- Review
- Open access
- Published:
Beyond the hype: a comprehensive exploration of CBD’s biological impacts and mechanisms of action
Journal of Cannabis Research volume 7, Article number: 24 (2025)
Abstract
Background
Cannabidiol (CBD) is the primary non-psychoactive component of cannabis. Consumption of CBD is increasing rapidly as it is federally legal and widely available in the United States, Europe, Mexico, Canada, and Asia. CBD is gaining traction in medical and biochemical research, though a comprehensive classification of CBD receptor interactions is yet to be elucidated.
Methods
A comprehensive literature search across PubMed, Web of Science, and Google Scholar identified studies reporting cannabidiol (CBD) interactions with receptors, enzymes, and biological processes. Eligible articles included cell culture, animal model, biochemical, and clinical studies. Findings were thematically synthesized by body system, emphasizing mechanisms and implications for health and disease.
Results
Herein, I compile the literature to date of known interactions between CBD and various receptors, enzymes, and processes. I discuss the impact of CBD exposure on multiple processes, including endocannabinoid receptors, ion channels, cytochrome 450 enzymes, inflammatory pathways, and sex hormone regulation. I explain the potential effects of CBD on psychiatric disorders, seizure activity, nausea and vomiting, pain sensation, thermal regulation, neuronal signaling, neurodegenerative diseases, reproductive aging, drug metabolism, inflammation, sex hormone regulation, and energy homeostasis.
Conclusions
Understanding how CBD functions and how it can interact with other recreational or pharmaceutical medications is necessary for proper clinical management of patients who consume CBD.
Introduction
Cannabis consumption is increasing rapidly in tandem with increased legalization and availability and decreased social stigma (Patrick et al. 2022). As of 2023, recreational cannabis consumption is legal in 24 states and medicinal cannabis is available in an additional 17 states in the United States (Marijuana legality by state - Updated Oct 1, 2023. DISA 2025). In the United States, roughly 18% of the adult population report consuming cannabis, making it the most consumed federally illicit substance (Results from the 2019 National Survey on Drug Use and Health (NSDUH): Key Substance Use and Mental Health Indicators in the United States | SAMHSA Publications and Digital Products n.d.). Cannabis products contain multiple component parts, including tetrahydrocannabinol (THC), cannabidiol (CBD), and minor cannabinoids and terpenes (Atakan 2012). CBD was removed from the federal schedule 1 drug classification in 2018 (Abernethy 2019) and is now widely available in gas stations and grocery stores in all U.S. states. CBD consumption is dramatically increasing both recreationally and medicinally (Goodman et al. 2022). CBD has multiple medicinal effects, including as a nausea reducing medication, an anxiety reducing medication, a sleep aid (Rapin et al. 2021), and one pharmaceutical CBD product, Epidiolex, is approved by the Food and Drug Administration (FDA) to treat severe childhood seizure (Abu-Sawwa et al. 2020). One inherent limitation with CBD research is the challenge of sourcing product that is pure, reliable, transparent in dosing, and available for various administration routes. As the landscape of synthetic and hemp-derived CBD product changes in the early 2020’s, access to product for research is an actively evolving field. Cannabidiol (CBD) has emerged as a highly popular and rapidly evolving area of research, with several comprehensive reviews published in recent years that explore its pharmacological properties and therapeutic potential. Notable reviews, such as those by Sideris and Doan (2024), Castillo-Arellano et al. (2023), and Vitale et al. (2021), have provided valuable insights into CBD’s effects, particularly its polypharmacological actions in neuropsychiatric conditions (Sideris and Doan 2024; Castillo-Arellano et al. 2023; Vitale et al. 2021). These reviews focus on the complex interactions between CBD and various receptors, contributing to its therapeutic effects in disorders like epilepsy, anxiety, and depression. However, this manuscript expands upon these existing reviews by broadening the scope to include not only neuropsychiatric conditions but also multiple disease states and symptoms. By presenting a comprehensive approach to CBD receptor activation across various pathologies, this review offers a more integrated understanding of how CBD can influence a wide range of therapeutic outcomes, making it a valuable addition to the growing body of CBD literature for both researchers and clinicians.
With increasing recreational and medicinal consumption of CBD, it is pertinent to understand the drug activity. There are many modalities in which a ligand may interact with a receptor. As discussed by Miller and colleagues, receptors may be ion channel receptors, enzyme linked receptors, G-protein-coupled receptors, or nuclear receptors (Miller and Lappin 2023). Ligands can bind with receptors directly, either by agonizing or antagonizing (Miller and Lappin 2023). Ligands can bind at the active site, or allosterically away from the active site (Miller and Lappin 2023). There are multiple methods a ligand may decrease activity of a receptor, including antagonism, inhibition, competitive inhibition, or inverse agonism (Miller and Lappin 2023). Certain substances may also impact receptor activity indirectly by suppressing or modulating activity, altering the kinetics of a reaction, or altering the expression of a receptor or another ligand (Miller and Lappin 2023). CBD affects multiple receptors in all of these ways. CBD was initially hypothesized to signal solely though the endocannabinoid system, though investigations have revealed functional interactions with Transient Potential Vanilloid 1 (TRPV1) (Costa et al. 2004) and the 5-hydroxytryptamine (5HT) receptors, or serotonin receptors (Rock et al. 2012). A previous review by de Almeida and colleagues has highlighted CBD binding on a subset of G-protein-coupled receptors and ion channels (Almeida and Devi 2020). The goal of this review is to compile the literature regarding various biological processes in which CBD in involved and to build on prior discussions of CBD pathways (Fig. 1).
Graphical Summary. This graphical abstract summarizes the CBD pathways discussed, including the endocannabinoid system, ion channels, cytochrome P450 enzymes, those involved in energy homeostasis, inflammatory pathways, apoptotic pathways, and sex hormone regulation. Additionally, this figure introduces the biological processes discussed herein, including psychiatric disorders, seizure activity, nausea and vomiting, pain sensation, thermal regulation, neuronal signaling, neurodegenerative diseases, reproductive aging, drug metabolism, inflammation, sex hormone regulation, and energy homeostasis. Created with BioRender.com
Methods
Search Strategy
I conducted a comprehensive literature search to compile evidence regarding cannabidiol (CBD) interactions with receptors, enzymes, and biological processes, organized by body systems. Searches were performed across PubMed, Web of Science, and Google Scholar for articles published through 2022. I included studies utilizing diverse methodologies, including cell culture, animal models, biochemical assays, and clinical research, to ensure a robust and holistic understanding of CBD receptor interactions. Search terms included combinations of the following keywords: "CBD," "cannabidiol," "receptors," "enzymes," "ion channels," "endocannabinoid system," "cytochrome P450," "inflammation," "hormones," and "neurotransmission." I screened titles and abstracts for relevance to the theme of CBD interactions with receptors and enzymes. Articles were included if they reported direct, indirect, or implied interactions of CBD with specific receptors, enzymes, or biological processes. Full-text articles available in English were retrieved for those meeting initial inclusion criteria.
Data Extraction and Synthesis
Data from eligible studies were extracted, including receptor/enzyme name, methodology (e.g., cell culture, animal model, clinical trial), and key findings related to CBD interactions. To facilitate thematic analysis, extracted data were grouped by body system (e.g., nervous, endocrine, immune) and categorized by the specific receptor or enzyme involved. Findings were narratively synthesized to highlight patterns, gaps, and implications for health and disease.
Quality Assessment
Given the narrative nature of this review, no formal quality assessment tools were applied. However, emphasis was placed on studies providing mechanistic insights, robust methodologies, or clinical relevance to ensure reliability in the synthesis.
Reporting
Relevant findings are presented in both narrative and tabular formats, organized by body system, to enhance clarity and accessibility.
CBD interacts with the endocannabinoid system
The endocannabinoid system encompasses a growing list of receptors that are bound by the endogenous cannabinoids, or endocannabinoids, 2-arachidonoyl glycerol (2-AG) (Sugiura et al. 1995) and arachidonoyl ethanolamide, or anandamide (AEA) (Felder et al. 1993). Though cannabis has been consumed for thousands of years (Bridgeman and Abazia 2017), it wasn’t until 1988 when Devane and colleagues characterized the first receptor than exogenous cannabinoids bound, called the cannabinoid receptor type 1 (CB1) (Devane et al. 1988). In 1992, the first endogenous cannabinoid that bound CB1 was isolated, called arachidonoylethanolamide, or anandamide (AEA) (Devane et al. 1992). In the last 30 years, the understanding of the endocannabinoid system has grown exponentially to include additional receptors, like transient receptors potential (TRP) channels (Costa et al. 2004), (Muller et al. 2019), and peroxisome proliferator activated receptors (PPAR) (O’Sullivan et al. 2009), (O’Sullivan 2016), as well as additional minor endocannabinoids like virodhamine (Porter et al. 2002) and 2-arachidonoyl glycerol ether (Hanuš et al. 2001). Further discussion on the function of the endocannabinoid system in the central nervous system was presented by Zou and Kumar (Zou and Kumar 2018) and Lu and Mackie (Lu and Mackie 2016).
CB1 and CB2 are activated by endogenous lipid-based retrograde neurotransmitters in the central and peripheral nervous system, including anandamide (AEA) (Felder et al. 1993) and 2-arachidonoylglyerol (2-AG) (Sugiura et al. 1995). CB1 is predominantly expressed in the central nervous system (Tissue expression of CNR1 - Summary - The Human Protein Atlas n.d) while CB2 is found in the peripheral nervous system and immune cells (Tissue expression of CNR2 - Summary - The Human Protein Atlas n.d), (Graham et al. 2010). CB1 and CB2 are G-protein-coupled receptors that confer intracellular signaling cascade activation when bound by ligands (Houston and Howlett 1998). CB1 and CB2 are activated by exogenous cannabinoid compounds such as THC (Shen and Thayer 1999). While CBD was initially theorized to activate CB1 and CB2 akin to the activation induced by THC, subsequent literature has debated this effect (McPartland et al. 2015) (Table 1, Fig. 2). Competitive binding affects downstream signaling by reducing receptor activation in a reversible manner, as higher concentrations of the endogenous ligand can outcompete the inhibitor and restore signaling. In contrast, non-competitive binding alters receptor function regardless of ligand concentration, often leading to partial or complete inhibition of downstream signaling by inducing conformational changes or disrupting signal transduction pathways. CBD has multiple effects on CB1 receptors, including inversely agonizing CB1 (Pertwee 2008) and serving as a negative allosteric modulator of CB1 (Laprairie et al. 2015), depending on the cellular context (Table 1, Fig. 2). For example, in HEK 293A cells that exogenously express CB1 receptors, and in a Huntington’s Disease model striatal cell line (STHdhQ7/Q7), application of CBD induced noncompetitive negative allosteric modulation of CB1 receptors with CB1 agonists (Laprairie et al. 2015) (Table 1, Fig. 2). CBD serves as an inverse agonist of CB1 at low levels in hCB2-CHO cells (Pertwee 2008) (Table 1, Fig. 2). Additionally, CBD alters the kinetics of internalization of CB1 receptors into the cell through β-arrestin recruitment (Table 1, Fig. 2) (Laprairie et al. 2015). CBD has an indirect effect on CB1 through antagonism of fatty acid amide hydrolase (FAAH) (Petrocellis et al. 2011), (Bisogno et al. 2001). FAAH breaks down the endocannabinoid anandamide (Kwilasz et al. 2014). By inhibiting FAAH, CBD can increase circulating anandamide levels (Hua et al. 2023), (Leweke et al. 2012), leading to prolonged activation of CB1 (Table 1, Fig. 2). CBD inversely activates CB2 (Thomas et al. 2007) (Table 1, Fig. 2). In a [ (Tissue expression of CNR2 - Summary - The Human Protein Atlas n.d) S]GTPγS binding assay using CHO cell membranes transfected with CB2 receptors (hCB2-CHO), 1 μM CBD showed a significantly lower KB than Ki, highlighting its function as an inverse agonist for CB2 (Thomas et al. 2007). Additionally, CBD induces a heterodimerization of CB2 with 5HT receptors (Pazos et al. 2013) (Table 1, Fig. 2). By regulating this heterodimer, CBD may be influential in neonatal hypoxic-ischemic brain damage (Pazos et al. 2013). Current work investigating the involvement of CBD and the endocannabinoid receptors is rapidly expanding, however there is still debate as to which of these effects are feasible at physiologic consumption levels of CBD.
CBD interacts with the endocannabinoid system. CBD interacts with multiple regulators of the endocannabinoid system, including the endocannabinoid receptors CB1 and CB2. Additionally, CBD indirectly interacts with CB1 via the enzyme FAAH and β-arrestin. CBD also regulates TRPV1 via interactions with CB1 and inhibits 5HT1A via interactions with CB2. Created with BioRender.com
CBD interacts with ion channels
Ion channels control the flow of charged ions, such as K+, Na+, Ca2+, and Cl− across the cell membrane. These ions regulate the cell membrane potential, which is critical in excitable tissues such as the brain, heart, and pancreas (Neher 1992). Cells within these tissues communicate through action potentials, chemical and electrical synapses, and gap junctions, all of which are mediated by the ion channels that control the membrane potential (Neher 1992). Altering the activity of these ion channels disrupts intercellular communication and can have downstream impacts on tissue function (Neher 1992).
CBD impacts processes involved in psychiatric disorders
CBD activates and antagonizes processes that are implicated in the development or management of psychiatric disorders, specifically serotonin receptors (Rock et al. 2012; Xiong et al. 2011; Yang et al. 2010) and G-protein-coupled receptors 3, 6, 12 and 55 (Laun et al. 2019; Laun and Song 2017; Lauckner et al. 2008) (Table 2, Fig. 3). GPRs 3, 6, and 12 are also called cannabinoid-related orphan receptors, because of their reactivity to endogenous and exogenous cannabinoids (Laun et al. 2019). CBD activates 5HT1A (Rock et al. 2012), a G-protein-coupled receptor that is heavily expressed in the brain, gastrointestinal tract, endocrine tissues, kidney, and muscles, among other tissues (Tissue expression of HTR1A - Summary - The Human Protein Atlas n.d) (Table 2, Fig. 3). In the central nervous system, dysregulation of 5HT1A signaling has harmful effects on cognition, mood and behavior disorders, depressive disorders, and panic disorders (Savitz et al. 2009; Akimova et al. 2009). CBD also indirectly decreases 5HT1A expression (Jenny et al. 2009; Jenny et al. 2010) (Table 2, Fig. 3). Under CBD exposure, tryptophan, the precursor to 5HT1A, preferentially follows the IDO1/2 pathway to tryptophan catabolism instead of conversion to 5HT1A (Jenny et al. 2009; Jenny et al. 2010) (Table 2, Fig. 3). CBD antagonizes another serotonin receptor, 5HT3A (Yang et al. 2010), which is expressed in the brain, digestive tract, pancreas, muscle, bone marrow, and lymphoid tissue (HTR3A protein expression summary - The Human Protein Atlas n.d) (Table 3, Fig. 3). Unlike the other 5HT receptors that couple to G-protein-coupled receptors, 5HT3A is a ligand-gated ion channel (Rodriguez Araujo et al. 2020). In the central nervous system, 5HT3A has been localized to pre- and post-synaptic nerve terminals in both excitatory and inhibitory neurons that release dopamine, cholecystokinin, and GABA (Engel et al. 2013). 5HT3A dysregulation has similar effects to 5HT1A in affecting mood disorders, as dysregulation of 5HT3A is implicated in depression, bipolar disorder, and post-traumatic stress disorder (Jang et al. 2015; Bétry et al. 2011). CBD inversely activates GPR3 and GPR6 (Laun and Song 2017) (Table 2, Fig. 3). GPR3 is expressed in the brain, endocrine tissues, muscle, respiratory system, and digestive tract (Tissue expression of GPR3 - Summary - The Human Protein Atlas n.d), and GPR6 is expressed in the brain and endocrine tissues (Tissue expression of GPR6 - Summary - The Human Protein Atlas n.d). Activation of GPR3 and GPR6 impact behavior, where activation of GPR3 mediates behavioral changes in stress response (Valverde et al. 2009), and GPR6 alters instrumental learning by regulating cyclic adenosine monophosphate (cAMP) production in striatal spiny neurons (Oeckl et al. 2014). GPR3 activation also modulates cocaine reinforcement (Tourino et al. 2012), suggesting it may play a role in risk for addiction disorders. Together, these interactions implicate a potential effect of CBD consumption on the development, progression, or management of psychiatric disorders.
CBD interacts with ion channels and G-protein coupled receptors. CBD interacts with multiple ion channels, including the TRP channels TRPM8, TRPV1, TRPV2, TRPV3, TRPV4, and TRPA1, serotonin receptor 5HT3A, sodium channels NaV1.1–1.7, L-type calcium channels, voltage-gated potassium channels KV7.2, KV7.3, KV4.3, and KV11.1, and glycine receptors. CBD interacts with the G protein-coupled receptors GPR3, GPR6, GPR12, and GPR55, the μ-opioid and ∂-opioid receptors, and interacts with receptors who couple with G protein coupled receptors for downstream signaling cascades, including 5HT1A serotonin receptors and D2 dopamine receptors. Created with BioRender.com
CBD impacts processes that modulate seizure activity
Epidiolex is the single United States Food and Drug Administration (FDA) approved CBD medication, used to treat seizures associated with Lennox-Gastaut Syndrome, Dravet Syndrome, or Tuberous Sclerosis Complex patients over the age of 18. CBD antagonizes GPR55 (Lauckner et al. 2008), a G protein-coupled receptor that is dysfunctional in epileptic patients (Rosenberg et al. 2023) (Table 2, Fig. 3). GPR55 is expressed in the brain, gastrointestinal tract, male reproductive tract, bone marrow, and lymphoid tissues (GPR55 protein expression summary - The Human Protein Atlas n.d). By indirectly blocking the malfunctioning GPR55 in epileptic patients, CBD can significantly reduce seizure episodes (Rosenberg et al. 2023). Additional ion channels involved in seizure activity modulation are KV7.2 and KV7.3 (Miceli et al. 2015), which CBD activates (Zhang et al. 2022) (Table 2, Fig. 3). CBD inhibits KV4.3 (Marois et al. 2020), a potassium channel whose dysregulation via genetic mutation causes epilepsy (Smets et al. 2015) and KV11.1 (Marois et al. 2020), a potassium channel whose dysregulation induces seizures (Keller et al. 2009). CBD inhibits voltage-gated sodium channels, NaV1.1–1.7 (Ghovanloo et al. 2018) (Table 2, Fig. 3). Genetic disruptions in NaV channels induce seizure activity due to alterations in action potential propagation, as discussed by Menezes and colleagues (Menezes et al. 2020). In genetic seizure disorders, pharmaceutical NaV channel agonists or antagonists like carbamazepine and lamotrigine can be therapeutic in mitigating seizure activity by restabilizing action potential propagation (Catterall 2014). Because CBD is an effective seizure-reducing medication, and it is federally approved for treatment of childhood seizures (Abu-Sawwa et al. 2020), the mechanistic understanding of how CBD can mitigate seizure activity is an active area of investigation.
CBD impacts receptors that regulate temperature sensitivity
CBD may impact temperature sensation by agonizing or antagonizing transient potential receptors, or TRPs (Table 2, Fig. 3) (Petrocellis et al. 2011; Anand et al. 2020; Petrocellis et al. 2012). CBD activates five TRP channels, including TRPV1, TRPV2, TRPV3, TRPV4, and TRPA1, and antagonizes TRPM8 (Petrocellis et al. 2011; Anand et al. 2020; Qin et al. 2008; Petrocellis et al. 2012; Petrocellis et al. 2008) (Table 2, Fig. 3). TRPV1 and TRPV2 sense high thermal stimuli, including stimuli above 42 °C and above 52 °C, respectively (Samanta et al. 2018). TRPM8 and TRPA1 sense low thermal stimuli, including temperatures 25–34 °C and below 17 °C, respectively (Samanta et al. 2018). TRPV1 is expressed in the brain, liver, gallbladder, pancreas, muscle, and reproductive tissues, TRPV2 is expressed in the brain, endocrine tissues, respiratory system, gastrointestinal tract, liver, gallbladder, and pancreas, among other tissues, TRPM8 is expressed in the liver, gallbladder, and male reproductive tissues, and TRPA1 is expressed in the gastrointestinal tract, liver, gallbladder, kidney, bladder (TRPA1 protein expression summary - The Human Protein Atlas n.d; Tissue expression of TRPV1 - Summary - The Human Protein Atlas n.d; TRPV2 protein expression summary - The Human Protein Atlas n.d; TRPM8 protein expression summary - The Human Protein Atlas n.d). TRPV3 is expressed predominantly in the gastrointestinal tract and skin, with lower expression levels in the muscle, reproductive tissues, brain, and endocrine tissues (TRPV3 protein expression summary - The Human Protein Atlas n.d). TRPV4 is expressed in the brain, endocrine tissues, gastrointestinal tract, pancreas, reproductive tissues, and muscle tissues, among others (TRPV4 protein expression summary - The Human Protein Atlas n.d). Dysregulation of TRP channels can alter thermal pain sensation, as discussed by Cortright and colleagues (Cortright et al. 2007). Exposure to TRP agonists can increase thermal pain sensitivity (Cortright et al. 2007). In fact, intrauterine CBD exposure increases sensitivity to thermal pain in adult male offspring in a TRPV1 dependent manner (Swenson et al. 2023). The agonism of these six receptors by CBD signals that CBD consumption could potentially mediate thermal sensitivity.
CBD content may modulate cannabis-induced hyperemesis
Following prolonged consumption of cannabis, some patients experience severe refractory nausea and vomiting called cannabis hyperemesis (Perisetti et al. 2020). The mechanism of cannabis hyperemesis is yet to be elucidated, though one working mechanistic theory is that prolonged THC exposure induces TRPV1 hypersensitization in the enteric and vagal neurons (Sharkey 2022). Interestingly, many patients with cannabis hyperemesis report relief following a hot shower (Perisetti et al. 2020), suggesting potential involvement of TRP receptors. Because TRPV1 is responsive to high heat (Samanta et al. 2018), and because it is expressed in the area postrema of the medulla, along gastric enteric and vagal nerves, and on cutaneous receptors in the dermis and epidermis (Tissue expression of TRPV1 - Summary - The Human Protein Atlas n.d), researchers postulate that repetitive TRPV1 activation may cause vomiting from overexcitation, or relief from the vomiting under acute activation of the hot shower. As CBD activates TRPV1, its role in inducing or mediating cannabis hyperemesis is theoretical and a key point for future investigations.
CBD impacts processes that regulate nausea and vomiting
CBD has gained traction recently as an anti-emetic, or nausea-reducing, medication. One potential mechanism by which CBD may inhibit nausea and vomiting is through antagonism of 5HT3A receptors (Yang et al. 2010; Theriot et al., n.d) (Table 2, Fig. 3). In the enteric nervous system that lines the gastrointestinal tract, 5HT3A receptor antagonists inhibit the gastrointestinal activity in nausea and vomiting (Browning 2015). Additionally, it is theorized that 5HT3A antagonism in the area postrema in the brainstem, known as the vomiting center, decreases the nausea and vomiting response in small mammals (Higgins et al. 1989). In rodent studies, CBD suppresses 0.1% saccharin solution induced vomiting in Asian house shrews (S. Murinus) and conditioned gaping (a measure of rodent nausea) in rats due to indirect agonism of 5HT1A somatodendritic autoreceptors in the dorsal raphe nucleus (Rock et al. 2012). CBD is increasingly consumed as an anti-emetic medication, particularly by chemotherapy patients, pregnant patients, and migraine patients (O’Brien 2022; Baron 2018). As CBD inhibits CYP 450 enzymes that metabolize pharmaceuticals (Smith and Gruber 2023), (Doohan et al. 2021), (Qian et al. 2019) understanding the impacts of CBD usage in chemotherapy is critical. Additionally, understanding the potential impact of CBD on fetal development, and the impact of co-consumption of CBD with migraine medications, would help inform these patients to the safety, risks, or drug-drug interactions that are possible with CBD. As CBD does not cause many of the side effects that accompany other nausea medications, like constipation and headache (Tincello and Johnstone 1996), it is a promising area of clinical investigation.
CBD impacts processes that regulate pain sensation
CBD impacts multiple processes that regulate pain sensation, including ion channels that sense painful stimuli, opioid receptors, and enzymes which regulate the breakdown of pain medications. CBD activates five TRP channels, including TRPV1, TRPV2, TRPV3, TRPV4, and TRPA1, and antagonizes TRPM8 (Table 2, Fig. 3) (Petrocellis et al. 2011; Anand et al. 2020; Qin et al. 2008; Petrocellis et al. 2012; Petrocellis et al. 2008). TRPV1 and TRPA1 antagonists are under current clinical trials as medications to reduce inflammatory, neuropathic, and visceral pain conditions (Gunthorpe and Chizh 2009; Giorgi et al. 2019). In addition to the thermal pain sensation mediated by TRP channels, CBD also impacts receptors that mediate nociceptive pain. CBD is an allosteric modulator of both μ-opioid receptors and ∂-opioid receptors at high concentrations, altering the efficacy of which opioid agonists bind or dissociate from the receptors (Vaysse et al. 1987; Kathmann et al. 2006) (Table 2, Fig. 3). Though CBD does allosterically modulate these opioid receptors, the authors who published on this interaction discuss how it is unlikely that CBD would induce these interactions at physiologically relevant levels (Kathmann et al. 2006). Kathmann discusses how the half maximal effective concentration (EC50) needed for CBD to interact with opioid receptors is likely 100 times higher than what can be consumed in a standard oral dose of CBD (Kathmann et al. 2006). This discussion was later supported by human pharmacokinetic studies which show plasma CBD metabolite levels following various levels of CBD ingestion, as discussed by Ujváry and colleagues (Ujváry and Hanuš 2016). Additionally, serotonin receptors such as 5HT1A regulate neuropathic pain conditions such as migraine and fibromyalgia (Leone et al. 1998; Tour et al. 2022). By activating 5HT1A, CBD inhibits paclitaxel-induced neuropathic pain (Ward et al. 2014). In the context of pain management, CBD may also impact the effectiveness of standard medications, including codeine, hydrocodone, oxycodone, fentanyl, meperidine, methadone, buprenorphine, and tramadol, all of which are metabolized by the cytochrome p450 (CYP) enzyme group (Table 3, Fig. 4) (Doohan et al. 2021; Interactions Between Cannabinoids and Cytochrome P450-Metabolized Drugs - Full Text View - ClinicalTrials.gov n.d). By antagonizing or competitively inhibiting the CYP enzymes (Doohan et al. 2021; Qian et al. 2019; Yamaori et al. 2011 Yamaori et al. 2011), it is possible that CBD co-consumption with narcotics will increase the narcotic half-life in the system, increasing pain management, but also increasing risk of overdose. In clinical studies investigating the beneficial role of CBD in pain management, co-consumption of CBD with opioids allowed patients to decrease opioid dose while maintaining effective levels of pain relief, though it is only speculated that this could be through a CYP enzyme inhibition mechanism (Capano et al. 2020). CBD antagonizes CYP2D6 (Qian et al. 2019; Yamaori et al. 2011), which metabolizes opioids (Yamaori et al. 2011) (Table 3, Fig. 4). By inhibiting this enzyme, CBD may hinder the breakdown of opioids and alter the half-life of the opioids in the bloodstream. CBD may also alter the metabolism of painkillers ketamine and methadone via antagonizing CYP2B6 (Doohan et al. 2021; Qian et al. 2019) (Table 3, Fig. 4). CBD inversely activates GPR3 (Laun et al. 2019), and GPR3 knockout mice show increased pain sensitivity and reduced response to morphine, highlighting the role of GPR3 in pain sensation (Ruiz-Medina et al. 2011) (Table 2, Fig. 3). CBD is an allosteric modulator of GlyRs (Ahrens et al. 2009), which mediate pain processing and pain hypersensitivity (Moraga-Cid et al. 2020) (Table 2, Fig. 3). CBD also activates KV7.2/3 (Zhang et al. 2022), whose activation in sensory nociceptive neurons mediates how Aδ peripheral nerves respond to noxious heat as discussed by Brown and Colleagues (Brown and Passmore 2009) (Table 2, Fig. 3). Together, these data implicate how consumption of CBD can alter pain sensation, which can have long-lasting impacts on pain tolerance, pain sensitivity, and consumption of pain-reducing medications.
CBD impacts neuronal function
CBD consumption may mediate neuronal function by agonizing or antagonizing multiple ion channels that maintain neuronal membrane potential, including potassium channels, sodium channels, and serotonin receptors, as well as G-protein-coupled receptors. CBD interacts with multiple ion channels, all of which have the potential to mediate neuronal signaling by altering neuronal membrane potential. CBD activates voltage-gated potassium channels KV7.2 and KV7.3 (Table 2, Fig. 3) (Zhang et al. 2022). As discussed by Brown and colleagues, KV7.2 and KV7.3 are expressed in the nervous system and regulate neuronal excitability (Brown and Passmore 2009). In sympathetic neurons, KV7.2/3 activity mediates repetitive discharges and conversion from phasic to tonic firing, and in hippocampal pyramidal neurons, KV7.2/3 activity mediates repetitive discharges of the neuron (Brown and Passmore 2009). By agonizing these ion channels and altering these repetitive discharges, CBD could be altering neuronal communication. CBD also inhibits NaV1.1–1.7 channels and L-type and T-type calcium channels (Ghovanloo et al. 2018; Isaev et al. 2022; Ross et al. 2008; Ghovanloo and Ruben 2022; Ali et al. 2015) all of which regulate action potential propagation and subsequent neuron communication (Grider et al. 2023) (Table 2, Fig. 3). In fact, fetal CBD exposure reduces the excitability of the prefrontal cortex and cognition in females (Swenson et al. 2023). By disrupting sodium and potassium channels which regulate neuronal communication, CBD may have lasting impacts on neuronal network structure and subsequent function.
CBD inversely activates GPRs 3, 6, and 12 (Table 2, Fig. 3) (Laun et al. 2019). The exact mechanism of these receptors is yet to be classified, but they have proposed mechanisms based on structural similarity to other receptors (Laun and Song 2017). GPRs 3, 6, and 12 are structurally similar to cannabinoid receptors, and the involvement of CBD with these receptors has been under recent investigation (Laun and Song 2017). GPR12 is expressed in the brain, eye, and gastrointestinal tract (GPR12 protein expression summary - The Human Protein Atlas n.d). All three receptors impact neurological functioning, where activation of GPR3, GPR6, and GPR12 mediates neurite outgrowth (Tanaka et al. 2007) and activation of GPR3, GPR6 and GPR12 regulates neuronal survival (Tanaka et al. 2014; Full article: Towards a better understanding of the cannabinoid-related orphan receptors GPR3, GPR6, and GPR12 n.d). CBD is an allosteric modulator of GlyRs (Ahrens et al. 2009), which are ligand-gated ion channels that regulate motor coordination, respiratory control, and muscle tone by controlling action potential activity (Moraga-Cid et al. 2020) (Table 2, Fig. 3).
CBD activates 5HT1A, 5HT3A, and D2 dopamine receptors (Rock et al. 2012; Yang et al. 2010; Seeman 2016), which mediate neurotransmitter release and neuronal communication (Altieri et al. 2012; Fields et al. 1990; Bhatt et al. 2021; Wu and Hablitz 2005). 5HT1A and 5HT3A play crucial roles in mediating neuronal signaling (Altieri et al. 2012; Bhatt et al. 2021). The agonism of 5HT1A receptors hyperpolarizes the neuron, leading to a decrease in action potential propagation (Sprouse and Aghajanian 1986). As action potential propagation increases neurite outgrowth (Fields et al. 1990), CBD may hinder neurite outgrowth and subsequent neuronal connections. 5HT3A receptor antagonism is under current investigation for its therapeutic effects in depression models (Bhatt et al. 2021). CBD antagonizes 5HT3A (Yang et al. 2010), meaning CBD may play a role in depressive disorders. CBD is a partial agonist of D2 dopamine receptors (Table 2, Fig. 3) (Seeman 2016). As agonism of D2 dopamine receptors increases neurite outgrowth (Todd 1992), CBD exposure may subsequently increase outgrowth.
These receptors, including the 5HT1A, 5HT3A, TRP channels, D2 dopamine receptors, KV7.2, KV7.3, NaV1.1–1.7, and L-type and T-type calcium channels, may have synergistic or oppositional effects upon CBD binding. The receptors and channels mediate many components of action potential propagation, including the influx of sodium ions during depolarization and the efflux of potassium during repolarization and hyperpolarization. Some receptors and channels have the ability to mediate activity of other receptors or channels often through the alteration of the membrane potential. Sodium channels, including NaV1.1–1.7, are responsible for the inward flux of sodium ions that depolarize a neuron (Eijkelkamp et al. 2012). Inhibition of these channels by CBD would decrease neuronal excitability by preventing membrane voltage from raising above baseline. 5HT1A agonism opens potassium channels (allowing potassium influx and repolarization/hyperpolarization) and closing calcium channels (hindering depolarization) (Ehrengruber et al. 1997; Penington et al. 1991; Albert and Vahid-Ansari 2019). By agonizing 5HT1A receptors (Rock et al. 2012), CBD may additionally decrease action potential propagation by hyperpolarizing the cell (Albert and Vahid-Ansari 2019). In some neuronal subtypes, calcium channels serve as upstream moderators of action potential propagation by dictating membrane potential (Iosub et al. 2015). For example, in the calcium-induced calcium release in inner hair cells, L type calcium channel activity induces the opening of potassium channels during repolarization (Iosub et al. 2015). Similarly, agonism of T type calcium channels induces depolarization when the neurons are in a hyperpolarized state (Cain and Snutch 2010). T type calcium channels are also involved in the repolarization and hyperpolarization of membrane potential, as they activate voltage-gated potassium channels that allow an outward flux of positively charged potassium ions (Cain and Snutch 2010). By inhibiting these calcium channels, CBD may further decrease depolarization. Voltage-gated potassium channels, including KV7.2 and 7.3, respond to changes in membrane potential to allow the outward flux of potassium ions during the latter stage of the action potential (Estacion et al. 2023). This outward flux of potassium pushes the membrane voltage back to baseline, and later to hyperpolarization (Estacion et al. 2023). By agonizing these channels, CBD may additionally decrease action potential propagation. In opposition, many of the TRP receptors induce depolarization upon activation (Gees et al. 2010), including when activated specifically by CBD (Kowalski et al. 2020). Similarly, D2 dopamine receptor agonism induces depolarization (Wu and Hablitz 2005).
CBD interacts with receptors that affect neurodegenerative disease progression and symptom management
CBD is under investigation for its impact on treating symptoms of neurodegenerative diseases, such as Parkinson’s disease and Alzheimer's disease (Bhunia et al. 2022). Neurodegeneration is a complex process regulated by many receptors, some of which are bound by CBD. As discussed by Bhunia and colleagues, CB1R, CB2R, PPARγ, 5HT1A, A2A-R, and TRPV1 all have neuroprotective effects (Table 2, Fig. 3) (Bhunia et al. 2022). In addition to these receptors, CBD also inversely activates GPR3 and GPR12 (Table 2, Fig. 3) (Laun et al. 2019; Laun and Song 2017). GPR3 and GPR6 have shown an impact on Alzheimer’s disease progression by regulating amyloid beta production (Huang et al. 2022), and activation of GPR6 modulates Parkinson’s disease progression by regulating striatal dopamine production (Brice et al. 2021). CBD is currently under investigation in clinical trials for symptom management in Parkinson’s Disease and Alzheimer’s disease, and in disease progression using animal model studies though the mechanism behind potential benefits has not yet been defined (Chagas et al. 2014; Chesworth et al. 2022; Hao and Feng 2021; McManus et al. 2021; Almeida et al. 2023; Faria et al. 2020; Zhang et al. 2022).
CBD antagonizes CYP enzymes which metabolize pharmaceuticals
CBD may impact pharmacologic drug metabolism by antagonizing CYP enzymes (Doohan et al. 2021; Qian et al. 2019; Yamaori et al. 2011 Yamaori et al. 2011) (Table 3, Fig. 4), the largest regulator of drug metabolism (Zhao et al. 2021). CYP enzymes are predominantly expressed in the liver, but are also present in the kidney, placenta, adrenal gland, gastrointestinal tract, and skin (Zhao et al. 2021). Additionally, CYP enzymes are critical to produce cholesterol, steroids, prostacyclins, and thromboxane A2 (Rendic and Guengerich 2018). CBD has two primary effects on CYP enzymes that suppress CYP enzyme activity: antagonism and competitive inhibition (Smith and Gruber 2023; Doohan et al. 2021; Qian et al. 2019; Yamaori et al. 2011 Yamaori et al. 2011) (Table 3, Fig. 4). CBD competitively inhibits CYPs 3A4, 3A7, and 3A5 (Doohan et al. 2021; Yamaori et al. 2011) (Table 3, Fig. 4). CYP3A4 breaks down small foreign organic molecules (xenobiotics) that are common prescription medications, such as clarithromycin, erythromycin, diltiazem, itraconazole, ketoconazole, ritonavir, and verapamil (Sweeney and Bromilow 2006). By competitively inhibiting CYP3A4 (Smith and Gruber 2023), CBD can interfere with drug metabolism, increasing the half-life of the drug. During fetal liver development, CYP3A7 is the predominant CYP, while CYP3A4 takes over during postnatal development (Li and Lampe 2019). CYP3A7 hydroxylates testosterone and dehydroepiandrosterone 3-sulphate, which is involved in the formation of estradiol during pregnancy (CYP3A7 Gene - Cytochrome P450 Family 3 Subfamily A Member 7 n.d). By inhibiting CYP3A7 (Yamaori et al. 2011), CBD may have impacts on estradiol creation or maintenance. Unlike the majority of CYP enzymes that function in the liver, CYP3A5 metabolizes endogenous steroids and xenobiotics in extrahepatic tissues, including the lung, kidney, prostrate, breast and leukocytes (Lamba et al. 2002). This activity highlights potential risks of co-consuming CBD with common pharmaceutical or recreational medications as CBD may alter drug metabolism and subsequent activity.
CBD antagonizes CYP2C9, CYP1A1, CYP1A2, CYP1B1, CYP2D6, CYP2B6, and CYP2J2 (Smith and Gruber 2023; Doohan et al. 2021; Qian et al. 2019; Yamaori et al. 2011 Yamaori et al. 2011) (Table 3, Fig. 4). CYP2C9 is the predominant metabolizer of the blood clot prevention medication Warfarin (Dean and Warfarin Therapy and VKORC1 and CYP Genotype. In: Pratt VM, Scott SA, Pirmohamed M, Esquivel B, Kattman BL, Malheiro AJ, eds. Medical Genetics Summaries. National Center for Biotechnology Information (US) 2012). By antagonizing CYP2C9, CBD impairs the degradation of Warfarin, impacting blood clotting (Grayson et al. 2017; Cortopassi 2020; Hsu and Painter 2020). CYP1A1 is critical in cancer regulation because it metabolizes carcinogens into epoxide intermediates which are less detrimental (Androutsopoulos et al. 2009). CBD increases CYP1A1 expression in a Hep2G cell line and antagonizes the enzyme activity (Qian et al. 2019; Yamaori et al. 2015). CYP1A2 metabolizes endogenous compounds including retinols, melatonin, steroids, uroporphyrinogen, and arachidonic acid, as well as recreational and pharmaceutical drugs including phenacetin, caffeine, clozapam, tacrine, propranolol, and mexiletine (Zhou et al. 2009). CYP1A2 also metabolizes precarcinogens, including aflatoxins, mycotoxins, and nitrosamines (Zhou et al. 2009). By antagonizing CYP1A2 (Qian et al. 2019), CBD exposure could alter the breakdown of these substances. CYP1B1 metabolizes exogenous compounds akin to other CYP enzymes, while also metabolizing endogenous compounds such as estrogen, arachidonic acid, melatonin, and retinoids (Li et al. 2017). CYP2D6 metabolizes pharmaceutical medications including antidepressants, neuroleptics, some antiarrhythmics, lipophilic β-adrenoceptor blockers and opioids (Bertilsson et al. 2002). CBD inhibits CYP2C19 (Doohan et al. 2021; Qian et al. 2019), an enzyme that metabolizes multiple pharmaceutical drugs including citalopram, clomipramine, clopidrogrel, diazepam, omeprazole (Jiang et al. 2013). CYP2B6 is responsible for metabolizing pharmaceuticals including artemisinin, bupropion, cyclophosphamide, efavirenz, ketamine, and methadone (Zanger and Klein 2013). CYP2J2 metabolizes many pharmaceuticals, including antihistamines (terfenadine, ebastine, and astemizole), anticancer agents (doxorubicin and tamoxifen), and immunosuppressants (cyclosporine) (Solanki et al. 2018). Combined, these interactions demonstrate CBD is not inert, and CBD consumption can alter metabolism of many substances. Because of these interactions, healthcare providers and pharmacists should inquire about patient CBD consumption.
CBD impacts the breakdown of exogenous cannabinoids
CBD hinders the breakdown of tetrahydrocannabinol (Zamarripa et al. 2023), or THC, the primary psychoactive component of marijuana by inhibiting CYP2C9, CYP2D6, and CYP3A4 (Doohan et al. 2021; Qian et al. 2019; Yamaori et al. 2011; Ng et al. 2023). CBD is the second most common cannabinoid included in marijuana products, followed by minor cannabinoids like cannabinol, cannabichromene, cannabigerol, cannabinolic acid, and cannabidivarin, among others (Walsh et al. 2021). CBD antagonizes CYP2C9 and CYP2C19 (Doohan et al. 2021; Qian et al. 2019; Jiang et al. 2013), which are the predominant metabolizers of exogenous cannabinoids (Bland et al. 2005) (Table 3, Fig. 4). Through this mechanism, CBD exposure could slow the metabolism of other exogenous cannabinoids, increasing their half-life and therefore increasing the length of symptomatology from the psychoactive components.
CBD may alter the regulation of sex hormones
By interacting with multiple regulators of sex hormone production or metabolism, CBD may alter sex hormone production or levels. CBD antagonizes aromatase (Almada et al. 2020), an enzyme that converts testosterone to estrogen (Brodie et al. 1999) (Table 4, Fig. 5). Inhibition of aromatase during fetal development can be harmful, as it decreases the production of estrogens that are required for pregnancy maintenance and offspring sexual development (Tiboni and Ponzano 2016). Because of this, many aromatase inhibitor pharmaceuticals are contraindicated during pregnancy (Tiboni and Ponzano 2016). CBD antagonizes progesterone 17 hydroxylase (Watanabe et al. 2005), which hydroxylates pregnenolone and progesterone (precursors to aldosterone), to form 17-hydroxypregnenolone and 17-hydroxyprogesterone (precursors to cortisol) (Chormanski and Muzio 2023) (Table 4, Fig. 5). CYP3A7 hydroxylates testosterone and dehydroepiandrosterone 3-sulphate, a critical process in the production of estriol during pregnancy (CYP3A7 Gene - Cytochrome P450 Family 3 Subfamily A Member 7 n.d). By inhibiting CYP3A7 (Yamaori et al. 2011), CBD may have impacts on estriol creation or maintenance. CBD antagonizes CYP1B1 (Qian et al. 2019) (Table 4, Fig. 5). CYP1B1 metabolizes exogenous compounds akin to other CYP enzymes, while also metabolizing endogenous compounds such as estrogen, arachidonic acid, melatonin, and retinoids (Li et al. 2017). CBD inversely activates GPR3 and GPR12 (Table 2, Fig. 5) (Laun et al. 2019). GPR3 and GPR12 have roles in female reproduction through ovarian aging, where both receptors maintain meiotic arrest of oocytes and premature ovarian aging (Hinckley et al. 2005). By antagonizing or inhibiting this complex of processes, CBD could be altering sex hormone pathways.
CBD impacts processes that mediate metabolic homeostasis
CBD affects energy homeostasis and metabolism via multiple mechanisms. CBD accumulates in fat, muscle, and liver following consumption, however, females showed higher accumulation in the muscle and liver compared to males (Child and Tallon 2022). As discussed by Wiciński and colleagues, CBD impacts multiple metabolic processes, including in maintaining glucose homeostasis, regulating adipose tissue insulin sensitivity, maintaining low density lipid (LDL) and high density lipid (HDL) profiles, hypertension, and in the treatment of metabolic syndrome in clinical studies (Wiciński et al. 2023). CBD activates PPARγ (O’Sullivan 2016) (Table 5, Fig. 6). PPARγ activation promotes fatty acid uptake, triglyceride formation and storage in lipid droplets (Montaigne et al. 2021). This activation in turn increases insulin sensitivity and glucose metabolism (PPARδ regulates glucose metabolism and insulin sensitivity | PNAS n.d). As such, CBD may impact insulin sensitivity and glucose tolerance via PPARγ. PPARγ is expressed in the brain, gastrointestinal tract, liver, gallbladder, kidney, reproductive tissues, and lymphoid tissues, among others (Tissue expression of PPARG - Summary - The Human Protein Atlas n.d). CBD also increases lipolysis, the metabolic process by which triglycerols break down into glycerol and free fatty acids (Caldari-Torres et al. 2023) (Table 5, Fig. 6). In the liver, PPARγ activity regulates lipid accumulation, lipid uptake, triaglycerol storage, and the formation of lipid droplets (Wang et al. 2020). In both human and mouse cultured mesenchymal stromal stem cells (MSCs), PPARγ agonism by CBD increased lipid accumulation and increased the expression of adipogenic genes, markers of adipogenic differentiation (Chang et al. 2022). Also in MSCs, CBD restores adipogenesis and chondrogenesis following lipopolysaccharide exposure (Ruhl et al. 2018). In murine skeletal muscle, PPARγ agonism increases adiponectin production and serves as a protective factor against systemic insulin resistance (Amin et al. 2010). Combined, the effects of CBD on PPARγ on metabolic outcomes may differ depending on the dose and the location of the receptor, increasing adiposity and lipid accumulation, or by impacting insulin resistance.
CBD both activates and antagonizes TRPV1 depending on the concentration (Muller et al. 2019; Anand et al. 2020). TRPV1 regulates multiple metabolic processes depending on tissue (Luo et al. 2012). TRPV1 is expressed in the brain, liver, gallbladder, pancreas, muscle, and reproductive tissues (Tissue expression of TRPV1 - Summary - The Human Protein Atlas n.d). In the muscle, TRPV1 agonism by capsaicin increases PGC-1α expression, increases expression of genes involved in fatty acid oxidation and mitochondrial respiration, and increased oxidative fibers (Luo et al. 2012). Additionally, in vivo TRPV1 agonism enhances exercise endurance and prevents high fat diet induced metabolic disorders (Luo et al. 2012). TRPV1−/− mice show decreased calcitonin gene related peptide (CGRP) production in the sensory neurons that innervate the pancreas, leading to improved insulin secretion and metabolic health (Riera et al. 2014). CBD inversely activates both GPR3 and GPR12 (Laun and Song 2017) (Table 2, Fig. 3). GPR3 and GPR12 both regulate obesity and energy balance (Bjursell et al. 2006; Godlewski et al. 2015). GPR12 knockout mice have changes in body composition, including increased body weight and fat mass, coupled with metabolic disorders including decreased respiratory exchange ratio, hepatic steatosis, and dyslipidemia (Bjursell et al. 2006). GPR3 knockout mice have late-onset obesity (Godlewski et al. 2015). These interactions suggest a potential mechanism by which CBD could improve metabolic homeostasis. Further studies are needed to understand the combinatorial effect of CBD on PPARs, TRPs, and GPCRs, as the metabolic impacts appear to be contradictory to each other. However, it is biologically plausible that differing tissues would have different responses to CBD exposure, leading to a net change or net neutral in overall metabolic efficiency.
CBD mediates anti-inflammatory processes
CBD benefits chemotherapy patients, pain patients, and people with neurodegenerative disorders by serving as an anti-inflammatory agent (Sholler et al. 2020). As discussed by Atalay and colleagues (Atalay et al. 2019), Pereira and colleagues (Pereira et al. 2021), and Jîtcă and colleagues (Jîtcă et al. 2023), CBD inhibits reactive oxygen species (ROS) production and produces an antioxidative defense. CBD activates caspases 8 and 9 (Massi et al. 2006), which subsequently induces the intrinsic apoptotic pathways (Massi et al. 2006) (Table 6, Fig. 7). CBD antagonizes the lipoxygenase pathway (Massi et al. 2008). The lipoxygenase pathway is a pro-carcinogenic pathway which, when active, generates proinflammatory mediations including leukotrienes and lipoxins (Wisastra and Dekker 2014) (Table 6, Fig. 7).
CBD impacts inflammatory and apoptotic pathways. CBD interacts with multiple components of inflammatory and apoptotic pathways. CBD antagonizes TLR4, lipoxygenase, interleukins 1α, 1β, 6, 18, TNFα, NFκB, NLRP3, and caspase 1. CBD activates caspases 8 and 9, G protein-coupled receptors GPR3, 6, and 12, PPARγ, ADORA2A, ERK1/2, and MAPK. CBD inversely activates GPR 3, 6, and 12, and allosterically modulates P2X7. Created with BioRender.com
CBD antagonizes multiple pro-inflammatory processes, leading to a subsequent decrease in inflammation. CBD decreases levels of TNF-α, NF-κB, TLR4 and NLRP3 (Yndart Arias et al. 2023; Suryavanshi et al. 2022; Chen et al. 2023) (Table 6, Fig. 7), the activation of which produce proinflammatory cytokines (Blevins et al. 2022) and decreases proinflammatory cytokines IL-1β, IL-6, IL-8 (Suryavanshi et al. 2022; Dinarello 2000; Hoffmann et al. 2002; Xing et al. 1998). CBD suppresses caspase 1 (Yndart Arias et al. 2023), decreasing the pyroptosis pathway and subsequent immune cell activation (Molla et al. 2020) (Table 6, Fig. 7). CBD indirectly modulates ADORA2A (Mecha et al. 2013) (Table 6, Fig. 6) via inhibition of the adenosine transporter, increasing adenosine levels which can in turn activate ADORA2A (Pandolfo et al. 2011), which inhibits inflammation in microglia (Yuan et al. 2022). NLRP3 is suppressed by CBD (Yndart Arias et al. 2023; Suryavanshi et al. 2022) (Table 6, Fig. 7). Additionally, CBD modulates P2X7 receptors (Liu et al. 2020), which are a second signal for NLRP3 inflammasome activation and subsequent IL-1β release by decreasing calcium efflux (Table 6, Fig. 7) (Liu et al. 2020). CBD activates PPARγ (O’Sullivan 2016), which subsequently inhibits the release of inflammatory cytokines (Jiang et al. 1998) (Table 6, Fig. 7).
CBD interacts with the mitogen activated protein kinase (MAPK) pathway (Hwang et al. 2017), a signal transduction pathway that regulates gene expression, mitosis, apoptosis, and differentiation (Cargnello and Roux 2011) (Table 6, Fig. 7). CBD increases ERK1/2 and p38 activity within the MAPK pathway (Hwang et al. 2017; Vrechi et al. 2021) (Table 6, Fig. 7). ERK1/2, when activated, inhibits apoptosis and subsequently increases the rates of conversion from one cell type to another cell type (metaplasia) and increases rates of tumor development, as discussed by Mebratu and colleagues (Mebratu and Tesfaigzi 2009). Activation of p38, another component of the MAPK pathway, increases biosynthesis of proinflammatory cytokines (Xiao et al. 2002). Increased activity of both ERK1/2 and p38 increase the phosphorylation and subsequent activity of ternary complex factor (TCF) and serum response factor (SRF) (Vickers et al. 2004). Activation of TCF and SRF downregulate apoptotic pathways, similar to the effects of ERK1/2 activation (Vickers et al. 2004). In a tumor microenvironment, inhibition of apoptosis furthers tumor progression (Gadiyar et al. 2020). Additionally, CBD reduces the potency with which endogenous (2‐AG) and exogenous (THC) cannabinoids signal through the ERK1/2 pathway (Laprairie et al. 2015). CBD both activates and inversely activates GPR12 (Laun et al. 2019; Laun and Song 2017), which increases cell survival and protein kinase signaling to increase cell proliferation (Table 6, Fig. 7) (Lu et al. 2012). GPR12 agonism increases keratin 8 phosphorylation (Park et al. 2016). Phosphorylation of keratin 8 increases tumor cell migration, which contributes to metastatic capabilities of tumor cells (Busch et al. 2012). By agonizing components of the MAPK pathway and GPR12, CBD may increase tumor cell survival and migration. Henshaw and colleagues demonstrated that in vivo animal model and clinical studies validate the in vitro studies, as CBD consumption decreased proinflammatory cytokines in > 90% of studies reviewed (Henshaw et al. 2021).
How can CBD be interacting with so many processes?
Though not yet fully mechanistically understood, there are multiple potential reasons as to why CBD is able to bind with multiple different receptors, enzymes, and ion channels in different pathways. The first primary distinction is that CBD serves as a ligand to some receptors directly, but participates in allosteric binding with many other receptors, as discussed. Previous studies demonstrate that individual ligands act as allosteric modulators for multiple receptors, dramatically increasing the number of biological effects a single ligand can have (Wang et al. 2009). Additionally, some receptors may have multiple binding sites to allow the receptor to interact with multiple ligands (Ma et al. 2002; Alhosaini et al. 2021). In this case, a smaller number of the receptor binding sites would need to be functionally able to bind to CBD in order to produce the same effect on the receptor. In consort with having multiple binding sites, some receptors are considered promiscuous receptors and regularly bind multiple ligands of different structures (Alhosaini et al. 2021; Gilberg et al. 2019). Some receptor pathways have multiple receptor subtypes or isoforms that produce the same downstream effect (Baker and Hill 2007), further increasing the likelihood that CBD could chemically interact with the pathway. When interacting with complex signal transduction pathways, CBD may indirectly induce multiple downstream effects by agonizing or antagonizing an upstream receptor. In this case, CBD may indirectly impact multiple processes without needing to directly interact with the downstream intermediates. Similarly, CBD may interact with systems that have high levels of redundancy, or similar downstream processes (Mantovani 2018). By having multiple upstream pathways induce a downstream effect, this increases the likelihood that CBD may structurally interact with one of the receptors. Lastly, some receptors may undergo conformational changes upon ligand binding (Frimurer et al. 2003). By changing the structure of the binding site, receptors may conform to a structure that CBD is capable of interacting with, only after binding of another ligand (Kondra et al. 2022). However, the morphological structure of these receptors are still being classified (Reggio 2010). As CBD gains significant traction in research, further studies are needed to understand how CBD specifically is able to interact with so many pathways.
In addition to CBD’s ability to bind with many receptors and interact with so many pathways, CBD may have an additional indirect impact on receptor activity by impacting the membrane fluidity of the cell the receptors are present on (Watkins 2019). Because CBD is highly lipophilic, it’s interactions with the lipid bilayer of cells has been under recent investigation. Nelson and colleagues propose that this impact on membrane fluidity has a direct impact on CBD’s promiscuity to receptors (Nelson et al. 2020). Watkins proposes that CBD can increase membrane fluidity, and subsequently change the conformation and gating kinetics of channels embedded in the membrane (Watkins 2019). Further studies are needed to elucidate the connections between CBD, membrane fluidity, and channel activity.
Effects of acute CBD exposure may differ from chronic exposure
CBD may be consumed in acute settings (for example, for a sleep aid, a nausea suppressant, etc.), or chronically (Epidiolex prescriptions for seizure, etc.). Little is known about the differential effects of acute versus chronic CBD exposure. Receptors may have differential activity depending on acute or chronic exposure (Jacobson et al. 1996). For example, a receptor may activate readily upon acute exposure, though under chronic exposure, the receptor may become overactivated and subsequently become downregulated either through decreased expression levels or cell surface presentation (Posner and Laporte 2010). Conversely, other receptors may continue to signal at maximum capacity despite chronic activation (Jacobson et al. 1996). For example, when CBD activates one receptor, I may see an upregulation of downstream signaling cascades. If that receptor is downregulated, CBD may activate the receptor, but the long-term output would mimic that CBD antagonized the receptor because the receptor was downregulated and no longer signaling or weakly signaling.
Biological effects of CBD are likely dose dependent
The activation of some CBD receptors varies depending on the dose of CBD and the affinity of CBD for the receptor (Lucas et al. 2018). For example, CBD activates TRPV1 at high concentrations (10–30 mM) and inhibits TRPV1 at low concentrations (1 mM) in varying cell culture models (Muller et al. 2019; Anand et al. 2020). For many receptors, the threshold of interaction with CBD has yet to be defined. It is possible that at low doses, CBD binds and interacts with a subset of receptors, while at high doses it interacts with a different subset of receptors in addition to high-affinity receptors. Additionally, high-dose exposure has the potential to downregulate certain receptors, leading to decreased receptor expression and activity. Because the body of research on CBD varies in methodologies (cell culture, animal model, and concentration) the effects of CBD cannot be directly compared. Because of this, not all effects mentioned are likely to be found at all doses. Further research is necessary to investigate the differential effects of CBD at standard dosing protocols to be translationally relevant.
Consumption method affects pharmacodynamics of CBD
As discussed by Lucas and colleagues, the pharmacokinetics of CBD vary based on method of consumption (Lucas et al. 2018). Common methods of consumption of CBD include oral consumption in the form of gummies, foods, or oils, inhalation methods such as smoking or vaping, sublingual consumption of oils, topically in a lotion, or via transdermal application (Corroon and Phillips 2018). Sublingual consumption and inhalation methods have the most concentrated effect, as uptake of CBD is unimpeded (Lucas et al. 2018; Huestis 2007). Vaping products tend to be more concentrated than smoking products, leading to higher blood stream CBD levels (Lucas et al. 2018). Oral consumption of CBD products requires the CBD to undergo first pass metabolism in the liver, which causes a tenfold reduction in available CBD to be metabolized before entering the circulatory system (Franco et al. 2020). Because of this, peak metabolite concentration following oral consumption is significantly slower than that of smoking, vaping, or sublingual consumption (Lucas et al. 2018). Topical and transdermal applications lead to the lowest levels of circulating CBD and CBD metabolites (Lucas et al. 2018). In addition to the varying impact of method of consumption on pharmacokinetics, differences in metabolism and binding of CBD may differ from CBD metabolites. However, in the context of receptor activation, few studies elucidate the differential impact of CBD from the major metabolites, including 7-OH CBD, CBD-glucuronide, and 10-OH-7-COOH-CBD (Ujváry and Hanuš 2016). As each metabolite varies slightly in structure, receptor binding ability or affinities may differ (Ujváry and Hanuš 2016). These many metabolites may contribute to the mechanism by which CBD acts on such a wide variety of receptors, as each metabolite has a slightly different structure and can therefore interact as ligands to receptors with different binding sites.
Challenges and Limitations
CBD research faces several significant challenges that complicate the interpretation and application of findings. One major limitation is the difficulty in sourcing high-quality and standardized CBD for research purposes, which hinders replication and consistency across studies. Additionally, basic science and preclinical studies vary widely in dosing regimens and routes of administration, making it difficult to compare findings or translate them to human applications. In human studies, variability in cannabinoid formulations—ranging from pure CBD isolates to full-spectrum extracts with other cannabinoids—further complicates comparisons across trials.
A critical translational gap exists between in vitro and in vivo research, as many reported effects may not be achievable at physiologically relevant doses in humans. The lack of standardized dose–response studies makes it difficult to determine whether findings from basic science research hold clinical significance. Additionally, research often fails to distinguish between acute and chronic exposure, limiting our ability to predict long-term outcomes. Existing studies have tested a broad range of doses, from low doses (~ 5–25 mg/day) used in wellness products to high doses (300–1,500 mg/day) investigated in clinical trials for conditions such as epilepsy and anxiety. However, data on the effects of chronic, moderate-dose CBD use remain limited. Addressing these limitations requires carefully designed studies that evaluate CBD’s pharmacokinetics, bioavailability, and sustained effects across different dosing regimens and patient populations.
Future directions and implications for clinical practice
Future research must bridge the gap between preclinical findings and human applications by ensuring translational relevance in dosing, administration routes, and outcome measures. Studies should clearly document the sourcing and composition of CBD formulations to improve reproducibility and clinical applicability. Additionally, making research findings widely accessible is essential, as clinicians and researchers across various disciplines need accurate and transparent data to guide patient care. This is particularly important given that patients may use CBD off-label, recreationally, or as a prescribed treatment, necessitating a comprehensive and evidence-based understanding of its effects across different populations. For example, research must define safe co-administration guidelines and identify potential risks associated with long-term CBD use in polypharmacy settings given the impact on CYP enzymes. Given the widespread use of CBD across different patient populations, future studies should prioritize personalized CBD therapy, evaluating how genetic, metabolic, and environmental factors influence individual responses. Long-term safety trials are essential to guide clinical recommendations, regulatory policies, and patient education. Standardizing research methodologies, ensuring transparent reporting, and making findings accessible to healthcare providers will be key to integrating CBD into evidence-based clinical practice.
Conclusion
CBD is rapidly gaining traction both in the pharmaceutical industry and as a widely available supplement to aid common ailments like nausea or insomnia, to rare conditions like childhood epilepsy (Abu-Sawwa et al. 2020; Data and Statistics. April 22 2022). As CBD consumption is not regulated, patients may co-consume CBD with pharmaceutical medications. CBD’s interaction with multiple body systems, and its effects on drug metabolism pose potential risks to unsuspecting patients. Clinicians and clinical researchers should ask patients about CBD consumption and should educate patients on potential drug-drug interactions. This review compiles the many processes that CBD interacts with that can confer multiple impacts, including affecting nausea, insomnia, seizure, sex hormone regulation, drug metabolism, and inflammation. While CBD has many beneficial effects, many of these interactions also have the potential to confer harm, meaning that CBD consumption should be monitored, especially when co-consumed with pharmaceutical or recreational substances. Further research is needed to understand the interactions between the processes included herein, and the translation from cell culture or animal model studies into human consumption through clinical research studies.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- 2-AG:
-
2-Arachidonoylglyerol
- 5HT1A :
-
5-Hydroxytryptamine receptor 1A
- 5HT3A :
-
5-Hydroxytryptamine receptor 3A
- 17OHP:
-
Progesterone 17-hydroxylase
- ACOG:
-
American College of Obstetrics and Gynecology
- ADORA2A :
-
Adenosine A2A receptor
- AEA:
-
Anandamide
- Caspase:
-
1-Cysteinyl aspartate protease 1
- cAMP:
-
Cyclic adenosine monophosphate
- CBD:
-
Cannabidiol
- CBDV:
-
Cannabidivarin
- CBC:
-
Cannabichromene
- CBCV:
-
Cannabichromevarin
- CBG:
-
Cannabigerol
- CBDA:
-
Cannabidioloic acid
- CBGA:
-
Cannabigerolic acid
- CBGV:
-
Cannabigerovarin
- CBN:
-
Cannabinol
- CBNA:
-
Cannabinolic acid
- CB1 :
-
Cannabinoid receptor type 1
- CB2 :
-
Cannabinoid receptor type 2
- CPS:
-
Child Protective Services
- CYP:
-
Cytochrome p450 enzyme
- CYP3A5:
-
Cytochrome P450 3A5
- CYP3A7:
-
Cytochrome P450 3A7
- CYP3A4:
-
Cytochrome P450 3A4
- CYP2C9:
-
Cytochrome P450 2C9
- CYP1A1:
-
Cytochrome P450 1A1
- CYP1A2:
-
Cytochrome P450 1A2
- CYP1B1:
-
Cytochrome P450 1B1
- CYP2D6:
-
Cytochrome P450 2D6
- CYP2C19:
-
Cytochrome P450 2C19
- CYP2B6:
-
Cytochrome P450 2B6
- CYP2J2:
-
Cytochrome P450 2J2
- D2:
-
Dopamine receptors
- ERK1/2:
-
Extracellular signal-regulated kinase
- FAAH:
-
Fatty acid amine hydrolase
- FDA:
-
Food and Drug Administration
- GPR55:
-
G protein-coupled receptor 55.
- GlyRs:
-
Ligand-gated glycine receptors
- GPR3:
-
G protein-coupled receptor 3.
- GPR6:
-
G protein-coupled receptor 6.
- GPR12:
-
G protein-coupled receptor 12
- GPX:
-
Glutathione peroxidase
- IDO1/2:
-
Indoleamine-pyrrole 2,3-dioxygenase
- IL-1β:
-
Interleukin-1β
- IL-6:
-
Interleukin-6
- IL-8:
-
Interleukin-8
- KV7.2/3:
-
Potassium voltage-gated channel subfamily KQT member 2 and 3
- MAPK:
-
Mitogen activated protein kinase
- NaV1 :
-
Sodium channel protein type 1 subunit
- NF-κB:
-
Nuclear factor kappa B
- NIDA:
-
National Institute of Drug Abuse
- NLRP3:
-
Intracellular “NOD-like” receptor (NLR) family pyrin domain containing 3
- NVP:
-
Nausea and vomiting in pregnancy
- PGC-1α:
-
Peroxisome proliferator-activated receptor gamma coactivator 1-alpha
- PPARα:
-
Peroxisome proliferator-activated receptor alpha
- PPARγ:
-
Peroxisome proliferator-activated receptor gamma
- THC:
-
Tetrahydrocannabinol
- THCA:
-
Tetrahydrocannabinolic acid
- THCV:
-
Tetrahydrocannabivarin
- TRPA1:
-
Transient receptor potential cation channel subfamily A member 1
- TRPM8:
-
Transient receptor potential cation channel subfamily M member 8
- TRPV1:
-
Transient receptor potential cation channel subfamily V member 1
- TRPV2:
-
Transient receptor potential cation channel subfamily V member 2
- TNF-α:
-
Tumor necrosis factor α
- TLR4:
-
Toll like receptor 4
References
Alhosaini K, Azhar A, Alonazi A, Al-Zoghaibi F. GPCRs: The most promiscuous druggable receptor of the mankind. Saudi Pharm J. 2021;29(6):539–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jsps.2021.04.015.
Atalay S, Jarocka-Karpowicz I, Skrzydlewska E. Antioxidative and Anti-Inflammatory Properties of Cannabidiol. Antioxidants (Basel). 2019;9(1):21. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antiox9010021.
A Abernethy Hemp Production and the 2018 Farm Bill - 07/25/2019. FDA 2019 https://www.fda.gov/news-events/congressional-testimony/hemp-production-and-2018-farm-bill-07252019. Accessed 22 Nov 2022
Abu-Sawwa R, Scutt B, Park Y. Emerging Use of Epidiolex (Cannabidiol) in Epilepsy. J Pediatr Pharmacol Ther. 2020;25(6):485–99. https://doiorg.publicaciones.saludcastillayleon.es/10.5863/1551-6776-25.6.485.
Ahrens J, Demir R, Leuwer M. The Nonpsychotropic Cannabinoid Cannabidiol Modulates and Directly Activates Alpha-1 and Alpha-1-Beta Glycine Receptor Function. Pharmacology. 2009;83(4):217–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000201556.
Ali RM, Al Kury LT, Yang KHS, et al. Effects of cannabidiol on contractions and calcium signaling in rat ventricular myocytes. Cell Calcium. 2015;57(4):290–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ceca.2015.02.001.
Almada M, Amaral C, Oliveira A, et al. Cannabidiol (CBD) but not tetrahydrocannabinol (THC) dysregulate in vitro decidualization of human endometrial stromal cells by disruption of estrogen signaling. Reprod Toxicol. 2020;93:75–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.reprotox.2020.01.003.
DL Almeida de, LA Devi. 2020 Diversity of molecular targets and signaling pathways for CBD Pharmacol Res Perspect. 2020;8(6):e00682. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/prp2.682
Anand U, Jones B, Korchev Y, et al. CBD Effects on TRPV1 Signaling Pathways in Cultured DRG Neurons. J Pain Res. 2020;13:2269–78. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/JPR.S258433.
Akimova E, Lanzenberger R, Kasper S. The Serotonin-1A Receptor in Anxiety Disorders. Biol Psychiat. 2009;66(7):627–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopsych.2009.03.012.
Androutsopoulos VP, Tsatsakis AM, Spandidos DA. Cytochrome P450 CYP1A1: wider roles in cancer progression and prevention. BMC Cancer. 2009;9:187. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2407-9-187.
Atakan Z. Cannabis, a complex plant: different compounds and different effects on individuals. Ther Adv Psychopharmacol. 2012;2(6):241–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/2045125312457586.
de Almeida CMO, Brito MMC, Bosaipo NB, et al. The effect of cannabidiol for restless Legs Syndrome/Willis-Ekbom disease in Parkinson’s disease patients with REM sleep behavior disorder: a post Hoc exploratory analysis of phase 2/3 clinical trial. Cannabis Cannabinoid Res. 2023;8(2):374–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/can.2021.0158.
Amin RH, Mathews ST, Camp HS, Ding L, Leff T. Selective activation of PPARgamma in skeletal muscle induces endogenous production of adiponectin and protects mice from diet-induced insulin resistance. Am J Physiol Endocrinol Metab. 2010;298(1):E28-37. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/ajpendo.00446.2009.
Altieri SC, Garcia-Garcia AL, Leonardo ED, Andrews AM. Rethinking 5-HT1A Receptors: Emerging Modes of Inhibitory Feedback of Relevance to Emotion-Related Behavior. ACS Chem Neurosci. 2012;4(1):72–83. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/cn3002174.
Albert PR, Vahid-Ansari F. The 5-HT1A receptor: Signaling to behavior. Biochimie. 2019;161:34–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biochi.2018.10.015.
EP Baron. Medicinal Properties of Cannabinoids, Terpenes, and Flavonoids in Cannabis, and Benefits in Migraine, Headache, and Pain: An Update on Current Evidence and Cannabis Science. Headache J Head Face Pain 2018 58(7):1139-1186 https://doiorg.publicaciones.saludcastillayleon.es/10.1111/head.13345
Brice NL, Schiffer HH, Monenschein H, et al. Development of CVN424: a selective and novel GPR6 inverse agonist effective in models of Parkinson disease. J Pharmacol Exp Ther. 2021;377(3):407–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/jpet.120.000438.
Bisogno T, Brandi I, Moriello AS, Davis JB, Mechoulam R, Marzo VD. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. 2001. p. 134–845.
Bhatt S, Devadoss T, Manjula SN, Rajangam J. 5-HT3Receptor Antagonism: A Potential Therapeutic Approach for the Treatment of Depression and other Disorders. Curr Neuropharmacol. 2021;19(9):1545–59. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/1570159X18666201015155816.
Bridgeman MB, Abazia DT. Medicinal Cannabis: History, Pharmacology, And Implications for the Acute Care Setting. P T. 2017;42(3):180–8.
Bétry C, Etiévant A, Oosterhof C, Ebert B, Sanchez C, Haddjeri N. Role of 5-HT3 Receptors in the Antidepressant Response. Pharmaceuticals (Basel). 2011;4(4):603–29. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph4040603.
Busch T, Armacki M, Eiseler T, et al. Keratin 8 phosphorylation regulates keratin reorganization and migration of epithelial tumor cells. J Cell Sci. 2012;125(9):2148–59. https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jcs.080127.
Blevins HM, Xu Y, Biby S, Zhang S. The NLRP3 Inflammasome Pathway: A Review of Mechanisms and Inhibitors for the Treatment of Inflammatory Diseases. Front Aging Neurosci. 2022;14:879021. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnagi.2022.879021.
Bland TM, Haining RL, Tracy TS, Callery PS. CYP2C-catalyzed delta(9)-tetrahydrocannabinol metabolism: Kinetics, pharmacogenetics and interaction with phenytoin. Biochem Pharmacol. 2005;70(7):1096–103. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bcp.2005.07.007.
Brodie A, Lu Q, Long B. Aromatase and its inhibitors. J Steroid Biochem Mol Biol. 1999;69(1):205–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0960-0760(99)00051-5.
Browning KN. Role of central vagal 5-HT3 receptors in gastrointestinal physiology and pathophysiology. Front Neurosci. 2015;9:413. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fnins.2015.00413.
Bjursell M, Gerdin AK, Jönsson M, et al. G protein-coupled receptor 12 deficiency results in dyslipidemia and obesity in mice. Biochem Biophys Res Commun. 2006;348(2):359–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbrc.2006.07.090.
Bhunia S, Kolishetti N, Arias AY, Vashist A, Nair M. Cannabidiol for neurodegenerative disorders: a comprehensive review. Front Pharmacol. 2022;13. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2022.989717.
Baker JG, Hill SJ. Multiple GPCR conformations and signalling pathways: implications for antagonist affinity estimates. Trends Pharmacol Sci. 2007;28(8):374–81. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tips.2007.06.011.
Bertilsson L, Dahl ML, Dalén P, Al-Shurbaji A. Molecular genetics of CYP2D6: clinical relevance with focus on psychotropic drugs. Br J Clin Pharmacol. 2002;53(2):111–22. https://doiorg.publicaciones.saludcastillayleon.es/10.1046/j.0306-5251.2001.01548.x.
Brown DA, Passmore GM. Neural KCNQ (Kv7) channels. Br J Pharmacol. 2009;156(8):1185–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1476-5381.2009.00111.x.
Cortopassi J. Warfarin dose adjustment required after cannabidiol initiation and titration. Am J Health Syst Pharm. 2020;77(22):1846–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ajhp/zxaa268.
B Costa, G Giagnoni, C Franke, AE Trovato, M Colleoni. Vanilloid TRPV1 receptor mediates the antihyperalgesic effect of the nonpsychoactive cannabinoid, cannabidiol, in a rat model of acute inflammation Br J Pharmacol. 2004;143(2):247-250 https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.bjp.0705920
D Chormanski MR Muzio. 17-Hydroxylase Deficiency. In: StatPearls StatPearls Publishing 2023 http://www.ncbi.nlm.nih.gov/books/NBK546644/. Accessed 18 Oct 2023
Capano A, Weaver R, Burkman E. Evaluation of the effects of CBD hemp extract on opioid use and quality of life indicators in chronic pain patients: a prospective cohort study. Postgrad Med. 2020;132(1):56–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/00325481.2019.1685298.
Corroon J, Phillips JA. A Cross-Sectional Study of Cannabidiol Users. Cannabis and Cannabinoid Research. 2018;3(1):152–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/can.2018.0006.
Chagas MHN, Zuardi AW, Tumas V, et al. Effects of cannabidiol in the treatment of patients with Parkinson’s disease: An exploratory double-blind trial. J Psychopharmacol. 2014;28(11):1088–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0269881114550355.
Castillo-Arellano J, Canseco-Alba A, Cutler SJ, León F. The Polypharmacological Effects of Cannabidiol. Molecules. 2023;28(7):3271. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/molecules28073271.
Child RB, Tallon MJ. Cannabidiol (CBD) Dosing: Plasma Pharmacokinetics and Effects on Accumulation in Skeletal Muscle, Liver and Adipose Tissue. Nutrients. 2022;14(10):2101. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/nu14102101.
Cortright DN, Krause JE, Broom DC. TRP channels and pain. Biochim Biophys Acta (BBA) Mol Basis Dis. 2007;1772(8):978–88. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbadis.2007.03.003.
Chen H, Liu Y, Yu S, Li C, Gao B, Zhou X. Cannabidiol attenuates periodontal inflammation through inhibiting TLR4/NF-κB pathway. J Periodontal Res. 2023;58(4):697–707. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jre.13118.
Cargnello M, Roux PP. Activation and Function of the MAPKs and Their Substrates, the MAPK-Activated Protein Kinases. Microbiol Mol Biol Rev. 2011;75(1):50–83. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/MMBR.00031-10.
CYP3A7 Gene - Cytochrome P450 Family 3 Subfamily A Member 7 (n.d.). https://www.genecards.org/cgi-bin/carddisp.pl?gene=CYP3A7&keywords=cyp3a7.
C Caldari-Torres M Huang D Kasprovic Y Xu 2023 Cannabidiol treatment of proliferating 3T3-L1 pre-adipocytes affects mature cell size and expression of acyltransferases involved in lipid droplet synthesi Published online https://doiorg.publicaciones.saludcastillayleon.es/10.21203/rs.3.rs-2689027/v1
Chiu JW, Binte Hanafi Z, Chew LCY, Mei Y, Liu H. IL-1α Processing, Signaling and Its Role in Cancer Progression. Cells. 2021;10(1):92. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells10010092.
R Chesworth, D Cheng, C Staub, T Karl. Effect of long-term cannabidiol on learning and anxiety in a female Alzheimer’s disease mouse model. Front Pharmacol. 2022;13. https://www.frontiersin.org/articles/10.3389/fphar.2022.931384. Accessed 4 Sept 2023.
Catterall WA. Sodium channels, inherited epilepsy, and antiepileptic drugs. Annu Rev Pharmacol Toxicol. 2014;54:317–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-pharmtox-011112-140232.
Chang RC, Thangavelu CS, Joloya EM, Kuo A, Li Z, Blumberg B. Cannabidiol promotes adipogenesis of human and mouse mesenchymal stem cells via PPARγ by inducing lipogenesis but not lipolysis. Biochem Pharmacol. 2022;197:114910. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bcp.2022.114910.
Cain SM, Snutch TP. Contributions of T-type calcium channel isoforms to neuronal firing. Channels (Austin). 2010;4(6):475–82. https://doiorg.publicaciones.saludcastillayleon.es/10.4161/chan.4.6.14106.
Devane WA, Hanus L, Breuer A, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258(5090):1946–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/science.1470919.
WA Devane, FA Dysarz, MR Johnson, LS Melvin, AC Howlett. Determination and characterization of a cannabinoid receptor in rat brain Mol Pharmacol 1988;34(5):605-613
Data and Statistics. April 22 2022 https://www.cdc.gov/marijuana/data-statistics.htm. Accessed 31 Jul 2023
Dinarello CA. Proinflammatory Cytokines. Chest. 2000;118(2):503–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1378/chest.118.2.503.
Doohan PT, Oldfield LD, Arnold JC, Anderson LL. Cannabinoid Interactions with Cytochrome P450 Drug Metabolism: a Full-Spectrum Characterization. AAPS J. 2021;23(4):91. https://doiorg.publicaciones.saludcastillayleon.es/10.1208/s12248-021-00616-7.
L Dean. Warfarin therapy and VKORC1 and CYP Genotype. In: Pratt VM, Scott SA, Pirmohamed M, Esquivel B, Kattman BL, Malheiro AJ, eds. Medical genetics summaries. National Center for Biotechnology Information (US). 2012. http://www.ncbi.nlm.nih.gov/books/NBK84174/ . Accessed 22 Jun 2023.
Eijkelkamp N, Linley JE, Baker MD, et al. Neurological perspectives on voltage-gated sodium channels. Brain. 2012;135(Pt 9):2585–612. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/brain/aws225.
Ehrengruber MU, Doupnik CA, Xu Y, et al. Activation of heteromeric G protein-gated inward rectifier K+ channels overexpressed by adenovirus gene transfer inhibits the excitability of hippocampal neurons. Proc Natl Acad Sci. 1997;94(13):7070–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.94.13.7070.
Engel M, Smidt MP, van Hooft JA. The serotonin 5-HT3 receptor: a novel neurodevelopmental target. Front Cell Neurosci. 2013;7:76. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fncel.2013.00076.
Estacion M, Liu S, Cheng X, Dib-Hajj S, Waxman SG. Kv7-specific activators hyperpolarize resting membrane potential and modulate human iPSC-derived sensory neuron excitability. Front Pharmacol. 2023;14:1138556. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2023.1138556.
de Faria SM, de Morais FD, Tumas V, et al. Effects of acute cannabidiol administration on anxiety and tremors induced by a Simulated Public Speaking Test in patients with Parkinson’s disease. J Psychopharmacol. 2020;34(2):189–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0269881119895536.
Fields RD, Neale EA, Nelson PG. Effects of patterned electrical activity on neurite outgrowth from mouse sensory neurons. J Neurosci. 1990;10(9):2950–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.10-09-02950.1990.
Franco V, Gershkovich P, Perucca E, Bialer M. The Interplay Between Liver First-Pass Effect and Lymphatic Absorption of Cannabidiol and Its Implications for Cannabidiol Oral Formulations. Clin Pharmacokinet. 2020;59(12):1493–500. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40262-020-00931-w.
Frimurer TM, Peters GH, Iversen LF, Andersen HS, Møller NPH, Olsen OH. Ligand-Induced Conformational Changes: Improved Predictions of Ligand Binding Conformations and Affinities. Biophys J. 2003;84(4):2273–81.
Full article: Towards a better understanding of the cannabinoid-related orphan receptors GPR3, GPR6, and GPR12 n.d https://www.tandfonline.com/doi/full/10.1080/03602532.2018.1428616. Accessed 21 Jun 2023
Felder CC, Briley EM, Axelrod J, Simpson JT, Mackie K. WA Devane. Anandamide, an endogenous cannabimimetic eicosanoid, binds to the cloned human cannabinoid receptor and stimulates receptor-mediated signal transduction Proc Natl Acad Sci U S A. 1993;90(16):7656–60. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.90.16.7656.
Ghovanloo MR, Shuart NG, Mezeyova J, Dean RA, Ruben PC, Goodchild SJ. Inhibitory effects of cannabidiol on voltage-dependent sodium currents. J Biol Chem. 2018;293(43):16546–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.RA118.004929.
Grayson L, Vines B, Nichol K, Szaflarski JP. An interaction between warfarin and cannabidiol, a case report. Epilepsy Behav Case Rep. 2017;9:10–1. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ebcr.2017.10.001.
Gilberg E, Gütschow M, Bajorath J. Promiscuous Ligands from Experimentally Determined Structures, Binding Conformations, and Protein Family-Dependent Interaction Hotspots. ACS Omega. 2019;4(1):1729–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acsomega.8b03481.
GPR55 protein expression summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000135898-GPR55. Accessed 19 Jul 2023
Ghovanloo MR, Ruben PC. Cannabidiol and sodium channel pharmacology: general overview, mechanism, and clinical implications. Neuroscientist. 2022;28(4):318–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/10738584211017009.
Godlewski G, Jourdan T, Szanda G, et al. Mice lacking GPR3 receptors display late-onset obese phenotype due to impaired thermogenic function in brown adipose tissue. Sci Rep. 2015;5(1):14953. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep14953.
S Goodman, E Wadsworth, G Schauer, D Hammond. Use and Perceptions of Cannabidiol Products in Canada and in the United States Cannabis and Cannabinoid Research. 2022;7(3):355-364 https://doiorg.publicaciones.saludcastillayleon.es/10.1089/can.2020.0093
Gunthorpe MJ, Chizh BA. Clinical development of TRPV1 antagonists: targeting a pivotal point in the pain pathway. Drug Discov Today. 2009;14(1):56–67. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.drudis.2008.11.005.
Giorgi S, Nikolaeva-Koleva M, Alarcón-Alarcón D, Butrón L, González-Rodríguez S. Is TRPA1 Burning Down TRPV1 as Druggable Target for the Treatment of Chronic Pain? Int J Mol Sci. 2019;20(12):2906. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms20122906.
GPR12 protein expression summary - The Human Protein Atlas n.d. https://www.proteinatlas.org/ENSG00000132975-GPR12. Accessed 19 Jul 2023
Gadiyar V, Lahey KC, Calianese D, et al. Cell Death in the Tumor Microenvironment: Implications for Cancer Immunotherapy. Cells. 2020;9(10):2207. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells9102207.
Gees M, Colsoul B, Nilius B. The role of transient receptor potential cation channels in Ca2+ signaling. Cold Spring Harb Perspect Biol. 2010;2(10). https://doiorg.publicaciones.saludcastillayleon.es/10.1101/cshperspect.a003962.
MH Grider R Jessu R Kabir. Physiology, Action Potential. In: StatPearls. StatPearls Publishing 2023. http://www.ncbi.nlm.nih.gov/books/NBK538143/. Accessed 23 Oct 2023
Graham ES, Angel CE, Schwarcz LE, Dunbar PR, Glass M. Detailed characterisation of CB2 receptor protein expression in peripheral blood immune cells from healthy human volunteers using flow cytometry. Int J Immunopathol Pharmacol. 2010;23(1):25–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/039463201002300103.
Higgins GA, Kilpatrick GJ, Bunce KT, Jones BJ, Tyers MB. 5-HT3 receptor antagonists injected into the area postrema inhibit cisplatin-induced emesis in the ferret. Br J Pharmacol. 1989;97(1):247–55.
Huestis MA. Human Cannabinoid Pharmacokinetics. Chem Biodivers. 2007;4(8):1770–804. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/cbdv.200790152.
Hoffmann E, Dittrich-Breiholz O, Holtmann H, Kracht M. Multiple control of interleukin-8 gene expression. J Leukoc Biol. 2002;72(5):847–55.
Huang Y, Rafael Guimarães T, Todd N, et al. G protein–biased GPR3 signaling ameliorates amyloid pathology in a preclinical Alzheimer’s disease mouse model. Proc Natl Acad Sci U S A. 2022;119(40):e2204828119. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.2204828119.
A Hsu, NA Painter. Probable Interaction Between Warfarin and Inhaled and Oral Administration of Cannabis. J Pharm Pract. 2020;33(6):915-918 https://doiorg.publicaciones.saludcastillayleon.es/10.1177/0897190019854958.
Hinckley M, Vaccari S, Horner K, Chen R, Conti M. The G-protein-coupled receptors GPR3 and GPR12 are involved in cAMP signaling and maintenance of meiotic arrest in rodent oocytes. Dev Biol. 2005;287(2):249–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ydbio.2005.08.019.
Henshaw FR, Dewsbury LS, Lim CK, Steiner GZ. The Effects of Cannabinoids on Pro- and Anti-Inflammatory Cytokines: A Systematic Review of In Vivo Studies. Cannabis and Cannabinoid Research. 2021;6(3):177–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/can.2020.0105.
Hanuš L, Abu-Lafi S, Fride E, et al. 2-Arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci U S A. 2001;98(7):3662–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.061029898.
Hao F, Feng Y. Cannabidiol (CBD) enhanced the hippocampal immune response and autophagy of APP/PS1 Alzheimer’s mice uncovered by RNA-seq. Life Sci. 2021;264:118624. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2020.118624.
Houston DB, Howlett AC. Differential receptor–G-protein coupling evoked by dissimilar cannabinoid receptor agonists. Cell Signal. 1998;10(9):667–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0898-6568(98)00013-8.
Hua DYH, Hindocha C, Baio G. Effects of cannabidiol on anandamide levels in individuals with cannabis use disorder: findings from a randomised clinical trial for the treatment of cannabis use disorder. Transl Psychiatry. 2023;13:131. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41398-023-02410-9.
Hwang YS, Kim YJ, Kim MO, et al. Cannabidiol upregulates melanogenesis through CB1 dependent pathway by activating p38 MAPK and p42/44 MAPK. Chem Biol Interact. 2017;273:107–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cbi.2017.06.005.
HTR3A protein expression summary - The Human Protein Atlas.n.d https://www.proteinatlas.org/ENSG00000166736-HTR3A Accessed 19 Jul 2023
Isaev D, Shabbir W, Dinc EY, Lorke DE, Petroianu G, Oz M. Cannabidiol Inhibits Multiple Ion Channels in Rabbit Ventricular Cardiomyocytes Front Pharmacol. 2022;13:821758. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2022.821758.
Iosub R, Avitabile D, Grant L, Tsaneva-Atanasova K, Kennedy HJ. Calcium-induced calcium release during action potential firing in developing inner hair cells. Biophys J. 2015;108(5):1003–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bpj.2014.11.3489.
Interactions Between Cannabinoids and Cytochrome P450-Metabolized Drugs - Full Text View - ClinicalTrials.gov n.d https://clinicaltrials.gov/ct2/show/NCT04201197. Accessed 21 Jun 2023
Jiang C, Ting AT, Seed B. PPAR-γ agonists inhibit production of monocyte inflammatory cytokines. Nature. 1998;391(6662):82–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/34184.
Jenny M, Santer E, Pirich E, Schennach H, Fuchs D. Δ9-Tetrahydrocannabinol and cannabidiol modulate mitogen-induced tryptophan degradation and neopterin formation in peripheral blood mononuclear cells in vitro. J Neuroimmunol. 2009;207(1):75–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jneuroim.2008.12.004.
Jenny M, Schröcksnadel S, Überall F, Fuchs D. The potential role of cannabinoids in modulating serotonergic signaling by their influence on tryptophan metabolism. Pharmaceuticals (Basel). 2010;3(8):2647–60. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph3082647.
Jang KI, Lee SH, Huh HJ, Chae JH. Influence of the 5-HT3A Receptor Gene Polymorphism and Childhood Sexual Trauma on Central Serotonin Activity. PLoS One. 2015;10(12):e0145269. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0145269.
Jacobson KA, von Lubitz DKJE, Daly JW, Fredholm BB. Adenosine receptor ligands: differences with acute versus chronic treatment. Trends Pharmacol Sci. 1996;17(3):108–13.
Jiang R, Yamaori S, Okamoto Y, Yamamoto I, Watanabe K. Cannabidiol is a potent inhibitor of the catalytic activity of cytochrome P450 2C19. Drug Metab Pharmacokinet. 2013;28(4):332–8. https://doiorg.publicaciones.saludcastillayleon.es/10.2133/dmpk.DMPK-12-RG-129.
Jîtcă G, Ősz BE, Vari CE, Rusz CM, Tero-Vescan A, Pușcaș A. Cannabidiol: Bridge between Antioxidant Effect, Cellular Protection, and Cognitive and Physical Performance. Antioxidants (Basel). 2023;12(2):485. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/antiox12020485.
DI Keller J Grenier G Christé 2009 Characterization of novel KCNH2 mutations in type 2 long QT syndrome manifesting as seizures Can J Cardiol 25(8):455-462 https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0828-282X(09)70117-5
Kathmann M, Flau K, Redmer A, Tränkle C, Schlicker E. Cannabidiol is an allosteric modulator at mu- and delta-opioid receptors. Naunyn Schmied Arch Pharmacol. 2006;372(5):354–61. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00210-006-0033-x.
Kowalski CW, Ragozzino FJ, Lindberg JEM. Cannabidiol activation of vagal afferent neurons requires TRPA1. J Neurophysiol. 2020;124(5):1388–98. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/jn.00128.2020.
Kwilasz AJ, Abdullah RA, Poklis JL, Lichtman AH, Negus SS. Effects of the Fatty Acid Amide Hydrolase (FAAH) Inhibitor URB597 on Pain-Stimulated and Pain-Depressed Behavior in Rats. Behav Pharmacol. 2014;25(2):119–29. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/FBP.0000000000000023.
K O’Brien 2022 Cannabidiol (CBD) in Cancer Management Cancers 14 4 885 https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers14040885
Kondra S, Chen F, Chen Y, Chen Y, Collette CJ, Xu W. A study of a hierarchical structure of proteins and ligand binding sites of receptors using the triangular spatial relationship-based structure comparison method and development of a size-filtering feature designed for comparing different sizes of protein structures. Proteins Struct Function Bioinformatics. 2022;90(1):239–57. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/prot.26215.
Leweke FM, Piomelli D, Pahlisch F, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2(3):e94. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/tp.2012.15.
Lu HC, Mackie K. An introduction to the endogenous cannabinoid system. Biol Psychiatry. 2016;79(7):516–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.biopsych.2015.07.028.
Laun AS, Shrader SH, Brown KJ, Song ZH. GPR3, GPR6, and GPR12 as novel molecular targets: their biological functions and interaction with cannabidiol. Acta Pharmacol Sin. 2019;40(3):300–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41401-018-0031-9.
AS Laun, ZH Song. GPR3 and GPR6, novel molecular targets for cannabidiol. Biochem Biophys Res Commun. 2017;490(1);17-21. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbrc.2017.05.165.
Lauckner JE, Jensen JB, Chen HY, Lu HC, Hille B, Mackie K. GPR55 is a cannabinoid receptor that increases intracellular calcium and inhibits M current. Proc Natl Acad Sci U S A. 2008;105(7):2699–704. https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.0711278105.
Li H, Lampe JN. Neonatal cytochrome P450 CYP3A7: a comprehensive review of its role in development, disease, and xenobiotic metabolism. Arch Biochem Biophys. 2019;673:108078. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.abb.2019.108078.
Luo Z, Ma L, Zhao Z, et al. TRPV1 activation improves exercise endurance and energy metabolism through PGC-1α upregulation in mice. Cell Res. 2012;22(3):551–64. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/cr.2011.205.
Liu C, Ma H, Slitt AL, Seeram NP. Inhibitory Effect of Cannabidiol on the Activation of NLRP3 Inflammasome Is Associated with Its Modulation of the P2X7 Receptor in Human Monocytes. J Nat Prod. 2020;83(6):2025–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.jnatprod.0c00138.
Lucas CJ, Galettis P, Schneider J. The pharmacokinetics and the pharmacodynamics of cannabinoids. Br J Clin Pharmacol. 2018;84(11):2477–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bcp.13710.
Laprairie RB, Bagher AM, Kelly MEM, Denovan-Wright EM. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br J Pharmacol. 2015;172(20):4790–805. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bph.13250.
Li F, Zhu W, Gonzalez FJ. Potential role of CYP1B1 in the development and treatment of metabolic diseases. Pharmacol Ther. 2017;178:18–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pharmthera.2017.03.007.
Lu X, Zhang N, Meng B, Dong S, Hu Y. Involvement of GPR12 in the regulation of cell proliferation and survival. Mol Cell Biochem. 2012;366(1):101–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11010-012-1287-x.
Lamba JK, Lin YS, Schuetz EG, Thummel KE. Genetic contribution to variable human CYP3A-mediated metabolism. Adv Drug Deliv Rev. 2002;54(10):1271–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0169-409X(02)00066-2.
Leone M, Attanasio A, Croci D, et al. 5-HT1A receptor hypersensitivity in migraine is suggested by the m-chlorophenylpiperazine test. NeuroReport. 1998;9(11):2605–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/00001756-199808030-00033.
Lucas RM, Luo L, Stow JL. ERK1/2 in immune signalling. Biochem Soc Trans. 2022;50(5):1341–52. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/BST20220271.
Miceli F, Soldovieri MV, Ambrosino P. Early-onset epileptic encephalopathy caused by gain-of-function mutations in the voltage sensor of Kv7.2 and Kv7.3 potassium channel subunits. J Neurosci. 2015;35(9):3782–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.4423-14.2015.
Menezes LFS, Sabiá Júnior EF, Tibery DV, Carneiro LD, Schwartz EF. Epilepsy-Related Voltage-Gated Sodium Channelopathies: A Review. Front Pharmacol. 2020;11:1276. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2020.01276.
Montaigne D, Butruille L, Staels B. PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol. 2021;18(12):809–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41569-021-00569-6.
Massi P, Vaccani A, Bianchessi S, Costa B, Macchi P, Parolaro D. The non-psychoactive cannabidiol triggers caspase activation and oxidative stress in human glioma cells. Cell Mol Life Sci. 2006;63(17):2057–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00018-006-6156-x.
M Marois Le, V Ballet, C Sanson. Cannabidiol inhibits multiple cardiac ion channels and shortens ventricular action potential duration in vitro. Eur J Pharmacol. 2020;886:173542. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejphar.2020.173542.
EJ Miller, SL Lappin. Physiology, cellular receptor StatPearls StatPearls Publishing 2023 http://www.ncbi.nlm.nih.gov/books/NBK554403/. Accessed 22 Jun 2023
Moraga-Cid G, San Martín VP, Lara CO, et al. Modulation of glycine receptor single-channel conductance by intracellular phosphorylation. Sci Rep. 2020;10(1):4804. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-020-61677-w.
McManus K, Ash E, Harper D, Smith R, Gruber S, Forester B. Caring for Behavioral Symptoms of Dementia (CBD): a new investigation into cannabidiol for the treatment of anxiety and agitation in Alzheimer’s dementia. Am J Geriatr Psychiatry. 2021;29(4, Supplement):S110–1. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jagp.2021.01.107.
Mecha M, Feliú A, Iñigo PM, Mestre L, Carrillo-Salinas FJ, Guaza C. Cannabidiol provides long-lasting protection against the deleterious effects of inflammation in a viral model of multiple sclerosis: A role for A2A receptors. Neurobiol Dis. 2013;59:141–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbd.2013.06.016.
Massi P, Valenti M, Vaccani A, et al. 5-Lipoxygenase and anandamide hydrolase (FAAH) mediate the antitumor activity of cannabidiol, a non-psychoactive cannabinoid. J Neurochem. 2008;104(4):1091–100. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1471-4159.2007.05073.x.
Molla MD, Akalu Y, Geto Z, Dagnew B, Ayelign B, Shibabaw T. Role of Caspase-1 in the Pathogenesis of Inflammatory-Associated Chronic Noncommunicable Diseases. J Inflamm Res. 2020;13:749–64. https://doiorg.publicaciones.saludcastillayleon.es/10.2147/JIR.S277457.
Mebratu Y, Tesfaigzi Y. How ERK1/2 Activation Controls Cell Proliferation and Cell Death Is Subcellular Localization the Answer? Cell Cycle. 2009;8(8):1168–75.
Mantovani A. Redundancy and Robustness Versus Division of Labour and Specialization in Innate Immunity. Semin Immunol. 2018;36:28–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.smim.2017.12.006.
C Muller P Morales PH Reggio. Cannabinoid Ligands Targeting TRP Channels. Front Mol Neurosci 2019 11 https://www.frontiersin.org/articles/10.3389/fnmol.2018.00487. Accessed 22 Nov 2022
Marijuana legality by state - Updated Oct 1, 2023. DISA 2025 https://disa.com/marijuana-legality-by-state
Ma B, Shatsky M, Wolfson HJ, Nussinov R. Multiple diverse ligands binding at a single protein site: A matter of pre-existing populations. Protein Sci. 2002;11(2):184–97.
Neher E. Ion channels for communication between and within cells. Neuron. 1992;8(4):605–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0896-6273(92)90083-P.
Navarro G, Varani K, Lillo A, et al. Pharmacological data of cannabidiol- and cannabigerol-type phytocannabinoids acting on cannabinoid CB1, CB2 and CB1/CB2 heteromer receptors. Pharmacol Res. 2020;159. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.phrs.2020.104940.
Nelson KM, Bisson J, Singh G, et al. The Essential Medicinal Chemistry of Cannabidiol (CBD). J Med Chem. 2020;63(21):12137–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.jmedchem.0c00724.
Oeckl P, Hengerer B, Ferger B. G-protein coupled receptor 6 deficiency alters striatal dopamine and cAMP concentrations and reduces dyskinesia in a mouse model of Parkinson’s disease. Exp Neurol. 2014;257:1–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.expneurol.2014.04.010.
McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are cannabidiol and Δ9-tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. Br J Pharmacol. 2015;172(3):737–53. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bph.12944.
MR Pazos, N Mohammed, H Lafuente. Mechanisms of cannabidiol neuroprotection in hypoxic–ischemic newborn pigs: Role of 5HT1A and CB2 receptors Neuropharmacology. 2013;71:282-291 https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuropharm.2013.03.027
Pandolfo P, Silveirinha V, dos Santos-Rodrigues A, et al. Cannabinoids inhibit the synaptic uptake of adenosine and dopamine in the rat and mouse striatum. Eur J Pharmacol. 2011;655(1–3):38–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejphar.2011.01.013.
Pereira SR, Hackett B, O’Driscoll DN, Sun MC, Downer EJ. Cannabidiol modulation of oxidative stress and signalling. Neuronal Signal. 2021;5((3)):NS20200080. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/NS20200080.
Porter AC, Sauer JM, Knierman MD, et al. Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J Pharmacol Exp Ther. 2002;301(3):1020–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/jpet.301.3.1020.
Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Δ9-tetrahydrocannabinol, cannabidiol and Δ9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.bjp.0707442.
Park MK, Park S, Kim HJ, et al. Novel effects of FTY720 on perinuclear reorganization of keratin network induced by sphingosylphosphorylcholine: Involvement of protein phosphatase 2A and G-protein-coupled receptor-12. Eur J Pharmacol. 2016;775:86–95. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejphar.2016.02.024.
PPARδ regulates glucose metabolism and insulin sensitivity | PNAS n.d https://www.pnas.org/doi/abs/10.1073/pnas.0511253103. Accessed 22 Jun 2023
De Petrocellis L, Ligresti A, Moriello AS, et al. Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol. 2011;163(7):1479–94. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1476-5381.2010.01166.x.
Penington NJ, Kelly JS, Fox AP. A study of the mechanism of Ca2+ current inhibition produced by serotonin in rat dorsal raphe neurons. J Neurosci. 1991;11(11):3594–609. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.11-11-03594.1991.
L Petrocellis, De V Vellani, A Schiano-Moriello. Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J Pharmacol Exp Ther. 2008;325(3):1007-1015. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/jpet.107.134809.
Perisetti A, Gajendran M, Dasari CS, et al. Cannabis hyperemesis syndrome: an update on the pathophysiology and management. Ann Gastroenterol. 2020;33(6):571–8. https://doiorg.publicaciones.saludcastillayleon.es/10.20524/aog.2020.0528.
Posner BI, Laporte SA. Chapter 1 - Cellular Signalling: Peptide Hormones and Growth Factors. In: Martini L, ed. Progress in Brain Research. Neuroendocrinology: The Normal Neuroendocrine System. Elsevier. 2010;181:1–16. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0079-6123(08)81001-1.
M Patrick, J Schulenberg, R Miech, L Johnson, P O’Malley, J Bachman. Monitoring the future panel study annual report: national data on substance use among adults ages 19 to 60, (1976–2021). 2022 https://monitoringthefuture.org/wp-content/uploads/2022/09/mtfpanelreport2022.pdf
De Petrocellis L, Orlando P, Moriello AS, et al. Cannabinoid actions at TRPV channels: effects on TRPV3 and TRPV4 and their potential relevance to gastrointestinal inflammation. Acta Physiol (Oxf). 2012;204(2):255–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1748-1716.2011.02338.x.
Qian Y, Gurley BJ, Markowitz JS. The Potential for Pharmacokinetic Interactions Between Cannabis Products and Conventional Medications. J Clin Psychopharmacol. 2019;39(5):462–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1097/JCP.0000000000001089.
Qin N, Neeper MP, Liu Y, Hutchinson TL, Lubin ML, Flores CM. TRPV2 Is Activated by Cannabidiol and Mediates CGRP Release in Cultured Rat Dorsal Root Ganglion Neurons. J Neurosci. 2008;28(24):6231–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.0504-08.2008.
Ruiz-Medina J, Ledent C, Valverde O. GPR3 orphan receptor is involved in neuropathic pain after peripheral nerve injury and regulates morphine-induced antinociception. Neuropharmacology. 2011;61(1–2):43–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuropharm.2011.02.014.
Rapin L, Gamaoun R, El Hage C, Arboleda MF, Prosk E. Cannabidiol use and effectiveness: real-world evidence from a Canadian medical cannabis clinic. J Cannabis Res. 2021;3:19. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s42238-021-00078-w.
Ross HR, Napier I, Connor M. Inhibition of Recombinant Human T-type Calcium Channels by Δ9-Tetrahydrocannabinol and Cannabidiol. J Biol Chem. 2008;283(23):16124–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M707104200.
Rock E, Bolognini D, Limebeer C, et al. Cannabidiol, a non-psychotropic component of cannabis, attenuates vomiting and nausea-like behaviour via indirect agonism of 5-HT1A somatodendritic autoreceptors in the dorsal raphe nucleus. Br J Pharmacol. 2012;165(8):2620–34. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1476-5381.2011.01621.x.
Rendic S, Guengerich FP. Human cytochrome P450 enzymes 5–51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition—toxic effects and benefits. Drug Metab Rev. 2018;50(3):256–342. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/03602532.2018.1483401.
Results from the 2019 National Survey on Drug Use and Health (NSDUH): Key Substance Use and Mental Health Indicators in the United States | SAMHSA Publications and Digital Products n.d. https://store.samhsa.gov/product/key-substance-use-and-mental-health-indicators-in-the-united-states-results-from-the-2019-national-survey-on-Drug-Use-and-Health/PEP20-07-01-001 Accessed 30 Oct 2023
Rosenberg EC, Chamberland S, Bazelot M, et al. Cannabidiol modulates excitatory-inhibitory ratio to counter hippocampal hyperactivity. Neuron. 2023;111(8):1282-1300.e8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuron.2023.01.018.
Rodriguez Araujo N, Fabiani C, Mazzarini Dimarco A, Bouzat C, Corradi J. Orthosteric and Allosteric Activation of Human 5-HT3A Receptors. Biophys J. 2020;119(8):1670–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bpj.2020.08.029.
Reggio PH. Endocannabinoid Binding to the Cannabinoid Receptors: What Is Known and What Remains Unknown. Curr Med Chem. 2010;17(14):1468–86.
Riera CE, Huising MO, Follett P, et al. TRPV1 Pain Receptors Regulate Longevity and Metabolism by Neuropeptide Signaling. Cell. 2014;157(5):1023–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cell.2014.03.051.
Ruhl T, Kim BS, Beier JP. Cannabidiol restores differentiation capacity of LPS exposed adipose tissue mesenchymal stromal cells. Exp Cell Res. 2018;370(2):653–62. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.yexcr.2018.07.030.
Seeman P. Cannabidiol is a partial agonist at dopamine D2High receptors, predicting its antipsychotic clinical dose. Transl Psychiatry. 2016;6(10):e920. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/tp.2016.195.
Smith RT, Gruber SA. Contemplating cannabis? The complex relationship between cannabinoids and hepatic metabolism resulting in the potential for drug-drug interactions. Front Psychiatry. 2023;13:1055481. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fpsyt.2022.1055481.
A Samanta TET Hughes VY Moiseenkova-Bell 2018 Transient Receptor Potential (TRP) Channels Subcell Biochem 87:141-165 https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-981-10-7757-9_6
Sideris A, Doan LV. An Overview of Cannabidiol. Anesth Analg. 2024;138(1):54–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1213/ANE.0000000000006584.
O’Sullivan SE. An update on PPAR activation by cannabinoids. Br J Pharmacol. 2016;173(12):1899–910. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bph.13497.
O’Sullivan SE, Sun Y, Bennett AJ, Randall MD, Kendall DA. Time-dependent vascular actions of cannabidiol in the rat aorta. Eur J Pharmacol. 2009;612(1):61–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejphar.2009.03.010.
Sprouse JS, Aghajanian GK. (-)-Propranolol blocks the inhibition of serotonergic dorsal raphe cell firing by 5-HT1A selective agonists. Eur J Pharmacol. 1986;128(3):295–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0014-2999(86)90782-X.
KS Swenson LE Gomez Wulschner VM Hoelscher, . Fetal cannabidiol (CBD) exposure alters thermal pain sensitivity, problem-solving, and prefrontal cortex excitability. Mol Psychiatry 2023:1-17 https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41380-023-02130-y. Published online July 11
KA Sharkey. 11 - Proposed mechanisms of cannabinoid hyperemesis syndrome—how can cannabinoid pathways both relieve and cause vomiting? In: Hasler WL, Levinthal DJ, Venkatesan T, eds. Cyclic Vomiting Syndrome and Cannabinoid Hyperemesis. Academic Press 2022 175-200. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/B978-0-12-821597-5.00003-4
Sweeney BP, Bromilow J. Liver enzyme induction and inhibition: implications for anaesthesia. Anaesthesia. 2006;61(2):159–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1365-2044.2005.04462.x.
Solanki M, Pointon A, Jones B, Herbert K. Cytochrome P450 2J2: Potential Role in Drug Metabolism and Cardiotoxicity. Drug Metab Dispos. 2018;46(8):1053–65. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/dmd.117.078964.
Shen M, Thayer SA. Δ9-Tetrahydrocannabinol Acts as a Partial Agonist to Modulate Glutamatergic Synaptic Transmission between Rat Hippocampal Neurons in Culture Mol Pharmacol. 1999;55(1):8–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/mol.55.1.8.
Sholler DJ, Schoene L, Spindle TR. Therapeutic Efficacy of Cannabidiol (CBD): a Review of the Evidence From Clinical Trials and Human Laboratory Studies. Curr Addict Rep. 2020;7(3):405–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40429-020-00326-8.
Suryavanshi SV, Zaiachuk M, Pryimak N, Kovalchuk I, Kovalchuk O. Cannabinoids Alleviate the LPS-Induced Cytokine Storm via Attenuating NLRP3 Inflammasome Signaling and TYK2-Mediated STAT3 Signaling Pathways In Vitro. Cells. 2022;11(9):1391. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cells11091391.
Smets K, Duarri A, Deconinck T. First de novo KCND3 mutation causes severe Kv4.3 channel dysfunction leading to early onset cerebellar ataxia, intellectual disability, oral apraxia and epilepsy. BMC Med Gen. 2015;16(1):51. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12881-015-0200-3.
Ward SJ, McAllister SD, Kawamura R, Murase R, Neelakantan H, Walker EA. Cannabidiol inhibits paclitaxel-induced neuropathic pain through 5-HT(1A) receptors without diminishing nervous system function or chemotherapy efficacy. Br J Pharmacol. 2014;171(3):636–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/bph.12439.
Savitz J, Lucki I, Drevets WC. 5-HT1A receptor function in major depressive disorder. Prog Neurobiol. 2009;88(1):17–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pneurobio.2009.01.009.
Tincello DG, Johnstone MJ. Treatment of hyperemesis gravidarum with the 5-HT3 antagonist ondansetron (Zofran). Postgrad Med J. 1996;72(853):688–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/pgmj.72.853.688.
Tanaka S, Miyagi T, Dohi E, et al. Developmental expression of GPR3 in rodent cerebellar granule neurons is associated with cell survival and protects neurons from various apoptotic stimuli. Neurobiol Dis. 2014;68:215–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.nbd.2014.04.007.
Tanaka S, Ishii K, Kasai K, Yoon SO, Saeki Y. Neural Expression of G Protein-coupled Receptors GPR3, GPR6, and GPR12 Up-regulates Cyclic AMP Levels and Promotes Neurite Outgrowth *. J Biol Chem. 2007;282(14):10506–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M700911200.
Tourino C, Valjent E, Ruiz-Medina J, Herve D, Ledent C, Valverde O. The orphan receptor GPR3 modulates the early phases of cocaine reinforcement. Br J Pharmacol. 2012;167(4):892–904. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1476-5381.2012.02043.x.
J Theriot HR Wermuth JV Ashurst. Antiemetic Serotonin-5-HT3 Receptor Blockers. In: StatPearls. StatPearls Publishing 2023 n.d. http://www.ncbi.nlm.nih.gov/books/NBK513318/. Accessed 21 Jun 2023
T Ng V Gupta MC Keshock. Tetrahydrocannabinol (THC). In: StatPearls StatPearls Publishing 2023 http://www.ncbi.nlm.nih.gov/books/NBK563174/. Accessed 22 Jun 2023
T Sugiura, S Kondo, A Sukagawa. 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain Biochem Biophys Res Commun. 1995;215(1):89-97 https://doiorg.publicaciones.saludcastillayleon.es/10.1006/bbrc.1995.2437
Tissue expression of TRPV1 - Summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000196689-TRPV1/tissue. Accessed 22 Jan 2023.
Tiboni GM, Ponzano A. Fetal safety profile of aromatase inhibitors: Animal data. Reprod Toxicol. 2016;66:84–92. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.reprotox.2016.09.016.
TRPV3 protein expression summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000167723-TRPV3. Accessed 19 Jul 2023
TRPM8 protein expression summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000144481-TRPM8. Accessed 19 Jul 2023
Tissue expression of CNR1 - Summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000118432-CNR1/tissue. Accessed 25 Oct 2023
Tissue expression of PPARG - Summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000132170-PPARG/tissue. Accessed 22 Jan 2023
Tissue expression of CNR2 - Summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000188822-CNR2/tissue. Accessed 25 Oct 2023
Tissue expression of HTR1A - Summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000178394-HTR1A/tissue. Accessed 25 Jan 2023
Tissue expression of GPR3 - Summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000181773-GPR3/tissue. Accessed 19 Jul 2023
Tour J, Sandström A, Kadetoff D, Schalling M, Kosek E. The OPRM1 gene and interactions with the 5-HT1a gene regulate conditioned pain modulation in fibromyalgia patients and healthy controls. PLoS ONE. 2022;17(11). https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0277427.
TRPA1 protein expression summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000104321-TRPA1. Accessed 19 Jul 2023
Tissue expression of GPR6 - Summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000146360-GPR6/tissue. Accessed 19 Jul 2023
TRPV4 protein expression summary - The Human Protein Atlas n.d https://www.proteinatlas.org/ENSG00000111199-TRPV4. Accessed 19 Jul 2023.
Todd RD. Neural development is regulated by classical neurotransmitters: dopamine D2 receptor stimulation enhances neurite outgrowth. Biol Psychiatry. 1992;31(8):794–807. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0006-3223(92)90311-m.
Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG. Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol. 2007;150(5):613–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sj.bjp.0707133.
Ujváry I, Hanuš L. Human metabolites of cannabidiol: a review on their formation, biological activity, and relevance in therapy. Cannabis Cannabinoid Res. 2016;1(1):90–101. https://doiorg.publicaciones.saludcastillayleon.es/10.1089/can.2015.0012.
Vitale RM, Iannotti FA, Amodeo P. The (Poly)Pharmacology of Cannabidiol in Neurological and Neuropsychiatric Disorders: Molecular Mechanisms and Targets. Int J Mol Sci. 2021;22(9):4876. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms22094876.
Vaysse PJ, Gardner EL, Zukin RS. Modulation of rat brain opioid receptors by cannabinoids. J Pharmacol Exp Ther. 1987;241(2):534–9.
Valverde O, Célérier E, Baranyi M, et al. GPR3 Receptor, a Novel Actor in the Emotional-Like Responses. PLoS ONE. 2009;4(3). https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0004704.
Vrechi TAM, Leão AHFF, Morais IBM, et al. Cannabidiol induces autophagy via ERK1/2 activation in neural cells. Sci Rep. 2021;11(1):5434. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-021-84879-2.
Vickers ER, Kasza A, Kurnaz IA, et al. Ternary Complex Factor-Serum Response Factor Complex-Regulated Gene Activity Is Required for Cellular Proliferation and Inhibition of Apoptotic Cell Death. Mol Cell Biol. 2004;24(23):10340–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/MCB.24.23.10340-10351.2004.
Walsh KB, McKinney AE, Holmes AE. Minor Cannabinoids: Biosynthesis, Molecular Pharmacology and Potential Therapeutic Uses. Front Pharmacol. 2021;12:777804. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2021.777804.
Wang L, Martin B, Brenneman R, Luttrell LM, Maudsley S. Allosteric Modulators of G Protein-Coupled Receptors: Future Therapeutics for Complex Physiological Disorders. J Pharmacol Exp Ther. 2009;331(2):340–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/jpet.109.156380.
Wiciński M, Fajkiel-Madajczyk A, Kurant Z, et al. The Use of Cannabidiol in Metabolic Syndrome—An Opportunity to Improve the Patient’s Health or Much Ado about Nothing? J Clin Med. 2023;12(14):4620. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jcm12144620.
Wang Y, Nakajima T, Gonzalez FJ, Tanaka N. PPARs as Metabolic Regulators in the Liver: Lessons from Liver-Specific PPAR-Null Mice. Int J Mol Sci. 2020;21((6)):2061. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21062061.
Wisastra R, Dekker FJ. Inflammation, Cancer and Oxidative Lipoxygenase Activity are Intimately Linked. Cancers (Basel). 2014;6(3):1500–21. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers6031500.
Watanabe K, Motoya E, Matsuzawa N, et al. Marijuana extracts possess the effects like the endocrine disrupting chemicals. Toxicology. 2005;206(3):471–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tox.2004.08.005.
Wu J, Hablitz JJ. Cooperative activation of D1 and D2 dopamine receptors enhances a hyperpolarization-activated inward current in layer I interneurons. J Neurosci. 2005;25(27):6322. https://doiorg.publicaciones.saludcastillayleon.es/10.1523/JNEUROSCI.1405-05.2005.
Watkins AR. Cannabinoid interactions with ion channels and receptors. Channels (Austin). 2019;13(1):162–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/19336950.2019.1615824.
Xiong W, Koo BN, Morton R, Zhang L. Psychotropic and nonpsychotropic cannabis derivatives inhibit human 5-HT3A receptors through a receptor desensitization-dependent mechanism. Neuroscience. 2011;184:28–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.neuroscience.2011.03.066.
Xiao YQ, Malcolm K, Worthen GS, et al. Cross-talk between ERK and p38 MAPK Mediates Selective Suppression of Pro-inflammatory Cytokines by Transforming Growth Factor-β *. J Biol Chem. 2002;277(17):14884–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1074/jbc.M111718200.
Xing Z, Gauldie J, Cox G, et al. IL-6 is an antiinflammatory cytokine required for controlling local or systemic acute inflammatory responses. J Clin Invest. 1998;101(2):311–20.
Yamaori S, Ebisawa J, Okushima Y, Yamamoto I, Watanabe K. Potent inhibition of human cytochrome P450 3A isoforms by cannabidiol: role of phenolic hydroxyl groups in the resorcinol moiety. Life Sci. 2011;88(15–16):730–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2011.02.017.
Yamaori S, Okamoto Y, Yamamoto I, Watanabe K. Cannabidiol, a major phytocannabinoid, as a potent atypical inhibitor for CYP2D6. Drug Metab Dispos. 2011;39(11):2049–56. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/dmd.111.041384.
Yamaori S, Kinugasa Y, Jiang R, Takeda S, Yamamoto I, Watanabe K. Cannabidiol induces expression of human cytochrome P450 1A1 that is possibly mediated through aryl hydrocarbon receptor signaling in HepG2 cells. Life Sci. 2015;136:87–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2015.07.007.
Yndart Arias A, Kolishetti N, Vashist A, Madepalli L, Llaguno L, Nair M. Anti-inflammatory effects of CBD in human microglial cell line infected with HIV-1. Sci Rep. 2023;13(1):7376. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-023-32927-4.
Yang Y, Kim SC, Yu T, et al. Functional Roles of p38 Mitogen-Activated Protein Kinase in Macrophage-Mediated Inflammatory Responses. Mediators Inflamm. 2014;2014:352371. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2014/352371.
Yuan J, Chen L, Wang J, et al. Adenosine A2A Receptor Suppressed Astrocyte-Mediated Inflammation Through the Inhibition of STAT3/YKL-40 Axis in Mice With Chronic Cerebral Hypoperfusion-induced White Matter Lesions. Front Immunol. 2022;13:841290. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.841290.
Yang KH, Galadari S, Isaev D, Petroianu G, Shippenberg TS, Oz M. The Nonpsychoactive Cannabinoid Cannabidiol Inhibits 5-Hydroxytryptamine3A Receptor-Mediated Currents in Xenopus laevis Oocytes. J Pharmacol Exp Ther. 2010;333(2):547–54. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/jpet.109.162594.
Yamaori S, Kushihara M, Yamamoto I, Watanabe K. Characterization of major phytocannabinoids, cannabidiol and cannabinol, as isoform-selective and potent inhibitors of human CYP1 enzymes. Biochem Pharmacol. 2010;79(11):1691–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bcp.2010.01.028.
HXB Zhang, L Heckman, Z Niday. Cannabidiol activates neuronal Kv7 channels. eLife. 2022; 11:e73246. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.73246
U Zanger, K Klein. Pharmacogenetics of cytochrome P450 2B6 (CYP2B6): advances on polymorphisms, mechanisms, and clinical relevance. Front Genet. 2013;4. https://www.frontiersin.org/articles/10.3389/fgene.2013.00024. Accessed 19 Jul 2023.
Zhao M, Ma J, Li M, et al. Cytochrome P450 enzymes and drug metabolism in humans. Int J Mol Sci. 2021;22(23):12808. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms222312808.
Zamarripa CA, Spindle TR, Surujunarain R, et al. Assessment of Orally Administered Δ9-Tetrahydrocannabinol When Coadministered With Cannabidiol on Δ9-Tetrahydrocannabinol Pharmacokinetics and Pharmacodynamics in Healthy Adults: A Randomized Clinical Trial. JAMA Netw Open. 2023;6(2):e2254752. https://doiorg.publicaciones.saludcastillayleon.es/10.1001/jamanetworkopen.2022.54752.
Zhou SF, Yang LP, Wei MQ, Duan W, Chan E. Insights into the structure, function, and regulation of human cytochrome P450 1A2. Curr Drug Metab. 2009;10(7):713–29. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/138920009789895552.
Zhang Y, Li H, Jin S, et al. Cannabidiol protects against Alzheimer’s disease in C. elegans via ROS scavenging activity of its phenolic hydroxyl groups. Eur J Pharmacol. 2022;919:174829. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ejphar.2022.174829.
Zou S, Kumar U. Cannabinoid Receptors and the Endocannabinoid System: Signaling and Function in the Central Nervous System. Int J Mol Sci. 2018;19(3):833. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms19030833.
Acknowledgements
Not applicable
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
Karli Swenson conceptualized, wrote, and edited this manuscript. The author approves the final manuscript as submitted and agrees to be accountable for all aspects of the work.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Swenson, K. Beyond the hype: a comprehensive exploration of CBD’s biological impacts and mechanisms of action. J Cannabis Res 7, 24 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s42238-025-00274-y
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s42238-025-00274-y