1
|
DeVuono MV, Venkatesan T, Hillard CJ. Endocannabinoid signaling in stress, nausea, and vomiting. Neurogastroenterol Motil 2025; 37:e14911. [PMID: 39223918 PMCID: PMC11872018 DOI: 10.1111/nmo.14911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Classical antiemetics that target the serotonin system may not be effective in treating certain nausea and vomiting conditions like cyclic vomiting syndrome (CVS) and cannabinoid hyperemesis syndrome (CHS). As a result, there is a need for better therapies to manage the symptoms of these disorders, including nausea, vomiting, and anxiety. Cannabis is often used for its purported antiemetic and anxiolytic effects, given regulation of these processes by the endocannabinoid system (ECS). However, there is considerable evidence that cannabinoids can also produce nausea and vomiting and increase anxiety in certain instances, especially at higher doses. This paradoxical effect of cannabinoids on nausea, vomiting, and anxiety may be due to the dysregulation of the ECS, altering how it maintains these processes and contributing to the pathophysiology of CVS or CHS. PURPOSE The purpose of this review is to highlight the involvement of the ECS in the regulation of stress, nausea, and vomiting. We discuss how prolonged cannabis use, such as in the case of CHS or heightened stress, can dysregulate the ECS and affect its modulation of these functions. The review also examines the evidence for the roles of ECS and stress systems' dysfunction in CVS and CHS to better understand the underlying mechanisms of these conditions.
Collapse
Affiliation(s)
- Marieka V. DeVuono
- Department of Anatomy and Cell BiologySchulich School of Medicine & Dentistry, Western UniversityLondonOntarioCanada
| | - Thangam Venkatesan
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal MedicineThe Ohio State University College of MedicineColumbusOhioUSA
| | - Cecilia J. Hillard
- Department of Pharmacology and Toxicology and Neuroscience Research CenterMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
2
|
Sikder MM, Sasaki S, Miki Y, Nagasaki Y, Ohta KI, Hussain Z, Saiga H, Ohmura-Hoshino M, Hoshino K, Ueno M, Okada-Iwabu M, Murakami M, Ueda N, Uyama T. PLAAT5 as an N-acyltransferase responsible for the generation of anti-inflammatory N-acylethanolamines in testis. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159583. [PMID: 39592057 DOI: 10.1016/j.bbalip.2024.159583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 11/10/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
N-Acylethanolamines (NAEs) are a class of lipid mediators that exhibit anti-inflammatory and appetite-suppressive activities. Among them, palmitoylethanolamide (PEA) and arachidonoylethanolamide (AEA) bind to peroxisomal proliferator-activated receptor (PPAR) α and cannabinoid receptor CB1, respectively. N-Acyl-phosphatidylethanolamine (NAPE) as a precursor of NAEs is biosynthesized from membrane phospholipids by N-acyltransferases, which consist of group IVE cytosolic phospholipase A2ε (cPLA2ε) and PLAAT (phospholipase A and acyltransferase) family enzymes. While cPLA2ε is responsible for the production of NAEs not only in specific tissues, including muscle, skin, and the stomach, but also under pathological conditions, such as psoriasis and brain ischemia, the involvement of the PLAAT family in vivo remains unclear. Considering the specific expression of PLAAT5 in testes, we investigated the potential role of PLAAT5 in the formation of NAEs in testes using PLAAT5-deficient (Plaat5-/-) mice. High-performance liquid chromatography coupled with tandem mass spectrometry showed that PLAAT5 deficiency decreased the total level of NAEs by 61 %, with PEA and AEA being reduced by 64 % and 87 %, respectively. Following a treatment with cadmium chloride, an environmental toxin that induces testicular inflammation, the expression of inflammatory genes (Il6, Tnf, and Nos2) in testes was markedly higher in Plaat5-/- mice than in Plaat5+/+ mice, and their expression was attenuated by the administration of PEA and AEA. Furthermore, these anti-inflammatory effects were canceled by a co-treatment with the antagonists of PPARα or CB1. These results suggest that PLAAT5 is responsible for the biosynthesis of anti-inflammatory NAEs in testes.
Collapse
Affiliation(s)
| | - Sumire Sasaki
- Department of Biochemistry, Kagawa University School of Medicine, Kagawa, Japan
| | - Yoshimi Miki
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Nagasaki
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ken-Ichi Ohta
- Department of Anatomy and Neurobiology, Kagawa University School of Medicine, Kagawa, Japan
| | - Zahir Hussain
- Department of Biochemistry, Kagawa University School of Medicine, Kagawa, Japan
| | - Hiroyuki Saiga
- Department of Immunology, Kagawa University School of Medicine, Kagawa, Japan
| | - Mari Ohmura-Hoshino
- Department of Immunology, Kagawa University School of Medicine, Kagawa, Japan; Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Mie, Japan
| | - Katsuaki Hoshino
- Department of Immunology, Kagawa University School of Medicine, Kagawa, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Kagawa University School of Medicine, Kagawa, Japan
| | - Miki Okada-Iwabu
- Department of Biochemistry, Kagawa University School of Medicine, Kagawa, Japan
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Science, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Natsuo Ueda
- Department of Biochemistry, Kagawa University School of Medicine, Kagawa, Japan
| | - Toru Uyama
- Department of Biochemistry, Kagawa University School of Medicine, Kagawa, Japan.
| |
Collapse
|
3
|
Rock EM, Parker LA. The Role of Cannabinoids and the Endocannabinoid System in the Treatment and Regulation of Nausea and Vomiting. Curr Top Behav Neurosci 2024. [PMID: 39739175 DOI: 10.1007/7854_2024_554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Despite using the recommended anti-emetic treatments, control of nausea and vomiting is still an unmet need for cancer patients undergoing chemotherapy treatment. Few properly controlled clinical trials have evaluated the potential of exogenously administered cannabinoids or manipulations of the endogenous cannabinoid (eCB) system to treat nausea and vomiting. In this chapter, we explore the pre-clinical and human clinical trial evidence for the potential of exogenous cannabinoids and manipulations of the eCB system to reduce nausea and vomiting. Although there are limited high-quality human clinical trials, pre-clinical evidence suggests that cannabinoids and manipulations of the eCB system have anti-nausea/anti-emetic potential. The pre-clinical anti-nausea/anti-emetic evidence highlights the need for further evaluation of cannabinoids and manipulations of eCBs and other fatty acid amides in clinical trials.
Collapse
Affiliation(s)
- Erin M Rock
- Department of Psychology and Collaborative Neuroscience Graduate Program, University of Guelph, Guelph, ON, Canada
| | - Linda A Parker
- Department of Psychology and Collaborative Neuroscience Graduate Program, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
4
|
Rock EM, Limebeer CL, Smoum R, Mechoulam R, Parker LA. Evaluation of Sex Differences in the Potential of Δ 9-Tetrahydrocannabinol, Cannabidiol, Cannabidiolic Acid, and Oleoyl Alanine to Reduce Nausea-Induced Conditioned Gaping Reactions in Sprague-Dawley Rats. Cannabis Cannabinoid Res 2023; 8:1060-1068. [PMID: 35984924 DOI: 10.1089/can.2022.0158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Introduction: Cancer patients report nausea as a side effect of their chemotherapy treatment. Using the pre-clinical rodent model of acute nausea-lithium chloride (LiCl)-induced conditioned gaping-our group has demonstrated that exogenous cannabinoids may have antinausea potential. Materials and Methods: With the goal of evaluating the role of sex as a factor in pre-clinical research, we first compared the conditioned gaping reactions produced by varying doses of LiCl in male and female rats using the taste reactivity test (Experiment 1). Results: LiCl produced dose-dependent conditioned gaping similarly in male and female rats with the highest dose (127.2 mg/kg) producing robust conditioned gaping, with this dose used in subsequent experiments. Next, we examined the antinausea potential of THC (Experiment 2), CBD (Experiment 3), cannabidiolic acid (CBDA; Experiment 4) and oleoyl alanine (OlAla; Experiment 5) in both male and female rats. THC, CBD, CBDA, and OlAla dose dependently reduced conditioned gaping in both male and female rats in a similar manner. Conclusions: These results suggest that cannabinoids may be equally effective in treating nausea in both males and females.
Collapse
Affiliation(s)
- Erin M Rock
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, Canada
| | - Cheryl L Limebeer
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, Canada
| | - Reem Smoum
- Institute of Drug Research, Medical Faculty, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Raphael Mechoulam
- Institute of Drug Research, Medical Faculty, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Linda A Parker
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, Canada
| |
Collapse
|
5
|
Chen C, Wang W, Raymond M, Ahmadinejad F, Poklis JL, Em B, Gewirtz DA, Lichtman AH, Li N. Genetic Knockout of Fatty Acid Amide Hydrolase Ameliorates Cisplatin-Induced Nephropathy in Mice. Mol Pharmacol 2023; 103:230-240. [PMID: 36702548 PMCID: PMC10029825 DOI: 10.1124/molpharm.122.000618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/09/2022] [Accepted: 12/21/2022] [Indexed: 01/27/2023] Open
Abstract
Cisplatin is a potent first-line therapy for many solid malignancies, such as breast, ovarian, lung, testicular, and head and neck cancer. However, acute kidney injury (AKI) is a major dose-limiting toxicity in cisplatin therapy, which often hampers the continuation of cisplatin treatment. The endocannabinoid system, consisting of anandamide (AEA) and 2-arachidonoylglycerol and cannabinoid receptors, participates in different kidney diseases. Inhibition of fatty acid amide hydrolase (FAAH), the primary enzyme for the degradation of AEA and AEA-related N-acylethanolamines, elicits anti-inflammatory effects; however, little is known about its role in cisplatin nephrotoxicity. The current study tested the hypothesis that genetic deletion of Faah mitigates cisplatin-induced AKI. Male wild-type C57BL6 (WT) and Faah-/- mice were administered a single dose of intraperitoneal injection of cisplatin (30 mg/kg) and euthanatized 72 hours later. Faah-/- mice showed a reduction of cisplatin-induced blood urea nitrogen, plasma creatinine levels, kidney injury markers, and tubular damage in comparison with WT mice. The renal protection from Faah deletion was associated with enhanced tone of AEA-related N-acylethanolamines (palmitoylethanolamide and oleoylethanolamide), attenuated nuclear factor-κB/p65 activity, DNA damage markers p53 and p21, and decreased expression of the inflammatory cytokine interleukin-1β, as well as infiltration of macrophages and leukocytes in the kidneys. Notably, a selective FAAH inhibitor (PF-04457845) did not interfere with or perturb the antitumor effects of cisplatin in two head and neck squamous cell carcinoma cell lines, HN30 and HN12. Our work highlights that FAAH inactivation prevents cisplatin-induced nephrotoxicity in mice and that targeting FAAH could provide a novel strategy to mitigate cisplatin-induced nephrotoxicity. SIGNIFICANCE STATEMENT: Mice lacking the Faah gene are protected from cisplatin-induced inflammation, DNA damage response, tubular damage, and kidney dysfunction. Inactivation of FAAH could be a potential strategy to mitigate cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Chaoling Chen
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Weili Wang
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Marissa Raymond
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Fereshteh Ahmadinejad
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Justin L Poklis
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Brandon Em
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - David A Gewirtz
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
6
|
Guerra J, Naidoo V, Cacabelos R. Potential effects of cannabinoids on audiovestibular function: A narrative review. Front Pharmacol 2022; 13:1010296. [PMID: 36605398 PMCID: PMC9807921 DOI: 10.3389/fphar.2022.1010296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
The growing interest in the development of drugs that target the endocannabinoid system has extended to conditions that affect the audiovestibular pathway. The expression of cannabinoid (CB) receptors in that pathway has been widely demonstrated, indicating a therapeutic potential for drug development at this level. These medications may be beneficial for conditions such as noise-induced hearing loss, ototoxicity, or various forms of vertigo of central or peripheral origin. The therapeutic targets of interest include natural or synthetic compounds that act as CB1/CB2 receptor agonists/antagonists, and inhibitors of the endocannabinoid-degrading enzymes FAAH and MAGL. Furthermore, genetic variations implicated in the response to treatment and the development of related disorders such as epilepsy or migraine have been identified. Direct methods of administering these medications should be examined beyond the systemic strategy.
Collapse
Affiliation(s)
- Joaquin Guerra
- Neuro-Otolaryngology Unit, EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, Bergondo, Corunna, Spain,*Correspondence: Joaquin Guerra,
| | - Vinogran Naidoo
- Department of Neuroscience, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, Corunna, Spain
| | - Ramon Cacabelos
- Genomic Medicine, EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, Bergondo, Corunna, Spain
| |
Collapse
|
7
|
Bagues A, López-Tofiño Y, Llorente-Berzal Á, Abalo R. Cannabinoid drugs against chemotherapy-induced adverse effects: focus on nausea/vomiting, peripheral neuropathy and chemofog in animal models. Behav Pharmacol 2022; 33:105-129. [PMID: 35045012 DOI: 10.1097/fbp.0000000000000667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Although new drugs are being developed for cancer treatment, classical chemotherapeutic agents are still front-line therapies, despite their frequent association with severe side effects that can hamper their use. Cannabinoids may prevent or palliate some of these side effects. The aim of the present study is to review the basic research which has been conducted evaluating the effects of cannabinoid drugs in the treatment of three important side effects induced by classical chemotherapeutic agents: nausea and vomiting, neuropathic pain and cognitive impairment. Several published studies have demonstrated that cannabinoids are useful in preventing and reducing the nausea, vomits and neuropathy induced by different chemotherapy regimens, though other side effects can occur, such as a reduction of gastrointestinal motility, along with psychotropic effects when using centrally-acting cannabinoids. Thus, peripherally-acting cannabinoids and new pharmacological options are being investigated, such as allosteric or biased agonists. Additionally, due to the increase in the survival of cancer patients, there are emerging data that demonstrate an important cognitive deterioration due to chemotherapy, and because the cannabinoid drugs have a neuroprotective effect, they could be useful in preventing chemotherapy-induced cognitive impairment (as demonstrated through studies in other neurological disorders), but this has not yet been tested. Thus, although cannabinoids seem a promising therapeutic approach in the treatment of different side effects induced by chemotherapeutic agents, future research will be necessary to find pharmacological options with a safer profile. Moreover, a new line of research awaits to be opened to elucidate their possible usefulness in preventing cognitive impairment.
Collapse
Affiliation(s)
- Ana Bagues
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- High Performance Research Group in Experimental Pharmacology (PHARMAKOM-URJC)
- Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Yolanda López-Tofiño
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System NeuGut-URJC
| | - Álvaro Llorente-Berzal
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland
- Centre for Pain Research and Galway Neuroscience Centre, NCBES, National University of Ireland, Galway, Ireland
| | - Raquel Abalo
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), Alcorcón
- Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System NeuGut-URJC
- Grupo de Trabajo de Ciencias Básicas en Dolor y Analgesia de la Sociedad Española del Dolor, Madrid, Spain
| |
Collapse
|
8
|
Genovese T, Duranti A, D’Amico R, Fusco R, Impellizzeri D, Peritore AF, Crupi R, Gugliandolo E, Cuzzocrea S, Di Paola R, Siracusa R, Cordaro M. Fatty Acid Amide Hydrolase (FAAH) Inhibition Plays a Key Role in Counteracting Acute Lung Injury. Int J Mol Sci 2022; 23:2781. [PMID: 35269926 PMCID: PMC8910911 DOI: 10.3390/ijms23052781] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/26/2022] [Accepted: 03/01/2022] [Indexed: 02/01/2023] Open
Abstract
Acute lung injury (ALI) is a group of lung illnesses characterized by severe inflammation, with no treatment. The fatty acid amide hydrolase (FAAH) enzyme is an integral membrane protein responsible for the hydrolysis of the main endocannabinoids, such as anandamide (AEA). In pre-clinical pain and inflammation models, increasing the endogenous levels of AEA and other bioactive fatty acid amides (FAAs) via genetic deletion or the pharmacological inhibition of FAAH produces many analgesic benefits in several different experimental models. To date, nobody has investigated the role of FAAH inhibition on an ALI mouse model. Mice were subjected to a carrageenan injection and treated orally 1 h after with the FAAH inhibitor URB878 dissolved in a vehicle consisting of 10% PEG-400, 10% Tween-80 and 80% saline at different doses: The inhibition of FAAH activity was able to counteract not only the CAR-induced histological alteration, but also the cascade of related inflammatory events. URB878 clears the way for further studies based on FAAH inhibition in acute lung pathologies.
Collapse
Affiliation(s)
- Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino, Carlo Bo Piazza del Rinascimento 6, 61029 Urbino, Italy;
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Roberta Fusco
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (R.C.); (E.G.)
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (R.C.); (E.G.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (R.C.); (E.G.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres 31, 98166 Messina, Italy; (T.G.); (R.D.); (D.I.); (A.F.P.); (R.S.)
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| |
Collapse
|
9
|
Rock EM, Limebeer CL, Smoum R, Mechoulam R, Parker LA. Effect of oleoyl glycine and oleoyl alanine on lithium chloride induced nausea in rats and vomiting in shrews. Psychopharmacology (Berl) 2022; 239:377-383. [PMID: 34676441 DOI: 10.1007/s00213-021-06005-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
RATIONALE The fatty acid amide oleoyl glycine (OlGly) and its more stable methylated form oleoyl alanine (OlAla) reduce naloxone-precipitated morphine withdrawal (MWD)-induced conditioned gaping (nausea) responses in rats. In addition, OlGly has been shown to reduce lithium chloride (LiCl)-induced conditioned gaping in rats and vomiting in Suncus murinus (house musk shrews). OBJECTIVES Here, we compared the potential of these fatty acid amides to maintain their anti-nausea/anti-emetic effect over a delay. The following experiments examined the potential of a wider dose range of OlGly and OlAla to interfere with (1) LiCl-induced conditioned gaping in rats and (2) LiCl-induced vomiting in shrews, when administered 20 or 70 min prior to illness. RESULTS OlAla (1, 5, 20 mg/kg) reduced LiCl-induced conditioned gaping, with OlGly only effective at the high dose (20 mg/kg), with no effect of pretreatment delay time. At the high dose of 20 mg/kg, OlGly increased passive drips during conditioning suggesting a sedative effect. In shrews, both OlGly and OlAla (1, 5 mg/kg) suppressed LiCl-induced vomiting, with no effect of pretreatment delay. OlAla more effectively suppressed vomiting, with OlAla (5 mg/kg) also increasing the latency to the first vomiting reaction. CONCLUSIONS OlAla was more effective than OlGly in reducing both LiCl-induced gaping in rats and LiCl-induced vomiting in shrews. These findings provide further evidence that these fatty acid amides may be useful treatments for nausea and vomiting, with OlAla demonstrating superior efficacy.
Collapse
Affiliation(s)
- Erin M Rock
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, N1G2W1, Canada
| | - Cheryl L Limebeer
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, N1G2W1, Canada
| | - Reem Smoum
- Institute of Drug Research, Medical Facility, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Raphael Mechoulam
- Institute of Drug Research, Medical Facility, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Linda A Parker
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, N1G2W1, Canada.
| |
Collapse
|
10
|
Abstract
Cannabis use has been increasing in the United States and throughout the world. It is derived from one of the earliest plants cultivated by humans - Cannabis sativa. Cannabis (also called marijuana) is the most commonly used psychoactive substance worldwide. The cannabis plant has more than 400 chemicals, of which more than 100 cannabinoids (such as cannabigerol, cannabidiol, and cannabinol) have been identified. The endocannabinoid system (ECS) plays an essential role in the effects of cannabis on end organs. Although cannabis use has been reported for many decades, some of its unique adverse effects of nausea, vomiting, and abdominal pain, termed as cannabis hyperemesis syndrome (CHS), were noted recently. The legal status of cannabis in the United States has been rapidly changing from state to state. The incidence of CHS is expected to rise with rising access to cannabis in the United States. Furthermore, CHS is frequently underdiagnosed due to a lack of uniform criteria, subjective nature of symptoms, and overlap with cyclical vomiting syndrome (CVS). Understanding the ECS and its role in biphasic response (proemetic and antiemetic) of CHS is critical to explain its pathophysiology. As the use of cannabis increases globally, awareness of CHS is warranted for early recognition and prompt treatment to avoid complications. We describe the putative mechanism of CHS with an overview of the clinical features in these patients. Furthermore, we highlight the differences between CHS and CVS with important differentials to consider. We provide a narrative update on the current evidence on CHS pathophysiology, diagnosis, treatment, and identifying research gaps.
Collapse
Affiliation(s)
- Abhilash Perisetti
- Department of Gastroenterology and Hepatology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Hemant Goyal
- The Wright Center for Graduate Medical Education, Scranton, Pennsylvania, USA
| |
Collapse
|
11
|
Zhong W, Darmani NA. The HCN Channel Blocker ZD7288 Induces Emesis in the Least Shrew ( Cryptotis parva). Front Pharmacol 2021; 12:647021. [PMID: 33995059 PMCID: PMC8117105 DOI: 10.3389/fphar.2021.647021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/12/2021] [Indexed: 12/02/2022] Open
Abstract
Subtypes (1-4) of the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are widely expressed in the central and peripheral nervous systems, as well as the cells of smooth muscles in many organs. They mainly serve to regulate cellular excitability in these tissues. The HCN channel blocker ZD7288 has been shown to reduce apomorphine-induced conditioned taste aversion on saccharin preference in rats suggesting potential antinausea/antiemetic effects. Currently, in the least shew model of emesis we find that ZD7288 induces vomiting in a dose-dependent manner, with maximal efficacies of 100% at 1 mg/kg (i.p.) and 83.3% at 10 µg (i.c.v.). HCN channel subtype (1-4) expression was assessed using immunohistochemistry in the least shrew brainstem dorsal vagal complex (DVC) containing the emetic nuclei (area postrema (AP), nucleus tractus solitarius and dorsal motor nucleus of the vagus). Highly enriched HCN1 and HCN4 subtypes are present in the AP. A 1 mg/kg (i.p.) dose of ZD7288 strongly evoked c-Fos expression and ERK1/2 phosphorylation in the shrew brainstem DVC, but not in the in the enteric nervous system in the jejunum, suggesting a central contribution to the evoked vomiting. The ZD7288-evoked c-Fos expression exclusively occurred in tryptophan hydroxylase 2-positive serotonin neurons of the dorsal vagal complex, indicating activation of serotonin neurons may contribute to ZD7288-induced vomiting. To reveal its mechanism(s) of emetic action, we evaluated the efficacy of diverse antiemetics against ZD7288-evoked vomiting including the antagonists/inhibitors of: ERK1/2 (U0126), L-type Ca2+ channel (nifedipine); store-operated Ca2+ entry (MRS 1845); T-type Ca2+ channel (Z944), IP3R (2-APB), RyR receptor (dantrolene); the serotoninergic type 3 receptor (palonosetron); neurokinin 1 receptor (netupitant), dopamine type 2 receptor (sulpride), and the transient receptor potential vanilloid 1 receptor agonist, resiniferatoxin. All tested antiemetics except sulpride attenuated ZD7288-evoked vomiting to varying degrees. In sum, ZD7288 has emetic potential mainly via central mechanisms, a process which involves Ca2+ signaling and several emetic receptors. HCN channel blockers have been reported to have emetic potential in the clinic since they are currently used/investigated as therapeutic candidates for cancer therapy related- or unrelated-heart failure, pain, and cognitive impairment.
Collapse
Affiliation(s)
| | - N. A. Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
12
|
Bashashati M, Leishman E, Bradshaw H, Sigaroodi S, Tatro E, Bright T, McCallum R, Sarosiek I. Plasma endocannabinoids and cannabimimetic fatty acid derivatives are altered in gastroparesis: A sex- and subtype-dependent observation. Neurogastroenterol Motil 2021; 33:e13961. [PMID: 32779297 PMCID: PMC8018519 DOI: 10.1111/nmo.13961] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/23/2020] [Accepted: 07/20/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Gastroparesis (GP) is a motility disorder of the stomach presenting with upper gastrointestinal symptoms in the setting of delayed gastric emptying. Endocannabinoids are involved in the regulation of GI function including motility. However, their role in the pathophysiology of GP has not been sufficiently investigated. Our goal was to compare the circulating levels of endocannabinoids and cannabimimetic fatty acid derivatives in GP versus control subjects. METHODS The study compared plasma concentrations of endocannabinoids and their lipoamine and 2-acyl glycerol congeners, measured by high-pressure liquid chromatography/tandem mass spectrometry (HPLC-MS-MS), in adult patients with diabetic gastroparesis (DM-GP; n = 24; n = 16 female), idiopathic gastroparesis (ID-GP; n = 19; n = 11 female), diabetic patients without GP (DM; n = 19; n = 10 female), and healthy controls (HC; n = 18; n = 10 female). Data, presented as mean ± SEM, were analyzed with ANOVA (Sidak post hoc). KEY RESULTS Endocannabinoids anandamide (AEA: 0.5 ± 0.1 nMol/L) and 2-arachidonoyl glycerol (2-AG: 2.6 ± 0.7 nMol/L) were significantly lower in female DM-GP patients vs. DM females (AEA: 2.5 ± 0.7 nMol/L and 2-AG: 9.4 ± 3.3 nMol/L). Other monoacylglycerols including 2-palmitoyl glycerol and 2-oleoyl glycerol were also lower in female DM-GP patients compared to DM females. No changes were observed in men. CONCLUSIONS & INFERENCES Endocannabinoids and other fatty acid derivatives with cannabimimetic properties are reduced in female DM-GP patients. Since GP, particularly with diabetic etiology, is more prevalent among women and since cannabinoids are antiemetic, this decrease in levels may contribute to symptom development in these subjects. Targeting the endocannabinoid system may be a future therapeutic option in DM-GP patients.
Collapse
Affiliation(s)
- Mohammad Bashashati
- Division of Gastroenterology, Department of Internal Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Emma Leishman
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Heather Bradshaw
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Solmaz Sigaroodi
- Division of Gastroenterology, Department of Internal Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA,Department of Kinesiology, College of Health Sciences, University of Texas at El Paso, El Paso, TX, United States
| | - Eric Tatro
- Division of Gastroenterology, Department of Internal Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Tamis Bright
- Division of Endocrinology, Department of Internal Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Richard McCallum
- Division of Gastroenterology, Department of Internal Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Irene Sarosiek
- Division of Gastroenterology, Department of Internal Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| |
Collapse
|
13
|
Perisetti A, Gajendran M, Dasari CS, Bansal P, Aziz M, Inamdar S, Tharian B, Goyal H. Cannabis hyperemesis syndrome: an update on the pathophysiology and management. Ann Gastroenterol 2020; 33:571-578. [PMID: 33162734 PMCID: PMC7599351 DOI: 10.20524/aog.2020.0528] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Cannabis hyperemesis syndrome (CHS) is a form of functional gut-brain axis disorder characterized by bouts of episodic nausea and vomiting worsened by cannabis intake. It is considered as a variant of cyclical vomiting syndrome seen in cannabis users especially characterized by compulsive hot bathing/showers to relieve the symptoms. CHS was reported for the first time in 2004, and since then, an increasing number of cases have been reported. With cannabis use increasing throughout the world as the threshold for legalization becomes lower, its user numbers are expected to rise over time. Despite this trend, a strict criterion for the diagnosis of CHS is lacking. Early recognition of CHS is essential to prevent complications related to severe volume depletion. The recent body of research recognizes that patients with CHS impose a burden on the healthcare systems. Understanding the pathophysiology of the endocannabinoid system (ECS) remains central in explaining the clinical features and potential drug targets for the treatment of CHS. The frequency and prevalence of CHS change in accordance with the doses of tetrahydrocannabinol and other cannabinoids in various formulations of cannabis. CHS is unique in presentation, because of the cannabis’s biphasic effect as anti-emetic at low doses and pro-emetic at higher doses, and the association with pathological hot water bathing. In this narrative review, we elaborate on the role of the ECS, its management, and the identification of gaps in our current knowledge of CHS to further enhance its understanding in the future.
Collapse
Affiliation(s)
- Abhilash Perisetti
- Department of Gastroenterology and Hepatology, University of Arkansas for Medical Sciences, Little Rock, AR (Abhilash Perisetti)
| | - Mahesh Gajendran
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso (Mahesh Gajendran)
| | - Chandra Shekhar Dasari
- Department of Gastroenterology and Hepatology, Kansas City VA Medical Center (Chandra Shekhar Dasari)
| | - Pardeep Bansal
- Division of Gastroenterology, Moses Taylor Hospital and Reginal Hospital of Scranton, Scranton, PA (Pardeep Bansal)
| | - Muhammad Aziz
- Department of Internal Medicine, The University of Toledo, Toledo, OH (Muhammad Aziz)
| | - Sumant Inamdar
- Endoscopy Fellowship, University of Arkansas for Medical Sciences, Little Rock, AR (Sumant Inamdar, Benjamin Tharian)
| | - Benjamin Tharian
- Endoscopy Fellowship, University of Arkansas for Medical Sciences, Little Rock, AR (Sumant Inamdar, Benjamin Tharian)
| | - Hemant Goyal
- The Wright Center for Graduate Medical Education, Scranton, PA (Hemant Goyal), USA
| |
Collapse
|
14
|
Wooldridge LM, Ji L, Liu Y, Nikas SP, Makriyannis A, Bergman J, Kangas BD. Antiemetic Effects of Cannabinoid Agonists in Nonhuman Primates. J Pharmacol Exp Ther 2020; 374:462-468. [PMID: 32561684 PMCID: PMC7445860 DOI: 10.1124/jpet.120.265710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
Attenuating emesis elicited by both disease and medical treatments of disease remains a critical public health challenge. Although cannabinergic medications have been used in certain treatment-resistant populations, Food and Drug Administration-approved cannabinoid antiemetics are associated with undesirable side effects, including cognitive disruption, that limit their prescription. Previous studies have shown that a metabolically stable analog of the endocannabinoid anandamide, methanandamide (mAEA), may produce lesser cognitive disruption than that associated with the primary psychoactive constituent in cannabis, Δ9-tetrahydrocannabinol (Δ9-THC), raising the possibility that endocannabinoids may offer a therapeutic advantage over currently used medications. The present studies were conducted to evaluate this possibility by comparing the antiemetic effects of Δ9-THC (0.032-0.1 mg/kg) and mAEA (3.2-10.0 mg/kg) against nicotine- and lithium chloride (LiCl)-induced emesis and prodromal hypersalivation in squirrel monkeys. Pretreatment with 0.1 mg/kg Δ9-THC blocked nicotine-induced emesis and reduced hypersalivation in all subjects and blocked LiCl-induced emesis and reduced hypersalivation in three of four subjects. Pretreatment with 10 mg/kg mAEA blocked nicotine-induced emesis in three of four subjects and LiCl-induced emesis in one of four subjects and reduced both nicotine- and LiCl-induced hypersalivation. Antiemetic effects of Δ9-THC and mAEA were reversed by rimonabant pretreatment, providing verification of cannabinoid receptor type 1 mediation. These studies systematically demonstrate for the first time the antiemetic effects of cannabinoid agonists in nonhuman primates. Importantly, although Δ9-THC produced superior antiemetic effects, the milder cognitive effects of mAEA demonstrated in previous studies suggest that it may provide a favorable treatment option under clinical circumstances in which antiemetic efficacy must be balanced against side effect liability. SIGNIFICANCE STATEMENT: Emesis has significant evolutionary value as a defense mechanism against ingested toxins; however, it is also one of the most common adverse symptoms associated with both disease and medical treatments of disease. The development of improved antiemetic pharmacotherapies has been impeded by a paucity of animal models. The present studies systematically demonstrate for the first time the antiemetic effects of the phytocannabinoid Δ9-tetrahydrocannabinol and endocannabinoid analog methanandamide in nonhuman primates.
Collapse
Affiliation(s)
- Lisa M Wooldridge
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (J.B., B.D.K.); Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts (L.M.W., J.B., B.D.K.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., Y.L., S.P.N., A.M.)
| | - Lipin Ji
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (J.B., B.D.K.); Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts (L.M.W., J.B., B.D.K.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., Y.L., S.P.N., A.M.)
| | - Yingpeng Liu
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (J.B., B.D.K.); Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts (L.M.W., J.B., B.D.K.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., Y.L., S.P.N., A.M.)
| | - Spyros P Nikas
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (J.B., B.D.K.); Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts (L.M.W., J.B., B.D.K.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., Y.L., S.P.N., A.M.)
| | - Alexandros Makriyannis
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (J.B., B.D.K.); Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts (L.M.W., J.B., B.D.K.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., Y.L., S.P.N., A.M.)
| | - Jack Bergman
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (J.B., B.D.K.); Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts (L.M.W., J.B., B.D.K.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., Y.L., S.P.N., A.M.)
| | - Brian D Kangas
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (J.B., B.D.K.); Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts (L.M.W., J.B., B.D.K.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., Y.L., S.P.N., A.M.)
| |
Collapse
|
15
|
Abstract
Cannabinoid receptors, endocannabinoids and the enzymes responsible for their biosynthesis and degradation constitute the endocannabinoid system. In recent decades, the endocannabinoid system has attracted considerable interest as a potential therapeutic target in numerous pathological conditions. Its involvement in several physiological processes is well known, such as in energy balance, appetite stimulation, blood pressure, pain modulation, embryogenesis, nausea and vomiting control, memory, learning and immune response, among others, as well as in pathological conditions where it exerts a protective role in the development of certain disorders. As a result, it has been reported that changes in endocannabinoid levels may be related to neurological diseases such as Parkinson's disease, Huntington's disease, Alzheimer's disease and multiple sclerosis, as well as anorexia and irritable bowel syndrome. Alterations in the endocannabinoid system have also been associated with cancer, affecting the growth, migration and invasion of some tumours. Cannabinoids have been tested in several cancer types, including brain, breast and prostate cancers. Cannabinoids have shown promise as analgesics for the treatment of both inflammatory and neuropathic pain. There is also evidence for a role of the endocannabinoid system in the control of emotional states, and cannabinoids could prove useful in decreasing and palliating post-traumatic stress disorder symptoms and anxiolytic disorders. The role of the endocannabinoid system in addictions has also been examined, and cannabinoids have been postulated as alternative and co-adjuvant treatments in some abuse syndromes, mainly in ethanol and opioid abuses. The expression of the endocannabinoid system in the eye suggests that it could be a potential therapeutic target for eye diseases. Considering the importance of the endocannabinoid system and the therapeutic potential of cannabinoids in this vast number of medical conditions, several clinical studies with cannabinoid-based medications are ongoing. In addition, some cannabinoid-based medications have already been approved in various countries, including nabilone and dronabinol capsules for the treatment of nausea and vomiting associated with chemotherapy, dronabinol capsules for anorexia, an oral solution of dronabinol for both vomiting associated with chemotherapy and anorexia, a Δ9-tetrahydrocannabinol/cannabidiol oromucosal spray for pain related to cancer and for spasticity and pain associated with multiple sclerosis, and an oral solution of cannabidiol for Dravet and Lennox-Gastaut syndromes. Here, we review the available efficacy, safety and tolerability data for cannabinoids in a range of medical conditions.
Collapse
Affiliation(s)
- Ana Isabel Fraguas-Sánchez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramón y Cajal s/n, 28040 , Madrid, Spain
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Plaza Ramón y Cajal s/n, 28040 , Madrid, Spain. .,Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 , Madrid, Spain.
| |
Collapse
|
16
|
Ono H. [Hypothermic Action of Oseltamivir Not Dependent on Its Anti-influenza Virus Action]. YAKUGAKU ZASSHI 2019; 139:767-781. [PMID: 31061347 DOI: 10.1248/yakushi.18-00191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although the anti-influenza virus drug oseltamivir ameliorates the fever of influenza, adverse events related to its hypothermic effect have been reported. We found that oseltamivir causes dose-dependent hypothermia in normal mice, and have been studying the pharmacological mechanisms responsible for 12 years. Oseltamivir blocks nicotinic cholinergic transmission at sympathetic ganglia and reduces sympathetic modulation of brown adipose tissue (BAT), a heat generator. Oseltamivir was found to target the ion channels of nicotinic acetylcholine receptors, as demonstrated by patch clamp experiments with cells expressing the human α3β4 nicotinic receptor. Metabolized oseltamivir carboxylate, which inhibits the influenza virus neuraminidase, did not elicit hypothermia and ion channel suppression. Body temperature was decreased by intracerebroventricular administration of oseltamivir. Because this hypothermic effect was inhibited by dopamine D2 receptor blockade, it was suggested that oseltamivir centrally stimulates the D2 receptor. In Japan, the package inserts for oseltamivir and amantadine indicate very similar adverse neuropsychiatric reactions for the two drugs (abnormal behavior, consciousness disturbance, convulsion, delirium, delusion, hallucination). A literature search revealed that in some previous studies, oseltamivir and amantadine were shown to block the ion channel systems and activate the dopaminergic nervous system via several mechanisms. Therefore the similarity of the adverse reactions elicited by oseltamivir and amantadine was considered attributable to their similar pharmacological effects.
Collapse
Affiliation(s)
- Hideki Ono
- Laboratory of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, Musashino University.,Research Institute of Pharmaceutical Sciences, Musashino University
| |
Collapse
|
17
|
Wooldridge LM, Kangas BD. An assay of drug-induced emesis in the squirrel monkey (Saimiri sciureus). J Med Primatol 2019; 48:236-243. [PMID: 30968960 DOI: 10.1111/jmp.12411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/07/2019] [Accepted: 03/10/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Emesis has significant evolutionary value as a defense mechanism against ingested toxins; however, it is also one of the most common adverse symptoms associated with both disease and medical treatments of disease. The development of improved antiemetic pharmacotherapies has been impeded by a shortage of animal models. METHODS The present studies characterized the responses of the squirrel monkey to pharmacologically diverse emetic drugs. Subjects were administered nicotine (0.032-0.56 mg/kg), lithium chloride (150-250 mg/kg), arecoline (0.01-0.32 mg/kg), or apomorphine (0.032-0.32 mg/kg) and observed for emesis and prodromal hypersalivation. RESULTS Nicotine rapidly produced emesis and hypersalivation. Lithium chloride produced emesis with a longer time course without dose-dependent hypersalivation. Arecoline produced hypersalivation but not emesis. Apomorphine failed to produce emesis or hypersalivation. CONCLUSIONS The squirrel monkey is sensitive to drug-induced emesis by a variety of pharmacological mechanisms and is well-positioned to examine antiemetic efficacy and clinically important side effects of candidate antiemetic pharmacotherapies.
Collapse
Affiliation(s)
| | - Brian D Kangas
- Behavioral Biology Program, McLean Hospital, Belmont, Massachusetts.,Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Romano C, Dipasquale V, Rybak A, Comito D, Borrelli O. An overview of the clinical management of cyclic vomiting syndrome in childhood. Curr Med Res Opin 2018; 34:1785-1791. [PMID: 29484898 DOI: 10.1080/03007995.2018.1445983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 02/21/2018] [Accepted: 02/23/2018] [Indexed: 01/03/2023]
Abstract
This narrative review provides an update on cyclic vomiting syndrome pathogenesis, diagnosis and management, based upon studies published after the 2008 North American Society for Pediatric Gastroenterology, Hepatology and Nutrition (NASPGHAN) official recommendations. The review began with a comprehensive PubMed/Medline search for "cyclic vomiting syndrome", "periodic syndromes" and "pediatrics" from 2000 up to October 2017. Additional papers were identified by reviewing the re-ference lists of retrieved publications. Cyclic vomiting syndrome is a severe, debilitating disorder of the brain-gut axis with unclear pathogenesis, that significantly affects long-term quality of life of affected children and their families. The 2008 NASPGHAN recommendations defined the major clinical, diagnostic and therapeutic peculiarities. Over the last 10 years, advancements in pathogenesis and diagnostic criteria have been made, and new prophylactic and therapeutic strategies have been proposed. These aspects are discussed in this manuscript. For the pediatrician, the major aim is to have early clinical suspicion to avoid diagnostic delay and to start adequate, phase-related, symptom-tailored management.
Collapse
Affiliation(s)
- Claudio Romano
- a Division of Childhood Gastroenterology and Cystic Fibrosis, Department of Human Pathology in Adulthood and Childhood "G. Barresi" , University of Messina , Italy
| | - Valeria Dipasquale
- a Division of Childhood Gastroenterology and Cystic Fibrosis, Department of Human Pathology in Adulthood and Childhood "G. Barresi" , University of Messina , Italy
| | - Anna Rybak
- b Division of Neurogastroenterology and Motility, Department of Gastroenterology , Great Ormond Street Hospital and UCL , London , UK
| | - Donatella Comito
- a Division of Childhood Gastroenterology and Cystic Fibrosis, Department of Human Pathology in Adulthood and Childhood "G. Barresi" , University of Messina , Italy
| | - Osvaldo Borrelli
- b Division of Neurogastroenterology and Motility, Department of Gastroenterology , Great Ormond Street Hospital and UCL , London , UK
| |
Collapse
|
19
|
Abstract
Humans swallow a great variety and often large amounts of chemicals as nutrients, incidental food additives and contaminants, drugs, and inhaled particles and chemicals, thus exposing the gastrointestinal tract to many potentially toxic substances. It serves as a barrier in many cases to protect other components of the body from such substances and infections. Fortunately, the gastrointestinal tract is remarkably robust and generally is able to withstand multiple daily assaults by the chemicals to which it is exposed. Some chemicals, however, can affect one or more aspects of the gastrointestinal tract to produce abnormal events that reflect toxicity. It is the purpose of this chapter to evaluate the mechanisms by which toxic chemicals produce their deleterious effects and to determine the consequences of the toxicity on integrity of gastrointestinal structure and function. Probably because of the intrinsic ability of the gastrointestinal tract to resist toxic chemicals, there is a paucity of data regarding gastrointestinal toxicology. It is therefore necessary in many cases to extrapolate toxic mechanisms from infectious processes, inflammatory conditions, ischemia, and other insults in addition to more conventional chemical sources of toxicity.
Collapse
|
20
|
Bhandari S, Venkatesan T. Novel Treatments for Cyclic Vomiting Syndrome: Beyond Ondansetron and Amitriptyline. ACTA ACUST UNITED AC 2016; 14:495-506. [PMID: 27757817 DOI: 10.1007/s11938-016-0114-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT Cyclic vomiting syndrome (CVS) is a chronic functional gastrointestinal disorder that is characterized by episodic nausea and vomiting. Initially thought to only affect children, CVS in adults was often misdiagnosed with significant delays in therapy. Over the last decade, there has been a considerable increase in recognition of CVS in adults but there continues to be a lack of knowledge about management of this disorder. This paper seeks to provide best practices in the treatment of CVS and also highlight some novel therapies that have the potential in better treating this disorder in the future. Due to the absence of randomized control trials, we provide recommendations based on review of the available literature and expert consensus on the therapy of CVS. This paper will discuss prophylactic and abortive therapy and general measures used to treat an episode of CVS and also discuss pathophysiology as it pertains to novel therapy. Recent recognition of the association of chronic marijuana use with cyclic vomiting has led to the possibility of a new diagnosis called "Cannabinoid Hyperemesis Syndrome," which is indistinguishable from CVS. The treatment for this purported condition is abstinence from marijuana despite scant evidence that marijuana use is causative. Hence, this review will also discuss emerging data on the role for the endocannabinoid system in CVS and therapeutic agents targeting the endocannabinoid system, which offer the potential of transforming the care of these patients.
Collapse
Affiliation(s)
- Sanjay Bhandari
- Department of General Internal Medicine, Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA
| | - Thangam Venkatesan
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, 9200 W. Wisconsin Ave, Milwaukee, WI, 53226, USA.
| |
Collapse
|
21
|
Venkatesan T, Zadvornova Y, Raff H, Hillard CJ. Endocannabinoid-related lipids are increased during an episode of cyclic vomiting syndrome. Neurogastroenterol Motil 2016; 28:1409-18. [PMID: 27098832 PMCID: PMC5002231 DOI: 10.1111/nmo.12843] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 03/24/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND The endocannabinoid system and the hypothalamic-pituitary-adrenal axis are important neuromodulators of nausea and vomiting. This led us to hypothesize that patients with cyclic vomiting syndrome (CVS) have lower serum endocannabinoids (eCBs) and higher salivary cortisol and alpha amylase. METHODS Serum eCBs and related lipids, N-oleoylethanolamine (OEA) and N-palmitoylethanolamide (PEA), and salivary cortisol, and alpha amylase (index of sympathetic nervous system activity) were measured in 22 CVS patients (age 40 ± 11, female = 17) in the well and sick phases and 12 matched controls (age 37 ± 12, female = 10). KEY RESULTS Contrary to our hypothesis, serum concentrations of the eCBs were not different among the study groups. However, serum concentrations of OEA and PEA were significantly higher during the sick than well phase in CVS patients (p = 0.001 and p = 0.04). There were positive correlations between serum PEA and nausea scores in the sick phase (Pearson's rho = 0.48, p = 0.036) and between serum OEA and poor sleep quality in patients (Pearson's rho = 0.7, p = 0.0005). Salivary cortisol and alpha amylase were not different between patients and controls, but subgroup analysis revealed that both were significantly higher in marijuana users compared to non-users during the sick phase (p = 0.04 and 0.03, respectively). CONCLUSIONS & INFERENCES These data demonstrate that eCB-related lipids, OEA and PEA, are mobilized in the sick phase of CVS and are positively correlated with several of the symptoms of a CVS episode. These data also suggest the hypothesis that chronic marijuana use results in enhanced stress responses during CVS.
Collapse
Affiliation(s)
- Thangam Venkatesan
- Division of Gastroenterology and Hepatology, Department of Medicine, 9200 W. Wisconsin Ave., Milwaukee, WI 53226
| | - Yelena Zadvornova
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, 9200 W. Wisconsin Ave., Milwaukee WI 53226, Telephone: 414-955-7095,
| | - Hershel Raff
- Departments of Medicine, Surgery, and Physiology, Medical College of Wisconsin, Director, Endocrine Research Laboratory, Aurora St. Luke's Medical Center - Aurora Research Institute, 2801 W KK River Pkwy. Suite 245, Milwaukee WI 53215, Telephone: (414) 649-6411
- Fax: (414) 649-5747, or
| | - Cecilia J. Hillard
- Director of the Neuroscience Research Center and Professor of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, Telephone: (414) 955-8493 Phone / Fax: (414) 955-6057,
| |
Collapse
|
22
|
Rock EM, Parker LA. Cannabinoids As Potential Treatment for Chemotherapy-Induced Nausea and Vomiting. Front Pharmacol 2016; 7:221. [PMID: 27507945 PMCID: PMC4960260 DOI: 10.3389/fphar.2016.00221] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/11/2016] [Indexed: 12/20/2022] Open
Abstract
Despite the advent of classic anti-emetics, chemotherapy-induced nausea is still problematic, with vomiting being somewhat better managed in the clinic. If post-treatment nausea and vomiting are not properly controlled, anticipatory nausea—a conditioned response to the contextual cues associated with illness-inducing chemotherapy—can develop. Once it develops, anticipatory nausea is refractive to current anti-emetics, highlighting the need for alternative treatment options. One of the first documented medicinal uses of Δ9-tetrahydrocannabinol (Δ9-THC) was for the treatment of chemotherapy-induced nausea and vomiting (CINV), and recent evidence is accumulating to suggest a role for the endocannabinoid system in modulating CINV. Here, we review studies assessing the therapeutic potential of cannabinoids and manipulations of the endocannabinoid system in human patients and pre-clinical animal models of nausea and vomiting.
Collapse
Affiliation(s)
- Erin M Rock
- Department of Psychology and Collaborative Neuroscience Graduate Program, University of Guelph Guelph, ON, Canada
| | - Linda A Parker
- Department of Psychology and Collaborative Neuroscience Graduate Program, University of Guelph Guelph, ON, Canada
| |
Collapse
|
23
|
Kangas BD, Leonard MZ, Shukla VG, Alapafuja SO, Nikas SP, Makriyannis A, Bergman J. Comparisons of Δ9-Tetrahydrocannabinol and Anandamide on a Battery of Cognition-Related Behavior in Nonhuman Primates. J Pharmacol Exp Ther 2016; 357:125-33. [PMID: 26826191 PMCID: PMC4809315 DOI: 10.1124/jpet.115.228189] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 01/27/2016] [Indexed: 11/22/2022] Open
Abstract
The primary psychoactive ingredient of marijuana, Δ(9)-tetrahydrocannabinol (Δ(9)-THC), has medicinal value but also produces unwanted deleterious effects on cognitive function, promoting the search for improved cannabinergic therapeutics. The present studies used a battery of touchscreen procedures in squirrel monkeys to compare the effects of different types of cannabinergic drugs on several measures of performance including learning (repeated acquisition), cognitive flexibility (discrimination reversal), short-term memory (delayed matching-to-sample), attention (psychomotor vigilance), and motivation (progressive ratio). Drugs studied included the cannabinoid agonist Δ(9)-THC, fatty acid amide hydrolase (FAAH) inhibitor cyclohexylcarbamic acid 3-carbamoylbiphenyl-3-yl ester (URB597), and endocannabinoid anandamide and its stable synthetic analog methanandamide [(R)-(+)-arachidonyl-1'-hydroxy-2'-propylamide]. The effects of Δ(9)-THC and anandamide after treatment with the cannabinoid receptor type 1 inverse agonist/antagonist rimonabant [5-(4-chlorophenyl)-1-(2,4-dichloro-phenyl)-4-methyl-N-(piperidin-1-yl)-1Hpyrazole-3-carboxamide] and the FAAH inhibitor URB597, respectively, also were examined. The results showed the following: 1) Δ(9)-THC produced dose-related impairments of discrimination-based cognitive behavior with potency that varied across tasks (discriminative capability < learning < flexibility < short-term memory); 2) anandamide alone and URB597 alone were without effect on all endpoints; 3) anandamide following URB597 pretreatment and methanandamide had negligible effects on discriminative capability, learning, and reversal, but following large doses affected delayed matching-to-sample performance in some subjects; 4) all drugs, except anandamide and URB597, disrupted attention; and 5) progressive ratio breakpoints were generally unaffected by all drugs tested, suggesting little to no effect on motivation. Taken together, these data indicate that metabolically stable forms of anandamide may have lesser adverse effects on cognitive functions than Δ(9)-THC, possibly offering a therapeutic advantage in clinical settings.
Collapse
Affiliation(s)
- Brian D Kangas
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (B.D.K., J.B.); Preclinical Pharmacology Laboratory, McLean Hospital, Belmont, Massachusetts (B.D.K., M.Z.L., J.B.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (V.G.S., S.O.A., S.P.N., A.M.)
| | - Michael Z Leonard
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (B.D.K., J.B.); Preclinical Pharmacology Laboratory, McLean Hospital, Belmont, Massachusetts (B.D.K., M.Z.L., J.B.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (V.G.S., S.O.A., S.P.N., A.M.)
| | - Vidyanand G Shukla
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (B.D.K., J.B.); Preclinical Pharmacology Laboratory, McLean Hospital, Belmont, Massachusetts (B.D.K., M.Z.L., J.B.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (V.G.S., S.O.A., S.P.N., A.M.)
| | - Shakiru O Alapafuja
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (B.D.K., J.B.); Preclinical Pharmacology Laboratory, McLean Hospital, Belmont, Massachusetts (B.D.K., M.Z.L., J.B.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (V.G.S., S.O.A., S.P.N., A.M.)
| | - Spyros P Nikas
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (B.D.K., J.B.); Preclinical Pharmacology Laboratory, McLean Hospital, Belmont, Massachusetts (B.D.K., M.Z.L., J.B.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (V.G.S., S.O.A., S.P.N., A.M.)
| | - Alexandros Makriyannis
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (B.D.K., J.B.); Preclinical Pharmacology Laboratory, McLean Hospital, Belmont, Massachusetts (B.D.K., M.Z.L., J.B.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (V.G.S., S.O.A., S.P.N., A.M.)
| | - Jack Bergman
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts (B.D.K., J.B.); Preclinical Pharmacology Laboratory, McLean Hospital, Belmont, Massachusetts (B.D.K., M.Z.L., J.B.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (V.G.S., S.O.A., S.P.N., A.M.)
| |
Collapse
|
24
|
Parker LA, Niphakis MJ, Downey R, Limebeer CL, Rock EM, Sticht MA, Morris H, Abdullah RA, Lichtman AH, Cravatt BF. Effect of selective inhibition of monoacylglycerol lipase (MAGL) on acute nausea, anticipatory nausea, and vomiting in rats and Suncus murinus. Psychopharmacology (Berl) 2015; 232:583-93. [PMID: 25085768 DOI: 10.1007/s00213-014-3696-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/12/2014] [Indexed: 02/07/2023]
Abstract
RATIONALE To determine the role of the endocannabinoid, 2-arachodonyl glycerol (2-AG), in the regulation of nausea and vomiting. OBJECTIVE We evaluated the effectiveness of the potent selective monoacylglycerol lipase (MAGL) inhibitor, MJN110, which selectively elevates the endocannabinoid 2-AG, to suppress acute nausea and vomiting, as well as anticipatory nausea in rat and shrew models. METHODS The rat gaping models were used to evaluate the potential of MJN110 (5, 10, and 20 mg/kg, intraperitoneally [IP]) to suppress acute nausea produced by LiCl and of MJN110 (10 and 20 mg/kg, IP) to suppress anticipatory nausea elicited by a LiCl-paired context. The potential as well of MJN110 (10 and 20 mg/kg, IP) to suppress vomiting and contextually elicited gaping in the Suncus murinus was evaluated. RESULTS MJN110 suppressed acute nausea in rats, LiCl-induced vomiting in shrews and contextually-elicited anticipatory nausea in both rats (accompanied by elevation of 2-AG in the visceral insular cortex) and shrews. These effects were reversed by the CB1 antagonist/inverse agonist, SR141716. The MAGL inhibitor did not modify locomotion at any dose. An activity-based protein profiling analysis of samples of tissue collected from the visceral insular cortex in rats and whole brain tissues in shrews revealed that MJN110 selectively inhibited MAGL and the alternative 2-AG hydrolase, ABHD6. CONCLUSIONS MAGL inhibition by MJN110 which selectively elevates endogenous 2-AG has therapeutic potential in the treatment of acute nausea and vomiting as well as anticipatory nausea, a distressful symptom that is resistant to currently available treatments.
Collapse
Affiliation(s)
- Linda A Parker
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, N1G2W1, Canada,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rock EM, Kopstick RL, Limebeer CL, Parker LA. Tetrahydrocannabinolic acid reduces nausea-induced conditioned gaping in rats and vomiting in Suncus murinus. Br J Pharmacol 2014; 170:641-8. [PMID: 23889598 DOI: 10.1111/bph.12316] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 07/05/2013] [Accepted: 07/19/2013] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE We evaluated the anti-emetic and anti-nausea properties of the acid precursor of Δ(9) -tetrahydrocannabinol (THC), tetrahydrocannabinolic acid (THCA), and determined its mechanism of action in these animal models. EXPERIMENTAL APPROACH We investigated the effect of THCA on lithium chloride- (LiCl) induced conditioned gaping (nausea-induced behaviour) to a flavour, and context (a model of anticipatory nausea) in rats, and on LiCl-induced vomiting in Suncus murinus. Furthermore, we investigated THCA's ability to induce hypothermia and suppress locomotion [rodent tasks to assess cannabinoid1 (CB1 ) receptor agonist-like activity], and measured plasma and brain THCA and THC levels. We also determined whether THCA's effect could be blocked by pretreatment with SR141716 (SR, a CB1 receptor antagonist). KEY RESULTS In rats, THCA (0.05 and/or 0.5 mg·kg(-1) ) suppressed LiCl-induced conditioned gaping to a flavour and context; the latter effect blocked by the CB1 receptor antagonist, SR, but not by the 5-hydroxytryptamine-1A receptor antagonist, WAY100635. In S. murinus, THCA (0.05 and 0.5 mg·kg(-1) ) reduced LiCl-induced vomiting, an effect that was reversed with SR. A comparatively low dose of THC (0.05 mg·kg(-1) ) did not suppress conditioned gaping to a LiCl-paired flavour or context. THCA did not induce hypothermia or reduce locomotion, indicating non-CB1 agonist-like effects. THCA, but not THC was detected in plasma samples. CONCLUSIONS AND IMPLICATIONS THCA potently reduced conditioned gaping in rats and vomiting in S. murinus, effects that were blocked by SR. These data suggest that THCA may be a more potent alternative to THC in the treatment of nausea and vomiting.
Collapse
Affiliation(s)
- E M Rock
- Department of Psychology, University of Guelph, Guelph, ON, Canada
| | | | | | | |
Collapse
|
26
|
Schlosburg JE, Kinsey SG, Ignatowska-Jankowska B, Ramesh D, Abdullah RA, Tao Q, Booker L, Long JZ, Selley DE, Cravatt BF, Lichtman AH. Prolonged monoacylglycerol lipase blockade causes equivalent cannabinoid receptor type 1 receptor-mediated adaptations in fatty acid amide hydrolase wild-type and knockout mice. J Pharmacol Exp Ther 2014; 350:196-204. [PMID: 24849924 PMCID: PMC4109488 DOI: 10.1124/jpet.114.212753] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 05/20/2014] [Indexed: 11/22/2022] Open
Abstract
Complementary genetic and pharmacological approaches to inhibit monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH), the primary hydrolytic enzymes of the respective endogenous cannabinoids 2-arachidonoylglycerol (2-AG) and N-arachidonoylethanolamine, enable the exploration of potential therapeutic applications and physiologic roles of these enzymes. Complete and simultaneous inhibition of both FAAH and MAGL produces greatly enhanced cannabimimetic responses, including increased antinociception, and other cannabimimetic effects, far beyond those seen with inhibition of either enzyme alone. While cannabinoid receptor type 1 (CB1) function is maintained following chronic FAAH inactivation, prolonged excessive elevation of brain 2-AG levels, via MAGL inhibition, elicits both behavioral and molecular signs of cannabinoid tolerance and dependence. Here, we evaluated the consequences of a high dose of the MAGL inhibitor JZL184 [4-nitrophenyl 4-(dibenzo[d][1,3]dioxol-5-yl(hydroxy)methyl)piperidine-1-carboxylate; 40 mg/kg] given acutely or for 6 days in FAAH(-/-) and (+/+) mice. While acute administration of JZL184 to FAAH(-/-) mice enhanced the magnitude of a subset of cannabimimetic responses, repeated JZL184 treatment led to tolerance to its antinociceptive effects, cross-tolerance to the pharmacological effects of Δ(9)-tetrahydrocannabinol, decreases in CB1 receptor agonist-stimulated guanosine 5'-O-(3-[(35)S]thio)triphosphate binding, and dependence as indicated by rimonabant-precipitated withdrawal behaviors, regardless of genotype. Together, these data suggest that simultaneous elevation of both endocannabinoids elicits enhanced cannabimimetic activity but MAGL inhibition drives CB1 receptor functional tolerance and cannabinoid dependence.
Collapse
Affiliation(s)
- Joel E Schlosburg
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Steven G Kinsey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Bogna Ignatowska-Jankowska
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Divya Ramesh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Rehab A Abdullah
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Qing Tao
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Lamont Booker
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Jonathan Z Long
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Benjamin F Cravatt
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia (J.E.S., B.I.-J., D.R., R.A.A., Q.T., L.B., D.E.S., A.H.L.); Department of Psychology, West Virginia University, Morgantown, West Virginia (S.G.K.); and The Skaggs Institute for Chemical Biology and Department of Chemical Physiology (J.Z.L., B.F.C.), and Committee on the Neurobiology of Addictive Disorders (J.E.S.), The Scripps Research Institute, La Jolla, California
| |
Collapse
|
27
|
Zheng Y, Wang XL, Mo FF, Li M. Dexamethasone alleviates motion sickness in rats in part by enhancing the endocannabinoid system. Eur J Pharmacol 2014; 727:99-105. [PMID: 24508383 DOI: 10.1016/j.ejphar.2014.01.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 01/22/2014] [Accepted: 01/24/2014] [Indexed: 01/09/2023]
Abstract
Low-dose dexamethasone has been widely used for the prevention of nausea and vomiting after chemotherapy and surgical procedures and to treat motion sickness due to its minimal adverse effects, but the mechanisms underlying its anti-motion sickness effects are poorly understood. Previous studies have demonstrated that the endocannabinoid system is suppressed by motion sickness but stimulated by dexamethasone. The aim of the present study was to determine whether dexamethasone has an anti-motion sickness effect in rats and to elucidate the mechanism of this action. We used HPLC-MS/MS to measure the plasma concentrations of anandamide and 2-arachidonoylglycerol+1-arachidonoylglycerol, and we employed real-time quantitative PCR (qRT-PCR) and/or Western blot analysis to assay the expression of N-acylphosphatidyl-ethanolamine hydrolyzing phospholipase D, sn-1-selective diacylglycerol lipase, fatty acid hydrolase, monoacylglycerol lipase and endocannabinoid CB1 receptor in the dorsal vagal complex and stomach of rats exposed to a motion sickness protocol. The results showed that dexamethasone lowered the motion sickness index and restored the levels of endogenous cannabinoids and the expression of the endocannabinoid CB1 receptor, which declined after the induction of motion sickness, in the dorsal vagal complex and stomach.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Military Hygiene, Faculty of Naval Medicine, Second Military Medical University, 800 Xiang Yin Road, Shanghai, China; Department of Nutrition, Tong Ren Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 1111 Xian Xia Road, Shanghai, China.
| | - Xiao-Li Wang
- Department of Military Hygiene, Faculty of Naval Medicine, Second Military Medical University, 800 Xiang Yin Road, Shanghai, China.
| | - Feng-Feng Mo
- Department of Military Hygiene, Faculty of Naval Medicine, Second Military Medical University, 800 Xiang Yin Road, Shanghai, China.
| | - Min Li
- Department of Military Hygiene, Faculty of Naval Medicine, Second Military Medical University, 800 Xiang Yin Road, Shanghai, China.
| |
Collapse
|
28
|
Attenuation of anticipatory nausea in a rat model of contextually elicited conditioned gaping by enhancement of the endocannabinoid system. Psychopharmacology (Berl) 2014; 231:603-12. [PMID: 24043345 DOI: 10.1007/s00213-013-3282-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/28/2013] [Indexed: 12/28/2022]
Abstract
RATIONALE Enhancement of the endocannabinoid (EC) system may reduce anticipatory nausea (AN). OBJECTIVES The experiments evaluated the potential of the dual fatty acid amide hydrolase (FAAH)/monoacylglycerol lipase (MAGL) inhibitor, JZL195, on its own and combined with anandamide (AEA) and 2-arachidonoyl glycerol (2-AG) to reduce contextually elicited gaping, a measure of AN in rats. METHODS Following four context lithium chloride (LiCl) pairings, rats were injected with vehicle (VEH) or JZL195 (10 mg kg(-1), intraperitoneally) 105 min before an injection of VEH, 2-AG (1.25 mg kg(-1)), or AEA (5.0 mg kg(-1)). Fifteen minutes later, all rats were placed in the LiCl-paired context for 5 min and in a different context for a 15-min locomotor test. Whole brains were extracted for EC analysis. The potential of the CB1 antagonist, SR141716, to reverse the suppression of AN by both JZL195 and AEA and of the CB2 antagonist, AM630, to reverse the suppression of AN by JZL195 was then evaluated. RESULTS JZL195 suppressed gaping and elevated AEA, palmitoylethanolamine, and oleoylethanolamide. As the suppression of gaping was reversed by SR141716, but not by AM630, the effect was CB1 mediated. The suppressive effect of JZL195 on gaping, as well as elevation of AEA and 2-AG, was amplified by pretreatment with either AEA or 2-AG. On its own, AEA, but not 2-AG, also suppressed gaping-an effect that was also prevented by CB1 antagonism. CONCLUSIONS JZL195 reduces AN primarily by acting as a FAAH inhibitor, but MAGL inhibition is also indicated.
Collapse
|
29
|
Sharkey KA, Darmani NA, Parker LA. Regulation of nausea and vomiting by cannabinoids and the endocannabinoid system. Eur J Pharmacol 2014; 722:134-46. [PMID: 24184696 PMCID: PMC3883513 DOI: 10.1016/j.ejphar.2013.09.068] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/22/2013] [Accepted: 09/27/2013] [Indexed: 12/13/2022]
Abstract
Nausea and vomiting (emesis) are important elements in defensive or protective responses that animals use to avoid ingestion or digestion of potentially harmful substances. However, these neurally-mediated responses are at times manifested as symptoms of disease and they are frequently observed as side-effects of a variety of medications, notably those used to treat cancer. Cannabis has long been known to limit or prevent nausea and vomiting from a variety of causes. This has led to extensive investigations that have revealed an important role for cannabinoids and their receptors in the regulation of nausea and emesis. With the discovery of the endocannabinoid system, novel ways to regulate both nausea and vomiting have been discovered that involve the production of endogenous cannabinoids acting centrally. Here we review recent progress in understanding the regulation of nausea and vomiting by cannabinoids and the endocannabinoid system, and we discuss the potential to utilize the endocannabinoid system in the treatment of these frequently debilitating conditions.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1.
| | - Nissar A Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA
| | - Linda A Parker
- Department of Psychology, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
30
|
O'Brien LD, Limebeer CL, Rock EM, Bottegoni G, Piomelli D, Parker LA. Anandamide transport inhibition by ARN272 attenuates nausea-induced behaviour in rats, and vomiting in shrews (Suncus murinus). Br J Pharmacol 2013; 170:1130-6. [PMID: 23991698 PMCID: PMC3949659 DOI: 10.1111/bph.12360] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/12/2013] [Accepted: 08/20/2013] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND AND PURPOSE To understand how anandamide transport inhibition impacts the regulation of nausea and vomiting and the receptor level mechanism of action involved. In light of recent characterization of an anandamide transporter, fatty acid amide hydrolase-1-like anandamide transporter, to provide behavioural support for anandamide cellular reuptake as a facilitated transport process. EXPERIMENTAL APPROACH The systemic administration of the anandamide transport inhibitor ARN272 ([(4-(5-(4-hydroxy-phenyl)-3,4-diaza-bicyclo[4.4.0]deca-1(6),2,4,7,9-pentaen-2-ylamino)-phenyl)-phenylamino-methanone]) was used to evaluate the prevention of LiCl-induced nausea-induced behaviour (conditioned gaping) in rats, and LiCl-induced emesis in shrews (Suncus murinus). The mechanism of how prolonging anandamide availability acts to regulate nausea in rats was explored by the antagonism of cannabinoid 1 (CB1) receptors with the systemic co-administration of SR141716. KEY RESULTS The systemic administration of ARN272 produced a dose-dependent suppression of nausea-induced conditioned gaping in rats, and produced a dose-dependent reduction of vomiting in shrews. The systemic co-administration of SR141716 with ARN272 (at 3.0 mg·kg(-1)) in rats produced a complete reversal of ARN272-suppressed gaping at 1.0 mg·kg(-1). SR141716 alone did not differ from the vehicle solution. CONCLUSIONS AND IMPLICATIONS These results suggest that anandamide transport inhibition by the compound ARN272 tonically activates CB1 receptors and as such produces a type of indirect agonism to regulate toxin-induced nausea and vomiting. The results also provide behavioural evidence in support of a facilitated transport mechanism used in the cellular reuptake of anandamide.
Collapse
Affiliation(s)
- L D O'Brien
- Department of Psychology and Collaborative Neuroscience Program, University of GuelphGuelph, ON, Canada
| | - C L Limebeer
- Department of Psychology and Collaborative Neuroscience Program, University of GuelphGuelph, ON, Canada
| | - E M Rock
- Department of Psychology and Collaborative Neuroscience Program, University of GuelphGuelph, ON, Canada
| | - G Bottegoni
- Drug Discovery and Development, Instituto Italiano di TechnologiaGenova, Italy
| | - D Piomelli
- Drug Discovery and Development, Instituto Italiano di TechnologiaGenova, Italy
- Department of Anatomy and Neurobiology, University of CaliforniaIrvine, CA, USA
| | - L A Parker
- Department of Psychology and Collaborative Neuroscience Program, University of GuelphGuelph, ON, Canada
| |
Collapse
|
31
|
Sticht MA, Rock EM, Parker LA. 2-arachidonoylglycerol interferes with lithium-induced vomiting in the house musk shrew, Suncus murinus. Physiol Behav 2013; 120:228-32. [PMID: 23958470 DOI: 10.1016/j.physbeh.2013.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 07/30/2013] [Accepted: 08/09/2013] [Indexed: 10/26/2022]
Abstract
The role of the endocannabinoid system in vomiting has been previously studied using several animal species. These investigations have clearly demonstrated an anti-emetic role for the eCB, anandamide, in these animal models; however, research concerning the role of 2-arhachidonoylglycerol (2AG) has been less clear. The aim of the present study was to assess the effects of exogenous 2AG administration in the house musk shrew, Suncus murinus. In Experiment 1, shrews were injected with vehicle or 2AG (1, 2, 5, 10 mg/kg) 15 min prior to behavioral testing in which the frequency of vomiting episodes was observed. In Experiment 2, shrews were pre-treated with 2AG (2, 5 mg/kg) prior to being administered the emetic drug, lithium chloride (LiCl). It was found that 2AG alone did not induce emesis, but interfered with vomiting in response to LiCl administration. The anti-emetic effects of 2AG in Suncus murinus do not appear to be mediated by CB1 receptors, as concomitant pretreatment with the CB1 receptor antagonist, SR141716, did not reverse the suppressive effects of 2AG. These results confirm that manipulations that increase levels of 2AG exert anti-emetic effects in the house musk shrew.
Collapse
Affiliation(s)
- M A Sticht
- Department of Psychology and Neuroscience Graduate Program, University of Guelph, Guelph, Ontario, Canada
| | | | | |
Collapse
|
32
|
Abstract
The endocannabinoid signaling system regulates diverse physiologic processes and has attracted considerable attention as a potential pharmaceutical target for treating diseases, such as pain, anxiety/depression, and metabolic disorders. The principal ligands of the endocannabinoid system are the lipid transmitters N-arachidonoylethanolamine (anandamide) and 2-arachidonoylglycerol (2-AG), which activate the two major cannabinoid receptors, CB1 and CB2. Anandamide and 2-AG signaling pathways in the nervous system are terminated by enzymatic hydrolysis mediated primarily by the serine hydrolases fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), respectively. In this review, we will discuss the development of FAAH and MAGL inhibitors and their pharmacological application to investigate the function of anandamide and 2-AG signaling pathways in preclinical models of neurobehavioral processes, such as pain, anxiety, and addiction. We will place emphasis on how these studies are beginning to discern the different roles played by anandamide and 2-AG in the nervous system and the resulting implications for advancing endocannabinoid hydrolase inhibitors as next-generation therapeutics.
Collapse
Affiliation(s)
- Jacqueline L Blankman
- Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | |
Collapse
|
33
|
Ono H, Iwajima Y, Nagano Y, Chazono K, Maeda Y, Ohsawa M, Yamamoto S. Reduction in sympathetic nerve activity as a possible mechanism for the hypothermic effect of oseltamivir, an anti-influenza virus drug, in normal mice. Basic Clin Pharmacol Toxicol 2013; 113:25-30. [PMID: 23398656 DOI: 10.1111/bcpt.12058] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 01/31/2013] [Indexed: 11/28/2022]
Abstract
Oseltamivir, an anti-influenza virus drug, has strong antipyretic effects in mice (Biological and Pharmaceutical Bulletin, 31, 2008, 638) and patients with influenza. In addition, hypothermia has been reported as an adverse event. The prodrug oseltamivir is converted to oseltamivir carboxylate (OC), an active metabolite of influenza virus neuraminidase. In this study, core body temperature was measured in mice, and oseltamivir and OC were administered intracerebroventricularly (i.c.v.) or intraperitoneally (i.p). Low i.c.v. doses of oseltamivir and OC dose-dependently produced hypothermia. Zanamivir (i.c.v.), another neuraminidase inhibitor, did not produce hypothermia. These results suggested that the hypothermic effects of oseltamivir (i.p. and i.c.v.) and OC (i.c.v.) are not due to neuraminidase inhibition. OC (i.p.) did not lower body temperature. Although mecamylamine (i.c.v.) blocked the hypothermic effect of nicotine-administered i.c.v., the hypothermic effects of oseltamivir and OC (i.c.v.) were not blocked by mecamylamine (i.c.v.). The effect of oseltamivir (i.p.) was markedly increased by s.c.-pre-administered mecamylamine and also hexamethonium, a peripherally acting ganglionic blocker, suggesting their potentiating interaction at peripheral sites. The hypothermic effect of nicotine (i.c.v.) was decreased by lower doses of oseltamivir (i.c.v.), suggesting the anti-nicotinic action of oseltamivir. These results suggest that oseltamivir (i.p.) causes hypothermia through depression of sympathetic temperature regulatory mechanisms via inhibition of nicotinic receptor function and through unknown central mechanisms.
Collapse
Affiliation(s)
- Hideki Ono
- Laboratory of CNS Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
34
|
Schicho R, Storr M. Targeting the endocannabinoid system for gastrointestinal diseases: future therapeutic strategies. Expert Rev Clin Pharmacol 2012; 3:193-207. [PMID: 22111567 DOI: 10.1586/ecp.09.62] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cannabinoids extracted from the marijuana plant (Cannabis sativa) and synthetic cannabinoids have numerous effects on gastrointestinal (GI) functions. Recent experimental data support an important role for cannabinoids in GI diseases. Genetic studies in humans have proven that defects in endocannabinoid metabolism underlie functional GI disorders. Mammalian cells have machinery, the so-called endocannabinoid system (ECS), to produce and metabolize their own cannabinoids in order to control homeostasis of the gut in a rapidly adapting manner. Pharmacological manipulation of the ECS by cannabinoids, or by drugs that raise the levels of endogenous cannabinoids, have shown beneficial effects on GI pathophysiology. This review gives an introduction into the functions of the ECS in the GI tract, highlights the role of the ECS in GI diseases and addresses its potential pharmacological exploitation.
Collapse
Affiliation(s)
- Rudolf Schicho
- Division of Gastroenterology, Department of Medicine, University of Calgary, 6D25, TRW Building, 3280 Hospital Drive NW, Calgary T2N 4N1, AB, Canada.
| | | |
Collapse
|
35
|
Sticht MA, Long JZ, Rock EM, Limebeer CL, Mechoulam R, Cravatt BF, Parker LA. Inhibition of monoacylglycerol lipase attenuates vomiting in Suncus murinus and 2-arachidonoyl glycerol attenuates nausea in rats. Br J Pharmacol 2012; 165:2425-35. [PMID: 21470205 DOI: 10.1111/j.1476-5381.2011.01407.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE To evaluate the role of 2-arachidonoyl glycerol (2AG) in the regulation of nausea and vomiting using animal models of vomiting and of nausea-like behaviour (conditioned gaping). EXPERIMENTAL APPROACH Vomiting was assessed in shrews (Suncus murinus), pretreated with JZL184, a selective monoacylglycerol lipase (MAGL) inhibitor which elevates endogenous 2AG levels, 1 h before administering the emetogenic compound, LiCl. Regulation of nausea-like behaviour in rats by exogenous 2AG or its metabolite arachidonic acid (AA) was assessed, using the conditioned gaping model. The role of cannabinoid CB(1) receptors, CB(2) receptors and cyclooxygenase (COX) inhibition in suppression of vomiting or nausea-like behaviour was assessed. KEY RESULTS JZL184 dose-dependently suppressed vomiting in shrews, an effect prevented by pretreatment with the CB(1) receptor inverse agonist/antagonist, AM251. In shrew brain tissue, JZL184 inhibited MAGL activity in vivo. In rats, 2AG suppressed LiCl-induced conditioned gaping but this effect was not prevented by AM251 or the CB(2) receptor antagonist, AM630. Instead, the COX inhibitor, indomethacin, prevented suppression of conditioned gaping by 2AG or AA. However, when rats were pretreated with a high dose of JZL184 (40 mg·kg(-1) ), suppression of gaping by 2AG was partially reversed by AM251. Suppression of conditioned gaping was not due to interference with learning because the same dose of 2AG did not modify the strength of conditioned freezing to a shock-paired tone. CONCLUSIONS AND IMPLICATIONS Our results suggest that manipulations that elevate 2AG may have anti-emetic or anti-nausea potential. LINKED ARTICLES This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visit http://dx.doi.org/10.1111/bph.2011.163.issue-7.
Collapse
Affiliation(s)
- Martin A Sticht
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
36
|
Armirotti A, Romeo E, Ponzano S, Mengatto L, Dionisi M, Karacsonyi C, Bertozzi F, Garau G, Tarozzo G, Reggiani A, Bandiera T, Tarzia G, Mor M, Piomelli D. β-Lactones Inhibit N-acylethanolamine Acid Amidase by S-Acylation of the Catalytic N-Terminal Cysteine. ACS Med Chem Lett 2012; 3:422-6. [PMID: 24900487 DOI: 10.1021/ml300056y] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 04/06/2012] [Indexed: 12/11/2022] Open
Abstract
The cysteine amidase N-acylethanolamine acid amidase (NAAA) is a member of the N-terminal nucleophile class of enzymes and a potential target for anti-inflammatory drugs. We investigated the mechanism of inhibition of human NAAA by substituted β-lactones. We characterized pharmacologically a representative member of this class, ARN077, and showed, using high-resolution liquid chromatography-tandem mass spectrometry, that this compound forms a thioester bond with the N-terminal catalytic cysteine in human NAAA.
Collapse
Affiliation(s)
- Andrea Armirotti
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Elisa Romeo
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Stefano Ponzano
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Luisa Mengatto
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Mauro Dionisi
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Claudia Karacsonyi
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Fabio Bertozzi
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Gianpiero Garau
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Glauco Tarozzo
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Angelo Reggiani
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Tiziano Bandiera
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
| | - Giorgio Tarzia
- Dipartimento
di Scienze Biomolecolari, Università degli Studi di Urbino “Carlo Bo”, Piazza del Rinascimento
6, I-61029 Urbino, Italy
| | - Marco Mor
- Pharmaceutical
Department, University of Parma, 43124
Parma, Italy
| | - Daniele Piomelli
- Department of Drug
Discovery and Development, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy
- Departments of Pharmacology, University of California, Irvine, 360 MSRII, California 92697-4625,
United States
| |
Collapse
|
37
|
Rock EM, Bolognini D, Limebeer CL, Cascio MG, Anavi-Goffer S, Fletcher PJ, Mechoulam R, Pertwee RG, Parker LA. Cannabidiol, a non-psychotropic component of cannabis, attenuates vomiting and nausea-like behaviour via indirect agonism of 5-HT(1A) somatodendritic autoreceptors in the dorsal raphe nucleus. Br J Pharmacol 2012; 165:2620-34. [PMID: 21827451 PMCID: PMC3423241 DOI: 10.1111/j.1476-5381.2011.01621.x] [Citation(s) in RCA: 173] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 07/04/2011] [Accepted: 07/15/2011] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE To evaluate the hypothesis that activation of somatodendritic 5-HT(1A) autoreceptors in the dorsal raphe nucleus (DRN) produces the anti-emetic/anti-nausea effects of cannabidiol (CBD), a primary non-psychoactive cannabinoid found in cannabis. EXPERIMENTAL APPROACH The potential of systemic and intra-DRN administration of 5-HT(1A) receptor antagonists, WAY100135 or WAY100635, to prevent the anti-emetic effect of CBD in shrews (Suncus murinus) and the anti-nausea-like effects of CBD (conditioned gaping) in rats were evaluated. Also, the ability of intra-DRN administration of CBD to produce anti-nausea-like effects (and reversal by systemic WAY100635) was assessed. In vitro studies evaluated the potential of CBD to directly target 5-HT(1A) receptors and to modify the ability of the 5-HT(1A) agonist, 8-OH-DPAT, to stimulate [(35) S]GTPγS binding in rat brainstem membranes. KEY RESULTS CBD suppressed nicotine-, lithium chloride (LiCl)- and cisplatin (20 mg·kg(-1) , but not 40 mg·kg(-1) )-induced vomiting in the S. murinus and LiCl-induced conditioned gaping in rats. Anti-emetic and anti-nausea-like effects of CBD were suppressed by WAY100135 and the latter by WAY100635. When administered to the DRN: (i) WAY100635 reversed anti-nausea-like effects of systemic CBD, and (ii) CBD suppressed nausea-like effects, an effect that was reversed by systemic WAY100635. CBD also displayed significant potency (in a bell-shaped dose-response curve) at enhancing the ability of 8-OH-DPAT to stimulate [(35) S]GTPγS binding to rat brainstem membranes in vitro. Systemically administered CBD and 8-OH-DPAT synergistically suppressed LiCl-induced conditioned gaping. CONCLUSIONS AND IMPLICATIONS These results suggest that CBD produced its anti-emetic/anti-nausea effects by indirect activation of the somatodendritic 5-HT(1A) autoreceptors in the DRN. LINKED ARTICLES This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visit http://dx.doi.org/10.1111/bph.2011.163.issue-7.
Collapse
Affiliation(s)
- E M Rock
- Department of Psychology and Neuroscience Graduate Program, University of Guelph, Guelph, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Shiina T, Shima T, Suzuki Y, Wörl J, Shimizu Y. Neural regulation of esophageal striated muscle in the house musk shrew (Suncus murinus). Auton Neurosci 2012; 168:25-31. [PMID: 22285704 DOI: 10.1016/j.autneu.2012.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 01/05/2012] [Accepted: 01/08/2012] [Indexed: 12/21/2022]
Abstract
In the present study, we characterized the neural regulation of esophageal striated muscle in Suncus murinus (a house musk shrew; "suncus" used as a laboratory name), which was compared with that in the rat. The tunica muscularis consists of striated muscle in the suncus esophagus. An isolated segment of the suncus esophagus was placed in an organ bath and the contractile responses were recorded using a force transducer. Electrical stimulations to vagus nerves induced contractile responses in the esophageal segment. Treatment with α-bungarotoxin, a blocker of nicotinic acetylcholine receptors, blocked the vagally mediated contractions of the suncus esophagus. D-tubocurarine and succinylcholine, typical antagonists of nicotinic acetylcholine receptors, also inhibited the suncus esophageal contractions, while higher concentrations of the agents were required rather than concentrations for producing an equivalent block in the rat. We used capsaicin, a stimulator of small-caliber afferent neurons, for activating the peripheral neural network. The reagent inhibited the vagally mediated twitch contractions of striated muscle in the suncus esophagus, which was reversed by pretreatment with a nitric oxide synthase inhibitor, N(G)-nitro-L-arginine methyl ester. Application of a nitric oxide donor, diethylamine NONOate diethylammonium salt, mimicked capsaicin-induced inhibition. The results suggest that motility of the suncus esophagus, which consists of striated muscles, is regulated by vagal cholinergic neurons. The local neural network including capsaicin-sensitive neurons and intrinsic nitrergic neurons can modify the vagally mediated motility in the suncus esophagus. In addition, nicotinic acetylcholine receptors of the suncus esophagus might be pharmacologically distinct from those of rodent esophagi.
Collapse
Affiliation(s)
- Takahiko Shiina
- Laboratory of Physiology, Department of Basic Veterinary Science, The United Graduate School of Veterinary Sciences, Gifu University, Japan.
| | | | | | | | | |
Collapse
|
39
|
Alternative targets within the endocannabinoid system for future treatment of gastrointestinal diseases. CANADIAN JOURNAL OF GASTROENTEROLOGY = JOURNAL CANADIEN DE GASTROENTEROLOGIE 2011; 25:377-83. [PMID: 21876860 DOI: 10.1155/2011/953975] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Many beneficial effects of herbal and synthetic cannabinoids on gut motility and inflammation have been demonstrated, suggesting a vast potential for these compounds in the treatment of gastrointestinal disorders. These effects are based on the so-called 'endocannabinoid system' (ECS), a cooperating network of molecules that regulate the metabolism of the body's own and of exogenously administered cannabinoids. The ECS in the gastrointestinal tract quickly responds to homeostatic disturbances by de novo synthesis of its components to maintain homeostasis, thereby offering many potential targets for pharmacological intervention. Of major therapeutic interest are nonpsychoactive cannabinoids or compounds that do not directly target cannabinoid receptors but still possess cannabinoid-like properties. Drugs that inhibit endocannabinoid degradation and raise the level of endocannabinoids are becoming increasingly promising alternative therapeutic tools to manipulate the ECS.
Collapse
|
40
|
Investigating the effect of emetic compounds on chemotaxis in Dictyostelium identifies a non-sentient model for bitter and hot tastant research. PLoS One 2011; 6:e24439. [PMID: 21931717 PMCID: PMC3169598 DOI: 10.1371/journal.pone.0024439] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 08/10/2011] [Indexed: 01/05/2023] Open
Abstract
Novel chemical entities (NCEs) may be investigated for emetic liability in a range of unpleasant experiments involving retching, vomiting or conditioned taste aversion/food avoidance in sentient animals. We have used a range of compounds with known emetic /aversive properties to examine the possibility of using the social amoeba, Dictyostelium discoideum, for research into identifying and understanding emetic liability, and hence reduce adverse animal experimentation in this area. Twenty eight emetic or taste aversive compounds were employed to investigate the acute (10 min) effect of compounds on Dictyostelium cell behaviour (shape, speed and direction of movement) in a shallow chemotaxic gradient (Dunn chamber). Compound concentrations were chosen based on those previously reported to be emetic or aversive in in vivo studies and results were recorded and quantified by automated image analysis. Dictyostelium cell motility was rapidly and strongly inhibited by four structurally distinct tastants (three bitter tasting compounds--denatonium benzoate, quinine hydrochloride, phenylthiourea, and the pungent constituent of chilli peppers--capsaicin). In addition, stomach irritants (copper chloride and copper sulphate), and a phosphodiesterase IV inhibitor also rapidly blocked movement. A concentration-dependant relationship was established for five of these compounds, showing potency of inhibition as capsaicin (IC(50) = 11.9 ± 4.0 µM) > quinine hydrochloride (IC(50) = 44.3 ± 6.8 µM) > denatonium benzoate (IC(50) = 129 ± 4 µM) > phenylthiourea (IC(50) = 366 ± 5 µM) > copper sulphate (IC(50) = 1433 ± 3 µM). In contrast, 21 compounds within the cytotoxic and receptor agonist/antagonist classes did not affect cell behaviour. Further analysis of bitter and pungent compounds showed that the effect on cell behaviour was reversible and not cytotoxic, suggesting an uncharacterised molecular mechanism of action for these compounds. These results therefore demonstrate that Dictyostelium has potential as a non-sentient model in the analysis of the molecular effects of tastants, although it has limited utility in identification of emetic agents in general.
Collapse
|
41
|
Parker LA, Rock EM, Limebeer CL. Regulation of nausea and vomiting by cannabinoids. Br J Pharmacol 2011; 163:1411-22. [PMID: 21175589 PMCID: PMC3165951 DOI: 10.1111/j.1476-5381.2010.01176.x] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 11/11/2010] [Accepted: 11/17/2010] [Indexed: 12/18/2022] Open
Abstract
Considerable evidence demonstrates that manipulation of the endocannabinoid system regulates nausea and vomiting in humans and other animals. The anti-emetic effect of cannabinoids has been shown across a wide variety of animals that are capable of vomiting in response to a toxic challenge. CB(1) agonism suppresses vomiting, which is reversed by CB(1) antagonism, and CB(1) inverse agonism promotes vomiting. Recently, evidence from animal experiments suggests that cannabinoids may be especially useful in treating the more difficult to control symptoms of nausea and anticipatory nausea in chemotherapy patients, which are less well controlled by the currently available conventional pharmaceutical agents. Although rats and mice are incapable of vomiting, they display a distinctive conditioned gaping response when re-exposed to cues (flavours or contexts) paired with a nauseating treatment. Cannabinoid agonists (Δ(9) -THC, HU-210) and the fatty acid amide hydrolase (FAAH) inhibitor, URB-597, suppress conditioned gaping reactions (nausea) in rats as they suppress vomiting in emetic species. Inverse agonists, but not neutral antagonists, of the CB(1) receptor promote nausea, and at subthreshold doses potentiate nausea produced by other toxins (LiCl). The primary non-psychoactive compound in cannabis, cannabidiol (CBD), also suppresses nausea and vomiting within a limited dose range. The anti-nausea/anti-emetic effects of CBD may be mediated by indirect activation of somatodendritic 5-HT(1A) receptors in the dorsal raphe nucleus; activation of these autoreceptors reduces the release of 5-HT in terminal forebrain regions. Preclinical research indicates that cannabinioids, including CBD, may be effective clinically for treating both nausea and vomiting produced by chemotherapy or other therapeutic treatments.
Collapse
Affiliation(s)
- Linda A Parker
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, Guelph, Ontario, N1G 2W1, Canada. DA-9789
| | | | | |
Collapse
|
42
|
Järbe TUC, LeMay BJ, Vemuri VK, Vadivel SK, Zvonok A, Makriyannis A. Central mediation and differential blockade by cannabinergics of the discriminative stimulus effects of the cannabinoid CB1 receptor antagonist rimonabant in rats. Psychopharmacology (Berl) 2011; 216:355-65. [PMID: 21369753 PMCID: PMC3727221 DOI: 10.1007/s00213-011-2226-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 02/06/2011] [Indexed: 10/18/2022]
Abstract
RATIONALE Discovery of an endocannabinoid signaling system launched the development of the blocker rimonabant, a cannabinoid CB1 receptor (CB(1)R) antagonist/inverse agonist. Due to untoward effects, this medication was withdrawn and efforts have been directed towards discovering chemicals with more benign profiles. OBJECTIVE This study aims to comparatively evaluate new ligands using a rimonabant discriminated drinking aversion procedure. METHODS Rats discriminated between rimonabant (5.6 mg/kg) and vehicle. The 30 min saccharin (0.1%) drinking after rimonabant pretreatment was followed by injection of lithium chloride (120 mg/kg) in the experimental animals. After vehicle pretreatment, experimental animals were given i.p. NaCl (10 ml/kg). Postdrinking treatment for controls was NaCl, irrespective of pretreatment condition (rimonabant or vehicle). RESULTS The centrally acting neutral CB(1)R antagonist AM4113, but not the limited brain penetrating CB(1)R neutral antagonist AM6545, substituted for rimonabant. The CB(1)R agonists THC (1-10 mg/kg), AM1346 (1-10 mg/kg) did not substitute. The rimonabant-induced conditioned suppression of saccharin drinking was attenuated when CB(1)R agonists AM5983 (0.01-1 mg/kg) and THC (10 mg/kg), but not the CB(1)R agonist AM1346 (0.1-18 mg/kg), were combined with rimonabant (5.6 mg/kg). By varying the injection-to-test interval, we gauged the relative duration of the cueing effects of rimonabant, and the in vivo functional half-life was estimated to be approximately 1.5 h. CONCLUSION A neutral CB(1)R antagonist (AM4113) produced cueing effects similar to those of rimonabant and generalization likely was centrally mediated. The functional cueing effects of rimonabant are relatively short-acting, pharmacologically selective, and differentially blocked by cannabinergics.
Collapse
|
43
|
Interaction between non-psychotropic cannabinoids in marihuana: effect of cannabigerol (CBG) on the anti-nausea or anti-emetic effects of cannabidiol (CBD) in rats and shrews. Psychopharmacology (Berl) 2011; 215:505-12. [PMID: 21243485 DOI: 10.1007/s00213-010-2157-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 12/19/2010] [Indexed: 02/08/2023]
Abstract
RATIONALE The interaction between two non-psychotropic cannabinoids, cannabidiol (CBD) and cannabigerol (CBG), which have been reported to act as a 5-hydroxytryptamine 1A (5-HT(1A)) agonist and antagonist, respectively, was evaluated. OBJECTIVE To evaluate the potential of CBG to reverse the anti-nausea, anti-emetic effects of CBD. MATERIALS AND METHODS In experiment 1, rats were pre-treated with CBG (0.0, 1, 5, and 10 mg/kg, ip), 15 min prior to being treated with CBD (experiment 1a: VEH or 5 mg/kg, ip) or 8-OH-DPAT (experiment 1b: VEH or 0.01 mg/kg, ip). Thirty minutes later, all rats received a pairing of 0.1% saccharin solution and LiCl (20 ml/kg of 0.15 M, ip). Seventy-two hours later, the rats received a drug-free taste reactivity test with saccharin to evaluate the effects of the treatments on the establishment of conditioned gaping reactions (a model of nausea). As well, conditioned saccharin avoidance was measured. In experiment 2, Suncus murinus were injected with CBG (5 mg/kg, ip) or VEH 15 min prior to CBD (5 mg/kg) or VEH and 30 min later were injected with LiCl (60 ml/kg of 0.15 M, i.p.), and the number of vomiting episodes were measured. RESULTS CBD (5 mg/kg) suppressed conditioned gaping in rats and vomiting in shrews, which were reversed by pre-treatment with all doses of CBG. CBG also prevented the anti-nausea effects of 8-OH-DPAT. CONCLUSIONS Interactions between moderate doses of CBG and CBD may oppose one another at the 5-HT(1A) receptor in the regulation of nausea and vomiting.
Collapse
|
44
|
Huang D, Meyers K, Henry S, De la Torre F, Horn CC. Computerized detection and analysis of cancer chemotherapy-induced emesis in a small animal model, musk shrew. J Neurosci Methods 2011; 197:249-58. [PMID: 21392533 DOI: 10.1016/j.jneumeth.2011.02.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 02/25/2011] [Accepted: 02/28/2011] [Indexed: 11/17/2022]
Abstract
Vomiting is a common side effect of cancer chemotherapy and many drug treatments and diseases. In animal studies, the measurement of vomiting usually requires direct observation, which is time consuming and often lacks temporal precision. Musk shrews have been used to study the neurobiology of emesis and have a rapid emetic episode (∼1 s for a sequence of retching and expulsion). The aim of the current study was to develop a method to automatically detect and characterize emetic episodes induced by the cancer chemotherapy agent cisplatin. The body contour in each video frame was tracked and normalized to a parameterized shape basis. The tracked shape was projected to a feature space that maximized the shape variations in the consecutive frames during retching. The resulting one dimensional projection was sufficient to detect most emetic episodes in the acute (peak at 2h) and delayed (peak at 54 h) phases after cisplatin treatment. Emetic episodes were relatively invariant in the number of retches (∼6.2), duration (∼1.2s), inter-retch interval (∼198 ms), and amplitude during the 72 h after cisplatin treatment. This approach should open a new vista into emesis research to permit tracking and analysis of emesis in a small animal model and facilitate the development of new antiemetic therapies. These results also yield a better understanding of the brain's central pattern generator for emesis and indicate that the retching response in the musk shrew (at ∼5.4 Hz) is the fastest ever recorded in a free-moving animal.
Collapse
Affiliation(s)
- Dong Huang
- Carnegie Mellon University, Robotics Institute, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
45
|
Percie du Sert N, Ho WSV, Rudd JA, Andrews PLR. Cannabinoid-induced reduction in antral pacemaker frequency: a telemetric study in the ferret. Neurogastroenterol Motil 2010; 22:1257-66, e324. [PMID: 20731777 DOI: 10.1111/j.1365-2982.2010.01581.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The gastric myoelectric activity (GMA) is the electrical pacesetter potential, which drives gastric motility. Cannabinoids have broad-spectrum antiemetic and antinauseant activity. Paradoxically, they inhibit intestinal peristalsis and reduce gastric motility but their effect on GMA remains unknown. METHODS Ferrets were surgically implanted with radiotelemetry transmitters to record GMA, body temperature and heart rate. The effect of WIN 55,212-2 (1 mg kg(-1), i.p.), an agonist at the cannabinoid type 1 and 2 receptors was examined in conscious, unrestrained ferrets. WIN 55,212-2 was also compared to the anandamide upregulator URB 597 (5 mg kg(-1), i.p.) for a potential to modulate the emetic response and behavioral changes induced by apomorphine (0.25 mg kg(-1), s.c.). KEY RESULTS WIN 55,212-2 decreased GMA frequency (8.1 ± 0.4 cpm, compared to 9.6 ± 0.1 cpm in vehicle-treated animals, n = 6, P < 0.01). Apomorphine induced 9.0 ± 1.6 emetic episodes, WIN 55,212-2 inhibited the emetic response (3.3 ± 1.0 episodes, n = 6, P < 0.05) but URB 597 had no effect (9.0 ± 1.5 episodes). Apomorphine-induced hyperactivity in vehicle-treated animals (6.5 ± 3.6-16.6 ± 4.9 active behavior counts, n = 6, P < 0.01), which was reduced by WIN 55,212-2 (5.0 ± 1.5 counts, n = 6, P < 0.05). CONCLUSIONS & INFERENCES WIN 55,212-2 demonstrated clear antiemetic efficacy, which extends the broad-spectrum antiemetic efficacy of cannabinoids to dopamine receptor agonists in the ferret. Our results, however, suggest a more limited spectrum of action for URB 597. WIN 55,212-2 decreased the frequency of the antral electrical pacemaker, which reveals new insights into the mechanism regulating the decrease in motility induced by cannabinoids.
Collapse
Affiliation(s)
- N Percie du Sert
- Division of Basic Medical Sciences, St George's University of London, London, UK.
| | | | | | | |
Collapse
|
46
|
Darmani NA. Mechanisms of Broad-Spectrum Antiemetic Efficacy of Cannabinoids against Chemotherapy-Induced Acute and Delayed Vomiting. Pharmaceuticals (Basel) 2010; 3:2930-2955. [PMID: 27713384 PMCID: PMC4034105 DOI: 10.3390/ph3092930] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 08/26/2010] [Accepted: 08/30/2010] [Indexed: 01/02/2023] Open
Abstract
Chemotherapy-induced nausea and vomiting (CINV) is a complex pathophysiological condition and consists of two phases. The conventional CINV neurotransmitter hypothesis suggests that the immediate phase is mainly due to release of serotonin (5-HT) from the enterochromaffin cells in the gastrointestinal tract (GIT), while the delayed phase is a consequence of release of substance P (SP) in the brainstem. However, more recent findings argue against this simplistic neurotransmitter and anatomical view of CINV. Revision of the hypothesis advocates a more complex, differential and overlapping involvement of several emetic neurotransmitters/modulators (e.g. dopamine, serotonin, substance P, prostaglandins and related arachidonic acid derived metabolites) in both phases of emesis occurring concomitantly in the brainstem and in the GIT enteric nervous system (ENS) [1]. No single antiemetic is currently available to completely prevent both phases of CINV. The standard antiemetic regimens include a 5-HT₃ antagonist plus dexamethasone for the prevention of acute emetic phase, combined with an NK1 receptor antagonist (e.g. aprepitant) for the delayed phase. Although NK1 antagonists behave in animals as broad-spectrum antiemetics against different emetogens including cisplatin-induced acute and delayed vomiting, by themselves they are not very effective against CINV in cancer patients. Cannabinoids such as D⁸-THC also behave as broad-spectrum antiemetics against diverse emetic stimuli as well as being effective against both phases of CINV in animals and patients. Potential side effects may limit the clinical utility of direct-acting cannabinoid agonists which could be avoided by the use of corresponding indirect-acting agonists. Cannabinoids (both phyto-derived and synthetic) behave as agonist antiemetics via the activation of cannabinoid CB₁ receptors in both the brainstem and the ENS emetic loci. An endocannabinoid antiemetic tone may exist since inverse CB₁ agonists (but not the corresponding silent antagonists) cause nausea and vomiting.
Collapse
Affiliation(s)
- Nissar A Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
47
|
Darmani NA. Cannabinoid-Induced Hyperemesis: A Conundrum-From Clinical Recognition to Basic Science Mechanisms. Pharmaceuticals (Basel) 2010; 3:2163-2177. [PMID: 27713347 PMCID: PMC4036650 DOI: 10.3390/ph3072163] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 06/25/2010] [Accepted: 06/29/2010] [Indexed: 12/14/2022] Open
Abstract
Cannabinoids are used clinically on a subacute basis as prophylactic agonist antiemetics for the prevention of nausea and vomiting caused by chemotherapeutics. Cannabinoids prevent vomiting by inhibition of release of emetic neurotransmitters via stimulation of presynaptic cannabinoid CB₁ receptors. Cannabis-induced hyperemesis is a recently recognized syndrome associated with chronic cannabis use. It is characterized by repeated cyclical vomiting and learned compulsive hot water bathing behavior. Although considered rare, recent international publications of numerous case reports suggest the contrary. The syndrome appears to be a paradox and the pathophysiological mechanism(s) underlying the induced vomiting remains unknown. Although some traditional hypotheses have already been proposed, the present review critically explores the basic science of these explanations in the clinical setting and provides more current mechanisms for the induced hyperemesis. These encompass: (1) pharmacokinetic factors such as long half-life, chronic exposure, lipid solubility, individual variation in metabolism/excretion leading to accumulation of emetogenic cannabinoid metabolites, and/or cannabinoid withdrawal; and (2) pharmacodynamic factors including switching of the efficacy of Δ⁸-THC from partial agonist to antagonist, differential interaction of Δ⁸-THC with Gs and Gi signal transduction proteins, CB₁ receptor desensitization or downregulation, alterations in tissue concentrations of endocannabinoid agonists/inverse agonists, Δ⁸-THC-induced mobilization of emetogenic metabolites of the arachidonic acid cascade, brainstem versus enteric actions of Δ⁸-THC, and/or hypothermic versus hyperthermic actions of Δ⁸-THC. In addition, human and animal findings suggest that chronic exposure to cannabis may not be a prerequisite for the induction of vomiting but is required for the intensity of emesis.
Collapse
Affiliation(s)
- Nissar A Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, CA, USA.
| |
Collapse
|
48
|
Maccarrone M, Gasperi V, Catani MV, Diep TA, Dainese E, Hansen HS, Avigliano L. The Endocannabinoid System and Its Relevance for Nutrition. Annu Rev Nutr 2010; 30:423-40. [DOI: 10.1146/annurev.nutr.012809.104701] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Mauro Maccarrone
- Department of Biomedical Sciences, University of Teramo, Teramo, Italy; ,
- European Center for Brain Research (CERC)/Santa Lucia Foundation, Rome, Italy
| | - Valeria Gasperi
- Department of Experimental Medicine and Biochemical Sciences, University of Rome, Tor Vergata, Rome, Italy; , ,
| | - Maria Valeria Catani
- Department of Experimental Medicine and Biochemical Sciences, University of Rome, Tor Vergata, Rome, Italy; , ,
| | - Thi Ai Diep
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmaceutical Sciences, University of Copenhagen, Copenhagen, Denmark; ,
| | - Enrico Dainese
- Department of Biomedical Sciences, University of Teramo, Teramo, Italy; ,
| | - Harald S. Hansen
- Department of Pharmacology and Pharmacotherapy, Faculty of Pharmaceutical Sciences, University of Copenhagen, Copenhagen, Denmark; ,
| | - Luciana Avigliano
- Department of Experimental Medicine and Biochemical Sciences, University of Rome, Tor Vergata, Rome, Italy; , ,
| |
Collapse
|
49
|
Choukèr A, Kaufmann I, Kreth S, Hauer D, Feuerecker M, Thieme D, Vogeser M, Thiel M, Schelling G. Motion sickness, stress and the endocannabinoid system. PLoS One 2010; 5:e10752. [PMID: 20505775 PMCID: PMC2873996 DOI: 10.1371/journal.pone.0010752] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2010] [Accepted: 04/19/2010] [Indexed: 12/02/2022] Open
Abstract
Background A substantial number of individuals are at risk for the development of motion sickness induced nausea and vomiting (N&V) during road, air or sea travel. Motion sickness can be extremely stressful but the neurobiologic mechanisms leading to motion sickness are not clear. The endocannabinoid system (ECS) represents an important neuromodulator of stress and N&V. Inhibitory effects of the ECS on N&V are mediated by endocannabinoid-receptor activation. Methodology/Principal Findings We studied the activity of the ECS in human volunteers (n = 21) during parabolic flight maneuvers (PFs). During PFs, microgravity conditions (<10−2 g) are generated for approximately 22 s which results in a profound kinetic stimulus. Blood endocannabinoids (anandamide and 2-arachidonoylglycerol, 2-AG) were measured from blood samples taken in-flight before start of the parabolic maneuvers, after 10, 20, and 30 parabolas, in-flight after termination of PFs and 24 h later. Volunteers who developed acute motion sickness (n = 7) showed significantly higher stress scores but lower endocannabinoid levels during PFs. After 20 parabolas, blood anandamide levels had dropped significantly in volunteers with motion sickness (from 0.39±0.40 to 0.22±0.25 ng/ml) but increased in participants without the condition (from 0.43±0.23 to 0.60±0.38 ng/ml) resulting in significantly higher anandamide levels in participants without motion sickness (p = 0.02). 2-AG levels in individuals with motion sickness were low and almost unchanged throughout the experiment but showed a robust increase in participants without motion sickness. Cannabinoid-receptor 1 (CB1) but not cannabinoid-receptor 2 (CB2) mRNA expression in leucocytes 4 h after the experiment was significantly lower in volunteers with motion sickness than in participants without N&V. Conclusions/Significance These findings demonstrate that stress and motion sickness in humans are associated with impaired endocannabinoid activity. Enhancing ECS signaling may represent an alternative therapeutic strategy for motion sickness in individuals who do not respond to currently available treatments.
Collapse
Affiliation(s)
- Alexander Choukèr
- Department of Anesthesiology, Ludwig-Maximilians-University, Munich, Germany
| | - Ines Kaufmann
- Department of Anesthesiology, Ludwig-Maximilians-University, Munich, Germany
| | - Simone Kreth
- Department of Anesthesiology, Ludwig-Maximilians-University, Munich, Germany
| | - Daniela Hauer
- Department of Anesthesiology, Ludwig-Maximilians-University, Munich, Germany
| | - Matthias Feuerecker
- Department of Anesthesiology, Ludwig-Maximilians-University, Munich, Germany
| | - Detlef Thieme
- Institute of Doping Analysis and Sports Biochemistry, Dresden, Germany
| | - Michael Vogeser
- Department of Clinical Chemistry, Ludwig-Maximilians-University, Munich, Germany
| | - Manfred Thiel
- Department of Anaesthesiology and Intensive Care, Medical Faculty Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Gustav Schelling
- Department of Anesthesiology, Ludwig-Maximilians-University, Munich, Germany
- * E-mail:
| |
Collapse
|
50
|
Izzo AA, Sharkey KA. Cannabinoids and the gut: new developments and emerging concepts. Pharmacol Ther 2010; 126:21-38. [PMID: 20117132 DOI: 10.1016/j.pharmthera.2009.12.005] [Citation(s) in RCA: 309] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 12/24/2009] [Indexed: 12/11/2022]
Abstract
Cannabis has been used to treat gastrointestinal (GI) conditions that range from enteric infections and inflammatory conditions to disorders of motility, emesis and abdominal pain. The mechanistic basis of these treatments emerged after the discovery of Delta(9)-tetrahydrocannabinol as the major constituent of Cannabis. Further progress was made when the receptors for Delta(9)-tetrahydrocannabinol were identified as part of an endocannabinoid system, that consists of specific cannabinoid receptors, endogenous ligands and their biosynthetic and degradative enzymes. Anatomical, physiological and pharmacological studies have shown that the endocannabinoid system is widely distributed throughout the gut, with regional variation and organ-specific actions. It is involved in the regulation of food intake, nausea and emesis, gastric secretion and gastroprotection, GI motility, ion transport, visceral sensation, intestinal inflammation and cell proliferation in the gut. Cellular targets have been defined that include the enteric nervous system, epithelial and immune cells. Molecular targets of the endocannabinoid system include, in addition to the cannabinoid receptors, transient receptor potential vanilloid 1 receptors, peroxisome proliferator-activated receptor alpha receptors and the orphan G-protein coupled receptors, GPR55 and GPR119. Pharmacological agents that act on these targets have been shown in preclinical models to have therapeutic potential. Here, we discuss cannabinoid receptors and their localization in the gut, the proteins involved in endocannabinoid synthesis and degradation and the presence of endocannabinoids in the gut in health and disease. We focus on the pharmacological actions of cannabinoids in relation to GI disorders, highlighting recent data on genetic mutations in the endocannabinoid system in GI disease.
Collapse
Affiliation(s)
- Angelo A Izzo
- Department of Experimental Pharmacology, University of Naples Federico II and Endocannabinoid Research Group, Naples, Italy.
| | | |
Collapse
|