Table 3.
Class | Compound | Experimental Model | Findings | Reference |
---|---|---|---|---|
CBG | CBG | Mouse model of intestinal inflammation induced with the intracolonic administration of DNBS | Anti-inflammatory effect associated with the downregulation of inflammatory cytokines interleukin-1β, interleukin-10, and interferon-γ and reduction in iNOS expression. | [77] |
CBG CBGA CBGV |
In vitro HEK-293 cells stably overexpressing rat recombinant TRPV3 or TRPV4 |
CBGV and CBGA desensitize TRPV3 to the action of carvacrol at concentrations of EC50 = 0.8 and 7.4 µM. CBGV, CBGA, and CBG desensitize TRPV4 to the action of 4α-phorbol-12,13-didecanoate(4α-PDD) with EC50 values of 1.3–5.4 µM. These compounds desensitize TRPV3 and TRPV4 channels at lower doses than those at which they stimulate these channels. |
[76] | |
CBG CBGV |
HEK-293 cells stably overexpressing human TRPV1 | CBG and CBGV stimulated and desensitized human TRPV1. | [72] | |
CBG CBGA |
COX-1 enzyme, purified from ram seminal vesicles and COX-2 enzyme, purified from sheep placental cotyledons |
Inhibition of more than 30% of COX -1 and 292 COX -2 in a concentration-dependent manner. | [78] | |
CBG | Computational model of α2A, α2B, and α2C isoforms of murine and human 304 adrenoceptors | Affinity for the receptor appeared higher than that of the α2-adrenergic receptor agonist clonidine. | [152] | |
CBG | CBG | Mouse skin melanoma cells | Significant antitumor activity (inhibitory concentration (ICs0) = 31.31 gg/mL) in in vitro assay. | [117] |
CBG | Human oral epithelioid carcinoma 308 cell lines (KB) and NIH 3T3 fibroblasts | CBG exhibited the highest growth inhibitory activity against the cancer cell lines. | [118] | |
CBG | HEK-293 encoding the rat TRPM8 and overexpressing high levels of TRPM8 | Potent TRPM8 antagonist (IC50 = 0.16 ± 0.02). | [72] | |
CBG | CBG | Two human colon adenocarcinoma cell lines (Caco-2 and HCT 116, ATCC); Mouse azoxymethane (AOM) model of colon carcinogenesis | CBG inhibits the growth of CRC cells mainly via a pro-apoptotic mechanism and hinders the development and the growth of colon carcinogenesis in vivo. | [111] |
Mouse brain membranes | CBG activates α2-adrenoreceptors and blocks 5-HT1A receptors, antagonizing the 5-HT1A receptor agonist R-(+)-8-hydroxy-2-(di-n-propylamino) tetralin. | [86] | ||
CBG | TMEV (Thaler’s murine encephalomyelitis virus)-induced demyelinating disease (TMEV-IDD) in SJL/J mice | Anti-inflammatory and neuroprotective effects through the inhibition of IL-1β and IL-6 cytokines, and downregulation of PGE2 synthesis. CBG and CBG-quinone inhibited the microglia inflammatory response, protected neurons from toxic insults. |
[124] | |
CBG | CBG | Mouse model of Huntington’s disease (HD), created using 3-Nitropropionate i.p. repeated administration | Neuroprotective effects by downregulating the proinflammatory markers COX-2, 367 iNOS, IL-6, and TNF-α, by preventing neuronal degradation, downregulating disease-associated genes SgKL and CD44, and normalizing specific protein-1 levels. | [125] |
CBG | In vitro model of neuro inflammation on NSC-34 motor neurons | Pretreatment with CBG (7.5 μM) improved viability in treated cells through the inhibition of cell apoptosis, reduction in IL-1β, TNF-α, IFN-γ, and PPAR-γ proinflammatory protein levels, reduction in oxidative stress, and upregulation of Nrf-2 levels. | [74] | |
CBG | MC65 human neuron-like cell lines treated to induce intra-neuronal Alzheimer’s disease cell alterations | CBG blocked cell death, reduced oxidative damage, and prevented neurons from accumulating toxic β-amyloid protein. | [126] | |
CBG | Male Lister hooded rats | Doses between 120 and 140 mg/ kg of CBG induced a dose-dependent increase in food intake, increased the number of meals taken, decreased the latency until the first meal, and improved locomotor activity. | [128] | |
CBG | Standard S. aureus strain (ATCC 25923) and a clinical isolate (XU212) MRSA strain |
Antibacterial properties. | [110] | |
Methicillin-resistant S. aureus 404 (MRSA) strain; murine systemic infection model caused by MRSA |
In vitro disruption of the cytoplasmatic membrane of MRSA. In vivo efficacy against MRSA. |
[129] | ||
CBG | Keratinocyte proliferation assay | CBG had an inhibitory action on keratinocyte proliferation in a CB1/CB2 receptor-independent manner. | [130] | |
Human keratinocytes (HaCaT cells) |
CBG acted as a transcriptional repressor controlling cell proliferation and differentiation through a mechanism that involved increasing DNA methylation on the keratin-10 gene. | [131] | ||
CBG CBGA |
Human recombinant and pig kidney aldose reductase | Both compounds showed statistically significant ALR2 inhibitory activity by being able to interact with the major active site of the enzyme. | [79] | |
CBG | CBG | HEK-293 cells stably overexpressing human TRPV1 | Stimulates and desensitizes TRPV1 channels with an of EC50 = 21.0 ± 1.25. | [72] |
Colon cancer cells and normal colon cell lines | Cytotoxic activity on colon cancer cells, but reduced activity on normal colon cell lines. | [120] | ||
CBGV | HEK-293 cells encoding the rat TRPV2 and expressing high levels of TRPV2 | Antagonizes TRPV2 channels with an EC50 = 1.7 μM. | [72] | |
CBD | CBD | Murine (mouse) model of depression | CBD reduced immobility time in mice undergoing forced swimming test, the effect being similar to that produced by antidepressants such as imipramine. | [85] |
CBD | Mouse model of autism spectrum disorders | 10–20 mg/kg acute administration of CBD determined an improvement in social behavior. | [89] | |
CBD | Alzheimer’s disease mouse model | 20 mg/kg sub-chronic administration of CBD reversed cognitive deficits in object recognition memory and social recognition memory. | [153] | |
CBD | PTSD determined by yohimbine HCl (Tocris) administration in Wistar rats | 10 mg/kg acute administration came with therapeutic benefits for post-traumatic stress disorder symptoms. | [154] | |
CBD | Human breast cancer cell lines MDA-MB231 and MDA-MB436 | Significantly decreased Id-1 expression in metastatic breast cancer cells, leading to the downregulation of tumor aggressiveness. | [155] | |
CBD | CBDV | HEK-293 cells stably overexpressing human TRPV1 HEK-293 cells encoding the rat TRPV2 and expressing high levels of TRPV2 HEK-293 cells over- expressingTRPA1 HEK-293 encoding the rat TRPM8 and overexpressing high levels of TRPM8 |
Stimulates TRPV1 channels. Stimulates TRPV2 channels. Stimulates TRPA1 channels. Antagonizes TRPM8 channels. |
[72] |
hGPR55-HEK293 cells | Antagonizes GPR55 channels. | [148] | ||
CBD | 43-day-old rats received d,l-AMPH (4 mg/kg, i.p.) or vehicle in the conditioned place preference (CPP) paradigm (8 days), when each experimental group was re-assigned to receive CBD at two different doses (5 or 10 mg/kg, i.p) or control, for 5 days | CBD treatment prevented amphetamine relapse behavior in rats that had previously exhibited amphetamine-conditioned place preference, modulated immunoreactivity of dopaminergic targets in the prefrontal cortex and ventral striatum, areas with major involvement in drug dependence. CBD maintains dopamine transport levels. |
[27] | |
CBD | Mouse genetic model of Dravet syndrome (DS) | CBD reduced the frequency, severity, and duration of spontaneous seizures through the antagonization of GPR55 receptors. | [89] | |
Mecp2 mutant mice, a model of Rett syndrome (RTT) | CBDV rescues recognition memory deficits in Mecp2 mutant mice and delays the appearance of neurological defects. | [149] | ||
Mouse model for Rett syndrome, caused by mutations in the MECP2 gene | CBDV proved to attenuate brain alterations, restore the compromised general status, increase sociability, and partially restore motor coordination in treated mice. Molecularly, CBDV has antagonistic properties on GPR55. | [144] | ||
CBD | Double AD transgenic mouse model (APP/PS1) | CBD inhibited tau hyperphosphorylation and reduced Aβ production. | [26] | |
CBD | CBD | Wistar rat model of neuropathic pain (Bennet and Xie’s NP model (1988)) | CBD modulates chronic neuropathic pain and depression-specific behavior by activating 5-HT1A and CB1 receptors in the prefrontal cortex. | [83] |
CBDV | Autism-like behavior models through prenatal valproic acid exposure in rats | CBDV ameliorated behavioral abnormalities, restored hippocampal endocannabinoid signaling, and decreased neuroinflammation. | [149] | |
CBDV | In vitro model of ischemic stroke obtained by exposing cells to ischemic conditions through oxygen–glucose deprivation | CBDV has neuroprotective and anti-inflammatory properties. | [151] | |
CBDV | IBD mouse model of DNBS- and DSS-induced colitis | CBDV (orally or intraperitoneally) reduced the specific signs of colon inflammation–neutrophil infiltration, and increased colon weight and intestinal permeability. | [91] | |
Human colonic tissues from children with active ulcerative colitis | In vitro treatment with CBGV produced a significant reduction in the proinflammatory cytokine levels (IL-1β). | |||
CBDA | Mouse model of Dravet syndrome (Scn1aRX/+ mice) | CBDA exhibited significant anticonvulsant properties through a mechanism that could involve the 5-HT1A, GPR55, or TRPV1 receptors. | [136] | |
Rodent models of carrageenan-induced inflammatory pain | I.p. administration of CBDA at 60 min before carrageenan produced anti-inflammatory and anti-hyperalgesia effects. | [135] | ||
MDA-MB-231 breast cancer cell model | CBDA inhibited cell migration through a mechanism that is supposed to involve the activation of RhoA and through the inhibition of cAMP-dependent protein kinase A. | [94] | ||
Rat models of acute lithium chloride-induced nausea | CBDA suppresses nausea and vomiting in rats through the activation of the serotonin 1A receptor (5-HT1A). | [92,93] | ||
THC | Δ9-THC | Murine model of concanavalin A (ConA)-induced hepatitis | Intraperitoneal administration of THC inhibited hepatitis by significant decrease in liver enzymes and reduced liver tissue injury. THC treatment significantly suppressed inflammatory cytokines in ConA-induced hepatitis. | [156] |
Δ9-THC | Splenocytes of C57BL/6 mice | In vitro THC treatment significantly reduced proliferative response to mitogens, including anti-CD3 monoclonal antibodies (mAbs), concanavalin A (Con A), and lipopolysaccharide (LPS). | [157] | |
Δ9-THC | Sprague Dawley male rats | Δ9-THC therapy inhibited acetylcholinesterase, reduced amyloid-β levels and hippocampal neurogenesis, and induced brain-derived neurotrophic factor release through mixed CB1 and CB2 modulation. | [9,117] | |
Δ9-THC | Genes encoding human, mouse, and rat TRPV2 | Δ9-THC is a potent TRPV2 agonist. | [101] | |
Δ8-THC | Water-deprived albino rats | Groups treated with 5.0 and 10.0 mg/kg of Δ8-THC reduced intake of food at 1 day post-injection. | [158] | |
THCV | Rat recombinant TRPV3- and TRPV4-expressing HEK-293 cells | Stimulates TRPV3 with high efficacy (50–70% of the effect of ionomycin) and potency (EC50 = 3.7 μM) and TRPV4 with moderate-high efficacy (30–60% of the effect of ionomycin) and potency (EC50 = 0.9–6.4 μM) [76]. |
[76] | |
Δ9-THCA | HEK-293T, Neuro-2a (N2a), STHdh Q7/Q7, and STHdh Q111/Q111 cells, which express either a wild-type or a mutated form of the huntingtin protein | Δ9-THCA activated PPARγ and increased mitochondrial mass in neuroblastoma N2a cells and prevented cytotoxicity induced by serum deprivation in STHdh Q111/Q111 cells and by mutHtt-q94 in N2a cells. Δ9-THCA showed potent neuroprotective activity, worth consideration for the treatment of Huntington’s disease and possibly other neurodegenerative and neuroinflammatory diseases. |
[104] | |
Δ9-THCA-A | Mouse model of HFD significantly induced obesity | Administration of Δ9-THCA-A reduced fat mass and body weight gain, markedly ameliorating glucose intolerance and insulin resistance, and largely preventing liver steatosis, adipogenesis, and macrophage infiltration in fat tissues. | [159] |
CBG, cannabigerol; CB1, cannabinoid receptor 1; CB2, cannabinoid receptor 1; TRPM8, Transient Receptor Potential Melastatin-8; TRPV1, vanilloid receptor 1; α2-Adrenoceptor, alpha-2-Adrenoceptor; IL-1β, interleukin-1β; TNF-α, tumor necrosis factor alpha; IFN-γ, interferon gamma; PPAR-γ, peroxisome proliferator-activated receptor gamma; Nrf-2 levels, nuclear factor E2-related factor 2; TRPA1, transient receptor potential ankyrin 1; TRPV3, transient receptor potential vanilloid-3; TRPV4, transient receptor potential vanilloid-type 4; iNOS expression, inducible nitric oxide synthase expression; CBGV, cannabigerovarin; CBGA, cannabigerolic acid; CBD, cannabidiol; CBDV, cannabidivarin; CBDA, cannabidiolic acid; COX-1, COX-2, Cyclooxygenase-1, Cyclooxygenase-2; SOD, superoxide dismutase; PLA2, Phospholipase A2; MAGL, monoacylglycerol lipase; PPARα/γ, peroxisome proliferator-activated receptors α/γ; GPR55, G protein-coupled receptor 55; CBDV, cannabidivarin; TRPV2, transient receptor potential vanilloid 2; GPR6, G Protein-Coupled Receptor 6; DAGLα, diacylglycerol lipase-alpha; AEA, N-arachidonoylethanolamine (anandamide); 5HT1A, 5-hydroxytryptamine receptor 1A; cAMP protein kinase A, cyclic adenosine monophosphate protein kinase A; THC, tetrahydrocannabinol; Δ9-THC, Δ9-trans-tetrahydrocannabinol; MDSCs, myeloid-derived suppressor cells; AchE, acetylcholinesterase; Δ8-THC, Δ8-trans-tetrahydrocannabinol; THCA-A, tetrahydrocannabinolic acid; IBD, inflammatory bowel disease; ConA, concanavalin A; mAbs, monoclonal antibodies; LPS, lipopolysaccharide; i.p., intraperitoneal; DNBS, dinitrobenzene sulphonic acid; DSS, dextran sulfate sodium; AD, Alzheimer’s disease.