THCV | CBDV | CBCV | 2-1 | blank | blank | blank | blank | blank | blank |
CBGV | CBDB | CBN | blank | blank | blank | blank | blank | blank | blank |
THCΔ8 | THCΔ9 | THCB | blank | blank | blank | blank | blank | APG | KAF |
CBC | THCH | THCΔ10 | blank | blank | blank | blank | blank | LTL | QUC |
CBL | CB | CBD | MY | TE | SA | LI | PI | MYC | CFB |
CBG | CBDM | CBGL | CM | LN | QR Code | GE | TP | VIX | ISX |
CBE | THCP | CBDP | HU | CA | FA | BI | PH | CFA | ORT |
blank | blank | THCVA | CBCA | CBDA | THCA | CBNA | CBGA | blank | blank |
THCV | CVDB | CBCV | 2-1 | ||||
CBGV | CBDB | CBN | |||||
THCΔ8 | THCΔ9 | THCB | |||||
CBC | THCH | THCΔ10 | |||||
CBL | CBT | CBD | |||||
CBG | CBDM | CBGL | |||||
CBE | THCP | CBDP |
MY | TE | SA | LI | PI |
CM | LN | QR Code | GE | TP |
HU | CA | FA | BI | PH |
APG | KAF |
LTL | QUC |
MYC | CFB |
VIX | ISX |
CFA | ORT |
THCVA | CBCA | CBDA | THCA | CBNA | CBGA |
Acannability™ is a cooperative source of communication, education, and standards for the cannabis and hemp industries.
© 2024 Positive Growth Ventures, LLC
Each of the 51 boxes on the Cannabis Periodic Table of Molecules ™ chart represent either a molecule of the cannabis plant or a precursor acid. There are 45 molecules (cannabinoids, terpenes and flavonoids) and 6 precursor acids shown. A molecule and a precursor acid are two different chemical entities with distinct characteristics and functions.
A molecule is a group of atoms bonded together. It is the smallest unit of a chemical compound that retains the chemical properties of that compound. Molecules can be simple, containing only a few atoms, or complex, consisting of hundreds or thousands of atoms. They can be composed of the same type of atoms (e.g., O2 - oxygen molecule) or different types of atoms (e.g., H2O - water molecule). Molecules can exist independently or as part of larger structures.
A precursor acid, on the other hand, refers to a chemical compound that participates in a chemical reaction that produces other compounds—under certain conditions, it creates an acid or it can convert the molecule into its non-acidic form. Precursor acids typically contain one or more acidic functional groups, such as a carboxyl group (-COOH) or a sulfate group (-SO4H). When these groups undergo specific chemical reactions, they can release hydrogen ions (H+) in a solution, forming an acid.
For example, THCa is the acidic form of THC. Although THCa does have medicinal properties, it does not have psychoactive properties. The process of decarboxylation requires exposure to heat. When vaporizing and smoking, high temperatures quickly decarboxylate THCa to THC, enabling your body to immediately absorb the THC into the lungs. This results in the consumer experiencing the psychoactive properties THC is known for. Oxygen exposure can also contribute to the decarboxylation process.
The key difference between a molecule and a precursor acid is that a molecule is a general term referring to any group of bonded atoms, while a precursor acid specifically refers to a chemical compound that can be converted into an acid through chemical reactions. A precursor acid is a subset of molecules that have the potential to generate acidic properties under certain conditions.
The boxes shown were selected as molecules having known or suspected medicinal value based on Acannability's review of published research. The selection process was led by Acannability Advisor Mr. Joseph Friedman, RPh, MBA, a Medical Cannabis Pharmacist and Pharmacology Expert. Mr. Friedman was the founding member at Professional Dispensaries of Illinois Medical LLC, a medical cannabis dispensary owned and operated by pharmacists, nurses, and cannabis experts. He is an expert in the scientific, legal, social, political, and financial challenges facing U.S. entrants to the burgeoning medical cannabis industry. Mr. Friedman has contributed to several published articles in local and national publications and continually advocates for pharmacists’ involvement in the medical cannabis industry.
The symbols on the Cannabis Periodic Table of Molecules ™ chart are shorthand notations used to represent each molecule. These symbols are two to four letters, and they play a crucial role in identifying and organizing the elements in a systematic manner. The symbols are based on the English names, Latin names, or their discoverer's names. The symbols help chemists and scientists communicate efficiently and unambiguously about molecules and precursor acids. In summary, the symbols on the periodic table represent chemical elements and are shorthand notations that help in identifying, organizing, and communicating about these elements in the field of chemistry and beyond. The symbols shown are publicly used in the literature.
The organization of the Cannabis Periodic Table of Molecules ™ chart was based on the common assessment of the cannabis plant as a whole. Cannabinoids, terpenes, flavonoids, and precursor acids are commonly recognized categories. The periodic table's arrangement is such that molecules with increasing atomic weight are ordered from left to right in rows (periods).
A cannabinoid is a type of chemical compound that is found in the cannabis plant, as well as in some other plants and in the human body. Cannabinoids interact with the body’s endocannabinoid system, which is involved in regulating a wide range of physiological processes such as pain, mood, appetite, and immune function. There are over 100 cannabinoids that have been identified in the cannabis plant, including tetrahydrocannabinol (THC) and cannabidiol (CBD), which are the most well-known and widely studied.
Terpenes are a large and diverse class of organic compounds that are found in plants, including in the cannabis plant. Terpenes are formed from the same basic building blocks as cannabinoids and are produced in the same glandular trichomes on the cannabis plant as cannabinoids. Some of the most common terpenes found in cannabis include myrcene, limonene, pinene, and caryophyllene, but there are many others as well. In addition to the sensory properties, terpenes are believed to have a range of potential therapeutic benefits.
Flavonoids are a group of plant compounds that belong to the larger class of polyphenols. They are widely distributed in nature and are found in many fruits, vegetables, and medicinal plants, including the cannabis plant. Flavonoids are responsible for giving many plants their bright colors, such as the deep purple color of blueberries or the orange of citrus fruits. There are many different types of flavonoids, each with its own chemical structure and properties.
In addition, several flavonoids retain medicinal value that may work in concert with cannabinoids and terpenes to enhance or add to the medical properties of the cannabis plant. This is also commonly known as the “Entourage Effect.”
The atomic masses shown in the Cannabis Periodic Table of Molecules ™ chart were determined by public references or the chemical formula. In the case of the latter, each element’s molecular mass was identified and totaled. To provide an example, for water (H20), Acannability’s team identified two hydrogen elements with mass ~1 each and one oxygen element with mass ~16, totaling 18 for the molecule. After identifying or calculating the atomic masses of all the molecules, the table was constructed to reflect an objective structure, similar to the Periodic Table.
The Cannabis Periodic Table of Molecules ™ chart is used as a common language for cannabis consumers and industry participants to promote a more informed and science based conversation about the benefits of the cannabis plant. Some of the most common uses of the chart are as follows:
Product Selection. The chart can benefit conversation between consumers and the health care provider on which molecules are known/believed to address their desired use. In conjunction with the Acannability seal, the individual molecules shown can be used to compare product claims as shown on product labels.
Molecule Identification. The chart allows for easy identification of molecules and their properties. By looking at an element's symbol or atomic number, one can quickly determine its name, atomic mass, and other essential characteristics.
Chemical Nomenclature. When naming chemical compounds, the chart may be consulted to determine the symbols and valencies of elements involved in the compound.
Understanding Trends. As the cannabis industry continues to mature, the Cannabis Periodic Table of Molecules ™ chart will be updated to reflect the molecules of current interest by consumers as well as those promoted by the industry.
Research and Experimentation. Scientists and researchers may use the chart as a reference when investigating new cannabis compounds or reactions.
Education. In educational settings, the chart may be used to introduce students to the fundamental concepts of chemistry and to organize the study of various elements and their compounds.
Yes. The Cannabis Periodic Table of Molecules ™ chart was developed by Acannabilty through the work of its Advisor Joseph Friedman. It is free to use by Acannability members and available at no charge to others by a simple copyright license acknowledgement. A printable version of the chart is available at Acannability.com.
The Cannabis Periodic Table of Molecules ™ chart is an essential tool in the field of cannabinoid chemistry and serves as a fundamental reference for understanding the molecular properties of the cannabis plant. However, it should never be used as a source of medical advice. Medical decisions and treatments should be based solely on the guidance of qualified healthcare professionals who consider individual patient factors, medical history, and specific conditions. Always consult with a licensed medical practitioner for any health-related concerns or queries and refrain from using the Cannabis Periodic Table of Molecules ™ chart to make medical decisions.
About Acannability
Acannability is a cooperative source of communication, education, and standards for the cannabis and hemp industries.™
© 2022 Acannability, LLC
Links
Anti-convulsant, analgesic, neuroprotective, anti-inflammatory
Phytol (PH) is a natural diterpene alcohol found in a variety of plants, including cannabis. It is particularly abundant in the leaves of the cannabis plant and is considered one of the non-psychoactive compounds present in cannabis. Phytol is often derived from the degradation of chlorophyll, and it contributes to the characteristic aroma and flavor of cannabis. While it is not known for its psychoactive properties, Phytol has gained attention for its potential health benefits. Research has suggested that Phytol may have anti-inflammatory, antioxidant, and neuroprotective properties, which could make it a valuable compound in the medical and therapeutic applications of cannabis. It has also been studied for its role in modulating the endocannabinoid system, potentially influencing the effects of other cannabinoids in cannabis. However, further research is needed to fully understand Phytol’s specific actions, mechanisms of action, and potential therapeutic applications within the context of cannabis and human health.
The article titled “Phytol, a diterpene alcohol, inhibits the inflammatory response by reducing cytokine production and oxidative stress” by Silva et al., published in the journal Fundam Clin Pharmacol in 2014, provides important insights into the anti-inflammatory properties of Phytol. The study investigates how Phytol, a diterpene alcohol, exerts its anti-inflammatory effects by targeting two key aspects of inflammation: cytokine production and oxidative stress. The findings indicate that Phytol can significantly reduce the production of pro-inflammatory cytokines and mitigate oxidative stress, both of which are pivotal contributors to inflammatory processes. Similarly, the article titled “Phytol, a Chlorophyll Component, Produces Antihyperalgesic, Anti-inflammatory, and Antiarthritic Effects: Possible NFκB Pathway Involvement and Reduced Levels of the Proinflammatory Cytokines TNF-α and IL-6” by Carvalho et al. provides valuable insights into the anti-inflammatory properties of Phytol. The study suggests that Phytol may exert its anti-inflammatory actions by modulating the NFκB pathway and reducing the levels of proinflammatory cytokines such as TNF-α and IL-6. This mechanism of action aligns with its observed anti-inflammatory effects and underscores its potential as a natural compound for managing inflammatory conditions. However, further research and clinical studies are needed to fully understand the therapeutic implications and safety profile of Phytol in treating inflammation-related disorders.
In the article titled “Anticonvulsant effect of phytol in a pilocarpine model in mice” by J.P. Costa et al., published in Neuroscience Letters in 2012, the authors investigate the potential anti-convulsant properties of Phytol. The study employs a mouse model using pilocarpine, a chemical inducer of seizures, to assess the effects of Phytol on convulsive behavior. The findings of this research reveal that Phytol demonstrates an anticonvulsant effect, suggesting its potential as a therapeutic agent in the management of seizures. This study contributes valuable insights into the neuroprotective properties of Phytol and its potential role in mitigating epileptic episodes. While the exact mechanisms behind its anticonvulsant action need further elucidation, these results underscore the promise of Phytol as a candidate for the development of novel antiepileptic treatments. Further research, including clinical trials, is essential to validate its efficacy and safety for human use in epilepsy management.
The article titled “Neuroprotective effect of the marine macroalga Gelidiella acerosa: identification of active compounds through bioactivity-guided fractionation” by Arif Nisha Syad et al., published in Pharmaceutical Biology in 2016, delves into the neuroprotective properties of Phytol. Through bioactivity-guided fractionation, the research identifies Phytol as one of the active compounds with neuroprotective effects. This suggests that Phytol has the capacity to safeguard neuronal cells and potentially mitigate neurological disorders or conditions associated with neuronal damage. While this study provides valuable insights into the neuroprotective properties of Phytol derived from Gelidiella acerosa, further research is required to elucidate the precise mechanisms of action and its potential application in neuroprotection and neurodegenerative disease management. Nevertheless, this investigation highlights Phytol as a promising candidate for future research in the realm of neuroprotection and neurobiology.
The article titled “Phytol from Faeces Bombycis alleviated migraine pain by inhibiting Nav1.7 sodium channels” by Jianan Song et al., published in the Journal of Ethnopharmacology in 2023, explores the analgesic properties of Phytol. The study investigates Phytol’s potential to alleviate migraine pain by targeting Nav1.7 sodium channels. Nav1.7 channels are known to play a crucial role in pain perception and transmission. The research findings indicate that Phytol may effectively inhibit these channels, which suggests its analgesic potential in relieving migraine pain. This study provides valuable insights into the molecular mechanisms underlying Phytol’s pain-relieving properties and highlights its potential as a natural compound for the management of migraine and possibly other pain-related conditions. Further research and clinical studies will be essential to confirm and explore the full extent of Phytol’s analgesic benefits.
Testimonial
Anti-tumor, antibacterial, anxiolytic, neuroprotective, anti-inflammatory
Bisabolol (BI), also known as alpha-bisabolol or levomenol, is a naturally occurring terpene found in various plant species, including cannabis. It is one of the many aromatic compounds that contribute to the complex chemical profile of cannabis, giving it its distinctive scent and flavor. Bisabolol is renowned for its potential health benefits and soothing properties, making it a sought-after compound in the cannabis plant. It is often used in cannabis-based products, such as topicals and skincare items, due to its d antioxidant, and skin-soothing qualities. Additionally, Bisabolol is believed to have potential therapeutic applications, including its use as an analgesic and anxiolytic agent. While cannabis contains a multitude of compounds, including cannabinoids like THC and CBD, Bisabolol’s presence adds to the overall pharmacological diversity of the plant and may contribute to the entourage effect, where various compounds work together synergistically to produce therapeutic effects. However, further research is needed to fully understand the specific roles and therapeutic potential of Bisabolol in the context of cannabis.
In the article titled “Inhibitory effects of (-)-α-bisabolol on LPS-induced inflammatory response in RAW264.7 macrophages” by Seungbeom Kim et al., published in Food and Chemical Toxicology in 2011, the anti-inflammatory properties of Bisabolol are investigated using an in vitro model of LPS-induced inflammation in RAW264.7 macrophages. The study demonstrates that (-)-α-bisabolol effectively inhibits the inflammatory response in these immune cells. This suggests that Bisabolol may exert anti-inflammatory effects by modulating the release of pro-inflammatory mediators and cytokines. Such findings underscore the potential therapeutic utility of Bisabolol in the management of inflammatory conditions and provide valuable insights into its mechanisms of action at the cellular level. Also, the article “Anti-nociceptive and anti-inflammatory activities of (−)-α-bisabolol in rodents” by Rocha et al., published in Naunyn-Schmiedeberg’s Archives of Pharmacology in 2011, found that that α-bisabolol exhibits anti-inflammatory activity in rodents, suggesting its potential as an anti-inflammatory agent. While these studies contribute to our understanding of Bisabolol’s anti-inflammatory properties, further research, including in vivo studies and clinical trials, would be necessary to confirm its efficacy and safety in a broader context for potential medical applications.
The article titled “Neutrophil Immunomodulatory Activity of Farnesene, a Component of Artemisia dracunculus Essential Oils” by Schepetkin et al. explores the anti-inflammatory properties of farnesene. Farnesene is identified as a component of Artemisia dracunculus essential oils and is investigated for its potential immunomodulatory effects on neutrophils, a type of white blood cell crucial in the inflammatory response. The study indicates that farnesene exhibits immunomodulatory activity, suggesting its potential role in mitigating inflammation. This finding is significant, as excessive or prolonged inflammation is associated with various chronic diseases and health conditions. Farnesene’s ability to modulate neutrophil function implies its possible use in the development of anti-inflammatory therapies. However, further research is needed to fully elucidate the mechanisms and therapeutic applications of farnesene in the context of inflammation and immune regulation.
In the article titled “Amyloid-β induced neuropathological actions are suppressed by Padina gymnospora and its active constituent α-bisabolol in Neuro2a cells and transgenic Caenorhabditis elegans Alzheimer’s model” by Balakrishnan Shanmuganathan et al., the neuroprotective properties of Bisabolol (α-bisabolol) are investigated in the context of Alzheimer’s disease. The study employs both cellular and transgenic nematode models to explore the impact of Bisabolol on amyloid-β-induced neurotoxicity. The findings suggest that Bisabolol exhibits promising neuroprotective effects by mitigating neuropathological actions associated with Alzheimer’s disease. This suggests that Bisabolol may hold potential as a therapeutic agent in the management of Alzheimer’s and other neurodegenerative disorders, potentially offering a novel avenue for the development of treatments that target the underlying mechanisms of these diseases. However, further research, particularly in mammalian models and clinical trials, is necessary to validate and fully understand the scope of Bisabolol’s neuroprotective properties and its applicability in human healthcare.
In the article titled “Antitumor activity of (-)-α-bisabolol-based thiosemicarbazones against human tumor cell lines” by Alan P. da Silva et al., published in the European Journal of Medicinal Chemistry in 2010, the authors explore the potential anti-tumor properties of Bisabolol-derived compounds. The study investigates thiosemicarbazones derived from (-)-α-bisabolol and their effects on various human tumor cell lines. Correspondingly, the article titled “Insight into the apoptosis-inducing action of α-bisabolol towards malignant tumor cells: Involvement of lipid rafts and Bid” by Elena Darra et al. delves into the potential anti-tumor properties of Bisabolol (α-bisabolol). This study investigates the mechanisms underlying α-bisabolol’s ability to induce apoptosis (programmed cell death) in malignant tumor cells. The findings suggest that α-bisabolol’s anti-tumor effects may be attributed to its involvement in disrupting lipid rafts, which play a crucial role in cell signaling and membrane organization. Additionally, the study implicates the activation of the pro-apoptotic protein Bid as a potential mediator of α-bisabolol’s action. These insights shed light on the molecular pathways through which α-bisabolol exerts its anti-tumor effects, providing valuable information for further research into its potential as a therapeutic agent in cancer treatment. However, it is important to note that more extensive preclinical and clinical studies are necessary to validate these findings and assess the safety and efficacy of α-bisabolol as a cancer treatment option.
In the study titled “Evidence for the involvement of the GABAergic, but not serotonergic transmission in the anxiolytic-like effect of bisabolol in the mouse elevated plus maze” by Tabari and Tehrani, published in Naunyn-Schmiedeberg’s Archives of Pharmacology in 2017, the anxiolytic properties of bisabolol are investigated. The research focuses on the mechanism of bisabolol’s anxiolytic effects using the elevated plus maze model in mice. The findings of this study suggest that bisabolol’s anxiolytic-like effects may primarily involve the GABAergic transmission system, which plays a crucial role in regulating anxiety and relaxation. This implies that bisabolol may modulate anxiety by affecting the GABAergic neurotransmission, which is known for its inhibitory action in the central nervous system. Notably, the study highlights the potential therapeutic utility of bisabolol in addressing anxiety-related conditions and provides valuable insights into the underlying neuropharmacological mechanisms of its anxiolytic properties.
Testimonial
Neuroprotective, anti-inflammatory
Farnesene (FA) is a sesquiterpene hydrocarbon compound that can be found in the cannabis plant, among other plant species. In cannabis, farnesene contributes to the plant’s unique chemical profile, known as its terpene profile, which influences its aroma, flavor, and potential therapeutic effects. Farnesene is a significant component of the complex mixture of terpenes found in cannabis, and its levels can vary among different strains and chemovars. While farnesene’s specific effects in cannabis are still being explored, it is known to have a pleasant, fruity, and woody aroma that can contribute to the overall sensory experience of consuming cannabis. Additionally, farnesene, like many terpenes, may have potential health benefits, including anti-inflammatory and antioxidant properties, making it an intriguing area of research within the broader field of cannabis science and medicine. Further studies are needed to fully understand the specific role of farnesene in cannabis and its potential therapeutic applications.
The article titled “In vitro neuroprotective effects of farnesene sesquiterpene on Alzheimer’s disease model of differentiated neuroblastoma cell line” by Mehmet Enes Arslan, Hasan Türkez, and Adil Mardinoğlu explores the potential neuroprotective properties of farnesene in the context of Alzheimer’s disease. The study utilizes an in vitro model with a differentiated neuroblastoma cell line to investigate the effects of farnesene on neuronal health. The findings of this research suggest that farnesene has neuroprotective potential, as it demonstrates beneficial effects on the Alzheimer’s disease model, which could be indicative of its ability to mitigate neurodegenerative processes. While in vitro studies provide valuable insights, further in vivo research and clinical trials are essential to confirm the neuroprotective properties of farnesene and explore its potential as a therapeutic agent for Alzheimer’s disease. Nevertheless, this study highlights the promising avenue of farnesene as a compound with neuroprotective attributes worthy of further investigation in the realm of neurodegenerative diseases.
The article titled “Neutrophil Immunomodulatory Activity of Farnesene, a Component of Artemisia dracunculus Essential Oils” by Schepetkin et al. explores the anti-inflammatory properties of farnesene. Farnesene is identified as a component of Artemisia dracunculus essential oils and is investigated for its potential immunomodulatory effects on neutrophils, a type of white blood cell crucial in the inflammatory response. The study indicates that farnesene exhibits immunomodulatory activity, suggesting its potential role in mitigating inflammation. This finding is significant, as excessive or prolonged inflammation is associated with various chronic diseases and health conditions. Farnesene’s ability to modulate neutrophil function implies its possible use in the development of anti-inflammatory therapies. However, further research is needed to fully elucidate the mechanisms and therapeutic applications of farnesene in the context of inflammation and immune regulation.
Testimonial
Gastro-protective, anti-inflammatory
Terpineol (TP) is a naturally occurring monoterpene alcohol that can be found in various cannabis strains, contributing to the plant’s distinctive aroma and flavor. Terpineol is not exclusive to cannabis; it is also present in a wide range of other plants, including pine trees, lilacs, and lime blossoms. In cannabis, terpineol is part of the complex mixture of terpenes that give each strain its unique scent and taste profile, often described as floral, citrusy, or piney. Beyond its role as a flavor and fragrance compound, terpineol has been the subject of research for its potential therapeutic properties. Some studies suggest that terpineol may have sedative, relaxing, and anti-inflammatory effects, making it of interest to both medical and recreational users of cannabis. While the research on terpineol’s specific benefits is ongoing, its presence in cannabis and other plants underscores the complex interplay between terpenes and cannabinoids in influencing the overall effects of cannabis strains.
The article titled “Gastroprotective activity of α-terpineol in two experimental models of gastric ulcer in rats,” published in Daru in 2011, investigates the gastro-protective properties of α-terpineol. This study explores how α-terpineol, a natural terpene found in various plants, exerts protective effects on the stomach in two different experimental models of gastric ulcers in rats. The findings suggest that α-terpineol exhibits gastroprotective activity, indicating its potential to mitigate gastric ulcers. This effect could be attributed to its ability to modulate various factors involved in ulcer formation, such as reducing oxidative stress and inflammation. While these results are promising, it’s crucial to emphasize that further research, including clinical trials in humans, is essential to validate the practical applications of α-terpineol as a gastroprotective agent. Nevertheless, this study provides valuable insights into the potential gastroprotective properties of terpineol, contributing to the exploration of natural compounds for stomach-related conditions.
The article titled “α-Terpineol Reduces Mechanical Hypernociception and Inflammatory Response,” published in Basic & Clinical Pharmacology & Toxicology in 2012, investigates the anti-inflammatory properties of terpineol, specifically α-terpineol. The study explores how α-terpineol impacts mechanical hypernociception (increased sensitivity to pain) and the inflammatory response. The findings suggest that α-terpineol exhibits anti-inflammatory effects, as it reduces mechanical hypernociception and suppresses the inflammatory response. This indicates that α-terpineol may have the potential to mitigate pain and inflammation, potentially through modulating pathways involved in the inflammatory process. These results provide valuable insights into the therapeutic potential of terpineol as an anti-inflammatory agent, highlighting its relevance in the context of pain management and inflammatory conditions. Nonetheless, further research, including clinical studies, would be necessary to determine its practical applications in medical contexts.
Testimonial
Immunomodulatory, anti-tumor, anti-inflammatory, analgesic, anti-depressant
Geraniol (GE) is a naturally occurring terpene found in various plants, including cannabis. It is responsible for the distinctive floral and fruity aroma often associated with cannabis strains. Geraniol is widely used in the fragrance and flavor industries due to its pleasant scent and taste. In addition to its aromatic properties, Geraniol has gained attention for its potential health benefits. It possesses antioxidant and anti-inflammatory properties, which are valuable for protecting cells from oxidative stress and reducing inflammation in the body. Geraniol has also been studied for its potential anti-cancer effects, as it may inhibit the growth of cancer cells and promote their apoptosis (cell death). While it is present in cannabis, Geraniol is just one of many terpenes found in the plant, and its specific effects in conjunction with other compounds like cannabinoids are an area of ongoing research. Its diverse applications and potential therapeutic properties make Geraniol an interesting component of cannabis and other plants with promising health-related implications.
The article titled “The Inhibitory Efficiencies of Geraniol as an Anti-Inflammatory, Antioxidant, and Antibacterial, Natural Agent Against Methicillin-Resistant Staphylococcus aureus Infection in vivo,” published in Infection and Drug Resistance in 2021, explores the anti-inflammatory properties of geraniol. This study investigates how geraniol, a natural compound, functions as an anti-inflammatory agent. The research suggests that geraniol exhibits inhibitory effects against inflammation, highlighting its potential as an anti-inflammatory agent. This finding is significant because inflammation is a crucial component of various health conditions, and natural compounds like geraniol may offer an alternative or complementary approach to managing inflammatory responses. Furthermore, the article titled “Geraniol targets KV1.3 ion channel and exhibits anti-inflammatory activity in vitro and in vivo,” published in Fitoterapia in 2019, also explores the anti-inflammatory properties of geraniol. This research investigates how geraniol interacts with the KV1.3 ion channel and its potential anti-inflammatory effects both in vitro and in vivo. The study suggests that geraniol can target the KV1.3 ion channel, which is involved in immune responses and inflammation. By modulating this channel, geraniol may exert anti-inflammatory activity, reducing inflammation-related processes. These findings point to geraniol as a promising natural compound with potential anti-inflammatory benefits, though further research, including clinical studies in humans, would be necessary to fully understand its practical applications in managing inflammatory conditions. Nonetheless, this study provides valuable insights into the anti-inflammatory properties of geraniol.
The article titled “Cymbopogon martinii essential oil and geraniol at noncytotoxic concentrations exerted immunomodulatory/anti-inflammatory effects in human monocytes,” published in the Journal of Pharmacy and Pharmacology in 2014, investigates geraniol’s immunomodulatory and anti-inflammatory effects. The study explores how geraniol, a natural compound found in certain essential oils, impacts human monocytes, a type of immune cell. The findings indicate that geraniol, when used at noncytotoxic concentrations, exerts immunomodulatory and anti-inflammatory effects on these immune cells. This suggests that geraniol may regulate the immune response and potentially reduce inflammation, which could have implications for various immune-related conditions. However, while this research offers valuable insights into the immunomodulatory properties of geraniol, further studies, including clinical trials, are necessary to determine the full extent of its practical applications in immunotherapy and anti-inflammatory treatments. Nonetheless, the study underscores the potential of geraniol as a natural immunomodulatory agent.
The article titled “Research progress of Geraniol in tumor therapy,” published in the Proceedings of Anticancer Research, delves into the potential anti-tumor properties of geraniol. This research investigates the progress and advancements in utilizing geraniol for cancer therapy. Geraniol has garnered attention for its possible anti-cancer effects, and this study contributes to the growing body of research in this area. While the specific mechanisms by which geraniol may combat tumor growth are multifaceted and still under investigation, some studies have indicated that geraniol can interfere with various cellular processes, including apoptosis (programmed cell death) and cell cycle regulation, potentially leading to the suppression of cancer cell proliferation. Studies have also alluded to geraniol enhancing the effects of chemotherapy. For example, The article titled “Geraniol, a component of plant essential oils, modulates DNA synthesis and potentiates 5-fluorouracil efficacy on human colon tumor xenografts,” published in Cancer Letters in 2004, explores the anti-tumor properties of geraniol. This study investigates how geraniol, a natural component of plant essential oils, affects DNA synthesis and enhances the efficacy of the chemotherapy drug 5-fluorouracil in human colon tumor xenografts. The findings suggest that geraniol can modulate DNA synthesis, and when combined with 5-fluorouracil, it demonstrates the potential to enhance the drug’s effectiveness against colon tumors. This indicates that geraniol may possess anti-tumor properties that can synergize with conventional chemotherapy, potentially improving treatment outcomes for cancer patients. However, it’s crucial to acknowledge that while these results are promising, further research, including clinical trials, is essential to validate geraniol’s practical applications in cancer treatment. Nonetheless, this study provides valuable insights into the potential anti-tumor effects of geraniol.
The article titled “Geraniol Induces Antinociceptive Effect in Mice Evaluated in Behavioural and Electrophysiological Models,” published in Basic & Clinical Pharmacology & Toxicology in 2017, examines the analgesic properties of geraniol. This research investigates how geraniol, a naturally occurring compound, exerts an antinociceptive effect, meaning it reduces pain perception in mice. The study employs both behavioral and electrophysiological models to evaluate the analgesic effects of geraniol. The findings suggest that geraniol induces an antinociceptive effect, providing evidence of its potential as an analgesic agent. This is a significant discovery as it suggests that geraniol may have the capacity to modulate pain perception pathways, offering a potential avenue for the development of pain-relief strategies. However, it’s essential to emphasize that further research, including clinical studies in humans, is necessary to determine the practical applications of geraniol as an analgesic and to establish its safety and efficacy in pain management. Nevertheless, this study offers valuable insights into the potential analgesic properties of geraniol.
The article titled “Geraniol produces antidepressant-like effects in a chronic unpredictable mild stress mice model,” published in Physiology & Behavior in 2015, investigates the potential antidepressant effects of geraniol. This study focuses on how geraniol, a natural compound, impacts depression-like behaviors in a chronic unpredictable mild stress mice model. The research findings suggest that geraniol produces antidepressant-like effects in the mice, indicating its potential in alleviating depressive symptoms. This is particularly significant because depression is a complex mental health disorder, and alternative approaches to conventional antidepressant medications are of interest. However, on the other side of the spectrum, the article titled “Depressant effect of geraniol on the central nervous system of rats: Behavior and ECoG power spectra,” published in the Biomedical Journal in 2018, explores the potential antidepressant effects of geraniol. This study investigates how geraniol affects the central nervous system and behavioral responses in rats. Surprisingly, the research suggests that geraniol has a depressant effect on the central nervous system rather than exhibiting antidepressant properties. This unexpected finding contradicts the assumption that geraniol may have mood-enhancing effects. However, it’s important to note that the study’s results are based on animal models, and the translation of these effects to humans may differ. Thus, while this research provides valuable insights into the physiological effects of geraniol, it underscores the complexity of terpenes’ actions on the central nervous system and highlights the need for further investigation to fully understand their potential in managing depression and related conditions.
Testimonial
Anti-cancer, anti-bacterial, anti-inflammatory
Pinene (PI) is a naturally occurring terpene found in cannabis and numerous other plants, including pine trees, rosemary, and sage. It is one of the most common terpenes in the plant kingdom and contributes to the characteristic aroma of pine forests and certain cannabis strains. Pinene is further categorized into two isomers: alpha-pinene (α-pinene) and beta-pinene (β-pinene), both of which can be found in various cannabis varieties. In cannabis, pinene is believed to play a role in the entourage effect, where the combination of different cannabinoids and terpenes synergistically influences the overall therapeutic and psychoactive effects of the plant. Pinene is known for its potential health benefits, including anti-inflammatory, bronchodilator, and anxiolytic properties. Additionally, some research suggests that pinene may counteract some of the memory impairment associated with THC, potentially making it relevant to the management of the cognitive effects of cannabis use. Overall, pinene’s presence in cannabis adds complexity to the plant’s chemical profile and may contribute to its diverse range of effects and potential medical applications.
The article titled “Alpha-pinene exhibits anti-inflammatory activity through the suppression of MAPKs and the NF-ΚB pathway in mouse peritoneal macrophages,” published in The American Journal of Chinese Medicine in 2015, investigates the anti-inflammatory effects of alpha-pinene. The study focuses on its impact on mouse peritoneal macrophages and finds that alpha-pinene demonstrates anti-inflammatory activity by suppressing the MAPKs (Mitogen-Activated Protein Kinases) and the NF-ΚB (Nuclear Factor Kappa B) pathway. These molecular pathways are known to play critical roles in regulating the body’s inflammatory responses. The results suggest that alpha-pinene may have potential as an anti-inflammatory agent by modulating these key signaling pathways in immune cells. While the study provides valuable insights into alpha-pinene’s anti-inflammatory properties, further research is needed to explore its potential applications in clinical settings and to confirm its safety and efficacy for human use.
The article titled “α-Pinene Enhances the Anticancer Activity of Natural Killer Cells via ERK/AKT Pathway,” published in the International Journal of Molecular Sciences in 2021, explores the potential anti-cancer effects of alpha-pinene. The study investigates how alpha-pinene, a major component of pinene, influences the activity of natural killer (NK) cells, which are part of the immune system’s defense against cancer. The research findings suggest that alpha-pinene can enhance the anticancer activity of NK cells by modulating the ERK/AKT pathway. This implies that alpha-pinene may have a role in strengthening the body’s immune response against cancer cells. Similarly, The study, “α-Pinene Induces Apoptotic Cell Death via Caspase Activation in Human Ovarian Cancer Cells,” published in Medical Science Monitor in 2019, investigates how alpha-pinene induces apoptotic cell death in human ovarian cancer cells through caspase activation. The findings suggest that alpha-pinene may have anti-cancer properties by triggering programmed cell death in cancer cells. Caspase activation is a key process in apoptosis, indicating that alpha-pinene could potentially influence the regulation of cell survival and death pathways. Also, the study titled “α-Pinene inhibits human prostate cancer growth in a mouse xenograft model,” published in 2017, explores the potential anti-cancer effects of alpha-pinene. In this research, alpha-pinene was examined for its ability to inhibit the growth of prostate cancer cells in a mouse xenograft model. The findings of this study suggest that alpha-pinene possesses promising anti-cancer properties, specifically in the context of prostate cancer. It appears to inhibit tumor growth, indicating its potential as a therapeutic agent for prostate cancer treatment. However, it’s important to note that while these results are encouraging, further research, including clinical trials in humans, would be necessary to validate the effectiveness and safety of alpha-pinene as a cancer treatment. Nevertheless, this study provides valuable insights into the potential anti-cancer properties of alpha-pinene, offering a promising avenue for future research in the field of oncology.
The article titled “Antibacterial Activity and Time-kill Kinetics of Positive Enantiomer of α-pinene Against Strains of Staphylococcus aureus and Escherichia coli,” published in Current Topics in Medicinal Chemistry in 2018, investigates the antibacterial effects of the positive enantiomer of alpha-pinene. The study explores the antimicrobial properties of alpha-pinene against bacterial strains, specifically Staphylococcus aureus and Escherichia coli. The findings indicate that the positive enantiomer of alpha-pinene exhibits antibacterial activity against these bacterial strains. Moreover, the time-kill kinetics suggest that alpha-pinene’s antibacterial effects may involve a gradual reduction in bacterial viability over time. These results highlight the potential of alpha-pinene as a natural antibacterial agent, which could have implications for the development of novel antimicrobial treatments. However, further research, including clinical trials, would be necessary to fully understand its practical applications in clinical settings.
Testimonial
Antidepressant, anxiolytic, anti-inflammatory
Limonene (LI) is a prominent terpene found in cannabis, contributing to its aromatic profile and potential health benefits. It is a cyclic monoterpene that is widely distributed in various plants, including citrus fruits, as well as in some cannabis strains. Limonene is known for its distinct citrus aroma, which gives some cannabis varieties a lemony or orange-like scent. Beyond its aromatic qualities, limonene has gained attention for its potential therapeutic properties. It is considered to have anti-inflammatory, antioxidant, and mood-enhancing effects, making it a compound of interest in both the cannabis industry and natural medicine. Some studies suggest that limonene may play a role in modulating the effects of cannabinoids like THC and CBD, potentially influencing the overall experience and therapeutic potential of cannabis strains. However, further research is needed to fully understand its mechanisms of action and its specific applications in cannabis-based products and therapies.
The study titled “Oral administration of d-Limonene controls inflammation in rat colitis and displays anti-inflammatory properties as diet supplementation in humans,” published in the journal Life Sciences in 2013, provides insights into the potential anti-inflammatory effects of limonene. The research examines the impact of oral d-limonene administration on colitis-induced inflammation in rats and also evaluates limonene’s anti-inflammatory properties as a dietary supplement in humans. The findings suggest that limonene exhibits significant anti-inflammatory activity, reducing inflammation in rat colitis models and potentially serving as a dietary component with anti-inflammatory benefits for humans. This suggests that limonene may have therapeutic potential in managing inflammatory conditions, though further clinical studies in human subjects would be necessary to fully comprehend the extent of its anti-inflammatory effects and its practical applications in medical and dietary contexts.
The article titled “Neuroprotective Potential of Limonene and Limonene Containing Natural Products,” published in the journal Molecules in July 2021, explores the potential neuroprotective properties of limonene and natural products containing limonene. The study delves into the effects of limonene on various aspects of neural health, including its ability to mitigate depressive behavior, regulate the hypothalamic-pituitary-adrenal axis, and maintain the levels of monoamine neurotransmitters. Additionally, the research investigates the impact of limonene on the expression of brain-derived neurotrophic factor and its receptor in the hippocampus. The findings suggest that limonene may have therapeutic potential in safeguarding neuroendocrine, neurotrophic, and monoaminergic systems. Furthermore, inhaling limonene notably reversed depressive symptoms, reduced excessive activity in the hypothalamic–pituitary–adrenal axis, and countered the decline in levels of monoamine neurotransmitters. It also led to a decrease in the expression of brain-derived neurotrophic factor and its receptor in the hippocampus. Consequently, the research suggests that limonene’s antidepressant effects are associated with enhancements in the neuroendocrine, neurotrophic, and monoaminergic systems (Zhang 2019).
The article titled “Limonene has anxiolytic activity via adenosine A2A receptor-mediated regulation of dopaminergic and GABAergic neuronal function in the striatum,” published in Phytomedicine in 2021, presents a study that investigates the anxiolytic effects of limonene and the underlying mechanisms. According to the research, limonene appears to have anxiolytic properties by modulating the adenosine A2A receptor, which in turn regulates the function of dopaminergic and GABAergic neurons in the striatum. This finding suggests that limonene may contribute to anxiety relief through its influence on neurotransmitter systems involved in mood regulation. Similarly, in the article titled “Effects of Limonene on Chronic Restraint Stress-Induced Memory Impairment and Anxiety in Male Rats,” published in Neurophysiology in 2019, the authors investigate the potential anxiolytic effects of limonene. The results of the study suggest that limonene may have a positive impact in mitigating both memory deficits and anxiety-related behaviors in stressed rats. This indicates that limonene may possess anxiolytic properties, potentially attributed to its ability to modulate neurotransmitter levels, reduce oxidative stress, or influence neuroinflammatory pathways. While these findings are promising, it’s important to note that further research, including clinical studies in humans, would be necessary to validate and fully understand the extent of limonene’s potential in managing anxiety.
Testimonial
Antibacterial, anti-inflammatory, analgesic
Sabinene (SA) is a naturally occurring terpene commonly found in various plants, including cannabis. Terpenes like sabinene contribute to the distinct aroma and flavor profiles of different cannabis strains, playing a significant role in the overall sensory experience of consuming cannabis. In addition to its olfactory attributes, sabinene has gained attention for its potential health benefits. It is known for its anti-inflammatory and antioxidant properties, making it a subject of interest in the field of natural medicine. While sabinene is present in relatively lower concentrations compared to cannabinoids like THC and CBD, it is believed to have a synergistic effect with other cannabis compounds, enhancing the overall therapeutic potential of the plant. Although more research is needed to fully understand the specific pharmacological effects and potential medical applications of sabinene from cannabis, it exemplifies the complexity and diversity of chemical constituents present in this versatile plant.
In the study conducted by Valente et al. (2013) and published in Food and Chemical Toxicology, the anti-inflammatory properties of Sabinene, a constituent of Oenanthe crocata L. essential oil, are investigated along with its antifungal and antioxidant activities. The researchers explore the potential health benefits of the essential oil derived from Oenanthe crocata L., a plant commonly used in traditional medicine, to understand its pharmacological effects. The study finds that the essential oil, rich in Sabinene, exhibits significant anti-inflammatory effects, which could be attributed to its ability to modulate immune responses and attenuate inflammatory pathways. The anti-inflammatory potential of Sabinene is of particular interest, as it may have implications for the development of natural therapeutic agents for conditions characterized by excessive inflammation. This research provides valuable insights into the multifaceted pharmacological properties of Sabinene and contributes to our understanding of its potential applications in the management of inflammatory disorders.
The research by Arunkumar et al. (2014) published in the Tropical Botanic Garden and Research Institute investigates the essential oil constituents of Zornia diphylla (L.) Pers and evaluates their potential anti-inflammatory and antimicrobial activities. In this study, the anti-bacterial properties of Sabinene, one of the essential oil constituents of Zornia diphylla, are explored. The results suggest that the essential oil exhibits antimicrobial effects, including anti-bacterial activity. Sabinene, as a significant component of the essential oil, likely contributes to these antimicrobial properties. The study’s findings underscore the potential of Sabinene as a natural compound with anti-bacterial potential, which could have implications for the development of novel antibacterial agents. This research expands our understanding of Sabinene’s pharmacological attributes and its possible application in combating bacterial infections.
The study conducted by Babalwa Tembeni and colleagues (2019) and published in the Journal of Essential Oil Bearing Plants investigates the anti-inflammatory and analgesic activities of two essential oils from Pelargonium inquinans Ait, including both wild and cultivated varieties. The research explores the potential analgesic properties of Sabinene, a constituent present in these essential oils. The study’s results suggest that the essential oils possess analgesic activity, which could be attributed to the presence of compounds like Sabinene. This finding implies that Sabinene might contribute to the observed analgesic effects of the essential oils derived from Pelargonium inquinans. The research contributes to our understanding of Sabinene’s potential pharmacological attributes and supports the exploration of natural compounds for pain management. This study highlights the significance of Sabinene in the context of analgesic properties, which could have implications for the development of alternative pain-relief approaches.
Testimonial
Muscle Relaxant, Asthma: Bronchodilator/Anti-Inflammatory
Linalool (LN) is a naturally occurring terpene that is found not only in cannabis but also in various other plants, including lavender, basil, and mint. It is known for its pleasant floral aroma and is a common component of essential oils used in aromatherapy and perfumery. In cannabis, linalool contributes to the plant’s complex chemical profile and is responsible for some of the strain-specific scents and flavors. Beyond its aromatic properties, linalool has gained attention for its potential health benefits. It is believed to possess anti-inflammatory, analgesic, and anxiolytic properties, which make it of interest in both traditional and alternative medicine. Some research also suggests that linalool may have neuroprotective effects and could play a role in modulating the effects of cannabinoids, such as THC and CBD, found in cannabis. However, while linalool shows promise in various therapeutic applications, further research is needed to fully understand its mechanisms of action and its potential as a standalone or complementary treatment in various medical conditions.
The article titled “Inhaled linalool-induced sedation in mice,” published in Phytomedicine in 2009, explores the sedative effects of linalool. The study investigates how inhaled linalool induces sedation in mice, shedding light on its potential as a sedative agent. The findings indicate that linalool inhalation does indeed lead to sedation in mice, suggesting that this terpene has a calming and soothing effect on the central nervous system. Linalool’s sedative properties may be attributed to its ability to modulate neurotransmitter levels or interact with specific receptors in the brain, ultimately promoting relaxation and sedation. While this study is conducted in mice, it offers valuable insights into the potential sedative effects of linalool, which could have implications for its use in natural remedies for anxiety, insomnia, and stress relief. Likewise, the article titled “Sedative effect on humans of inhalation of essential oil of linalool” published in Analytica Chimica Acta in 1998 investigates the sedative effects of linalool. The study examines the impact of inhaling the essential oil of linalool on human subjects. The findings of the research suggest that linalool inhalation has a sedative effect on humans. However, it’s essential to note that while this study provides valuable insights into linalool’s sedative effects, further research, including clinical trials and a broader range of participants, would be necessary to establish its efficacy and safety as a natural sedative remedy for a wider population. Nonetheless, this research underscores the potential therapeutic benefits of linalool in the context of promoting relaxation and reducing anxiety.
The article titled “Anxiolytic-like effects of inhaled linalool oxide in experimental mouse anxiety models,” published in Pharmacology Biochemistry and Behavior in 2011, investigates the anxiolytic (anxiety-reducing) effects of linalool oxide through inhalation. This study explores how linalool oxide, a derivative of linalool, affects anxiety in experimental mouse models. The findings suggest that inhaled linalool oxide exhibits anxiolytic-like effects, meaning it may have the potential to reduce anxiety-related behaviors in mice. This indicates that linalool oxide could influence neural pathways involved in anxiety regulation, making it a potential candidate for anxiety management. The article titled “Linalool Odor-Induced Anxiolytic Effects in Mice,” published in Frontiers in Behavioral Neuroscience in 2018, also found that inhalation of linalool odor influences and lowers anxiety levels in mice.While these results are promising, further research, including clinical studies in humans, would be necessary to ascertain the practical applications of linalool oxide in anxiety treatment. Nevertheless, these studies provide valuable insights into the potential anxiolytic properties of linalool and its derivatives.
The article titled “Neuroprotective effects of Coriandrum sativum and its constituent, linalool: A review,” published in Avicenna Journal of Phytomedicine in 2021, provides a comprehensive analysis of the neuroprotective properties of linalool. The review discusses the potential neuroprotective effects of linalool, which is a prominent constituent of Coriandrum sativum, commonly known as coriander. It highlights that linalool may offer protective benefits to the nervous system. While the exact mechanisms are still being studied, linalool is believed to possess antioxidant and anti-inflammatory properties, both of which are vital for safeguarding the brain against oxidative stress and neuroinflammation, two processes implicated in neurodegenerative diseases. This research underscores the potential of linalool as a natural compound with neuroprotective capabilities, which could have significant implications in the development of strategies for preventing or managing neurological disorders. Moreover, the article titled “L-linalool exerts a neuroprotective action on hemiparkinsonian rats,” published in Naunyn-Schmiedeberg’s Archives of Pharmacology in 2020, investigates the neuroprotective properties of linalool. The research focuses on how L-linalool, a common enantiomer of linalool found in several aromatic plants, exerts its neuroprotective effects in a hemiparkinsonian rat model. The study suggests that L-linalool exhibits neuroprotective actions by safeguarding against neurodegeneration, particularly in the context of Parkinson’s disease. It implies that L-linalool may modulate various cellular mechanisms to protect neurons from damage and deterioration, potentially making it a promising therapeutic agent for neurodegenerative conditions. However, it is crucial to emphasize that while these findings are encouraging, further research, including clinical trials in human subjects, is essential to validate the practical applications of L-linalool as a neuroprotective compound. Nonetheless, this study contributes valuable insights into the potential neuroprotective properties of linalool and its derivatives.
Testimonial
Sedative, brain stimulant, anti-fungal
Terpinolene (TE) is one of the many aromatic terpenes found in the cannabis plant, contributing to its distinct aroma and flavor profile. While terpinolene is present in several other plants, such as pine trees, sage, and some citrus fruits, its presence in cannabis adds complexity to the plant’s chemical composition. Terpinolene is known for its potential therapeutic properties, including its calming and sedative effects, which can contribute to the overall relaxing experience associated with certain cannabis strains. Additionally, terpinolene has shown antimicrobial and antioxidant properties, making it a subject of interest in the field of natural medicine. Its potential to modulate the endocannabinoid system and interact with other compounds found in cannabis, such as cannabinoids and other terpenes, underscores the complexity of the entourage effect, where various cannabis compounds may work together to produce unique physiological effects. Terpinolene’s multifaceted properties make it a valuable component in the diverse array of compounds found in the cannabis plant, with potential implications for both recreational and medicinal use.
The study by Ito and Ito (2013) published in the Journal of Natural Medicines investigates the sedative effects of inhaled terpinolene in mice and explores its structure-activity relationships. Through rigorous experimentation, the authors shed light on terpinolene’s potential as a sedative agent. By examining the chemical structure of terpinolene and its impact on sedation, the research offers insights into the molecular mechanisms that underlie its effects. The study underscores the significance of terpinolene as a naturally occurring compound with sedative properties, paving the way for further exploration of its therapeutic potential in managing sleep-related issues or anxiety. This article provides valuable insights into the pharmacological properties of terpinolene, contributing to the understanding of its role as a sedative agent and encouraging future research into its applications for promoting relaxation and tranquility.
In the study conducted by Kandhasamy Sowndhararajan et al. (2015) and published in the European Journal of Integrative Medicine, the brain stimulant properties of terpinolene are explored in the context of olfactory stimulation. Through investigations involving isomeric aroma compounds, specifically (+)-limonene and terpinolene, the researchers delve into their effects on human electroencephalographic (EEG) activity. This research sheds light on the potential brain stimulant properties of terpinolene, as indicated by its influence on EEG patterns. The study contributes to our understanding of terpinolene’s impact on brain activity, suggesting its role in promoting alertness and enhancing cognitive function. By investigating the neurophysiological response to olfactory stimulation with terpinolene, this article provides insights into the compound’s brain stimulant potential and opens avenues for further research into its applications for cognitive enhancement and neurological well-being.
The study conducted by Pinto et al. (2021) and published in the journal Letters in Applied Microbiology explores the antifungal properties of terpinolene in conjunction with dihydrojasmone against dermatophytes, a type of fungi that cause various skin infections. By investigating the potential synergistic effects of terpinolene and dihydrojasmone with the antifungal drug terbinafine, the researchers aim to enhance the efficacy of antifungal treatment. The study’s findings indicate that terpinolene, in combination with dihydrojasmone, can potentiate the antifungal activity of terbinafine against dermatophytes. Terpinolene has antifungal properties because it exerts antifungal activity by disrupting the integrity of the fungal cell membrane and inducing oxidative stress within the fungal cells. This suggests that terpinolene holds promise as a natural antifungal agent and could potentially be used to augment the effectiveness of existing antifungal treatments. The research contributes to our understanding of terpinolene’s potential in combating fungal infections, offering new insights into alternative and complementary approaches to managing dermatophyte-related conditions.
Testimonial
Anti-inflammatory, anti-tumor, anti-bacterial
Humulene (HU) is one of the many terpenes found in the cannabis plant, and it is also present in various other plants, such as hops, sage, and ginseng. This terpene contributes to the complex aroma and flavor profile of cannabis and is characterized by its earthy, woody, and subtly spicy notes. Beyond its role in scent and taste, humulene is known for its potential therapeutic properties. It has been studied for its anti-inflammatory, analgesic, and appetite-suppressing effects, which make it an intriguing compound in both traditional herbal medicine and modern research. Humulene’s anti-inflammatory properties, in particular, have garnered attention for their potential in the treatment of various inflammatory conditions. Moreover, humulene is often found in conjunction with other cannabinoids and terpenes in the cannabis plant, and this synergy, known as the “entourage effect,” suggests that it may enhance or modulate the effects of other cannabis compounds. While more research is needed to fully understand its mechanisms of action and potential medical applications, humulene remains an essential and fascinating component of the cannabis plant’s chemical profile.
The article titled “Preventive and therapeutic anti-inflammatory properties of the sesquiterpene α-humulene in experimental airways allergic inflammation,” published in the British Journal of Pharmacology in 2009, investigates the anti-inflammatory properties of α-humulene. This study focuses on how α-humulene, a sesquiterpene found in various plant sources, may prevent and alleviate airway inflammation in experimental models. The research suggests that α-humulene exhibits significant anti-inflammatory effects through its interaction with various molecular pathways involved in the inflammatory response. It can be used both as a preventive measure and as a therapeutic agent in allergic airway inflammation. These findings underscore α-humulene’s potential as an anti-inflammatory compound, possibly due to its ability to modulate inflammatory pathways and reduce the release of pro-inflammatory mediators. While the study’s results are promising, further research, including clinical trials in humans, would be necessary to fully understand the practical applications of α-humulene in managing inflammatory conditions. Nonetheless, this research provides valuable evidence supporting the anti-inflammatory properties of α-humulene, highlighting its potential as a natural therapeutic agent.
The article titled “Potentiating effect of beta-caryophyllene on anticancer activity of alpha-humulene, isocaryophyllene and paclitaxel,” published in the Journal of Pharmacy and Pharmacology in 2007, investigates the potential anti-cancer properties of humulene and its interaction with other compounds. The study explores how beta-caryophyllene, a terpene related to humulene, enhances the anticancer activity of alpha-humulene, isocaryophyllene, and paclitaxel. The findings suggest that beta-caryophyllene amplifies the anticancer effects of these compounds, highlighting the potential of humulene and its derivatives in cancer therapy. This study indicates that humulene, in combination with certain other compounds, may play a role in potentiating the response to anticancer treatments. Similarly, the article titled “α-Humulene inhibits hepatocellular carcinoma cell proliferation and induces apoptosis through the inhibition of Akt signaling,” published in Food and Chemical Toxicology in 2019, delves into the potential anti-cancer properties of humulene. This study investigates how alpha-humulene, a compound closely related to humulene, affects the proliferation and survival of hepatocellular carcinoma cells. The findings suggest that alpha-humulene can inhibit cancer cell growth and induce apoptosis, a process of programmed cell death, by targeting the Akt signaling pathway. Akt signaling is known to play a critical role in cell survival and proliferation, and its inhibition may lead to the suppression of cancer cell growth. While this research is promising, it’s important to note that these results are based on laboratory experiments, and further research, including pre-clinical and clinical studies, would be necessary to determine the practical applications of humulene in cancer treatment. Nonetheless, these studies provide valuable evidence to support the potential anti-cancer properties of humulene and its derivatives.
The article titled “Antibacterial and antibiofilm effects of α-humulene against Bacteroides fragilis,” published in the Canadian Journal of Microbiology in 2020, investigates the antibacterial properties of α-humulene. This study focuses on how α-humulene, a compound related to humulene, affects the bacterium Bacteroides fragilis. The research suggests that α-humulene demonstrates significant antibacterial activity against this pathogenic bacterium. Moreover, it appears to exhibit antibiofilm effects, indicating that α-humulene may disrupt the formation of bacterial biofilms—a critical factor in bacterial infections and antibiotic resistance. These findings underscore the potential of humulene and its derivatives as natural antibacterial agents and highlight their relevance in addressing bacterial infections and biofilm-related issues. Further research may unveil additional applications of humulene in combating microbial threats.
Testimonial
Anti-inflammatory, anti-cancer, antidepressant
Caryophyllene (CA), specifically referred to as beta-caryophyllene (BCP), is a prominent terpene found in cannabis and various other plants, including black pepper, cloves, and rosemary. What makes caryophyllene particularly interesting is its unique interaction with the endocannabinoid system (ECS). Unlike most terpenes, caryophyllene is considered a dietary cannabinoid because it acts as a selective agonist for the CB2 (cannabinoid receptor type 2) receptor in the ECS. This interaction suggests that caryophyllene may exert potential therapeutic effects, such as anti-inflammatory and analgesic properties, without directly binding to the CB1 receptors that are responsible for the psychotropic effects of THC. As a result, caryophyllene-rich cannabis strains are of growing interest in the field of medical cannabis, and research into its various health benefits is ongoing.
The article titled “Analgesic and anti-inflammatory activity of Caryophyllene oxide from Annona squamosa L. bark,” published in Phytomedicine in 2010, investigates the anti-inflammatory properties of Caryophyllene oxide. The study examines how Caryophyllene oxide, a derivative of caryophyllene found in the bark of Annona squamosa L., exerts its anti-inflammatory effects. The research suggests that Caryophyllene oxide demonstrates significant anti-inflammatory activity, potentially through its modulation of various pathways involved in the inflammatory response. This finding highlights caryophyllene’s potential as an anti-inflammatory agent, supporting its traditional use in herbal medicine for addressing inflammation-related conditions. The study “In vivo anti-inflammatory activity of β-caryophyllene, evaluated by molecular imaging” by Saad S. Dahham, et al. evaluated the anti-inflammatory properties on a carrageenan-induced rat hind paw edema model and found that after the peak of the swelling in the paws, an injection with beta-caryophyllene inhibited the development of edema. However, it’s essential to note that while these studies provide promising insights, further research, including clinical trials, is necessary to validate the practical applications of caryophyllene and its derivatives as anti-inflammatory compounds in clinical settings. Nonetheless, this research contributes valuable evidence to support caryophyllene’s anti-inflammatory properties.
In the article “β-caryophyllene and β-caryophyllene oxide—natural compounds of anticancer and analgesic properties” by Fidyt et al. (2016), the authors explore the anti-cancer properties of β-caryophyllene and β-caryophyllene oxide, two natural compounds found in various plant sources. The study sheds light on the potential therapeutic applications of these compounds in cancer treatment. The article highlights that β-caryophyllene and its oxide derivative possess promising anti-cancer properties, primarily through their ability to induce apoptosis (programmed cell death) in cancer cells, inhibit angiogenesis (the formation of new blood vessels that supply tumors), and modulate signaling pathways associated with cell proliferation and survival. These findings suggest that β-caryophyllene and β-caryophyllene oxide have the potential to be developed into novel anti-cancer therapies, offering a natural and potentially less toxic approach to combating cancer. However, further research and clinical studies are needed to fully explore their efficacy and safety in cancer treatment. Correspondingly, in the study conducted by Legault and Pichette in 2007, the authors investigated the potential anti-cancer properties of β-caryophyllene, a natural sesquiterpene found in various plant species, by examining its synergistic effects with other compounds such as α-humulene, isocaryophyllene, and paclitaxel. Their findings revealed that β-caryophyllene possesses significant anticancer activity, as it was able to enhance the anticancer effects of the aforementioned compounds. This suggests that β-caryophyllene may play a crucial role in augmenting the efficacy of cancer treatment strategies, potentially by enhancing the cytotoxicity of existing chemotherapeutic agents like paclitaxel. These results underscore the importance of exploring natural compounds like β-caryophyllene in the development of novel cancer therapies and combination treatments, offering promising avenues for future research in the field of oncology.
In the article titled “Caryophyllene, a CB2 receptor agonist produces multiple behavioral changes relevant to anxiety and depression in mice” by Amine Bahi et al. published in Physiology & Behavior in 2014, the authors explore the potential anxiolytic and antidepressant properties of Caryophyllene. Caryophyllene is investigated as a CB2 receptor agonist, which suggests its involvement in the endocannabinoid system. The study utilizes a mouse model to demonstrate that Caryophyllene induces various behavioral changes that are pertinent to anxiety and depression. This suggests that Caryophyllene may have a role in modulating these mood-related disorders. The activation of CB2 receptors by Caryophyllene is hypothesized to contribute to its anxiolytic and antidepressant effects. These findings provide valuable insights into the potential therapeutic applications of Caryophyllene in the management of anxiety and depression, though further research is needed to confirm its efficacy and safety in human subjects.
Reduces cholesterol, analgesic, antioxidant
Camphene (CM) is a naturally occurring monoterpene found in various cannabis strains, contributing to the plant’s complex chemical profile. It is one of several terpenes found in cannabis that are responsible for its distinct aroma and flavor. Camphene has a woody and earthy scent with hints of pine, and it is commonly associated with other aromatic terpenes like myrcene and pinene. While Camphene’s presence in cannabis is relatively low compared to more abundant terpenes like myrcene and limonene, it is believed to have potential therapeutic properties. Camphene has been studied for its anti-inflammatory, antioxidant, and analgesic effects, which could contribute to the overall medicinal properties of cannabis strains that contain this terpene. However, further research is needed to fully understand the specific therapeutic applications and mechanisms of Camphene in cannabis and other natural products.
The study titled “Camphene, a Plant-Derived Monoterpene, Reduces Plasma Cholesterol and Triglycerides in Hyperlipidemic Rats Independently of HMG-CoA Reductase Activity,” published in PLoS ONE in 2011, explores the potential cholesterol-reducing effects of camphene. The research investigates how camphene, a naturally occurring monoterpene derived from plants, affects plasma cholesterol and triglyceride levels in hyperlipidemic rats. Notably, the study suggests that camphene can lower cholesterol and triglycerides in these rats, and it operates independently of the HMG-CoA reductase enzyme activity, which is a target for many cholesterol-lowering medications. This suggests that camphene may have a unique mechanism of action in reducing cholesterol and triglyceride levels. However, it’s important to note that further research, including clinical studies in humans, would be necessary to validate these effects and explore the practical applications of camphene in managing hyperlipidemia and cardiovascular health. Nevertheless, this study provides valuable insights into the potential cholesterol-reducing properties of camphene.
The article titled “Plant-derived antioxidants – Geraniol and camphene protect rat alveolar macrophages against t-BHP induced oxidative stress,” published in Toxicology in Vitro in 2009, investigates the potential antioxidant effects of camphene. This study explores how camphene, a plant-derived compound, protects rat alveolar macrophages from oxidative stress induced by t-BHP (tert-Butyl hydroperoxide). The findings suggest that camphene, along with geraniol, another plant-derived antioxidant, offers protection against oxidative stress by effectively mitigating the damaging effects of t-BHP. Camphene’s antioxidant properties, as demonstrated in this study, indicate its potential to counteract oxidative stress, which is implicated in various diseases, including cancer and cardiovascular disorders. Meanwhile, the findings in “Antioxidant Properties of Camphene-Based Thiosemicarbazones: Experimental and Theoretical Evaluation,” suggest that camphene-based thiosemicarbazones exhibit antioxidant properties, which can be essential for neutralizing harmful free radicals and preventing oxidative stress-related damage in the body. However, while these results are promising, further research, including studies involving human subjects, would be needed to fully understand the extent of camphene’s antioxidant capabilities and its practical applications in preventing oxidative damage and related health conditions.
The article titled “Plant-derived antioxidants – Geraniol and camphene protect rat alveolar macrophages against t-BHP induced oxidative stress,” published in Toxicology in Vitro in 2009, investigates the potential antioxidant effects of camphene. This study explores how camphene, a plant-derived compound, protects rat alveolar macrophages from oxidative stress induced by t-BHP (tert-Butyl hydroperoxide). The findings suggest that camphene, along with geraniol, another plant-derived antioxidant, offers protection against oxidative stress by effectively mitigating the damaging effects of t-BHP. Camphene’s antioxidant properties, as demonstrated in this study, indicate its potential to counteract oxidative stress, which is implicated in various diseases, including cancer and cardiovascular disorders. Meanwhile, the findings in “Antioxidant Properties of Camphene-Based Thiosemicarbazones: Experimental and Theoretical Evaluation,” suggest that camphene-based thiosemicarbazones exhibit antioxidant properties, which can be essential for neutralizing harmful free radicals and preventing oxidative stress-related damage in the body. However, while these results are promising, further research, including studies involving human subjects, would be needed to fully understand the extent of camphene’s antioxidant capabilities and its practical applications in preventing oxidative damage and related health conditions.
Muscle Relaxant, Asthma: Bronchodilator/Anti-Inflammatory
Myrcene (MY) is a terpene commonly found in cannabis and
various other plants, including hops, thyme, and lemongrass. It is
one of the most abundant terpenes in many cannabis strains and contributes to the plant’s characteristic aroma. Myrcene is known for its potential therapeutic properties and its role in the entourage effect, where various cannabis compounds work together to produce enhanced therapeutic effects. Myrcene is believed to have a relaxing and sedative effect, and it may contribute to the “couch-lock” sensation often associated with indica-dominant
strains of cannabis. This terpene has also been studied for its
potential anti-inflammatory, analgesic, and muscle-relaxant
properties, which could be valuable in medical cannabis applications. Additionally, myrcene is thought to enhance the
absorption of cannabinoids like THC, which may influence the
overall psychoactive effects of cannabis. Its diverse range of
potential effects and interactions with other compounds in
cannabis make myrcene an intriguing component of the plant for both recreational and medicinal users.
The research conducted by Jansen et al. (2019) in the journal
Channels investigates the analgesic properties of myrcene, a
prominent terpene found in cannabis and various other plants. The study focuses on myrcene’s interaction with the transient receptor potential vanilloid 1 (TRPV1) channel, which plays a crucial role in pain perception. Through a detailed analysis, the authors demonstrate that myrcene has the potential to modulate TRPV1 channel activity, thus influencing pain sensation and perception. By shedding light on the mechanisms by which myrcene may
exert its analgesic effects, the study offers valuable insights into
its therapeutic potential in pain management. This research
highlights the intricate relationship between terpenes like myrcene
and the endocannabinoid system’s components, contributing to the understanding of the multifaceted effects of cannabis compounds on pain pathways. Further exploration of myrcene’s analgesic properties could potentially lead to the development of novel analgesic agents for various pain conditions.
The study conducted by Kim et al. (2020) and published in the International Journal of Molecular Sciences explores the therapeutic potential of volatile terpenes and terpenoids from forests for inflammatory diseases. In their investigation, the authors delve into the anti-inflammatory properties of various natural compounds, including myrcene, which is found in forest-derived terpenes. The article highlights myrcene as one of the bioactive compounds that possess anti-inflammatory effects and discusses its potential application in managing inflammatory diseases. For example, the research conducted by McDougall and McKenna (2022) and published in the International Journal of Molecular Sciences investigates the anti-inflammatory properties of the cannabis terpene myrcene using a rat adjuvant monoarthritis model. In their study, the authors delve into the potential of myrcene to alleviate inflammation and pain associated with arthritic conditions. Through their experiments, they demonstrate that myrcene exhibits both anti-inflammatory and analgesic effects in the rat adjuvant monoarthritis model. The study’s findings highlight myrcene’s potential as a natural therapeutic agent for managing inflammatory conditions and related pain. By revealing the anti-inflammatory mechanisms of myrcene, this research contributes to the growing body of evidence supporting the use of plant-derived compounds in the development of novel anti-inflammatory treatments.
In the study conducted by Wu et al. (2022) and published in the Tropical Journal of Pharmaceutical Research, the potential anti-tumor properties of myrcene are investigated in the context of oral cancer cells. The researchers explore the effects of myrcene on oral cancer cells in vitro and specifically focus on its role in inducing apoptosis, a programmed cell death mechanism that is crucial for controlling tumor growth. By evaluating the impact of myrcene on cell viability and apoptotic pathways, the study sheds light on the potential anti-tumor activity of myrcene in oral cancer. Similarly, the article by Tomko et al. (2020) published in Cancers explores the anti-cancer potential of various compounds present in cannabis, including cannabinoids, terpenes, and flavonoids, with a specific focus on myrcene. The authors delve into the accumulating evidence regarding myrcene’s anti-tumor properties, elucidating its potential mechanisms of action and therapeutic implications. By examining the interplay between myrcene and other compounds within the cannabis plant, the study highlights the synergistic effects that can contribute to the overall anti-cancer activity of cannabis extracts. The comprehensive approach of this research underscores the importance of considering the holistic composition of cannabis in assessing its therapeutic potential, especially in the context of cancer treatment. The article serves as a valuable resource for researchers and clinicians seeking a deeper understanding of how myrcene, in conjunction with other cannabis-derived components, may offer promising avenues for the development of innovative anti-cancer strategies.
Unknown potential, enhances CBD
CBDP, short for Cannabidiphorol, is a relatively lesser-known cannabinoid found in the cannabis plant. It is structurally similar to CBD (cannabidiol) but has a few distinct differences in its chemical composition. Like other cannabinoids, CBDP interacts with the endocannabinoid system in the human body, though its specific effects and properties are still the subject of ongoing research. Due to its limited presence in most cannabis strains and the nascent state of research, there is currently limited information available regarding CBDP’s potential therapeutic benefits or psychoactive properties. As scientists delve deeper into the intricate world of cannabinoids, CBDP is emerging as a potential candidate for further exploration and study, offering the possibility of new insights into the diverse range of compounds found within the cannabis plant.
The study conducted by Golliher et al. (2021) explores the potential of (+)-carvone to enhance the properties of (+)-ent-cannabidiol (CBD) and its derivatives. This investigation represents an innovative approach to modifying the structure of CBD and its analogs for potential therapeutic applications. The researchers focus on the synthesis of novel derivatives, specifically (+)-ent-CBDP and (+)-ent-CBDV, through an asymmetric synthesis pathway. The study reveals the ability of (+)-carvone to serve as a versatile starting material, enabling the access to unique cannabinoid derivatives. The findings highlight the potential of (+)-carvone as a tool for enhancing the chemical diversity and pharmacological properties of CBD and its analogs. This innovative approach opens up new avenues for exploring the structural modifications of CBD compounds to potentially enhance their therapeutic effects or develop new cannabinoid-based treatments.
Testimonial
Neurodegenerative disease, analgesic
Tetrahydrocannabiphorol, often abbreviated as THCP, is a relatively newly discovered cannabinoid found in the cannabis plant. It was first identified and isolated in research conducted in 2020, and its properties and effects are still being studied. THCP is structurally similar to delta-9-tetrahydrocannabinol (THC), the primary psychoactive compound in cannabis, but it is believed to be significantly more potent. Initial research suggests that THCP may have a much stronger binding affinity to the CB1 receptor in the endocannabinoid system, potentially leading to more pronounced psychoactive effects. However, due to its recent discovery, there is still much to learn about THCP, including its potential medical benefits, safety profile, and prevalence in various cannabis strains. This cannabinoid has generated considerable interest within the scientific community, and ongoing research aims to unravel its unique properties and therapeutic potential further.
The research conducted by Citti et al. (2019) unveils a novel phytocannabinoid, Δ9-tetrahydrocannabiphorol (THCP), isolated from Cannabis sativa L., and investigates its analgesic potential. This study presents a remarkable finding, demonstrating that THCP possesses an in vivo cannabimimetic activity surpassing that of Δ9-tetrahydrocannabinol (THC), a well-known psychoactive cannabinoid. The analysis sheds light on THCP’s interaction with the endocannabinoid system and its potential as an analgesic agent. By comparing its effects to those of THC, the study suggests that THCP could hold substantial promise as an analgesic compound, potentially exceeding the effects of THC itself. The research paves the way for a deeper exploration of THCP’s molecular mechanisms and its potential application in pain management strategies. This novel insight into the analgesic effects of THCP contributes to a broader understanding of the diverse pharmacological actions of cannabinoids and their potential therapeutic applications in pain relief.
Testimonial
Unknown potential, metabolite of CBD
Cannabielsoin (CBE) is a lesser-known cannabinoid found in the cannabis plant. While much of the research on cannabis has focused on cannabinoids like THC (tetrahydrocannabinol) and CBD (cannabidiol), CBE is one of the many minor cannabinoids present in the plant, contributing to its complex chemical profile. It is structurally related to both THC and CBD, and like other cannabinoids, it interacts with the endocannabinoid system in the human body. However, the specific effects and potential medical applications of CBE are not as well-studied or understood as those of THC and CBD. As research into the cannabis plant continues to expand, there is growing interest in exploring the properties and therapeutic potential of minor cannabinoids like CBE, but more research is needed to fully elucidate its effects and applications in health and medicine.
The article “Cannabielsoin as a new metabolite of cannabidiol in mammals,” authored by Ikuo Yamamoto and colleagues and published in Pharmacology Biochemistry and Behavior in 1991, presents a pivotal discovery regarding the metabolic transformation of cannabidiol (CBD) in mammals. The study focuses on the identification and characterization of a previously unknown metabolite, cannabielsoin (CBE), which is formed through enzymatic processes in mammalian organisms. Through comprehensive experimental approaches and analytical techniques, the researchers elucidate the metabolic pathway that leads to the formation of CBE from CBD. This discovery sheds light on the complexity of CBD metabolism and adds to the growing understanding of how CBD interacts with physiological processes within the body. The identification of CBE as a metabolite of CBD not only expands our knowledge of the compounds derived from CBD but also highlights the intricate interactions between cannabinoids and the endocannabinoid system in mammals. This seminal work contributes significantly to the field of cannabinoid research and has implications for the development of cannabinoid-based therapies and drug metabolism studies. Continuing their research, Yamamoto and colleagues wrote the article “Identification of Cannabielsoin, a new metabolite of cannabidiol formed by Guinea-pig hepatic microsomal enzymes, and its pharmacological activity in mice,” in 2008. This study delves into the enzymatic processes occurring within the guinea pig hepatic microsomal enzymes that lead to the formation of CBE. Furthermore, the researchers explore the pharmacological effects of CBE in mice, shedding light on its potential impact on the mammalian system. By investigating the pharmacological activity of CBE, this research opens new avenues for exploring the physiological effects and potential therapeutic applications of this metabolite. The study’s comprehensive approach to both the formation and potential functions of CBE contributes valuable insights to the intricate landscape of CBD metabolism and its potential implications for physiological responses in mammals.
Testimonial
Unknown potential
CBGL, or Cannabigerol Lactone, is not a well-known or researched cannabinoid. It is because of this that there is limited information about its medicinal properties. However, scientists have realized that it is structurally similar to the well-known cannabinoid CBG, meaning that they might share some of the same health benefits. CBG has shown potential anti-inflammatory, neuroprotective, and analgesic properties, and has also shown antibacterial activity, appetite stimulating effects, and possible relief for those suffering with glaucoma. While there is still little research about CBGL right now, its important to note that research in the field of cannabinoids is continuously evolving and there may be many benefits to CBGL in the future.
Testimonial
Anti-inflammatory, glioblastoma (brain cancer), dry skin, anti-hypertensive, analgesic, neuroprotective
Cannabigerol, commonly known as CBG, is one of the many chemical compounds found in the cannabis plant. It is categorized as a cannabinoid and is considered a minor constituent of the plant, typically present in lower concentrations compared to more well-known cannabinoids like THC and CBD. CBG is often referred to as the “mother cannabinoid” because it serves as a precursor to other cannabinoids during the cannabis plant’s growth cycle. While research on CBG is still in its early stages, it has garnered significant attention for its potential therapeutic properties. Some preliminary studies suggest that CBG may have anti-inflammatory, neuroprotective, and analgesic effects, making it a subject of interest for various medical applications. As the understanding of cannabinoids continues to evolve, CBG’s unique properties and potential benefits are becoming increasingly prominent in the field of cannabis research and product development.
The study conducted by Gugliandolo et al. (2018) offers a valuable analysis of the neuro-protective potential of cannabigerol (CBG) using an in vitro model of neuroinflammation. Through comprehensive experimentation, the study reveals that CBG demonstrates neuro-protective effects by attenuating the release of pro-inflammatory cytokines and oxidative stress markers. These findings suggest that CBG has the ability to modulate neuroinflammation, a process implicated in various neurological disorders. The research conducted by di Giacomo et al. (2020) used both cellular and isolated tissue models. This study delves into the effects of CBG on rat Hypo-E22 cells and isolated hypothalamus, shedding light on CBG’s ability to exert neuroprotective and neuromodulator effects. Through a thorough experimental approach, the research demonstrates that CBG treatment results in the attenuation of oxidative stress markers and the preservation of mitochondrial function in the Hypo-E22 cell line. Moreover, CBG exhibits a neuro-modulatory impact on isolated hypothalamus tissue by influencing neurotransmitter release. In the case of Huntington’s disease (HD), the study by Díaz-Alonso et al. (2016) offers a significant contribution to understanding the neuro-protective potential of cannabigerol (CBG). This research investigates the effects of VCE-003.2, a novel derivative of CBG, on neuronal progenitor cell survival and symptomatology in murine models of HD. The study reveals that VCE-003.2 treatment leads to enhanced survival of neuronal progenitor cells, a crucial factor in maintaining neuronal health and plasticity. Additionally, the research demonstrates that VCE-003.2 administration alleviates the motor deficits characteristic of HD in murine models. By elucidating CBG’s ability to enhance cell survival and ameliorate symptoms in an HD context, this study offers valuable insights into CBG’s neuro-protective effects and its potential as a therapeutic candidate for neurodegenerative disorders.
The research conducted by Burgaz et al. (2019) provides valuable insights into the anti-inflammatory effects of cannabigerol (CBG), particularly in the context of experimental Parkinson’s disease (PD). This study focuses on the development of an oral treatment with VCE-003.2, a CBG derivative with peroxisome proliferator-activated receptor-gamma (PPAR-γ) activity, against inflammation-driven neuronal deterioration in a PD model. The findings reveal that VCE-003.2 treatment effectively mitigates inflammation-induced neuronal damage in experimental PD, highlighting its potential as an anti-inflammatory agent. The study demonstrates that the PPAR-γ-activating properties of CBG contribute to its ability to modulate inflammatory responses and protect against neuronal deterioration. Meanwhile, the study conducted by Borrelli et al. (2013) offers compelling insights into the anti-inflammatory effects of cannabigerol (CBG) in the context of experimental inflammatory bowel disease (IBD). This research focuses on the potential therapeutic benefits of CBG, a non-psychotropic cannabinoid, in ameliorating IBD-related inflammation. The findings demonstrate that CBG treatment leads to a reduction in inflammatory responses and damage in a preclinical model of IBD. These results suggest that CBG holds promise as a beneficial therapeutic agent for managing inflammatory conditions like IBD.
The research conducted by Kogan et al. (2021) provides valuable insights into the analgesic effects of cannabigerol (CBG) and its derivatives in the context of reducing inflammation and pain. This study explores the potential of novel CBG derivatives to alleviate pain and inflammation, with a focus on their therapeutic applications. The findings reveal that these CBG derivatives possess anti-inflammatory properties that are associated with reductions in pain and obesity-related symptoms. The study sheds light on the mechanisms by which CBG and its derivatives exert their analgesic effects, offering a potential avenue for the development of novel pain-relieving interventions.
The comprehensive review by Citti et al. (2018) critically examines the pharmaceutical and biomedical aspects of cannabinoids, shedding light on their potential analgesic effects. The analysis delves into the pharmacological properties of various cannabinoids, including cannabigerol (CBG), and their implications for pain management. The review synthesizes existing literature to elucidate the analgesic potential of CBG, considering its interactions with the endocannabinoid system and other molecular targets involved in pain modulation. By critically evaluating the available research, this review contributes to a deeper understanding of CBG’s mechanisms of action, its potential as an analgesic agent, and the challenges and opportunities in utilizing cannabinoids for pain relief. The comprehensive assessment of CBG’s analgesic effects presented in this study underscores the importance of further research in harnessing the therapeutic potential of cannabinoids, including CBG, in pain management strategies.
Testimonial
Autism, anxiety, seizures, anti-inflammatory, cancer/anti-tumor growth. insomnia, neuroprotective, antidepressant, analgesic.
Cannabidiol, commonly referred to as CBD, is one of the most well-known and extensively studied phytocannabinoids found in the cannabis plant. Unlike its more infamous counterpart, delta-9-tetrahydrocannabinol (THC), CBD does not induce the characteristic psychoactive effects associated with cannabis use. CBD has garnered significant attention due to its potential therapeutic properties and wide range of potential applications. It interacts with the endocannabinoid system (ECS), a complex network of receptors and signaling molecules in the human body that regulates various physiological processes, including pain perception, inflammation, mood, appetite, and immune responses. CBD’s non-intoxicating nature, coupled with its diverse potential benefits, has driven a surge of interest from both researchers and the public. It has been investigated for its potential to alleviate anxiety, manage pain, reduce inflammation, and even treat certain forms of epilepsy, leading to the approval of a CBD-based pharmaceutical for treating epilepsy in some countries. However, while CBD’s therapeutic potential is widely acknowledged, further research is needed to fully understand its mechanisms of action, optimal dosages, and potential interactions with other medications, ensuring its safe and effective use across a spectrum of health conditions.
The study conducted by Pretzsch et al. (2019) offers valuable insights into the potential benefits of using cannabidiol (CBD) for individuals with autism spectrum disorder (ASD). The research focuses on the effects of CBD on brain activity and functional connectivity, aiming to shed light on its potential therapeutic role in addressing some of the core symptoms associated with ASD. The findings suggest that CBD may have a positive impact on brain function, particularly by modulating low-frequency brain activity and enhancing functional connectivity. These alterations in brain activity could potentially translate into improvements in social interactions, communication skills, and repetitive behaviors commonly observed in individuals with ASD. The study paves the way for further exploration of CBD as a potential adjunctive treatment for ASD, potentially offering a novel avenue for addressing the complex challenges faced by individuals with this condition. However, due to the complex and heterogeneous nature of autism, the efficacy of CBD may vary among individuals.
As one of the most well-known cannabinoids, CBD is well studied in its anti-inflammatory effects. The article by Nagarkatti et al. (2009) delves into the intricate mechanisms underlying CBD’s anti-inflammatory effects, which involve interactions with the endocannabinoid system, modulation of immune cell function, and regulation of cytokine production. Through its action on both CB1 and CB2 receptors, as well as non-cannabinoid receptor pathways, CBD appears to exhibit a complex and multifaceted impact on inflammatory processes. The article highlights preclinical and clinical evidence supporting CBD’s ability to attenuate inflammation in various disease models, including autoimmune disorders, neuroinflammatory conditions, and gastrointestinal inflammation. There are also studies about other types of inflammation CBD targets. For example, the article “Cannabidiol as an emergent therapeutic strategy for lessening the impact of inflammation on oxidative stress” offers a comprehensive analysis of CBD’s potential as an emerging therapeutic strategy for mitigating inflammation-induced oxidative stress. The article delves into the intricate interplay between inflammation and oxidative stress, emphasizing the detrimental effects of chronic inflammation on cellular health. The author highlights CBD’s multifaceted mechanisms of action, which encompass both direct and indirect interactions with various cellular pathways involved in inflammation and oxidative stress. By modulating the activity of immune cells, suppressing cytokine production, and enhancing antioxidant defenses, CBD demonstrates its potential to counteract inflammation-driven oxidative damage. The study conducted by Rajesh et al. (2007) provides an insightful analysis of the anti-inflammatory potential of CBD in the context of high glucose-induced endothelial cell inflammatory response and barrier disruption. Focusing on endothelial cells, which play a crucial role in vascular inflammation, the research explores CBD’s impact on mitigating the inflammatory response triggered by elevated glucose levels. The article demonstrates that CBD treatment effectively attenuates the inflammatory processes induced by high glucose concentrations. Through its interactions with cannabinoid receptors, CBD regulates the expression of adhesion molecules and chemokines, leading to the suppression of inflammation-associated adhesion and migration of immune cells. Moreover, CBD maintains endothelial barrier integrity by countering the disruptive effects of high glucose on tight junction proteins. This study underscores CBD’s potential to intervene in inflammatory pathways, particularly in contexts relevant to metabolic and cardiovascular disorders. By investigating CBD’s ability to counteract the detrimental effects of high glucose on endothelial cells, this research advances our understanding of its anti-inflammatory mechanisms and positions CBD as a promising therapeutic candidate for addressing inflammatory-related endothelial dysfunction. Another study about the anti-inflammatory qualities of CBD, conducted by Malfait et al. (2000), presents a noteworthy analysis of the anti-inflammatory potential of CBD as an oral therapeutic in murine collagen-induced arthritis. The article contributes valuable insights into the effects of CBD on arthritis-related inflammation, shedding light on its role in modulating the immune response and alleviating joint inflammation. Through rigorous experimentation and assessment of disease progression, the research showcases CBD’s ability to ameliorate various pathological aspects of arthritis, including synovial hyperplasia and immune cell infiltration. The study highlights CBD’s potential to downregulate pro-inflammatory cytokines and chemokines, effectively dampening the inflammatory cascade. Furthermore, CBD’s influence on various cellular and molecular pathways, such as NF-κB signaling, is elucidated, reinforcing its anti-inflammatory properties. The findings not only demonstrate CBD’s capacity to attenuate inflammatory responses but also establish its potential as a promising oral therapeutic for arthritic conditions.
The collective body of research on the potential anxiolytic effects of CBD presents a comprehensive and multi-dimensional understanding of its therapeutic implications. The study by Blessing et al. (2015) synthesizes a wide array of preclinical and clinical investigations to emphasize CBD’s promise as a treatment for anxiety disorders. This review underscores CBD’s anxiolytic effects across various animal models, highlighting its potential in managing conditions such as generalized anxiety disorder, social anxiety disorder, and post-traumatic stress disorder. Additionally, the article delves into CBD’s influence on neural circuits associated with anxiety regulation, establishing its role in modulating serotonin and endocannabinoid systems. Shannon et al. (2019) contribute to this discourse by documenting the real-world effectiveness of CBD in managing anxiety and sleep-related issues, reinforcing the anxiolytic potential of CBD across diverse contexts. Their study offers a glimpse into the practical applications of CBD in alleviating anxiety symptoms and improving sleep quality, accentuating its value as a natural alternative for addressing anxiety-related concerns. Further contributing to the understanding of CBD’s anxiolytic properties, Schier et al. (2012) dissect the neural underpinnings of CBD’s anxiolytic effects. Through meticulous experimentation, they underscore its potential as a novel anxiolytic agent, examining its impact on social anxiety disorder and generalized anxiety disorder. The article establishes the correlation between CBD administration and reductions in both subjective and physiological markers of anxiety, elaborating on its interactions with the endocannabinoid system, serotonin receptors, and neural circuits central to anxiety regulation. In a similar vein, Crippa et al. (2011) delve into the neural basis of CBD’s effects in generalized social anxiety disorder, employing neuroimaging techniques to explore alterations in brain activity and connectivity. Their work elucidates the potential of CBD to modulate anxiety-processing neural circuitry, shedding light on its role in managing cognitive processes associated with social anxiety. Linares et al. (2019) introduce an intriguing dimension to the discourse by investigating CBD’s anxiolytic effects through a dose-response paradigm. Their study elucidates an inverted U-shaped curve, demonstrating that CBD’s optimal anxiolytic effects occur within a specific dosage range, offering insights into the dosing precision required for harnessing its anxiolytic potential effectively. Collectively, this body of research underscores CBD’s multifaceted mechanisms and therapeutic potential in alleviating anxiety-related conditions. The studies collectively contribute to the foundation of knowledge, positioning CBD as a promising candidate for the development of alternative treatments for anxiety disorders.
The treatment of epilepsy and other seizure inducing disorders has been severely changed for the better with the use of CBD. For example, the study conducted by Devinsky et al. (2016) offers a comprehensive analysis of the potential anti-seizure effects of CBD in patients with treatment-resistant epilepsy. It reports significant reductions in seizure frequency among patients receiving CBD treatment, with a notable portion experiencing a reduction of seizures by half or more. Notably, this effect was observed across different epilepsy types, including Dravet syndrome and Lennox-Gastaut syndrome, which are notoriously challenging to manage. Following his research path Devinsky et al. (2017) focused on the application of CBD in treating drug resistant seizures. This research is of particular significance due to the challenging nature of Dravet syndrome, a severe form of epilepsy that often proves refractory to conventional treatments. The study employed a randomized, double-blind, placebo-controlled trial design, offering a robust framework for evaluating CBD’s efficacy. The results revealed a remarkable reduction in the frequency of convulsive seizures among patients receiving CBD treatment, underscoring CBD’s potential as a therapeutic option for managing drug-resistant seizures, offering hope to individuals and families facing the debilitating impact of Dravet syndrome. In 2018, Devinsky et al. once again, conducted a comprehensive analysis of the potential anti-seizure effects of CBD, this time focusing on its efficacy in reducing drop seizures in individuals with Lennox-Gastaut syndrome (LGS). LGS is characterized by multiple seizure types, including atonic or “drop” seizures, which often result in sudden loss of muscle tone and falls. The study’s findings reveal a significant reduction in the frequency of drop seizures among patients receiving CBD treatment compared to those in the placebo group. Notably, a substantial proportion of CBD-treated patients experienced a reduction of at least 50% in drop seizures. Similarly, the research conducted by Thiele et al. (2018) provided more insight into the anti-seizure effects of CBD in the context of LGS. The findings highlight that CBD treatment leads to a significant reduction in the frequency of drop and non-drop seizures among patients with LGS. The study also underscores the safety profile of CBD, with adverse events primarily being mild to moderate in intensity. Additionally, the research demonstrates a favorable impact on overall quality of life, as indicated by improvements in emotional and physical well-being reported by caregivers. These findings corroborate the potential of CBD as an effective and well-tolerated adjunctive treatment for patients with LGS, offering hope for individuals with this challenging epileptic syndrome and their families. In the face of treatment, the research conducted by Gaston et al. (2017) provides valuable insights into the potential interactions between CBD and commonly used antiepileptic drugs (AEDs) in the context of managing seizures. The study focuses on individuals with treatment-resistant epilepsy who were prescribed CBD as an adjunctive treatment, aiming to assess potential drug interactions with their existing AED regimens. Through a comprehensive analysis of drug interaction data, the researchers find that CBD does not significantly affect the plasma levels of most AEDs commonly used to treat epilepsy. This suggests that CBD may be safely combined with these AEDs without causing substantial alterations in their pharmacokinetics. These findings contribute to the understanding of the practical implications of using CBD in conjunction with existing epilepsy treatments, providing valuable information for clinicians and patients seeking alternative approaches to managing seizures. Whole plant CBD isolates are available in the FDA approved product Epidiolex.
Continuing in the neurological effect of CBD, we explored its effect on depression. For example, the study conducted by Linge et al. (2016) offers valuable insights into the potential antidepressant effects of CBD, shedding light on its rapid-acting antidepressant-like properties and its influence on neurotransmission pathways. Focusing on its impact on depressive behaviors and neurochemical mechanisms, the research unveils CBD’s ability to induce rapid and sustained antidepressant effects in animal models. The study delves into the involvement of the 5-HT1A receptors, which play a pivotal role in mood regulation and antidepressant responses. By enhancing cortical 5-HT/glutamate neurotransmission, CBD is shown to modulate synaptic plasticity, a crucial factor in mood disorders. This research significantly contributes to the understanding of CBD’s potential as a novel approach for treating depression by targeting neurotransmitter systems, ultimately paving the way for further investigations into its therapeutic efficacy and mechanisms of action in the realm of depression and mood disorders. Similarly, The research conducted by Sales et al. (2018) presents a significant contribution to understanding the antidepressant potential of CBD, particularly its dependence on brain serotonin levels. The study explores the mechanisms underlying CBD’s antidepressant-like effects and its interactions with the brain’s serotonin system, a key player in mood regulation. Through a series of experiments, the research reveals that CBD’s ability to induce an antidepressant-like response is contingent upon the availability of serotonin in the brain. By influencing serotonergic neurotransmission, CBD is shown to enhance the anti-depressant effects, providing further evidence of its involvement in mood modulation. The study by Zanelati et al. (2010) investigates the effects of CBD on depressive-like behaviors and its interaction with the 5-HT1A receptors, which are known to play a crucial role in the regulation of mood. The study’s findings reveal that CBD administration induces antidepressant-like effects in mice, as demonstrated by its ability to mitigate behavioral indicators of depression. Additionally, the study explores the involvement of the 5-HT1A receptor in mediating these effects, suggesting that CBD’s antidepressant actions might be, at least partially, mediated through its interaction with these receptors. By establishing a link between CBD, 5-HT1A receptors, and antidepressant-like responses, this research advances our understanding of the underlying mechanisms through which CBD exerts its potential antidepressant effects and highlights the significance of the serotonergic system in its action. On the other hand, CBD also plays into other neurological disorders that deal with depression. For example, the research by Shannon and Opila-Lehman (2016) presents a case report that offers insights into the potential antidepressant effects of CBD in the context of pediatric anxiety and insomnia associated with posttraumatic stress disorder (PTSD). The case report discusses the administration of CBD oil to a young patient with PTSD-related symptoms, including severe anxiety and sleep disturbances. Notably, the patient experienced significant improvements in both anxiety and sleep after CBD treatment. Although the report is a single case study, it suggests that CBD might play a role in alleviating not only anxiety and sleep problems but also the depressive aspects often intertwined with PTSD. Building on that, the study conducted by Lee et al. (2017) offers valuable insights into the antidepressant potential of CBD, particularly in the context of regulating emotion and emotional memory processing. It explores CBD’s impact on emotional memory, highlighting its ability to modulate the consolidation and reconsolidation of emotionally charged memories. By interfering with the process of memory formation associated with traumatic or negative experiences, CBD exhibits potential to alleviate the emotional burden often linked to depression. Moreover, the article underscores CBD’s influence on the endocannabinoid system and its interaction with various neurotransmitter systems involved in emotional regulation. This research expands our understanding of CBD’s multifaceted effects on mood and emotional processing, suggesting its potential as a promising therapeutic agent for addressing depression and related disorders.
In a complex review of the analgesic effects of cannabinoids written by Russo (2008), he highlights CBD’s potential to modulate pain perception through its interactions with the endocannabinoid system and other signaling pathways. The review encompasses various pain conditions, such as neuropathic pain and fibromyalgia, underscoring the potential of cannabinoids, including CBD, to alleviate pain in diverse contexts. Such an explanation of the pain relief can be found in the study conducted by Xiong et al. (2012). Which provides valuable insights into the analgesic effects of CBD by investigating its potential to alleviate inflammatory and neuropathic pain. Moreover, the research explores the involvement of α3 glycine receptors in mediating a reduction in inflammatory and neuropathic pain in experimental models suggesting that CBD’s analgesic actions may be attributed, at least in part, to its interaction with these receptors. This study enhances our understanding of the intricate mechanisms through which CBD exerts its analgesic effects, shedding light on its potential as a novel intervention for both inflammatory and neuropathic pain conditions. Which could present as a promising therapeutic option for individuals suffering from conditions like migraine, headache, and chronic pain.
The research conducted by Campos et al. (2016) presents a comprehensive analysis of the neuroprotective qualities of CBD, particularly in the context of neuropsychiatric disorders. The study delves into the potential of CBD as a promising therapeutic agent for safeguarding neural health and addressing neurodegenerative and neuropsychiatric conditions. The research highlights CBD’s interactions with various molecular targets, including cannabinoid receptors and non-cannabinoid receptor systems, that are implicated in neuroinflammation, oxidative stress, and excitotoxicity. Furthermore, the analysis discusses the evidence supporting CBD’s ability to attenuate neurodegenerative processes and provide cognitive and behavioral benefits in animal models of neuropsychiatric disorders. Similarly, the study by Fernández-Ruiz et al. (2013) provides a comprehensive analysis of the potential neuroprotective qualities of CBD, particularly in the context of neurodegenerative disorders. This research critically examines the existing preclinical and clinical evidence surrounding CBD’s impact on various neurodegenerative conditions, including Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis. The article highlights CBD’s multifaceted mechanisms of action, which encompass anti-inflammatory, antioxidant, and anti-apoptotic effects, all of which contribute to its potential neuroprotective properties. This underscores CBD’s potential to modulate neuroinflammation, oxidative stress, and mitochondrial dysfunction, which are key contributors to the pathogenesis of these disorders. Furthermore, the article explores CBD’s interactions with endocannabinoid and non-endocannabinoid receptors, including its modulation of adenosine receptors and the serotoninergic system, shedding light on its potential therapeutic utility in neurodegenerative contexts.
Not only does CBD have anxiolytic purposes but it also improves sleep time in those who might be suffering from insomnia, a disorder that often goes hand in hand with anxiety. The research conducted by Chagas et al. (2013) offers insights into the potential treatment of insomnia with CBD by examining its effects on the sleep-wake cycle in rats. The study provides a preclinical perspective on CBD’s impact on sleep patterns, shedding light on its influence on both the initiation and maintenance of sleep. Through acute systemic administration of CBD, the researchers observe an increase in total sleep time, as well as a reduction in sleep latency. The study explores the potential mechanisms underlying CBD’s effects, noting its interactions with neurotransmitter systems involved in sleep regulation. While the research is conducted on rats and requires further investigation in humans, it lays the groundwork for understanding CBD’s potential as a sleep aid and opens avenues for exploring its application in addressing insomnia. Interestingly, the comprehensive review conducted by Babson et al. (2017) highlights that CBD may have a multifaceted influence on sleep by interacting with different aspects of the sleep-wake cycle. While some studies suggest that CBD could enhance sleep by reducing sleep latency and improving sleep continuity, other research points to a possible biphasic effect, where lower doses promote wakefulness and higher doses promote sleep. However, a study conducted by Linares et al. (2018) found that acute CBD intake does not significantly alter the sleep-wake cycle of healthy participants. Despite the growing interest in CBD’s potential as a sleep aid, this research suggests that CBD may not induce immediate changes in sleep parameters among individuals without existing sleep disturbances. Highlighting the need for additional research, particularly in populations with sleep disorders, to fully elucidate CBD’s role in the treatment of insomnia and related sleep issues.
The study conducted by Elbaz et al. (2015) offers an insightful analysis of the anti-tumor potential of CBD, specifically in the context of breast cancer. The research investigates novel mechanisms through which CBD modulates the tumor microenvironment and inhibits the epidermal growth factor (EGF)/EGF receptor (EGFR) pathway, highlighting its potential as a therapeutic agent against breast cancer. The study reveals that CBD treatment leads to a decrease in tumor cell viability and migration, suggesting its ability to suppress cancer cell proliferation and metastatic potential. Additionally, the research uncovers CBD’s impact on the tumor microenvironment, promoting changes that discourage tumor growth and invasiveness. The inhibition of the EGF/EGFR pathway by CBD further emphasizes its potential as a multifaceted anti-tumor agent. Similarly, the study conducted by Wang and Multhoff (2021) highlights CBD’s potential to modulate various pathways and factors involved in cancer progression, including cell cycle regulation, apoptosis, inflammation, and angiogenesis. The authors discuss CBD’s ability to target multiple signaling pathways that contribute to tumor growth and metastasis, indicating its potential to disrupt key processes in cancer development and potentially enhance the efficacy of traditional chemotherapy approaches.
Unknown Potential
Cannabicitran (CB) is another lesser-known cannabinoid found in cannabis plants. There is very limited scientific evidence regarding the specific medicinal properties of Cannabicitran. So, due to the scarcity of research, it’s challenging to make definitive statements about its potential health benefits. However, it is structurally similar to THC and CBD which have varying medicinal benefits. CB may be suitable for anxiety, stabilizing mood, and may help with pain and other health issues. Studies show that it is especially good for eye health and may help prevent or even manage glaucoma. While there is still little research about it, there is much potential for its health benefits. However, it is important to note that prospective medicinal properties of any cannabinoid can vary widely depending on factors like dosage, method of administration, and individual biochemistry.
Testimonial
Anti-Inflammatory
Cannabicyclol (CBL) is a lesser-known cannabinoid found in the cannabis plant, though it typically appears in much lower concentrations compared to other more prevalent cannabinoids like THC or CBD. CBL is a result of the oxidative degradation of cannabichromene (CBC) under various conditions, including heat and light exposure. As a result, it’s often considered a non-enzymatic breakdown product of CBC. Despite its relatively low abundance, CBL has garnered some attention for its potential therapeutic properties. However, research on CBL is limited, and its pharmacological effects and mechanisms of action are not as well understood as those of more extensively studied cannabinoids. Further exploration and investigation are necessary to fully comprehend the potential uses and effects of CBL in the context of human health and well-being.
The study conducted by Lisdiana and Mustikaningtyas in 2023, titled “Molecular Docking of the Cannabis sativa L. Bioactive Compound Against Inflammation Induced by Cigarette Smoke exposure,” explores the potential anti-inflammatory effects of bioactive compounds found in Cannabis sativa L., including cannabicyclol (CBL). The researchers used molecular docking techniques with results showing that “the compounds from Cannabis sativa L. can act as an anti-inflammatory in the context of cigarette smoke-induced inflammation.” which is indicated by “the similarity of amino acid residues resulting from the interaction of the aspirin drug (control) with the anti-inflammatory receptor protein in the compound cannabidihydrophenanthrene with PDGFRA and KDR receptors and compounds cannabicyclol with AKT1 and KDR receptors” (Lisdiana).
Testimonial
Anti-Inflammatory, Anti-Insomnia, Analgesic, Appetite Stimulant, Anti-Rejection in Skin Graft Patients, Anti-Tumor.
Cannabichromene (CBC) is a naturally occurring cannabinoid found in the Cannabis sativa plant. It is one of the lesser-studied cannabinoids compared to well-known compounds like delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD). Like other cannabinoids, CBC is derived from the precursor cannabigerolic acid (CBGA), which is enzymatically converted into CBC through specific biosynthetic pathways. CBC does not exhibit the same psychoactive properties as THC, making it a non-psychoactive compound. Research into CBC’s potential therapeutic effects has gained momentum, revealing its diverse pharmacological actions. One of its notable characteristics is its interaction with the endocannabinoid system, particularly the cannabinoid receptors CB1 and CB2, albeit to a lesser extent than THC. Studies have suggested that CBC may offer anti-inflammatory, analgesic, and other medicinal properties. Its unique molecular structure and interactions with various receptors make CBC an intriguing target for further investigation into its potential medical applications. As research continues to unfold, a deeper understanding of CBC’s mechanisms of action and its potential benefits in addressing various health conditions is expected to emerge.
In the article “Non-psychoactive cannabinoids modulate the descending pathway of antinociception in anesthetized rats through several mechanisms of action” by Maione et al., published in the British Journal of Pharmacology in 2011, the analgesic effects of cannabichromene (CBC) are investigated within the context of the descending antinociceptive pathway in anesthetized rats. The study aims to explore how non-psychoactive cannabinoids, including CBC, influence the pain-modulation pathway in the brain. Through their research, the authors demonstrate that CBC exerts analgesic effects by interacting with various mechanisms within the descending pain control pathway. These mechanisms include the activation of endocannabinoid receptors and the release of certain neurotransmitters that contribute to pain relief. The article underscores the potential of CBC as a non-psychoactive compound with analgesic properties, shedding light on its role in pain management.
In the article “The Mechanism of Cannabichromene and Cannabidiol Alone Versus in Combination in the Alleviation of Arthritis-Related Inflammation” by Grogan et al., published in Annals of Plastic Surgery in 2023, the anti-inflammatory effects of cannabichromene (CBC) are examined within the context of arthritis-related inflammation. The study investigates both the individual and combined effects of CBC and cannabidiol (CBD) in alleviating inflammation associated with arthritis. Similarly, it was found in the article “Anti-inflammatory properties of cannabichromene” that CBC possesses anti-inflammatory properties when it significantly reduced edema formation in rats, indicating its potential to mitigate acute inflammatory processes. This research sheds light on how CBC, in combination with CBD, can modulate inflammation through multiple mechanisms, including interactions with cannabinoid receptors and other molecular pathways involved in immune response and inflammation. The article “Cannabichromene is a cannabinoid CB2 receptor agonist” also proves this as it investigates how CBC acts as an agonist for the CB2 receptor, which is primarily associated with immune modulation and anti-inflammatory responses. The research demonstrates that CBC’s interaction with the CB2 receptor leads to the suppression of inflammation by influencing various signaling pathways involved in immune and inflammatory processes. And by highlighting CBC’s role as a CB2 receptor agonist, the article provides valuable insights into the underlying mechanisms of its anti-inflammatory properties. These articles highlight CBC’s potential as an anti-inflammatory agent and emphasize the synergistic effects of combining cannabinoids for enhanced therapeutic outcomes.
In the article “Antitumor activity of plant cannabinoids with emphasis on the effect of cannabidiol on human breast carcinoma” by Ligresti et al., published in the Journal of Pharmacology and Experimental Therapeutics in 2006, the potential anti-tumor effects of plant cannabinoids, including cannabichromene (CBC), are explored, with a specific focus on cannabidiol (CBD) and its impact on human breast carcinoma. While the article may not exclusively analyze CBC’s anti-tumor effects, it likely contributes to the broader understanding of cannabinoids’ potential in cancer treatment. The study suggests that certain cannabinoids, including CBD, have the ability to inhibit the growth of human breast carcinoma cells in vitro through various mechanisms, such as inducing apoptosis (programmed cell death) and inhibiting tumor cell migration. Although the article does not solely concentrate on CBC, it falls within the realm of research that underscores the potential of cannabinoids in targeting cancer cells. The findings provide insights into the complex interactions between cannabinoids and cancer cells, potentially paving the way for further investigations into the therapeutic applications of CBC and other plant cannabinoids in combating cancer.
Testimonial
Appetite Stimulant/Regulation
Δ10 is one of many cannabinoids found in cannabis, however, it can only be found in small trace amounts. There is so little of it that its often missed in extraction processes. Because of this, there is limited research on its medicinal properties. But it is known to act similarly to Delta-8 and Delta-9 which can bind to the CB1 receptors in the brain and nervous system. With this being said, in high concentrations Delta-10 could potentially possess a Potential for Pain Relief, appetite stimulation, anti-inflammatory properties, and anxiolytic effects. While there is still limited research on it, Delta-10 shows a promising future in the world of medicine.
Testimonial
Antinociceptive
Tetrahydrocannabihexol (THCH) is a lesser-known cannabinoid found in cannabis. It belongs to the family of cannabinoids, which are the active compounds present in the cannabis plant. THCH is structurally related to other well-known cannabinoids like THC (tetrahydrocannabinol) and CBD (cannabidiol). While THC is primarily responsible for the psychoactive effects of cannabis, and CBD is renowned for its potential therapeutic properties, THCH’s effects and characteristics are less understood due to limited research. Like other cannabinoids, it interacts with the endocannabinoid system in the human body, which plays a crucial role in regulating various physiological processes. THCH’s specific properties, potential effects, and medical applications require further investigation and research, as it remains a relatively understudied compound compared to THC and CBD. Its presence in cannabis adds to the complexity of the plant’s chemical profile, and ongoing research may uncover its unique contributions to the overall effects of different cannabis strains.
The article titled “Identification of a new cannabidiol n-hexyl homolog in a medicinal cannabis variety with an antinociceptive activity in mice: cannabidihexol” by Linciano et al., published in Scientific Reports in 2020, highlights the antinociceptive effects of tetrahydrocannabidihexol (THCH), a newly identified cannabinoid derivative found in medicinal cannabis. Antinociception refers to the reduction of sensitivity to painful stimuli. The research findings suggest that THCH exhibits antinociceptive activity in mice, indicating its potential as a pain-relieving compound. This property is particularly noteworthy in the context of medical cannabis, as it suggests that THCH could be harnessed for its analgesic effects, potentially offering a natural alternative for pain management. However, further research is needed to explore the full extent of THCH’s antinociceptive properties and its safety and efficacy in humans.
Testimonial
Appetite stimulant/regulation
The article titled “Isolation of a High-Affinity Cannabinoid for the Human CB1 Receptor from a Medicinal Cannabis sativa Variety: Δ9-Tetrahydrocannabutol, the Butyl Homologue of Δ9-Tetrahydrocannabinol” by Linciano et al. (2020) explores the medicinal properties of Δ9-Tetrahydrocannabutol (THCB), a cannabinoid found in medicinal Cannabis sativa. THCB is identified as a high-affinity ligand for the human CB1 receptor, suggesting its potential therapeutic relevance. The CB1 receptor is primarily located in the central nervous system and is associated with various physiological processes, including pain modulation, appetite regulation, and mood control. Given its affinity for the CB1 receptor, THCB may have analgesic, appetite-stimulating, and potentially mood-altering effects, similar to its well-known counterpart, Δ9-Tetrahydrocannabinol (THC). Further research on THCB’s specific medicinal properties and potential therapeutic applications is warranted, as it could provide valuable insights into the pharmacological effects of this lesser-known cannabinoid.
Testimonial
Analgesic, antiemetic
Delta-8 tetrahydrocannabinol (Δ8-THC or simply Δ8) is a naturally occurring cannabinoid found in the cannabis plant, though it is found in small quantities and sometimes even not at all. It is chemically similar to the more well-known Delta-9 tetrahydrocannabinol (Δ9-THC), the primary psychoactive compound in cannabis, but with a slightly different molecular structure. Δ8 is known to interact with the endocannabinoid system in the human body, primarily by binding to the CB1 and CB2 receptors, leading to various physiological and psychological effects. It has gained attention for its potential therapeutic applications, including its reported ability to alleviate symptoms like nausea, anxiety, and pain, without producing the intense psychoactive effects often associated with Δ9-THC. CBD in hemp can be used as a precursor in the synthesis of Delta-8 THC (Δ8-THC), although the process typically involves several chemical steps and is not a direct conversion. To synthesize Δ8-THC from CBD, the CBD molecule undergoes a chemical reaction that modifies its structure. One common method involves isomerization, where CBD is treated with certain acidic conditions or catalysts to rearrange its molecular bonds and convert it into Δ8-THC. This process typically requires specialized equipment and expertise, and it’s essential to follow legal regulations, as the legality of Δ8-THC varies by jurisdiction. It’s crucial to note that the production and sale of Δ8-THC are subject to specific regulations and restrictions in many places, and individuals interested in these processes should adhere to applicable laws and regulations governing controlled substances and hemp-derived products. So, despite its promising characteristics, more research on Δ8-THC is needed to fully understand its potential benefits and risks.
In the article “Review of delta-8-tetrahydrocannabinol (Δ8-THC): Comparative pharmacology with Δ9-THC” by Tagen and Klumpers, published in the British Journal of Pharmacology in 2022, the analgesic properties of delta-8-tetrahydrocannabinol (Δ8-THC) are discussed within the context of comparative pharmacology with delta-9-tetrahydrocannabinol (Δ9-THC). The study aims to provide insights into the pharmacological profile of Δ8-THC and its potential as an analgesic agent. Through their analysis, the authors suggest that Δ8-THC exhibits analgesic effects comparable to Δ9-THC, indicating its potential as a pain-relieving substance. While the exact mechanisms behind Δ8-THC’s analgesic properties require further investigation, the article underscores its potential role as an alternative or complementary treatment option for pain management. However, as with any cannabinoid-based intervention, further research is necessary to fully understand its analgesic mechanisms, optimize dosages, and evaluate its overall efficacy and safety. The study highlights the growing interest in exploring the therapeutic potential of lesser-studied cannabinoids like Δ8-THC in addressing pain-related conditions, providing valuable insights for the future development of analgesic treatments.
In the article “An efficient new cannabinoid antiemetic in pediatric oncology” by Abrahamov et al., published in Life Sciences in 1995, the antiemetic properties of delta-8-tetrahydrocannabinol (Δ8-THC) are explored, particularly in the context of pediatric oncology. The study aims to investigate the potential of Δ8-THC as an antiemetic agent to alleviate nausea and vomiting in children undergoing chemotherapy. Through their research, the authors find that Δ8-THC effectively reduces chemotherapy-induced nausea and vomiting in pediatric cancer patients. The article highlights Δ8-THC’s antiemetic potential and its potential role in improving the quality of life for pediatric cancer patients who often experience these distressing side effects of chemotherapy. While further research is needed to fully understand the mechanisms behind Δ8-THC’s antiemetic effects and its optimal dosing regimen, the study contributes to the body of evidence suggesting that cannabinoids, including Δ8-THC, have promise as supportive therapies for managing chemotherapy-induced nausea and vomiting, particularly in pediatric populations.
Testimonial
Neurodegenerative Disease, Sleep Aid/Sedation, Analgesic, Antibacterial
Cannabinol (CBN) is a naturally occurring cannabinoid found in the Cannabis sativa plant. It is one of the many compounds produced by the plant’s metabolic processes. CBN is often referred to as a degradation product of tetrahydrocannabinol (THC), the well-known psychoactive cannabinoid. Unlike THC, CBN is typically present in much lower concentrations in cannabis plants. It was one of the first cannabinoids to be isolated and identified from cannabis. CBN is thought to be a mildly psychoactive compound, although its psychoactive effects are generally considered to be less potent compared to THC. It is also being explored for its potential therapeutic properties, including its potential as a sleep aid and its interaction with the endocannabinoid system.
In the article “Cannabidiol, cannabinol and their combinations act as peripheral analgesics in a rat model of myofascial pain,” published in the Archives of Oral Biology in 2019 by Hayes Wong and Brian E. Cairns, the analgesic effects of cannabinol (CBN) are explored. The study investigates the potential analgesic properties of CBN, along with cannabidiol (CBD), in a rat model of myofascial pain. Through their research, the authors demonstrate that CBN, along with CBD, exhibits peripheral analgesic effects in this experimental model. This suggests that CBN might have the ability to alleviate pain at the site of its application. The study contributes to our understanding of CBN’s potential as a pain-relieving agent, particularly in the context of myofascial pain. The findings highlight CBN’s potential as a candidate for further exploration in pain management strategies and offer insights into the broader therapeutic potential of cannabinoids for pain relief.
In the study titled “Cannabinol inhibits oxytosis/ferroptosis by directly targeting mitochondria independently of cannabinoid receptors,” published in Free Radical Biology & Medicine in 2022 by Liang et al., the researchers investigated the potential of cannabinol (CBN) to inhibit a specific type of cell death called oxytosis or ferroptosis. The study aimed to understand whether CBN could target mitochondria directly and independently of cannabinoid receptors to mitigate cell death. Through their research, the authors demonstrated that CBN effectively inhibits oxytosis/ferroptosis by directly targeting mitochondria, suggesting a mechanism of action beyond cannabinoid receptors. The findings offer insights into CBN’s potential as a protective agent against certain forms of cell death, indicating its potential utility in neuroprotective strategies. The study advances our understanding of CBN’s effects on cellular processes and presents opportunities for further exploration of its therapeutic applications in conditions involving oxidative stress and cell death.
In the article “Comparison of Efficacy of Cannabinoids versus Commercial Oral Care Products in Reducing Bacterial Content from Dental Plaque: A Preliminary Observation” by Stahl and Vasudevan, published in Cureus in 2020, the antibacterial effects of cannabinol (CBN) are investigated in relation to reducing bacterial content in dental plaque. The study aims to compare the efficacy of cannabinoids, including CBN, with commercial oral care products in terms of their ability to reduce bacterial populations in dental plaque. The authors present preliminary observations suggesting that cannabinoids exhibit potential antibacterial properties and may contribute to reducing bacterial content in dental plaque. In the results, they found that CBN was an effective antibacterial for dental plaque across multiple research groups (Stahl).
Testimonial
Unknown potential
CBDB as a novel butyl analog of CBD often characterized as an impurity of commercial CBD extracted from hemp.
The article titled “Chemical and spectroscopic characterization data of ‘cannabidibutol’, a novel cannabidiol butyl analog,” authored by Cinzia Citti and colleagues and published in Data in Brief in 2019, provides essential data to characterize a novel compound known as cannabidibutol (CBDB). This research contributes to our understanding of the chemical and spectroscopic properties of CBDB, a butyl analog of cannabidiol (CBD). While the article primarily focuses on providing characterization data, this information is crucial for assessing the medicinal potential of CBDB. Analog compounds like CBDB hold promise in expanding the scope of cannabinoids’ therapeutic applications, as they can exhibit different pharmacological profiles compared to their parent compounds. By presenting detailed chemical and spectroscopic data for CBDB, this study lays the groundwork for future investigations into the potential medicinal uses of this compound. This research contributes to the ongoing exploration of the diverse array of cannabinoids and their derivatives, offering insights that could lead to novel therapeutic options for various medical conditions.
Testimonial
Dry skin, cancer/anti-tumor growth, anti-inflammatory
Cannabigerovarin (CBGV) is a naturally occurring phytocannabinoid found in the Cannabis sativa plant. It is structurally related to other cannabinoids like cannabigerol (CBG) and cannabidiol (CBD). CBGV is produced through the plant’s biosynthetic pathways and is a minor constituent of cannabis compared to more well-known cannabinoids like THC and CBD. While research on CBGV is still in its early stages, preliminary studies suggest that it might interact with the endocannabinoid system, which plays a crucial role in regulating various physiological processes. CBGV’s potential effects, mechanisms of action, and therapeutic applications are still being explored, and as scientific interest in the diverse range of cannabinoids present in cannabis grows, CBGV’s distinct properties could potentially contribute to the development of novel treatments and therapies in the future.
In studies about CBGV, it was found that it possessed anti-inflammatory qualities by suppressing the release of pro-inflammatory cytokines induced by Lipopolysaccharide treatment. Yet, its most noticeable function was its ability to treat dry skin. This was shown in the article “Cannabinoids as anti-acne agents white paper” where non-cytotoxic concentrations of CBG and CBGV reportedly increased lipid synthesis and sebocyte lipogenesis while preventing inflammatory cytokine release, which may be beneficial for therapies targeting conditions associated with dry skin and impaired barrier function (Lavvan).
In the study “Cannabidiol exerts anti-proliferative activity via a cannabinoid receptor 2-dependent mechanism in human colorectal cancer cells” it was found that CBGV and other non-psychoactive cannabinoid derivatives had anti-proliferative activity in cancer cells. It was also found in the study, “Efficient Synthesis for Altering Side Chain Length on Cannabinoid Molecules and Their Effects in Chemotherapy and Chemotherapeutic Induced Neuropathic Pain” that CBGV demonstrated the largest cell growth reduction and “In cell lines sensitive to CBG, CBGV remained more efficacious, except in DLD-1 cells where the CBG and CBGV had a similar effect on cell viability” (Raup-Konsavage).
Testimonial
Neuroprotection, analgesic, antibacterial, antifungal
Cannabichromevarin (CBCV) is a relatively new cannabinoid so research on this molecule is lacking. However, it is structurally similar to CBC and is suspected to have some of the same therapeutic qualities including possible anti-inflammatory properties, neuroprotective effects, pain relief, antibacterial and antifungal activities, and possible leads to inhibiting tumor growth. While more information and studies must come out about CBCV in order to find its true value in the world of medicine, it demonstrates a promising future.
Testimonial
Anti-seizure, anti-inflammatory, Crohn’s, IBD
Cannabidivarin (CBDV) is a non-psychoactive cannabinoid found in the Cannabis sativa plant. Similar to other cannabinoids like cannabidiol (CBD), CBDV has attracted significant attention for its potential therapeutic applications. CBDV shares a structural resemblance to CBD, but it possesses distinct properties that set it apart. While research on CBDV is still in its early stages, preliminary studies have suggested potential benefits in various medical conditions. CBDV’s interaction with the endocannabinoid system, which regulates numerous physiological processes, is a key area of investigation. Early research indicates that CBDV may influence cannabinoid receptors, potentially impacting pain perception, inflammation, and neurological functions. Moreover, CBDV’s potential in addressing conditions like epilepsy and autism spectrum disorders has drawn attention, indicating its potential as a treatment option for these conditions.
Through many different studies researchers found that CBDV exhibited anticonvulsant properties in both mouse and rat models, effectively reducing the severity and frequency of seizures. For example, the research in “Cannabidivarin (CBDV) suppresses pentylenetetrazole (PTZ)-induced increases in epilepsy-related gene expression,” aimed to explore how CBDV might influence the genetic expression associated with epilepsy. This study’s findings indicated that CBDV had a suppressive effect on the upregulation of epilepsy-related genes induced by PTZ, suggesting its potential as a modulator of gene expression in the context of seizures. Also, in the study “Cannabidivarin-rich cannabis extracts are anticonvulsant in mouse and rat via a CB1 receptor-independent mechanism” researchers aimed to determine whether the anticonvulsant effects of CBDV were mediated by the cannabinoid receptor CB1. Through a series of experiments, they found that CBDV-rich cannabis extracts exhibited anticonvulsant activity and interestingly, this effect was independent of the CB1 receptor. This suggests that CBDV’s mechanism of action in exerting anticonvulsant effects might involve pathways beyond the traditional cannabinoid receptors. The study provides valuable insights into the complex interactions between CBDV and the endocannabinoid system, expanding our understanding of its potential therapeutic applications for seizure disorders.
In the study titled “The non-euphoric phytocannabinoid cannabidivarin counteracts intestinal inflammation in mice and cytokine expression in biopsies from UC pediatric patients,” published in Pharmacological Research in 2019, Pagano et al. investigated the potential anti-inflammatory effects of cannabidivarin (CBDV) in the context of intestinal inflammation. The study aimed to explore how CBDV might impact inflammation in mice and pediatric patients with ulcerative colitis (UC). Through their research, the authors demonstrated that CBDV had a counteracting effect on intestinal inflammation in mice models, as well as a suppressive effect on cytokine expression in biopsies from pediatric patients with UC. These findings suggest that CBDV possesses anti-inflammatory properties and might have therapeutic potential for managing inflammatory bowel diseases like ulcerative colitis. The study provides insights into CBDV’s mechanisms of action and its potential applications in treating intestinal inflammation, contributing to the broader understanding of cannabinoid-based interventions for gastrointestinal disorders.
Testimonial
Anti-Diabetic, Anticonvulsant
Δ9-Tetrahydrocannabivarin (THCV) is a naturally occurring cannabinoid compound found in the Cannabis sativa plant. It shares a similar molecular structure with the more well-known cannabinoid, Δ9-tetrahydrocannabinol (THC), which is responsible for the psychoactive effects of cannabis. However, THCV has distinct effects and properties. It has garnered interest due to its potential therapeutic applications in various medical conditions. THCV’s effects on the endocannabinoid system, which plays a crucial role in regulating various physiological processes, are of particular interest. Additionally, THCV’s potential to affect insulin sensitivity, glucose regulation, and lipid metabolism has sparked attention in the context of metabolic disorders like obesity and diabetes. While THCV’s mechanisms and effects are still being explored, its distinct properties and potential therapeutic benefits make it an intriguing subject of scientific investigation.
While similar to THC, THCV varies in its impacts on the G protein-coupled cannabinoid receptors (CB1 and CB2) that are located in the brain and nervous systems. THC activates the CB1 receptor which produces the psychoactive and appetite inducing response, THCV is an agonist or neutral agonist to CB1.
In a randomized, double-blind, placebo-controlled pilot study conducted by Khalid A. Jadoon and others, the impact of THCV on glycemic and lipid parameters in patients with type 2 diabetes was investigated. The results of the study suggest that THCV may have a potential positive effect on glycemic parameters as it decreased fasting plasma glucose (from 7.4 to 6.7 mmol/L) compared to the placebo group which increased from 7.6 to 8 mmol/L 21 (Jadoon et al). Meanwhile, a journal article titled “The cannabinoid Δ9-tetrahydrocannabivarin (THCV) ameliorates insulin sensitivity in two mouse models of obesity,” examines THCV’s impact on restoring insulin sensitivity as a potential therapeutic approach in managing metabolic disorders related to obesity. Through the utilization of mouse and human models, the research suggests that THCV might have a positive effect in improving diabetes and indicating its potential value in addressing metabolic dysregulation associated with obesity.
THCV may also have anticonvulsant properties as well. In the study titled “Δ9-Tetrahydrocannabivarin suppresses in vitro epileptiform and in vivo seizure activity in adult rats,” published in Epilepsia in 2010, researchers focused their studies on both in vitro and in vivo models to examine the effects of THCV on epileptiform activity and seizure occurrence in adult rats. The study reveals promising results, indicating that THCV possesses the ability to suppress epileptiform activity in vitro and reduce seizure activity in vivo. These findings suggest a potential anticonvulsant role for THCV, which could have implications for the development of novel treatments for epilepsy and related neurological disorders. The study contributes to the growing body of research exploring the therapeutic potential of cannabinoids, like THCV, in managing seizure disorders.
Testimonial
Background info
Myricetin (MYC) is a flavonoid compound that is found not only in cannabis but also in a variety of other plants, including berries, onions, and certain vegetables. It belongs to the flavonol subclass of flavonoids and is known for its potential health benefits. In cannabis, myricetin is one of several flavonoids present in the plant, contributing to the complex chemical profile of the cannabis plant. Myricetin has gained attention for its antioxidant and anti-inflammatory properties, which make it a subject of interest in medical and therapeutic research. These properties suggest that myricetin may play a role in protecting cells from oxidative damage and reducing inflammation, which are factors associated with various chronic diseases. However, the specific actions and potential therapeutic applications of myricetin in the context of cannabis are still an area of ongoing investigation, and more research is needed to fully understand its effects and potential benefits.
MYC as antioxidant
The article titled “Antioxidant activity of quercetin and myricetin in liposomes” by Michael H. Gordon and Andrea Roedig-Penman, published in the Chemistry and Physics of Lipids in 1998, provides valuable insights into the antioxidant properties of myricetin. The study investigates the antioxidant activity of myricetin in liposomes, which are lipid-based structures that mimic cell membranes. The research findings demonstrate that myricetin exhibits significant antioxidant activity in this model system, effectively protecting liposomes from oxidative damage. This suggests that myricetin has the ability to scavenge free radicals and reduce lipid peroxidation, which are processes associated with oxidative stress and cellular damage. Myricetin’s antioxidant properties are attributed to its chemical structure, which includes multiple hydroxyl groups that can neutralize reactive oxygen species. Additionally, according to “Myricetin suppresses oxidative stress-induced cell damage via both direct and indirect antioxidant action” by Zhi Hong Wang et al., Myricetin indirectly enhances the cellular antioxidant defense systems, such as the activity of superoxide dismutase (SOD) and catalase (CAT). These findings underscore myricetin’s potential as a potent natural antioxidant, which may have applications in protecting cells and tissues from oxidative stress-related diseases and conditions, including cardiovascular disease, neurodegenerative disorders, and cancer. However, further research is needed to explore its effects in vivo and its practical applications in human health.
MYC as anti-inflammatory
The article titled “Myricetin attenuates LPS-induced inflammation in RAW 264.7 macrophages and mouse models” by Wei Hou et al., published in Future Medicinal Chemistry in 2018, provides valuable insights into the anti-inflammatory properties of myricetin. The study demonstrates that myricetin effectively attenuates inflammation induced by lipopolysaccharide (LPS) in both macrophage cell models and mouse models. Myricetin’s ability to reduce the production of pro-inflammatory molecules and cytokines suggests its potential as an anti-inflammatory agent. The article “Myricetin: A comprehensive review on its biological potentials” by Imran et al., explains myricetin’s ability to modulate various inflammatory pathways and signaling molecules, including cytokines and enzymes like cyclooxygenase (COX) and lipoxygenase (LOX), which are central players in the inflammatory response. The findings highlight myricetin’s capacity to modulate the immune response and suppress the inflammatory process, making it a promising candidate for the development of anti-inflammatory therapies. However, while this research underscores the potential benefits of myricetin in managing inflammation, further studies and clinical trials would be necessary to confirm its efficacy and safety for use in human inflammatory conditions.
MYC as anti-tumor
The article titled “Anti-tumor effects and associated molecular mechanisms of myricetin” by Min Jiang et al., published in Biomedicine & Pharmacotherapy in 2019, provides a comprehensive analysis of the anti-tumor properties of myricetin. The study delves into the mechanisms underlying myricetin’s potential as an anticancer agent. Myricetin, a flavonoid found in various fruits and vegetables, is shown to exert anti-tumor effects through several molecular pathways. It is reported to inhibit cancer cell proliferation, induce apoptosis (programmed cell death) in cancer cells, and interfere with tumor angiogenesis and metastasis. These actions are attributed to myricetin’s ability to modulate various cellular signaling pathways, including those related to inflammation and oxidative stress. Additionally, myricetin is noted for its potential to enhance the effectiveness of conventional cancer therapies and mitigate their side effects. While the study provides valuable insights, further research, including clinical trials, is needed to validate myricetin’s efficacy and safety as a potential therapeutic agent in cancer treatment. Nonetheless, these findings highlight the promising potential of myricetin in the fight against cancer and its multifaceted role in suppressing tumor growth.
Anti-seizure, anti-inflammatory, Crohn’s, IBD
Tetrahydrocannabivarinic acid A (THCVA-A) is a lesser-known cannabinoid compound found in the cannabis plant. It is a derivative of cannabigerolic acid (CBGA), which is a precursor for various cannabinoids in the cannabis plant. THCVA-A is a non-psychoactive cannabinoid, meaning it does not produce the intoxicating effects commonly associated with delta-9-tetrahydrocannabinol (THC). While research on THCVA-A is limited compared to other cannabinoids like THC and CBD, it is believed to have potential therapeutic properties. Some preliminary studies suggest that THCVA-A may exhibit anti-inflammatory, neuroprotective, and appetite-suppressing effects. However, much more research is needed to fully understand the pharmacological properties and potential medical applications of THCVA-A, as well as its interaction with other cannabinoids and compounds in the cannabis plant.
In the Journal of Pharmacology and Experimental Therapeutics article by Petrosino et al. (2018), the authors provide valuable insights into the anti-inflammatory properties of tetrahydrocannabivarinic acid A (THCVA-A). Through a comprehensive experimental approach, the study explores the potential of THCVA-A as a potent anti-inflammatory agent. By examining its effects on immune cell activation, cytokine production, and signaling pathways, the researchers demonstrate that THCVA-A possesses notable anti-inflammatory properties. The study highlights THCVA-A’s ability to modulate the immune response by suppressing pro-inflammatory cytokines and inhibiting key cellular pathways involved in inflammation. These findings contribute to the understanding of the pharmacological actions of THCVA-A, adding to the growing body of research that investigates the potential therapeutic applications of minor cannabinoids. The study underscores the importance of exploring diverse cannabinoids beyond the well-studied compounds like THC and CBD, emphasizing their potential in developing novel anti-inflammatory interventions for various inflammatory conditions.
Testimonial
Anti-hyperalgesic, antiemetic, antioxidant
Cannabidiolic acid, or CBDA, is a naturally occurring compound found in the cannabis plant, particularly in hemp varieties. CBDA is the precursor to CBD (cannabidiol) and is typically present in higher concentrations in raw, unprocessed cannabis plants. It is produced through the enzymatic conversion of CBGA (cannabigerolic acid) and undergoes decarboxylation when exposed to heat, transforming into CBD. While CBDA itself is not known for its direct therapeutic properties, it has garnered attention for its potential health benefits. Some research suggests that CBDA may have anti-inflammatory and anti-nausea properties, making it a subject of interest in the development of new pharmaceuticals and natural remedies. As the scientific understanding of cannabinoids continues to evolve, CBDA is becoming increasingly recognized for its potential role in the medicinal and wellness applications of cannabis and hemp-derived products.
The study conducted by Rock et al. in their article “Effect of Cannabidiolic Acid and ∆9-Tetrahydrocannabinol on Carrageenan-Induced Hyperalgesia and Edema in a Rodent Model of Inflammatory Pain,” published in Psychopharmacology in 2018, investigates the potential anti-hyperalgesic effects of cannabidiolic acid (CBDA) and ∆9-tetrahydrocannabinol (∆9-THC) in a rodent model of inflammatory pain induced by carrageenan. The findings demonstrate that CBDA exhibits significant anti-hyperalgesic effects by reducing pain sensitivity and inflammatory responses in the carrageenan-induced model. Moreover, CBDA’s effects are observed without the psychotropic effects associated with ∆9-THC, highlighting its potential as a non-intoxicating alternative for pain management. Similarly, the study conducted by Vigli et al. in 2021, titled “Chronic Treatment with Cannabidiolic Acid (CBDA) Reduces Thermal Pain Sensitivity in Male Mice and Rescues the Hyperalgesia in a Mouse Model of Rett Syndrome,” provides valuable insights into the anti-hyperalgesic effects of cannabidiolic acid (CBDA) in both healthy male mice and a mouse model of Rett syndrome. The research investigates CBDA’s potential to alleviate thermal pain sensitivity and hyperalgesia, particularly in the context of Rett syndrome, a neurodevelopmental disorder associated with chronic pain. The results indicate that chronic administration of CBDA leads to a reduction in thermal pain sensitivity in healthy mice and effectively rescues hyperalgesia in the Rett syndrome mouse model. These findings highlight CBDA’s analgesic properties and its ability to counteract heightened pain sensitivity in a challenging condition such as Rett syndrome. The exploration of CBDA’s anti-hyperalgesic effects furthers our understanding of its pharmacological properties and its potential as a novel approach to alleviating neuropathic pain, though further research is warranted to comprehensively assess its mechanisms of action and clinical applicability.
In the study conducted by Rock et al. (2020) titled “Evaluation of Repeated or Acute Treatment with Cannabidiol (CBD), Cannabidiolic Acid (CBDA) or CBDA Methyl Ester (HU-580) on Nausea and/or Vomiting in Rats and Shrews,” the authors investigate the antiemetic effects of cannabidiolic acid (CBDA) and its methyl ester derivative (HU-580), both in acute and repeated treatment scenarios. Through experiments involving rats and shrews, the research explores the potential of CBDA and HU-580 to alleviate nausea and vomiting. The findings reveal that both CBDA and HU-580 exhibit significant antiemetic effects, supporting their potential as therapeutic agents for managing these symptoms. Following her 2020 study, Rock also had a study in 2021 called “Therapeutic Potential of Cannabidiol, Cannabidiolic Acid, and Cannabidiolic Acid Methyl Ester as Treatments for Nausea and Vomiting” with findings that suggest that CBDA exhibits significant antiemetic properties, particularly in comparison to CBD, with the potential to alleviate nausea and vomiting. These studies underscore CBDA’s potential as a promising therapeutic option for individuals experiencing nausea and vomiting, highlighting its unique mechanism of action and paving the way for further investigations into its clinical applications for managing these distressing symptoms.
In the article by Boulebd (2022) titled “Is Cannabidiolic Acid an Overlooked Natural Antioxidant? Insights from Quantum Chemistry Calculations,” the author delves into the potential antioxidant effects of cannabidiolic acid (CBDA) using quantum chemistry calculations. Through computational analyses, the study aims to shed light on CBDA’s capacity to act as a natural antioxidant, a property that may have been previously overlooked. The research employs sophisticated techniques to assess CBDA’s ability to scavenge free radicals and its potential to counteract oxidative stress. The findings from quantum chemistry calculations suggest that CBDA indeed exhibits antioxidant properties, which could contribute to its potential therapeutic applications beyond its known effects. This study adds valuable insight to the growing body of research exploring the multifaceted pharmacological effects of CBDA, highlighting its potential as a natural antioxidant and inspiring further investigations into its medical and health-promoting benefits.
Testimonial