Chem Soc Perkin trans 1: 2505-2508.
  7. Prisinzano TE (2005) Psychopharmacology of the hallucinogenic sage Salvia divinorum. Life Sci 78: 527-531.
  8. White KL, Robinson JE, Zhu H, DiBerto JF, Polepally PR, et al. (2015) The G protein-biased kappa-opioid receptor agonist RB-64 is analgesic with a unique spectrum of activities in vivo. J Pharmacol Exp Ther 352: 98-109.
  9. Harding WW, Tidgewell K, Byrd N, Cobb H, Dersch CM, et al. (2005) Neoclerodane diterpenes as a novel scaffold for mu opioid receptor ligands. J Med Chem 48: 4765-4771.
  10. Salaga M, Polepally PR, Sobczak M, Grzywacz D, Kamysz W, et al. (2014) Novel orally available salvinorin A analog PR-38 inhibits gastrointestinal motility and reduces abdominal pain in mouse models mimicking irritable bowel syndrome. The Journal of pharmacology and experimental therapeutics 350: 69-78.
  11. Keasling AW, Pandey P, Doerksen RJ, Pedrino GR, Costa EA, et al. (2019) Salvindolin elicits opioid system-mediated antinociceptive and antidepressant-like activities. J Psychopharmacol 33: 865-881.
  12. Tidgewell K, Groer CE, Harding WW, Lozama A, Schmidt M, et al. (2008) Herkinorin analogues with differential beta-arrestin-2 interactions. J Med Chem 51: 2421-2431.
  13. Paton KF, Biggerstaff A, Kaska S, Crowley RS, La Flamme1 AC, et al. (2020) Evaluation of biased and balanced salvinorin A analogs in preclinical models of pain. Front Neurosci 14: 765.
  14. Holden KG, Tidgewell K, Marquam A, Rothman RB, Navarro H, et al. (2007) Synthetic studies of neoclerodane diterpenes from Salvia divinorum: exploration of the 1-position. Bioorg Med Chem Lett 17: 6111-6115.
  15. Akins NS, Mohammed SF, Pandey P, Kim SJ, Mahdi F, et al. (2022) Alleviation of cocaine withdrawal and pertinent interactions between salvinorin-based antagonists and kappa opioid receptor. ChemRxiv doi: 10.26434/chemrxiv-2022-dpn5j-v2.
  16. Johnson MW, MacLean KA, Reissig CJ, Prisinzano TE, Griffiths RR (2011) Human psychopharmacology and dose-effects of salvinorin A, a kappa opioid agonist hallucinogen present in the plant Salvia divinorum. Drug Alcohol Depend 115: 150-155.
  17. Fantegrossi WE, Kugle KM, Valdes LJ, 3rd, Koreeda M, Woods JH (2005) Kappa-opioid receptor-mediated effects of the plant-derived hallucinogen, salvinorin A, on inverted screen performance in the mouse. Behav Pharmacol 16: 627-633.
  18. Hernandez-Bello R, Garcia-Rodriguez RV, Garcia-Sosa K, Pena-Rodriguez LM, Vazquez-Hernandez M, et al. (2015) Salvinorin A content in legal high products of Salvia divinorum sold in Mexico. Forensic Sci Int 249: 197-201.
  19. Butelman ER, Kreek MJ (2015) Salvinorin A, a kappa-opioid receptor agonist hallucinogen: pharmacology and potential template for novel pharmacotherapeutic agents in neuropsychiatric disorders. Front Pharmacol 6: 190.
  20. Tallent M, Dichter MA, Bell GI, Reisine T (1994) The cloned kappa opioid receptor couples to an N-type calcium current in undifferentiated PC-12 cells. Neurosci 63: 1033-1040.
  21. Stiefel KM, Merrifield A, Holcombe AO (2014) The claustrum's proposed role in consciousness is supported by the effect and target localization of Salvia divinorum. Frontiers in integrative neuroscience 8: 20.
  22. Chakraborty S, Majumdar S (2021) Natural products for the treatment of pain: chemistry and pharmacology of salvinorin A, mitragynine, and collybolide. Biochem 60: 1381-1400.
  23. Cruz A, Domingos S, Gallardo E, Martinho A (2017) A unique natural selective kappa-opioid receptor agonist, salvinorin A, and its roles in human therapeutics. Phytochemistry 137: 9-14.
  24. Taylor GT, Manzella F (2016) Kappa opioids, salvinorin A and major depressive disorder. Curr Neuropharmacol 14: 165-176.
  25. Wu L, Wu D, Chen J, Chen C, Yao T, et al. (2021) Intranasal salvinorin A improves neurological outcome in rhesus monkey ischemic stroke model using autologous blood clot. J Cereb Blood Flow Metab 41: 723-730.
  26. Paton KF, Kumar N, Crowley RS, Harper JL, Prisinzano TE, et al. (2017) The analgesic and anti-inflammatory effects of Salvinorin A analogue beta-tetrahydropyran Salvinorin B in mice. Eur J Pain 21: 1039-1050.
  27. Chen C, Cui X, Matsunaga F, Ma J, Ma N, et al. (2014) Salvinorin A decreases mortality and improves neurological outcome in a neonatal mouse hypoxia model. Transl Perioper Pain Med 1: 9-13.
  28. Chen C, Xi C, Liang X, Ma J, Su D, et al. (2016) The role of kappa opioid receptor in brain ischemia. Crit Care Med 44: e1219-e1225.
  29. Su D, Riley J, Armstead WM, Liu R (2012) Salvinorin A pretreatment preserves cerebrovascular autoregulation after brain hypoxic/ischemic injury via extracellular signal-regulated kinase/mitogen-activated protein kinase in piglets. Anesth Analg 114: 200-204.
  30. Wang Z, Ma N, Riley J, Armstead WM, Liu R (2012) Salvinorin A administration after global cerebral hypoxia/ischemia preserves cerebrovascular autoregulation via kappa opioid receptor in piglets PloS ONE 7: e41724.
  31. John TF, French LG, Erlichman JS (2006) The antinociceptive effect of salvinorin A in mice. Eur J Pharmacol 545: 129-133.
  32. Listos J, Merska A, Fidecka S (2011) Pharmacological activity of salvinorin A, the major component of Salvia divinorum. Pharmacol Rep 63: 1305-1309.
  33. Morani AS, Kivell B, Prisinzano TE, Schenk S (2009) Effect of kappa-opioid receptor agonists U69593, U50488H, spiradoline and salvinorin A on cocaine-induced drug-seeking in rats. Pharmacol Biochem Behav 94: 244-249.
  34. Wang Y, Tang K, Inan S, Siebert D, Holzgrabe U, et al. (2005) Comparison of pharmacological activities of three distinct kappa ligands (Salvinorin A, TRK-820 and 3FLB) on kappa opioid receptors in vitro and their antipruritic and antinociceptive activities in vivo. J Pharmacol Exp Ther 312: 220-230.
  35. Misilimu D, Li W, Chen D, Wei P, Huang Y, et al. (2021) Intranasal salvinorin a improves long-term neurological function via immunomodulation in a mouse ischemic stroke model. J Neuroimmune Pharmacol doi: 10.1007/s11481-021-10025-4.
  36. Salaga M, Polepally PR, Zakrzewski PK, Cygankiewicz A, Sobczak M, et al. (2014) Novel orally available salvinorin A analog PR-38 protects against experimental colitis and reduces abdominal pain in mice by interaction with opioid and cannabinoid receptors. Biochem Pharmacol 92: 618-626.
  37. Salaga M, Polepally PR, Sobczak M, Grzywacz D, Kamysz W, et al. (2014) Novel orally available salvinorin A analog PR-38 inhibits gastrointestinal motility and reduces abdominal pain in mouse models mimicking irritable bowel syndrome. J Pharmacol Exp Ther 350: 69-78.
  38. Salaga M, Polepally PR, Zielinska M, Marynowski M, Fabisiak A, et al. (2015) Salvinorin A analogues PR-37 and PR-38 attenuate compound 48/80-induced itch responses in mice. Br J Pharmacol 172: 4331-4341.
  39. Riley AP, Groer CE, Young D, Ewald AW, Kivell BM, et al. (2014) Synthesis and kappa-opioid receptor activity of furan-substituted salvinorin A analogues. J Med Chem 57: 10464-10475.
  40. Ewald AWM, Bosch PJ, Culverhouse A, Crowley RS, Neuenswander B, et al. (2017) The C-2 derivatives of salvinorin A, ethoxymethyl ether Sal B and beta-tetrahydropyran Sal B, have anti-cocaine properties with minimal side effects. Psychopharmacology 234: 2499-2514.
  41. Cui X, Xu X, Ju Z, Wang G, Xi C, et al. (2021) Herkinorin negatively regulates NLRP3 inflammasome to alleviate neuronal ischemic injury through activating Mu opioid receptor and inhibiting the NF-kappaB pathway. J Cell Biochem 9: 1085-1097.
  42. Gui X, Cui X, Wei H, Feng G, Zhang X, et al. (2017) cPKCgamma membrane translocation is involved in herkinorin-induced neuroprotection against cerebral ischemia/reperfusion injury in mice. Mol Med Rep 15: 221-227.