The Endocannabinoid System

The Endocannabinoid System and your body: Watch this YouTube video and discover how it works!

Endocannabinoid Deficiency Syndrome

Understanding the Endocannabinoid System of your body in Autism & Its Implications for Therapeutics

Your Endocannabinoid System Explained – Must see Video

Visualization of the endocannabinoid signaling system

 

 

What Is The Endocannabinoid System of the body and how does it work?

The endogenous cannabinoid system, named after the plant that led to its discovery, is perhaps the most important physiologic system involved in establishing and maintaining human health. Endocannabinoids and their receptors are found throughout the body: in the brain, organs, connective tissues, glands, and immune cells. In each tissue, the cannabinoid system performs different tasks, but the goal is always the same: homeostasis, the maintenance of a stable internal environment despite fluctuations in the external environment.

Cannabinoids promote homeostasis at every level of biological life, from the sub-cellular, to the organism, and perhaps to the community and beyond. Here’s one example: autophagy, a process in which a cell sequesters part of its contents to be self-digested and recycled, is mediated by the cannabinoid system. While this process keeps normal cells alive, allowing them to maintain a balance between the synthesis, degradation, and subsequent recycling of cellular products, it has a deadly effect on malignant tumor cells, causing them to consume themselves in a programmed cellular suicide. The death of cancer cells, of course, promotes homeostasis and survival at the level of the entire organism.   READ ENTIRE ARTICLE

 

Endocannabinoid system

The endocannabinoid system (ECS) is a group of endogenous cannabinoid receptorslocated in the mammalian brain and throughout the central and peripheral nervous systems, consisting of neuromodulatorylipids and their receptors. Known as “the body’s own cannabinoid system”,[1] the ECS is involved in a variety of physiological processes including appetite, pain-sensation,mood, and memory, and in mediating the psychoactive effects of cannabis.

Two primary endocannabinoid receptors have been identified: CB1, first cloned in 1990; CB2, cloned in 1993. CB1 receptors are found predominantly in the brain and nervous system, as well as in peripheral organs and tissues, and are the main molecular target of the endocannabinoid ligand (binding molecule), Anandamide, as well as itsmimetic phytocannabinoid, THC. One other main endocannabinoid is 2-Arachidonoylglycerol (2-AG) which is active at both cannabinoid receptors, along with its own mimetic phytocannabinoid, CBD. 2-AG and CBD are involved in the regulation of appetite, immune system functions and pain management.[1][2][3]

Basic overviewEdit

The endocannabinoid system, broadly speaking, includes:

The neurons, neural pathways, and other cells where these molecules, enzymes, and one or both cannabinoid receptor types are allcolocalized form the endocannabinoid system.

The endocannabinoid system has been studied using genetic and pharmacological methods. These studies have revealed that cannabinoids act as neuromodulators[5][6][7]for a variety of processes, including motor learning,[8] appetite,[9] and pain sensation,[10]among other cognitive and physical processes. The localization of the CB1 receptor in the endocannabinoid system has a very large degree of overlap with theorexinergic projection system, which mediates many of the same functions, both physical and cognitive.[11] Moreover, CB1 iscolocalized on orexin projection neurons in the lateral hypothalamus and many output structures of the orexin system,[11][12] where the CB1 and orexin receptor 1 (OX1) receptors physically and functionally join together to form the CB1–OX1 receptor heterodimer.[11][13][14]

Expression of receptorsEdit

For a more thorough description on receptor localization, see Cannabinoid receptor type 1 (CB1) andCannabinoid receptor type 2 (CB2).

Cannabinoid binding sites exist throughout the central and peripheral nervous systems. The two most relevant receptors for cannabinoids are the CB1 and CB2 receptors, which are expressed predominantly in the brain and immune system respectively.[15]Density of expression varies based on species and correlates with the efficacy that cannabinoids will have in modulating specific aspects of behavior related to the site of expression. For example, in rodents, the highest concentration of cannabinoid binding sites are in the basal ganglia and cerebellum, regions of the brain involved in the initiation and coordination of movement.[16] In humans, cannabinoid receptors exist in much lower concentration in these regions, which helps explain why cannabinoids possess a greater efficacy in altering rodent motor movements than they do in humans.

A recent analysis of cannabinoid binding in CB1 and CB2 receptor knockout mice found cannabinoid responsiveness even when these receptors were not being expressed, indicating that an additional binding receptor may be present in the brain.[16] Binding has been demonstrated by 2-arachidonoylglycerol(2-AG) on the TRPV1 receptor suggesting that this receptor may be a candidate for the established response.[17]

In addition to CB1 and CB2, certain orphan receptors are known to bind endocannabinoids as well, including GPR18,GPR55 (a regulator of neuroimmune function), and GPR119. CB1 has also been noted to form a functional human receptor heterodimer in orexin neurons with OX1, the CB1–OX1 receptor, which mediates feeding behavior and certain physical processes such as cannabinoid-induced pressor responseswhich are known to occur through signaling in the rostral ventrolateral medulla.[18][19]

Endocannabinoid synthesis, release, and degradationEdit

During neurotransmission, the pre-synaptic neuron releases neurotransmitters into thesynaptic cleft which bind to cognate receptors expressed on the post-synaptic neuron. Based upon the interaction between the transmitter and receptor, neurotransmitters may trigger a variety of effects in the post-synaptic cell, such as excitation, inhibition, or the initiation ofsecond messenger cascades. Based on the cell, these effects may result in the on-site synthesis of endogenous cannabinoidsanandamide or 2-AG by a process that is not entirely clear, but results from an elevation in intracellular calcium.[15] Expression appears to be exclusive, so that both types of endocannabinoids are not co-synthesized. This exclusion is based on synthesis-specific channel activation: a recent study found that in the bed nucleus of the stria terminalis, calcium entry through voltage-sensitive calcium channels produced an L-type current resulting in 2-AG production, while activation of mGluR1/5 receptors triggered the synthesis of anandamide.[17]

Evidence suggests that the depolarization-induced influx of calcium into the post-synaptic neuron causes the activation of an enzyme called transacylase. This enzyme is suggested to catalyze the first step of endocannabinoid biosynthesis by convertingphosphatidylethanolamine, a membrane-resident phospholipid, into N-acyl-phosphatidylethanolamine (NAPE). Experiments have shown that phospholipase D cleaves NAPE to yield anandamide.[20][21]This process is mediated by bile acids.[22] In NAPE-phospholipase D (NAPEPLD)-knockout mice, cleavage of NAPE is reduced in low calcium concentrations, but not abolished, suggesting multiple, distinct pathways are involved in anandamide synthesis.[23] The synthesis of 2-AG is less established and warrants further research.

Once released into the extracellular space by a putative endocannabinoid transporter, messengers are vulnerable to glial cellinactivation. Endocannabinoids are taken up by a transporter on the glial cell and degraded by fatty acid amide hydrolase (FAAH), which cleaves anandamide into arachidonic acidand ethanolamine or monoacylglycerol lipase(MAGL), and 2-AG into arachidonic acid and glycerol.[24] While arachidonic acid is a substrate for leukotriene and prostaglandinsynthesis, it is unclear whether this degradative byproduct has unique functions in the central nervous system.[25][26] Emerging data in the field also points to FAAH being expressed in postsynaptic neurons complementary to presynaptic neurons expressing cannabinoid receptors, supporting the conclusion that it is major contributor to the clearance and inactivation of anandamide and 2-AG after endocannabinoid reuptake.[16]A neuropharmacological study demonstrated that an inhibitor of FAAH (URB597) selectively increases anandamide levels in the brain of rodents and primates. Such approaches could lead to the development of new drugs with analgesic, anxiolytic-like and antidepressant-like effects, which are not accompanied by overt signs of abuse liability.[27]

Binding and intracellular effectsEdit

Cannabinoid receptors are G-protein coupled receptors located on the pre-synaptic membrane. While there have been some papers that have linked concurrent stimulation of dopamine and CB1 receptors to an acute rise in cyclic adenosine monophosphate (cAMP) production, it is generally accepted that CB1 activation via cannabinoids causes a decrease in cAMP concentration by inhibition of adenylyl cyclase and a rise in the concentration ofmitogen-activated protein kinase (MAP kinase).[4][16] The relative potency of different cannabinoids in inhibition of adenylyl cyclase correlates with their varying efficacy in behavioral assays. This inhibition of cAMP is followed by phosphorylation and subsequent activation of not only a suite of MAP kinases (p38/p42/p44), but also the PI3/PKB andMEK/ERK pathway (Galve-Roperh et al., 2002; Davis et al., 2005; Jones et al., 2005; Grahamet al., 2006). Results from rat hippocampalgene chip data after acute administration oftetrahydrocannabinol (THC) showed an increase in the expression of transcripts encoding myelin basic protein, endoplasmic proteins, cytochrome oxidase, and two cell adhesion molecules: NCAM, andSC1[disambiguation needed]; decreases in expression were seen in both calmodulin andribosomal RNAs (Kittler et al., 2000). In addition, CB1 activation has been demonstrated to increase the activity of transcription factors like c-Fos and Krox-24(Graham et al., 2006).

Binding and neuronal excitabilityEdit

The molecular mechanisms of CB1-mediated changes to the membrane voltage have also been studied in detail. CB1 agonists reduce calcium influx by blocking the activity of voltage-dependent N-, P/Q- and L-typecalcium channels.[28][29] In addition to acting on calcium channels, activation of Gi/o andGs, the two most commonly coupled G-proteins to cannabinoid receptors, has been shown to modulate potassium channelactivity. Recent studies have found that CB1activation specifically facilitates potassium ion flux through GIRKs, a family of potassium channels.[29] Immunohistochemistry experiments demonstrated that CB1 is co-localized with GIRK and Kv1.4 potassium channels, suggesting that these two may interact in physiological contexts.[30]

In the central nervous system, CB1 receptors influence neuronal excitability, reducing the incoming synaptic input.[31] This mechanism, known as presynaptic inhibition, occurs when a postsynaptic neuron releases endocannabinoids in retrograde transmission, which then bind to cannabinoid receptors on the presynaptic terminal. CB1receptors then reduce the amount of neurotransmitter released, so that subsequent excitation in the presynaptic neuron results in diminished effects on the postsynaptic neuron. It is likely that presynaptic inhibition uses many of the same ion channel mechanisms listed above, although recent evidence has shown that CB1receptors can also regulate neurotransmitter release by a non-ion channel mechanism, i.e. through Gi/o-mediated inhibition of adenylyl cyclase and protein kinase A.[32] Direct effects of CB1 receptors on membrane excitability have been reported, and strongly impact the firing of cortical neurons.[33] A series of behavioral experiments demonstrated that NMDAR, an ionotropicglutamate receptor, and the metabotropic glutamate receptors (mGluRs) work in concert with CB1 to induce analgesia in mice, although the mechanism underlying this effect is unclear.[citation needed]

Functions of the endocannabinoid systemEdit

MemoryEdit

Mice treated with tetrahydrocannabinol (THC) show suppression of long-term potentiation in the hippocampus, a process that is essential for the formation and storage of long-term memory.[34] These results concur with anecdotal evidence suggesting that smoking Cannabis impairs short-term memory.[35] Consistent with this finding, mice without the CB1 receptor show enhancedmemory and long-term potentiation indicating that the endocannabinoid system may play a pivotal role in the extinction of old memories. One study found that the high-dose treatment of rats with the synthetic cannabinoid HU-210over several weeks resulted in stimulation of neural growth in the rats’ hippocampusregion, a part of the limbic system playing a part in the formation of declarative andspatial memories, but did not investigate the effects on short-term or long-term memory.[36] Taken together, these findings suggest that the effects of endocannabinoids on the various brain networks involved in learning and memory may vary.

Role in hippocampal neurogenesisEdit

In the adult brain, the endocannabinoid system facilitates the neurogenesis of hippocampal granule cells.[36][37] In thesubgranular zone of the dentate gyrus, multipotent neural progenitors (NP) give rise to daughter cells that, over the course of several weeks, mature into granule cells whose axons project to and synapse onto dendrites on the CA3 region.[38] NPs in the hippocampus have been shown to possess fatty acid amide hydrolase (FAAH) and express CB1 and utilize 2-AG.[37] Intriguingly, CB1 activation by endogenous or exogenous cannabinoids promote NP proliferation and differentiation; this activation is absent in CB1knockouts and abolished in the presence of antagonist.[36][37]

Induction of synaptic depressionEdit

The inhibitory effects of cannabinoid receptor stimulation on neurotransmitter release have caused this system to be connected to various forms of depressant plasticity. A recent study conducted with the bed nucleusof the stria terminalis found that the endurance of the depressant effects was mediated by two different signaling pathways based on the type of receptor activated. 2-AG was found to act on presynaptic CB1receptors to mediate retrograde short-term depression (STD) following activation of L-type calcium currents, while anandamide was synthesized after mGluR5 activation and triggered autocrine signalling onto postsynapic TRPV1 receptors that inducedlong-term depression (LTD). Similar post-synaptic receptor dependencies were found in the striatum, but here both effects relied on presynaptic CB1 receptors.[17] These findings provide the brain a direct mechanism to selectively inhibit neuronal excitability over variable time scales. By selectively internalizing different receptors, the brain may limit the production of specific endocannabinoids to favor a time scale in accordance with its needs.

AppetiteEdit

Evidence for the role of the endocannabinoid system in food-seeking behavior comes from a variety of cannabinoid studies. Emerging data suggests that THC acts via CB1receptors in the hypothalamic nuclei to directly increase appetite.[39] It is thought that hypothalamic neurons tonically produce endocannabinoids that work to tightly regulate hunger. The amount of endocannabinoids produced is inversely correlated with the amount of leptin in the blood.[40] For example, mice without leptin not only become massively obese but express abnormally high levels of hypothalamic endocannabinoids as a compensatory mechanism.[9] Similarly, when these mice were treated with an endocannabinoid inverse agonists, such as rimonabant, food intake was reduced.[9] When the CB1 receptor is knocked out in mice, these animals tend to be leaner and less hungry than wild-type mice. A related study examined the effect of THC on the hedonic (pleasure) value of food and found enhanced dopamine release in thenucleus accumbens and increased pleasure-related behavior after administration of a sucrose solution.[41] A related study found that endocannabinoids affect taste perception in taste cells[42] In taste cells, endocannabinoids were shown to selectively enhance the strength of neural signaling for sweet tastes, whereas leptin decreased the strength of this same response. While there is need for more research, these results suggest that cannabinoid activity in the hypothalamus and nucleus accumbens is related to appetitive, food-seeking behavior.[39]

Energy balance & metabolismEdit

The endocannabinoid system has been shown to have a homeostatic role by controlling several metabolic functions, such as energy storage and nutrient transport. It acts on peripheral tissues such asadipocytes, hepatocytes, the gastrointestinal tract, the skeletal muscles and the endocrinepancreas. It has also been implied in modulating insulin sensitivity. Through all of this, the endocannabinoid system may play a role in clinical conditions, such as obesity,diabetes, and atherosclerosis, which may also give it a cardiovascular role.[43]

Stress responseEdit

While the secretion of glucocorticoids in response to stressful stimuli is an adaptive response necessary for an organism to respond appropriately to a stressor, persistent secretion may be harmful. The endocannabinoid system has been implicated in the habituation of the hypothalamic-pituitary-adrenal axis (HPA axis) to repeated exposure to restraint stress. Studies have demonstrated differential synthesis of anandamide and 2-AG during tonic stress. A decrease of anandamide was found along the axis that contributed to basal hypersecretion of corticosterone; in contrast, an increase of 2-AG was found in the amygdala after repeated stress, which was negatively correlated to magnitude of the corticosterone response. All effects were abolished by the CB1 antagonist AM251, supporting the conclusion that these effects were cannabinoid-receptor dependent.[44] These findings show that anandamide and 2-AG divergently regulate the HPA axis response to stress: while habituation of the stress-induced HPA axis via 2-AG prevents excessive secretion of glucocorticoids to non-threatening stimuli, the increase of basal corticosterone secretion resulting from decreased anandamide allows for a facilitated response of the HPA axis to novel stimuli.

Exploration, social behavior, and anxietyEdit

Prolonged, systemic exposure to cannabinoids has often been associated with anti-social effects. To investigate this theory, a cannabinoid receptor-knockout mouse study examined the effect that these receptors play on exploratory behavior. Researchers selectively targeted glutamatergic and GABAergic cortical interneurons and studied results in open field, novel object, and sociability tests. Eliminating glutamaterigic cannabinoid receptors led to decreased object exploration, social interactions, and increased aggressive behavior. In contrast, GABAergic cannabinoid receptor-knockout mice showed increased exploration of objects, socialization, and open field movement.[45] These contrasting effects reveal the importance of the endocannabinoid system in regulatinganxiety-dependent behavior. Results suggest that glutamatergic cannabinoid receptors are not only responsible for mediating aggression, but produce an anxiolytic-like function by inhibiting excessive arousal: excessive excitation produces anxiety that limited the mice from exploring both animate and inanimate objects. In contrast, GABAergic neurons appear to control an anxiogenic-like function by limiting inhibitory transmitter release. Taken together, these two sets of neurons appear to help regulate the organism’s overall sense of arousal during novel situations.

Immune functionEdit

Evidence suggests that endocannabinoids may function as both neuromodulators and immunomodulators in the immune system. Here, they seem to serve an autoprotective role to ameliorate muscle spasms, inflammation, and other symptoms ofmultiple sclerosis and skeletal muscle spasms.[4] Functionally, the activation of cannabinoid receptors has been demonstrated to play a role in the activation of GTPases in macrophages, neutrophils, andBM cells. These receptors have also been implicated in the proper migration of B cellsinto the marginal zone (MZ) and the regulation of healthy IgM levels.[46]Interestingly, some disorders seem to trigger an upregulation of cannabinoid receptors selectively in cells or tissues related to symptom relief and inhibition of disease progression, such as in that rodent neuropathic pain model, where receptors are increased in the spinal cord microglia, dorsal root ganglion, and thalmic neurons.[15]

Multiple sclerosisEdit

Historical records from ancient China and Greece suggest that preparations of Cannabis indica were commonly prescribed to ameliorate multiple sclerosis-like symptoms such as tremors and muscle pain. Modern research has confirmed these effects in a study on diseased mice, wherein both endogenous and exogenous agonists showed ameliorating effects on tremor and spasticity. It remains to be seen whether pharmaceutical preparations such as dronabinol have the same effects in humans.[47][48] Due to increasing use of medical Cannabis and rising incidence of multiple sclerosis patients who self-medicate with the drug, there has been much interest in exploiting the endocannabinoid system in the cerebellum to provide a legal and effective relief.[35] In mouse models of multiple sclerosis, there is a profound reduction and reorganization of CB1receptors in the cerebellum.[49] Serial sections of cerebellar tissue subjected toimmunohistochemistry revealed that this aberrant expression occurred during the relapse phase but returned to normal during the remitting phase of the disease.[49] Other studies suggest that CB1 agonists promote the survival of oligodendrocytes in vitro in the absence of growth and trophic factors; in addition, these agonist have been shown to promote mRNA expression of myelin lipid protein. (Kittler et al., 2000; Mollna-Holgado et al., 2002). Taken together, these studies point to the exciting possibility that cannabinoid treatment may not only be able to attenuate the symptoms of multiple sclerosis but also improve oligodendrocyte function (reviewed in Pertwee, 2001; Mollna-Holgado et al., 2002). 2-AG stimulates proliferation of amicroglial cell line by a CB2 receptor dependent mechanism, and the number of microglial cells is increased in multiple sclerosis.[50]

Female reproductionEdit

The developing embryo expresses cannabinoid receptors early in development that are responsive to anandamide secreted in the uterus. This signaling is important in regulating the timing of embryonic implantation and uterine receptivity. In mice, it has been shown that anandamide modulates the probability of implantation to the uterine wall. For example, in humans, the likelihood of miscarriage increases if uterine anandamide levels are too high or low.[51]These results suggest that intake of exogenous cannabinoids (e.g. marijuana) can decrease the likelihood for pregnancy for women with high anandamide levels, and alternatively, it can increase the likelihood for pregnancy in women whose anandamide levels were too low.[52][53]

Autonomic nervous systemEdit

Peripheral expression of cannabinoid receptors led researchers to investigate the role of cannabinoids in the autonomic nervous system. Research found that the CB1receptor is expressed presynaptically by motor neurons that innervate visceral organs. Cannabinoid-mediated inhibition of electric potentials results in a reduction in noradrenaline release from sympathetic nervous system nerves. Other studies have found similar effects in endocannabinoid regulation of intestinal motility, including the innervation of smooth muscles associated with the digestive, urinary, and reproductive systems.[16]

AnalgesiaEdit

At the spinal cord, cannabinoids suppress noxious-stimulus-evoked responses of neurons in the dorsal horn, possibly by modulating descending noradrenaline input from the brainstem.[16] As many of these fibers are primarily GABAergic, cannabinoid stimulation in the spinal column results in disinhibition that should increase noradrenaline release and attenuation of noxious-stimuli-processing in the periphery and dorsal root ganglion.

The endocannabinoid most researched in pain is palmitoylethanolamide. Palmitoylethanolamide is a fatty amine related to anandamide, but saturated and although initially it was thought that palmitoylethanolamide would bind to the CB1 and the CB2 receptor, later it was found that the most important receptors are the PPAR-alpha receptor, the TRPV receptor and the GRP55 receptor.[spelling?]Palmitoylethanolamide has been evaluated for its analgesic actions in a great variety of pain indications[54] and found to be safe and effective. Basically these data are proof of concept for endocannabinoids and related fatty amines to be therapeutically useful analgesics; palmitoylethanolamide is available under the brand names Normast and PeaPure as nutraceuticals.

Endocannabinoids are involved in placeboinduced analgesia responses.[55]

ThermoregulationEdit

Anandamide and N-arachidonoyl dopamine(NADA) have been shown to act on temperature-sensing TRPV1 channels, which are involved in thermoregulation.[56] TRPV1 is activated by the exogenous ligand capsaicin, the active component of chili peppers, which is structurally similar to endocannabinoids. NADA activates the TRPV1 channel with anEC50 of approximately of 50 nM.[clarify] The high potency makes it the putative endogenous TRPV1 agonist.[57] Anandamide has also been found to activate TRPV1 on sensory neuron terminals, and subsequently cause vasodilation.[16] TRPV1 may also be activated by methanandamide andarachidonyl-2′-chloroethylamide (ACEA).[4]

SleepEdit

Increased endocannabinoid signaling within the central nervous system promotes sleep-inducing effects. Intercerebroventricularadministration of anandamide in rats has been shown to decrease wakefulness and increase slow-wave sleep and REM sleep.[58]Administration of anandamide into the basal forebrain of rats has also been shown to increase levels of adenosine, which plays a role in promoting sleep and suppressing arousal.[59] REM sleep deprivation in rats has been demonstrated to increase CB1 receptor expression in the central nervous system.[60]Furthermore, anandamide levels possess acircadian rhythm in the rat, with levels being higher in the light phase of the day, which is when rats are usually asleep or less active, since they are nocturnal.[61]

Experimental use of CB1 -/- phenotypeEdit

Neuroscientists often utilize transgenic CB1knockout mice to discern novel roles for the endocannabinoid system. While CB1knockout mice are healthy and live into adulthood, there are significant differences between CB1 knockout and wild-type mice. When subjected to a high-fat diet, CB1knockout mice tend to be about sixty percent leaner and slightly less hungry than wildtype.[62] Compared to wildtype, CB1knockout mice exhibit severe deficits in motor learning, memory retrieval, and increased difficulty in completing the Morris water maze.[8][63][64] There is also evidence indicating that these knockout animals have an increased incidence and severity of strokeand seizure.[65][66]

ReferencesEdit

  1. ^ a b Grotenhermen, Franjo (23 Jul 2012).“The Therapeutic Potential of Cannabis and Cannabinoids”. Dtsch Arztebl Int. 109(PMC3442177): 495–501.doi:10.3238/arztebl.2012.0495.PMC 3442177. PMID 23008748.
  2. ^ Blázquez, C; Chiarlone, A; Bellocchio, L; Resel, E; Pruunsild, P; García-Rincón, D; Sendtner, M; Timmusk, T; Lutz, B; Galve-Roperh, I; Guzmán, M (20 February 2015).“The CB1 cannabinoid receptor signals striatal neuroprotection via a PI3K/Akt/mTORC1/BDNF pathway”. Cell Death and Differentiation 22 (10): 1618–1629.doi:10.1038/cdd.2015.11.
  3. ^ Russo, Ethan B (August 2011). “Taming THC: potential cannabis synergy and phytocannabinoid-terpenoid entourage effects”. British Journal of Pharmacology 163(7): 1344–1364. doi:10.1111/j.1476-5381.2011.01238.x. PMC 3165946.PMID 21749363.
  4. ^ a b c d Pertwee RG (April 2006). “The pharmacology of cannabinoid receptors and their ligands: an overview”. Int J Obes (Lond)30 (Suppl 1): S13–8.doi:10.1038/sj.ijo.0803272.PMID 16570099.
  5. ^ Fortin DA, Levine ES (2007). “Differential effects of endocannabinoids on glutamatergic and GABAergic inputs to layer 5 pyramidal neurons”. Cereb. Cortex 17 (1): 163–74. doi:10.1093/cercor/bhj133.PMID 16467564.
  6. ^ Good CH (2007). “Endocannabinoid-dependent regulation of feedforward inhibition in cerebellar Purkinje cells”. J. Neurosci. 27 (1): 1–3.doi:10.1523/JNEUROSCI.4842-06.2007.PMID 17205618.
  7. ^ Hashimotodani Y, Ohno-Shosaku T, Kano M (2007). “Presynaptic monoacylglycerol lipase activity determines basal endocannabinoid tone and terminates retrograde endocannabinoid signaling in the hippocampus”. J. Neurosci. 27 (5): 1211–9.doi:10.1523/JNEUROSCI.4159-06.2007.PMID 17267577.
  8. ^ a b Kishimoto Y, Kano M (2006). “Endogenous cannabinoid signaling through the CB1 receptor is essential for cerebellum-dependent discrete motor learning”. J. Neurosci. 26 (34): 8829–37.doi:10.1523/JNEUROSCI.1236-06.2006.PMID 16928872.
  9. ^ a b c Di Marzo V, Goparaju SK, Wang L, Liu J, Bátkai S, Járai Z, Fezza F, Miura GI, Palmiter RD, Sugiura T, Kunos G (April 2001). “Leptin-regulated endocannabinoids are involved in maintaining food intake”. Nature410 (6830): 822–5. doi:10.1038/35071088.PMID 11298451.
  10. ^ Cravatt BF, et al. (July 2001).“Supersensitivity to anandamide and enhanced endogenous cannabinoid signaling in mice lacking fatty acid amide hydrolase”.Proc. Natl. Acad. Sci. U.S.A. 98 (16): 9371–6.Bibcode:2001PNAS…98.9371C.doi:10.1073/pnas.161191698.JSTOR 3056353. PMC 55427.PMID 11470906.
  11. ^ a b c Flores A, Maldonado R, Berrendero F (2013). “Cannabinoid-hypocretin cross-talk in the central nervous system: what we know so far”. Front Neurosci 7: 256.doi:10.3389/fnins.2013.00256.PMC 3868890. PMID 24391536. Direct CB1-HcrtR1 interaction was first proposed in 2003 (Hilairet et al., 2003). Indeed, a 100-fold increase in the potency of hypocretin-1 to activate the ERK signaling was observed when CB1 and HcrtR1 were co-expressed … In this study, a higher potency of hypocretin-1 to regulate CB1-HcrtR1 heteromer compared with the HcrtR1-HcrtR1 homomer was reported (Ward et al., 2011b). These data provide unambiguous identification of CB1-HcrtR1 heteromerization, which has a substantial functional impact. … The existence of a cross-talk between the hypocretinergic and endocannabinoid systems is strongly supported by their partially overlapping anatomical distribution and common role in several physiological and pathological processes. However, little is known about the mechanisms underlying this interaction.
    Figure 1: Schematic of brain CB1 expression and orexinergic neurons expressing OX1 or OX2
    Figure 2: Synaptic signaling mechanisms in cannabinoid and orexin systems
    Figure 3: Schematic of brain pathways involved in food intake
  12. ^ Watkins BA, Kim J (2014). “The endocannabinoid system: directing eating behavior and macronutrient metabolism”.Front Psychol 5: 1506.doi:10.3389/fpsyg.2014.01506.PMC 4285050. PMID 25610411. CB1 is present in neurons of the enteric nervous system and in sensory terminals of vagal and spinal neurons in the gastrointestinal tract (Massa et al., 2005). Activation of CB1 is shown to modulate nutrient processing, such as gastric secretion, gastric emptying, and intestinal motility. … CB1 is shown to co-localize with the food intake inhibiting neuropeptide, corticotrophin-releasing hormone, in the paraventricular nucleus of the hypothalamus, and with the two orexigenic peptides, melanin-concentrating hormone in the lateral hypothalamus and with pre-pro-orexin in the ventromedial hypothalamus (Inui, 1999; Horvath, 2003). CB1 knockout mice showed higher levels of CRH mRNA, suggesting that hypothalamic EC receptors are involved in energy balance and may be able to mediate food intake (Cota et al., 2003). … The ECS works through many anorexigenic and orexigenic pathways where ghrelin, leptin, adiponectin, endogenous opioids, and corticotropin-releasing hormones are involved (Viveros et al., 2008).
  13. ^ Thompson MD, Xhaard H, Sakurai T, Rainero I, Kukkonen JP (2014). “OX1 and OX2 orexin/hypocretin receptor pharmacogenetics”. Front Neurosci 8: 57.doi:10.3389/fnins.2014.00057.PMC 4018553. PMID 24834023. OX1–CB1 dimerization was suggested to strongly potentiate orexin receptor signaling, but a likely explanation for the signal potentiation is, instead, offered by the ability of OX1 receptor signaling to produce 2-arachidonoyl glycerol, a CB1 receptor ligand, and a subsequent co-signaling of the receptors (Haj-Dahmane and Shen, 2005; Turunen et al., 2012; Jäntti et al., 2013). However, this does not preclude dimerization.
  14. ^ Jäntti MH, Mandrika I, Kukkonen JP (2014). “Human orexin/hypocretin receptors form constitutive homo- and heteromeric complexes with each other and with human CB1 cannabinoid receptors”. Biochem. Biophys. Res. Commun. 445 (2): 486–90.doi:10.1016/j.bbrc.2014.02.026.PMID 24530395. Orexin receptor subtypes readily formed homo- and hetero(di)mers, as suggested by significant BRET signals. CB1 receptors formed homodimers, and they also heterodimerized with both orexin receptors. … In conclusion, orexin receptors have a significant propensity to make homo- and heterodi-/oligomeric complexes. However, it is unclear whether this affects their signaling. As orexin receptors efficiently signal via endocannabinoid production to CB1 receptors, dimerization could be an effective way of forming signal complexes with optimal cannabinoid concentrations available for cannabinoid receptors.
  15. ^ a b c Pertwee RG (January 2008). “The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin”. Br. J. Pharmacol.153 (2): 199–215.doi:10.1038/sj.bjp.0707442.PMC 2219532. PMID 17828291.
  16. ^ a b c d e f g Elphick MR, Egertová M (March 2001). “The neurobiology and evolution of cannabinoid signalling”. Philos. Trans. R. Soc. Lond., B, Biol. Sci. 356 (1407): 381–408. doi:10.1098/rstb.2000.0787.PMC 1088434. PMID 11316486.
  17. ^ a b c Puente N, Cui Y, Lassalle O, Lafourcade M, Georges F, Venance L, Grandes P, Manzoni OJ (December 2011). “Polymodal activation of the endocannabinoid system in the extended amygdala”. Nat. Neurosci. 14 (12): 1542–7.doi:10.1038/nn.2974. PMID 22057189.
  18. ^ Ibrahim BM, Abdel-Rahman AA (2014).“Cannabinoid receptor 1 signaling in cardiovascular regulating nuclei in the brainstem: A review”. J Adv Res 5 (2): 137–45. doi:10.1016/j.jare.2013.03.008.PMC 4294710. PMID 25685481.
  19. ^ Ibrahim BM, Abdel-Rahman AA (2015). “A pivotal role for enhanced brainstem Orexin receptor 1 signaling in the central cannabinoid receptor 1-mediated pressor response in conscious rats”. Brain Res. 1622: 51–63. doi:10.1016/j.brainres.2015.06.011.PMID 26096126. Orexin receptor 1 (OX1R) signaling is implicated in cannabinoid receptor 1 (CB1R) modulation of feeding. Further, our studies established the dependence of the central CB1R-mediated pressor response on neuronal nitric oxide synthase (nNOS) and extracellular signal-regulated kinase1/2 (ERK1/2) phosphorylation in the RVLM. We tested the novel hypothesis that brainstem orexin-A/OX1R signaling plays a pivotal role in the central CB1R-mediated pressor response. Our multiple labeling immunofluorescence findings revealed co-localization of CB1R, OX1R and the peptide orexin-A within the C1 area of the rostral ventrolateral medulla (RVLM). Activation of central CB1R following intracisternal (i.c.) WIN55,212-2 (15μg/rat) in conscious rats caused significant increases in BP and orexin-A level in RVLM neuronal tissue. Additional studies established a causal role for orexin-A in the central CB1R-mediated pressor response
  20. ^ Okamoto Y, Morishita J, Tsuboi K, Tonai T, Ueda N (February 2004). “Molecular characterization of a phospholipase D generating anandamide and its congeners”. J. Biol. Chem. 279 (7): 5298–305.doi:10.1074/jbc.M306642200.PMID 14634025.
  21. ^ Liu J, Wang L, Harvey-White J, Osei-Hyiaman D, Razdan R, Gong Q, Chan AC, Zhou Z, Huang BX, Kim HY, Kunos G (September 2006). “A biosynthetic pathway for anandamide”. Proc. Natl. Acad. Sci. U.S.A.103 (36): 13345–50.Bibcode:2006PNAS..10313345L.doi:10.1073/pnas.0601832103.PMC 1557387. PMID 16938887.
  22. ^ Magotti P, Bauer I, Igarashi M, Babagoli M, Marotta R, Piomelli D, Garau G (2014). “Structure of Human N-Acylphosphatidylethanolamine-Hydrolyzing Phospholipase D: Regulation of Fatty Acid Ethanolamide Biosynthesis by Bile Acids”.Structure 24 (3): 598–604.doi:10.1016/j.str.2014.12.018.PMID 25684574.
  23. ^ Leung D, Saghatelian A, Simon GM, Cravatt BF (April 2006). “Inactivation of N-acyl phosphatidylethanolamine phospholipase D reveals multiple mechanisms for the biosynthesis of endocannabinoids”.Biochemistry 45 (15): 4720–6.doi:10.1021/bi060163l. PMC 1538545.PMID 16605240.
  24. ^ Pazos MR, Núñez E, Benito C, Tolón RM, Romero J (June 2005). “Functional neuroanatomy of the endocannabinoid system”. Pharmacol. Biochem. Behav. 81 (2): 239–47. doi:10.1016/j.pbb.2005.01.030.PMID 15936805.
  25. ^ Yamaguchi T, Shoyama Y, Watanabe S, Yamamoto T (January 2001). “Behavioral suppression induced by cannabinoids is due to activation of the arachidonic acid cascade in rats”. Brain Res. 889 (1–2): 149–54.doi:10.1016/S0006-8993(00)03127-9.PMID 11166698.
  26. ^ Brock TG (December 2005). “Regulating leukotriene synthesis: the role of nuclear 5-lipoxygenase”. J. Cell. Biochem. 96 (6): 1203–11. doi:10.1002/jcb.20662.PMID 16215982.
  27. ^ Clapper JR, Mangieri RA, Piomelli D (2009). “The endocannabinoid system as a target for the treatment of cannabis dependence”. Neuropharmacology 56 (Suppl 1): 235–43.doi:10.1016/j.neuropharm.2008.07.018.PMC 2647947. PMID 18691603.
  28. ^ Twitchell W, Brown S, Mackie K (1997). “Cannabinoids inhibit N- and P/Q-type calcium channels in cultured rat hippocampal neurons”. J. Neurophysiol. 78 (1): 43–50.PMID 9242259.
  29. ^ a b Guo J, Ikeda SR (2004). “Endocannabinoids modulate N-type calcium channels and G-protein-coupled inwardly rectifying potassium channels via CB1 cannabinoid receptors heterologously expressed in mammalian neurons”. Mol. Pharmacol. 65 (3): 665–74.doi:10.1124/mol.65.3.665.PMID 14978245.
  30. ^ Binzen U, Greffrath W, Hennessy S, Bausen M, Saaler-Reinhardt S, Treede RD (2006). “Co-expression of the voltage-gated potassium channel Kv1.4 with transient receptor potential channels (TRPV1 and TRPV2) and the cannabinoid receptor CB1 in rat dorsal root ganglion neurons”.Neuroscience 142 (2): 527–39.doi:10.1016/j.neuroscience.2006.06.020.PMID 16889902.
  31. ^ Freund TF, Katona I, Piomelli D (2003). “Role of endogenous cannabinoids in synaptic signaling”. Physiol. Rev. 83 (3): 1017–66. doi:10.1152/physrev.00004.2003 (inactive 2015-01-09). PMID 12843414.
  32. ^ Chevaleyre V, Heifets BD, Kaeser PS, Südhof TC, Purpura DP, Castillo PE (2007).“ENDOCANNABINOID-MEDIATED LONG-TERM PLASTICITY REQUIRES cAMP/PKA SIGNALING AND RIM1α”. Neuron 54 (5): 801–12. doi:10.1016/j.neuron.2007.05.020.PMC 2001295. PMID 17553427.
  33. ^ Bacci A, Huguenard JR, Prince DA (2004). “Long-lasting self-inhibition of neocortical interneurons mediated by endocannabinoids”. Nature 431 (7006): 312–6. Bibcode:2004Natur.431..312B.doi:10.1038/nature02913.PMID 15372034.
  34. ^ Hampson RE, Deadwyler SA (1999). “Cannabinoids, hippocampal function and memory”. Life Sci. 65 (6–7): 715–23.doi:10.1016/S0024-3205(99)00294-5.PMID 10462072.
  35. ^ a b Pertwee RG (2001). “Cannabinoid receptors and pain”. Prog. Neurobiol. 63 (5): 569–611. doi:10.1016/S0301-0082(00)00031-9. PMID 11164622.
  36. ^ a b c Jiang W, Zhang Y, Xiao L, Van Cleemput J, Ji SP, Bai G, Zhang X (2005).“Cannabinoids promote embryonic and adult hippocampus neurogenesis and produce anxiolytic- and antidepressant-like effects”.J. Clin. Invest. 115 (11): 3104–16.doi:10.1172/JCI25509. PMC 1253627.PMID 16224541.
  37. ^ a b c Aguado T, Monory K, Palazuelos J, Stella N, Cravatt B, Lutz B, Marsicano G, Kokaia Z, Guzmán M, Galve-Roperh I (2005). “The endocannabinoid system drives neural progenitor proliferation”. FASEB J. 19 (12): 1704–6. doi:10.1096/fj.05-3995fje.PMID 16037095.
  38. ^ Christie BR, Cameron HA (2006). “Neurogenesis in the adult hippocampus”.Hippocampus 16 (3): 199–207.doi:10.1002/hipo.20151. PMID 16411231.
  39. ^ a b Kirkham TC, Tucci SA (2006). “Endocannabinoids in appetite control and the treatment of obesity”. CNS Neurol Disord Drug Targets 5 (3): 272–92.doi:10.2174/187152706777452272.PMID 16787229.
  40. ^ Di Marzo V, Sepe N, De Petrocellis L, Berger A, Crozier G, Fride E, Mechoulam R (December 1998). “Trick or treat from food endocannabinoids?”. Nature 396 (6712): 636–7. Bibcode:1998Natur.396..636D.doi:10.1038/25267. PMID 9872309.
  41. ^ De Luca MA, Solinas M, Bimpisidis Z, Goldberg SR, Di Chiara G (July 2012).“Cannabinoid facilitation of behavioral and biochemical hedonic taste responses”.Neuropharmacology 63 (1): 161–8.doi:10.1016/j.neuropharm.2011.10.018.PMC 3705914. PMID 22063718.
  42. ^ Yoshida R, et al. (January 2010).“Endocannabinoids selectively enhance sweet taste”. Proc. Natl. Acad. Sci. U.S.A. 107(2): 935–9. Bibcode:2010PNAS..107..935Y.doi:10.1073/pnas.0912048107.JSTOR 40535875. PMC 2818929.PMID 20080779.
  43. ^ Bellocchio L, Cervino C, Pasquali R, Pagotto U (June 2008). “The endocannabinoid system and energy metabolism”. J. Neuroendocrinol. 20 (6): 850–7. doi:10.1111/j.1365-2826.2008.01728.x.PMID 18601709.
  44. ^ Hill MN, McLaughlin RJ, Bingham B, Shrestha L, Lee TT, Gray JM, Hillard CJ, Gorzalka BB, Viau V (May 2010).“Endogenous cannabinoid signaling is essential for stress adaptation”. Proc. Natl. Acad. Sci. U.S.A. 107 (20): 9406–11.Bibcode:2010PNAS..107.9406H.doi:10.1073/pnas.0914661107.PMC 2889099. PMID 20439721.
  45. ^ Häring M, Kaiser N, Monory K, Lutz B (2011). Burgess HA, ed. “Circuit specific functions of cannabinoid CB1 receptor in the balance of investigatory drive and exploration”. PLoS ONE 6 (11): e26617.Bibcode:2011PLoSO…626617H.doi:10.1371/journal.pone.0026617.PMC 3206034. PMID 22069458.
  46. ^ Basu S, Ray A, Dittel BN (December 2011). “Cannabinoid receptor 2 is critical for the homing and retention of marginal zone B lineage cells and for efficient T-independent immune responses”. J. Immunol. 187 (11): 5720–32. doi:10.4049/jimmunol.1102195.PMC 3226756. PMID 22048769.
  47. ^ Baker D, Pryce G, Croxford JL, Brown P, Pertwee RG, Huffman JW, Layward L (2000). “Cannabinoids control spasticity and tremor in a multiple sclerosis model”. Nature 404(6773): 84–7. doi:10.1038/35003583.PMID 10716447.
  48. ^ Baker D, Pryce G, Croxford JL, Brown P, Pertwee RG, Makriyannis A, Khanolkar A, Layward L, Fezza F, Bisogno T, Di Marzo V (2001). “Endocannabinoids control spasticity in a multiple sclerosis model”. FASEB J. 15(2): 300–2. doi:10.1096/fj.00-0399fje.PMID 11156943.
  49. ^ a b Cabranes A, Pryce G, Baker D, Fernández-Ruiz J (August 2006). “Changes in CB1 receptors in motor-related brain structures of chronic relapsing experimental allergic encephalomyelitis mice”. Brain Res.1107 (1): 199–205.doi:10.1016/j.brainres.2006.06.001.PMID 16822488.
  50. ^ Carrier EJ, Kearn CS, Barkmeier AJ, Breese NM, Yang W, Nithipatikom K, Pfister SL, Campbell WB, Hillard CJ (April 2004). “Cultured rat microglial cells synthesize the endocannabinoid 2-arachidonylglycerol, which increases proliferation via a CB2 receptor-dependent mechanism”. Mol. Pharmacol. 65 (4): 999–1007.doi:10.1124/mol.65.4.999.PMID 15044630.
  51. ^ Maccarrone M, Valensise H, Bari M, Lazzarin N, Romanini C, Finazzi-Agrò A (2000). “Relation between decreased anandamide hydrolase concentrations in human lymphocytes and miscarriage”. Lancet355 (9212): 1326–9. doi:10.1016/S0140-6736(00)02115-2. PMID 10776746.
  52. ^ Das SK, Paria BC, Chakraborty I, Dey SK (1995). “Cannabinoid ligand-receptor signaling in the mouse uterus”. Proc. Natl. Acad. Sci. U.S.A. 92 (10): 4332–6.Bibcode:1995PNAS…92.4332D.doi:10.1073/pnas.92.10.4332.PMC 41938. PMID 7753807.
  53. ^ Paria BC, Das SK, Dey SK (1995). “The preimplantation mouse embryo is a target for cannabinoid ligand-receptor signaling”.Proc. Natl. Acad. Sci. U.S.A. 92 (21): 9460–4.Bibcode:1995PNAS…92.9460P.doi:10.1073/pnas.92.21.9460.PMC 40821. PMID 7568154.
  54. ^ Hesselink JM (2012). “New Targets in Pain, Non-Neuronal Cells, and the Role of Palmitoylethanolamide”. The Open Pain Journal 5 (1): 12–23.doi:10.2174/1876386301205010012.
  55. ^ Colloca, Luana (2013-08-28). Placebo and Pain: From Bench to Bedside (1st ed.). Elsevier Science. pp. 11–12.ISBN 9780123979315.
  56. ^ Ross RA (November 2003).“Anandamide and vanilloid TRPV1 receptors”. Br. J. Pharmacol. 140 (5): 790–801. doi:10.1038/sj.bjp.0705467.PMC 1574087. PMID 14517174.
  57. ^ Huang SM, Bisogno T, Trevisani M, Al-Hayani A, De Petrocellis L, Fezza F, Tognetto M, Petros TJ, Krey JF, Chu CJ, Miller JD, Davies SN, Geppetti P, Walker JM, Di Marzo V (June 2002). “An endogenous capsaicin-like substance with high potency at recombinant and native vanilloid VR1 receptors”. Proc. Natl. Acad. Sci. U.S.A. 99 (12): 8400–5.Bibcode:2002PNAS…99.8400H.doi:10.1073/pnas.122196999.PMC 123079. PMID 12060783.
  58. ^ Murillo-Rodríguez E, Sánchez-Alavez M, Navarro L, Martínez-González D, Drucker-Colín R, Prospéro-García O (November 1998). “Anandamide modulates sleep and memory in rats”. Brain Res. 812 (1–2): 270–4.doi:10.1016/S0006-8993(98)00969-X.PMID 9813364.
  59. ^ Santucci V, Storme JJ, Soubrié P, Le Fur G (1996). “Arousal-enhancing properties of the CB1 cannabinoid receptor antagonist SR 141716A in rats as assessed by electroencephalographic spectral and sleep-waking cycle analysis”. Life Sci. 58 (6): PL103–10. doi:10.1016/0024-3205(95)02319-4. PMID 8569415.
  60. ^ Wang L, Yang T, Qian W, Hou X (January 2011). “The role of endocannabinoids in visceral hyposensitivity induced by rapid eye movement sleep deprivation in rats: regional differences”. Int. J. Mol. Med. 27 (1): 119–26.doi:10.3892/ijmm.2010.547.PMID 21057766.
  61. ^ Murillo-Rodriguez E, Désarnaud F, Prospéro-García O (May 2006). “Diurnal variation of arachidonoylethanolamine, palmitoylethanolamide and oleoylethanolamide in the brain of the rat”.Life Sci. 79 (1): 30–7.doi:10.1016/j.lfs.2005.12.028.PMID 16434061.
  62. ^ Ravinet Trillou C, Delgorge C, Menet C, Arnone M, Soubrié P (2004). “CB1 cannabinoid receptor knockout in mice leads to leanness, resistance to diet-induced obesity and enhanced leptin sensitivity”. Int. J. Obes. Relat. Metab. Disord. 28 (4): 640–8.doi:10.1038/sj.ijo.0802583.PMID 14770190.
  63. ^ Varvel SA, Lichtman AH (2002). “Evaluation of CB1 receptor knockout mice in the Morris water maze”. J. Pharmacol. Exp. Ther. 301 (3): 915–24.doi:10.1124/jpet.301.3.915.PMID 12023519.
  64. ^ Niyuhire F, Varvel SA, Martin BR, Lichtman AH (2007). “Exposure to marijuana smoke impairs memory retrieval in mice”. J. Pharmacol. Exp. Ther. 322 (3): 1067–75.doi:10.1124/jpet.107.119594.PMID 17586723.
  65. ^ Parmentier R, Ohtsu H, Djebbara-Hannas Z, Valatx JL, Watanabe T, Lin JS (September 2002). “Anatomical, physiological, and pharmacological characteristics of histidine decarboxylase knockout mice: evidence for the role of brain histamine in behavioral and sleep-wake control”. J. Neurosci. 22 (17): 7695–711. PMID 12196593.
  66. ^ Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, Cannich A, Azad SC, Cascio MG, Gutiérrez SO, van der Stelt M, López-Rodriguez ML, Casanova E, Schütz G, Zieglgänsberger W, Di Marzo V, Behl C, Lutz B (October 2003). “CB1 cannabinoid receptors and on-demand defense against excitotoxicity”. Science 302 (5642): 84–8.Bibcode:2003Sci…302…84M.doi:10.1126/science.1088208.PMID 14526074.

Further readingEdit

External linksEdit

Read in another language
Last edited 3 days ago by an anonymous user

Products for your Endocannabinoid system

Looking for a CBD Affiliate Program? Visit CBDPure to sign up.

Hempy CBD Oil © 2022 - - The sale, production and distribution of CBD oils/products derived from imported raw material industrial hemp is not in violation of the Controlled Substances Act (CSA). CBD, and even THC, when in industrial hemp oil, are legal. The importation of industrial hemp is lawful, as exempt parts of the “marijuana” plant such as industrial hemp fiber, stalk, seed and oils can be lawfully imported into the United States and these portions of an industrial hemp plant may enter into United States commerce. The DEA cannot regulate naturally-occurring CBD not contained within or derived from “marijuana,” i.e., non-psychoactive hemp products, because non-psychoactive hemp from the stalks and fibers of industrial hemp are not included in Schedule I. The DEA has no authority to regulate drugs that are not scheduled/defined. Frontier Theme