1
|
Bobrich M, Schwarz R, Ramer R, Borchert P, Hinz B. A simple LC-MS/MS method for the simultaneous quantification of endocannabinoids in biological samples. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1161:122371. [DOI: 10.1016/j.jchromb.2020.122371] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/30/2020] [Accepted: 09/02/2020] [Indexed: 01/15/2023]
|
2
|
Dato FM, Neudörfl JM, Gütschow M, Goldfuss B, Pietsch M. ω-Quinazolinonylalkyl aryl ureas as reversible inhibitors of monoacylglycerol lipase. Bioorg Chem 2019; 94:103352. [PMID: 31668797 DOI: 10.1016/j.bioorg.2019.103352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 02/07/2023]
Abstract
The serine hydrolase monoacylglycerol lipase (MAGL) is involved in a plethora of pathological conditions, in particular pain and inflammation, various types of cancer, metabolic, neurological and cardiovascular disorders, and is therefore a promising target for drug development. Although a large number of irreversible-acting MAGL inhibitors have been discovered over the past years, there are only few compounds known so far which inhibit the enzyme in a reversible manner. Therefore, much effort is put into the development of novel chemical entities showing reversible inhibitory behavior, which is thought to cause less undesired side effects. To explore a wide range of chemical structures as MAGL binders, we have applied a virtual screening approach by docking small molecules into the crystal structure of human MAGL (hMAGL) and envisaged a library of 45 selected compounds which were then synthesized. Biochemical investigations included the determination of the inhibitory potency on hMAGL and two related hydrolases, i.e. human fatty acid amide hydrolase (hFAAH) and murine cholesterol esterase (mCEase). The most promising candidates from theses analyses, i.e. three ω-quinazolinonylalkyl aryl ureas bearing alkyl spacers of three to five methylene groups, exhibited IC50 values of 20-41 µM and reversible, detergent-insensitive behavior towards hMAGL. Among these compounds, the inhibitor 1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(4-oxo-3,4-dihydroquinazolin-2-yl)butyl)urea (96) was selected for further kinetic characterization, yielding a dissociation constant Ki = 15.4 µM and a mixed-type inhibition with a pronounced competitive component (α = 8.94). This mode of inhibition was further supported by a docking experiment, which suggested that the inhibitor occupies the substrate binding pocket of hMAGL.
Collapse
Affiliation(s)
- Florian M Dato
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931 Cologne, Germany; Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939 Cologne, Germany
| | - Jörg-Martin Neudörfl
- Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939 Cologne, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Bernd Goldfuss
- Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939 Cologne, Germany.
| | - Markus Pietsch
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931 Cologne, Germany.
| |
Collapse
|
3
|
Chen Z, Mori W, Deng X, Cheng R, Ogasawara D, Zhang G, Schafroth MA, Dahl K, Fu H, Hatori A, Shao T, Zhang Y, Yamasaki T, Zhang X, Rong J, Yu Q, Hu K, Fujinaga M, Xie L, Kumata K, Gou Y, Chen J, Gu S, Bao L, Wang L, Lee Collier T, Vasdev N, Shao Y, Ma JA, Cravatt BF, Fowler C, Josephson L, Zhang MR, Liang SH. Design, Synthesis, and Evaluation of Reversible and Irreversible Monoacylglycerol Lipase Positron Emission Tomography (PET) Tracers Using a "Tail Switching" Strategy on a Piperazinyl Azetidine Skeleton. J Med Chem 2019; 62:3336-3353. [PMID: 30829483 PMCID: PMC6581563 DOI: 10.1021/acs.jmedchem.8b01778] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Monoacylglycerol lipase (MAGL) is a serine hydrolase that degrades 2-arachidonoylglycerol (2-AG) in the endocannabinoid system (eCB). Selective inhibition of MAGL has emerged as a potential therapeutic approach for the treatment of diverse pathological conditions, including chronic pain, inflammation, cancer, and neurodegeneration. Herein, we disclose a novel array of reversible and irreversible MAGL inhibitors by means of "tail switching" on a piperazinyl azetidine scaffold. We developed a lead irreversible-binding MAGL inhibitor 8 and reversible-binding compounds 17 and 37, which are amenable for radiolabeling with 11C or 18F. [11C]8 ([11C]MAGL-2-11) exhibited high brain uptake and excellent binding specificity in the brain toward MAGL. Reversible radioligands [11C]17 ([11C]PAD) and [18F]37 ([18F]MAGL-4-11) also demonstrated excellent in vivo binding specificity toward MAGL in peripheral organs. This work may pave the way for the development of MAGL-targeted positron emission tomography tracers with tunability in reversible and irreversible binding mechanisms.
Collapse
Affiliation(s)
- Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
- Department of Chemistry, School of Science, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Wakana Mori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Xiaoyun Deng
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Ran Cheng
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Daisuke Ogasawara
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Genwei Zhang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Michael A. Schafroth
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Kenneth Dahl
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Hualong Fu
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Akiko Hatori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Tuo Shao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yiding Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Tomoteru Yamasaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Xiaofei Zhang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Qingzhen Yu
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Kuan Hu
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Masayuki Fujinaga
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Lin Xie
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Katsushi Kumata
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Yuancheng Gou
- ChemShuttle, Inc., 1699 Huishan Blvd., Wuxi, Jiangsu, 214174, China
| | - Jingjin Chen
- ChemShuttle, Inc., 1699 Huishan Blvd., Wuxi, Jiangsu, 214174, China
| | - Shuyin Gu
- ChemShuttle, Inc., 1699 Huishan Blvd., Wuxi, Jiangsu, 214174, China
| | - Liang Bao
- ChemShuttle, Inc., 1699 Huishan Blvd., Wuxi, Jiangsu, 214174, China
| | - Lu Wang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Thomas Lee Collier
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Yihan Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Jun-An Ma
- Department of Chemistry, School of Science, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Benjamin F. Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Christopher Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
4
|
Szeremeta J, Karlsson J, Alhouayek M, Fowler CJ. Low mRNA expression and activity of monoacylglycerol lipase in human SH-SY5Y neuroblastoma cells. Prostaglandins Other Lipid Mediat 2019; 142:59-67. [PMID: 30978461 DOI: 10.1016/j.prostaglandins.2019.04.003] [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] [Received: 01/24/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 01/05/2023]
Abstract
Relatively little is known about the endocannabinoid system in human neuroblastoma cell lines. In the present study, we have investigated the expression of the genes coding for the enzymes involved in the synthesis and catabolism of endocannabinoids in the SH-SY5Y cell line. The expression of MGLL, the gene coding for the 2-arachidonoylglycerol hydrolytic enzyme monoacylglycerol lipase (MAGL), was found to be 85 and 340 fold lower than the expression levels for the genes coding for alpha/beta-hydrolase domain containing 6 and 12 (ABHD6, ABHD12), which are alternative hydrolytic enzymes for this endocannabinoid. In comparison, mRNA levels of MGLL were 1.5 fold higher than ABHD6 and 2 fold lower than the levels of ABHD12 in DU-145 human prostate cells. In functional assays, the hydrolysis of the 2-arachidonoylglycerol homologue 2-oleoylglycerol by intact SH-SY5Y cells was partially inhibited by the ABHD6 inhibitor WWL70, but not by the MAGL inhibitor JZL184, whereas the reverse was true in DU-145 cells. The combination of JZL184 + WWL70 did, however produce a significantly greater inhibition of 2-OG hydrolysis than seen with WWL70 alone in the SH-SY5Y cells. The low MGLL expression in the SH-SY5Y cells was not due to epigenetic silencing, since levels were not affected by treatment with the methylation inhibitor 5-aza-2'-deoxycytidine and/or the histone acetylase inhibitor trichostatin A. The low MGLL expression in SH-SY5Y cells should be taken into account when using these cells in experiments investigating the involvement of the endocannabinoid system in models of physiological and pathological processes.
Collapse
Affiliation(s)
- Janis Szeremeta
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Jessica Karlsson
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Mireille Alhouayek
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Christopher J Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden.
| |
Collapse
|
5
|
Dato FM, Sheikh M, Uhl RZ, Schüller AW, Steinkrüger M, Koch P, Neudörfl JM, Gütschow M, Goldfuss B, Pietsch M. ω-Phthalimidoalkyl Aryl Ureas as Potent and Selective Inhibitors of Cholesterol Esterase. ChemMedChem 2018; 13:1833-1847. [PMID: 30004170 DOI: 10.1002/cmdc.201800388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/06/2018] [Indexed: 11/09/2022]
Abstract
Cholesterol esterase (CEase), a serine hydrolase thought to be involved in atherogenesis and thus coronary heart disease, is considered as a target for inhibitor development. We investigated recombinant human and murine CEases with a new fluorometric assay in a structure-activity relationship study of a small library of ω-phthalimidoalkyl aryl ureas. The urea motif with an attached 3,5-bis(trifluoromethyl)phenyl group and the aromatic character of the ω-phthalimide residue were most important for inhibitory activity. In addition, an alkyl chain composed of three or four methylene groups, connecting the urea and phthalimide moieties, was found to be an optimal spacer for inhibitors. The so-optimized compounds 2 [1-(3,5-bis(trifluoromethyl)phenyl)-3-(3-(1,3-dioxoisoindolin-2-yl)propyl)urea] and 21 [1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(1,3-dioxoisoindolin-2-yl)butyl)urea] exhibited dissociation constants (Ki ) of 1-19 μm on the two CEases and showed either a competitive (2 on the human enzyme and 21 on the murine enzyme) or a noncompetitive mode of inhibition. Two related serine hydrolases-monoacylglycerol lipase and fatty acid amide hydrolase-were inhibited by ω-phthalimidoalkyl aryl ureas to a lesser extent.
Collapse
Affiliation(s)
- Florian M Dato
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany.,Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939, Cologne, Germany
| | - Miriam Sheikh
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| | - Rocky Z Uhl
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| | - Alexandra W Schüller
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany.,Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939, Cologne, Germany
| | - Michaela Steinkrüger
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| | - Peter Koch
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| | - Jörg-Martin Neudörfl
- Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939, Cologne, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Bernd Goldfuss
- Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939, Cologne, Germany
| | - Markus Pietsch
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| |
Collapse
|
6
|
Cheng R, Mori W, Ma L, Alhouayek M, Hatori A, Zhang Y, Ogasawara D, Yuan G, Chen Z, Zhang X, Shi H, Yamasaki T, Xie L, Kumata K, Fujinaga M, Nagai Y, Minamimoto T, Svensson M, Wang L, Du Y, Ondrechen MJ, Vasdev N, Cravatt BF, Fowler C, Zhang MR, Liang SH. In Vitro and in Vivo Evaluation of 11C-Labeled Azetidinecarboxylates for Imaging Monoacylglycerol Lipase by PET Imaging Studies. J Med Chem 2018; 61:2278-2291. [PMID: 29481079 PMCID: PMC5966020 DOI: 10.1021/acs.jmedchem.7b01400] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Monoacylglycerol lipase (MAGL) is the principle enzyme for metabolizing endogenous cannabinoid ligand 2-arachidonoyglycerol (2-AG). Blockade of MAGL increases 2-AG levels, resulting in subsequent activation of the endocannabinoid system, and has emerged as a novel therapeutic strategy to treat drug addiction, inflammation, and neurodegenerative diseases. Herein we report a new series of MAGL inhibitors, which were radiolabeled by site-specific labeling technologies, including 11C-carbonylation and spirocyclic iodonium ylide (SCIDY) radiofluorination. The lead compound [11C]10 (MAGL-0519) demonstrated high specific binding and selectivity in vitro and in vivo. We also observed unexpected washout kinetics with these irreversible radiotracers, in which in vivo evidence for turnover of the covalent residue was unveiled between MAGL and azetidine carboxylates. This work may lead to new directions for drug discovery and PET tracer development based on azetidine carboxylate inhibitor scaffold.
Collapse
Affiliation(s)
- Ran Cheng
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Wakana Mori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Longle Ma
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
| | - Mireille Alhouayek
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Akiko Hatori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Yiding Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Daisuke Ogasawara
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Gengyang Yuan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
- Department of Chemistry & Chemical Biology, Northeastern University, 360 Huntington Ave., Boston, MA, 02115, USA
| | - Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
| | - Xiaofei Zhang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
| | - Hang Shi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
| | - Tomoteru Yamasaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Lin Xie
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Katsushi Kumata
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Masayuki Fujinaga
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Yuji Nagai
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Takafumi Minamimoto
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Mona Svensson
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Lu Wang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
| | - Yunfei Du
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Mary Jo Ondrechen
- Department of Chemistry & Chemical Biology, Northeastern University, 360 Huntington Ave., Boston, MA, 02115, USA
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
| | - Benjamin F. Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, SR107 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Christopher Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
7
|
Schwarz R, Ramer R, Hinz B. Targeting the endocannabinoid system as a potential anticancer approach. Drug Metab Rev 2018; 50:26-53. [PMID: 29390896 DOI: 10.1080/03602532.2018.1428344] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The endocannabinoid system is currently under intense investigation due to the therapeutic potential of cannabinoid-based drugs as treatment options for a broad variety of diseases including cancer. Besides the canonical endocannabinoid system that includes the cannabinoid receptors CB1 and CB2 and the endocannabinoids N-arachidonoylethanolamine (anandamide) and 2-arachidonoylglycerol, recent investigations suggest that other fatty acid derivatives, receptors, enzymes, and lipid transporters likewise orchestrate this system as components of the endocannabinoid system when defined as an extended signaling network. As such, fatty acids acting at cannabinoid receptors (e.g. 2-arachidonoyl glyceryl ether [noladin ether], N-arachidonoyldopamine) as well as endocannabinoid-like substances that do not elicit cannabinoid receptor activation (e.g. N-palmitoylethanolamine, N-oleoylethanolamine) have raised interest as anticancerogenic substances. Furthermore, the endocannabinoid-degrading enzymes fatty acid amide hydrolase and monoacylglycerol lipase, lipid transport proteins of the fatty acid binding protein family, additional cannabinoid-activated G protein-coupled receptors, members of the transient receptor potential family as well as peroxisome proliferator-activated receptors have been considered as targets of antitumoral cannabinoid activity. Therefore, this review focused on the antitumorigenic effects induced upon modulation of this extended endocannabinoid network.
Collapse
Affiliation(s)
- Rico Schwarz
- a Institute of Pharmacology and Toxicology , Rostock University Medical Center , Rostock , Germany
| | - Robert Ramer
- a Institute of Pharmacology and Toxicology , Rostock University Medical Center , Rostock , Germany
| | - Burkhard Hinz
- a Institute of Pharmacology and Toxicology , Rostock University Medical Center , Rostock , Germany
| |
Collapse
|
8
|
Sierra S, Luquin N, Navarro-Otano J. The endocannabinoid system in cardiovascular function: novel insights and clinical implications. Clin Auton Res 2017; 28:35-52. [PMID: 29222605 DOI: 10.1007/s10286-017-0488-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022]
Abstract
RATIONALE Cardiovascular disease is now recognized as the number one cause of death in the world, and the size of the population at risk continues to increase rapidly. The dysregulation of the endocannabinoid (eCB) system plays a central role in a wide variety of conditions including cardiovascular disorders. Cannabinoid receptors, their endogenous ligands, as well as enzymes conferring their synthesis and degradation, exhibit overlapping distributions in the cardiovascular system. Furthermore, the pharmacological manipulation of the eCB system has effects on blood pressure, cardiac contractility, and endothelial vasomotor control. Growing evidence from animal studies supports the significance of the eCB system in cardiovascular disorders. OBJECTIVE To summarize the literature surrounding the eCB system in cardiovascular function and disease and the new compounds that may potentially extend the range of available interventions. RESULTS Drugs targeting CB1R, CB2R, TRPV1 and PPARs are proven effective in animal models mimicking cardiovascular disorders such as hypertension, atherosclerosis and myocardial infarction. Despite the setback of two clinical trials that exhibited unexpected harmful side-effects, preclinical studies are accelerating the development of more selective drugs with promising results devoid of adverse effects. CONCLUSION Over the last years, increasing evidence from basic and clinical research supports the role of the eCB system in cardiovascular function. Whereas new discoveries are paving the way for the identification of novel drugs and therapeutic targets, the close cooperation of researchers, clinicians and pharmaceutical companies is needed to achieve successful outcomes.
Collapse
Affiliation(s)
- Salvador Sierra
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Physiology and Biophysics, Molecular Medicine Research Building, Virginia Commonwealth University, 1220 East Broad Street, Richmond, VA, 23298, USA.
| | - Natasha Luquin
- Department of Medical Genomics, Royal Prince Alfred Hospital, Sydney, Australia
| | - Judith Navarro-Otano
- Neurology Service, Electromyography, Motor Control and Neuropathic Pain Unit, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Grabiec U, Dehghani F. N-Arachidonoyl Dopamine: A Novel Endocannabinoid and Endovanilloid with Widespread Physiological and Pharmacological Activities. Cannabis Cannabinoid Res 2017; 2:183-196. [PMID: 29082315 PMCID: PMC5627668 DOI: 10.1089/can.2017.0015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
N-arachidonoyl dopamine (NADA) is a member of the family of endocannabinoids to which several other N-acyldopamines belong as well. Their activity is mediated through various targets that include cannabinoid receptors or transient receptor potential vanilloid (TRPV)1. Synthesis and degradation of NADA are not yet fully understood. Nonetheless, there is evidence that NADA plays an important role in nociception and inflammation in the central and peripheral nervous system. The TRPV1 receptor, for which NADA is a potent agonist, was shown to be an endogenous transducer of noxious heat. Moreover, it has been demonstrated that NADA exerts protective and antioxidative properties in microglial cell cultures, cortical neurons, and organotypical hippocampal slice cultures. NADA is present in very low concentrations in the brain and is seemingly not involved in activation of the classical pathways. We believe that treatment with exogenous NADA during and after injury might be beneficial. This review summarizes the recent findings on biochemical properties of NADA and other N-acyldopamines and their role in physiological and pathological processes. These findings provide strong evidence that NADA is an effective agent to manage neuroinflammatory diseases or pain and can be useful in designing novel therapeutic strategies.
Collapse
Affiliation(s)
- Urszula Grabiec
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
10
|
Redmond WJ, Cawston EE, Grimsey NL, Stuart J, Edington AR, Glass M, Connor M. Identification of N-arachidonoyl dopamine as a highly biased ligand at cannabinoid CB1 receptors. Br J Pharmacol 2016; 173:115-27. [PMID: 26398720 PMCID: PMC4813372 DOI: 10.1111/bph.13341] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 09/13/2015] [Accepted: 09/17/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE N-arachidonyl dopamine (NADA) has been identified as a putative endocannabinoid, but there is little information about which signalling pathways it activates. The purpose of this study was to identify the signalling pathways activated by NADA in vitro. EXPERIMENTAL APPROACH Human or rat cannabinoid CB1 receptors were expressed in AtT20, CHO or HEK 293 cells. NADA displacement of radiolabelled cannabinoids, and CB1 receptor mediated activation of K channels or ERK phosphorylation, release of intracellular calcium ([Ca]i ) and modulation of adenylyl cyclase were measured in addition to NADA effects on CB1 receptor trafficking. KEY RESULTS At concentrations up to 30 μM, NADA failed to activate any signalling pathways via CB1 receptors, with the exception of mobilization of [Ca]i . The elevations of [Ca]i were insensitive to pertussis toxin, and reduced or abolished by blockers of Gq /11 -dependent processes including U73122, thapsigargin and a peptide antagonist of Gq /11 activation. Prolonged NADA incubation produced modest loss of cell surface CB1 receptors. The prototypical cannabinoid agonist CP55940 signalled as expected in all assays. CONCLUSIONS AND IMPLICATIONS NADA is an ineffective agonist at most canonical cannabinoid receptor signalling pathways, but did promote mobilization of [Ca]i via Gq -dependent processes and some CB1 receptor trafficking. This signalling profile is distinct from that of any known cannabinoid, and suggests that NADA may have a unique spectrum of effects in vivo. Our results also indicate that it may be possible to identify highly biased CB1 receptor ligands displaying a subset of the pharmacological or therapeutic effects usually attributed to CB1 ligands.
Collapse
Affiliation(s)
- William J. Redmond
- Department of Biomedical Sciences, Faculty of Medicine and Health SciencesMacquarie UniversityNSW2109Australia
| | - Erin E. Cawston
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain ResearchUniversity of AucklandAucklandNew Zealand
| | - Natasha L. Grimsey
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain ResearchUniversity of AucklandAucklandNew Zealand
| | - Jordyn Stuart
- Department of Biomedical Sciences, Faculty of Medicine and Health SciencesMacquarie UniversityNSW2109Australia
| | - Amelia R. Edington
- Department of Biomedical Sciences, Faculty of Medicine and Health SciencesMacquarie UniversityNSW2109Australia
| | - Michelle Glass
- Department of Pharmacology and Clinical Pharmacology, and Centre for Brain ResearchUniversity of AucklandAucklandNew Zealand
| | - Mark Connor
- Department of Biomedical Sciences, Faculty of Medicine and Health SciencesMacquarie UniversityNSW2109Australia
| |
Collapse
|
11
|
Del Carlo S, Manera C, Chicca A, Arena C, Bertini S, Burgalassi S, Tampucci S, Gertsch J, Macchia M, Saccomanni G. Development of an HPLC/UV assay for the evaluation of inhibitors of human recombinant monoacylglycerol lipase. J Pharm Biomed Anal 2015; 108:113-21. [DOI: 10.1016/j.jpba.2015.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 01/30/2015] [Accepted: 02/05/2015] [Indexed: 02/04/2023]
|
12
|
Savinainen JR, Kansanen E, Pantsar T, Navia-Paldanius D, Parkkari T, Lehtonen M, Laitinen T, Nevalainen T, Poso A, Levonen AL, Laitinen JT. Robust hydrolysis of prostaglandin glycerol esters by human monoacylglycerol lipase (MAGL). Mol Pharmacol 2014; 86:522-35. [PMID: 25140003 DOI: 10.1124/mol.114.094284] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The primary route of inactivation of the endocannabinoid 2-arachidonoylglycerol in the central nervous system is through enzymatic hydrolysis, mainly carried out by monoacylglycerol lipase (MAGL), along with a small contribution by the α/β-hydrolase domain (ABHD) proteins ABHD6 and ABHD12. Recent methodological progress allowing kinetic monitoring of glycerol liberation has facilitated substrate profiling of the human endocannabinoid hydrolases, and these studies have revealed that the three enzymes have distinct monoacylglycerol substrate and isomer preferences. Here, we have extended this substrate profiling to cover four prostaglandin glycerol esters, namely, 15-deoxy-Δ(12,14)-prostaglandin J2-2-glycerol (15d-PGJ2-G), PGD2-G, PGE2-G, and PGF2 α-G. We found that the three enzymes hydrolyzed the tested substrates, albeit with distinct rates and preferences. Although human ABHD12 (hABHD12) showed only marginal activity toward PGE2-G, hABHD6 preferentially hydrolyzed PGD2-G, and human MAGL (hMAGL) robustly hydrolyzed all four. This was particularly intriguing for MAGL activity toward 15d-PGJ2-G whose hydrolysis rate rivaled that of the best monoacylglycerol substrates. Molecular modeling studies combined with kinetic analysis supported favorable interaction with the hMAGL active site. Long and short MAGL isoforms shared a similar substrate profile, and hMAGL hydrolyzed 15d-PGJ2-G also in living cells. The ability of 15d-PGJ2-G to activate the canonical nuclear factor erythroid 2-related factor (Nrf2) signaling pathway used by 15d-PGJ2 was assessed, and these studies revealed for the first time that 15d-PGJ2 and 15d-PGJ2-G similarly activated Nrf2 signaling as well as transcription of target genes of this pathway. Our study challenges previous claims regarding the ability of MAGL to catalyze PG-G hydrolysis and extend the MAGL substrate profile beyond the classic monoacylglycerols.
Collapse
Affiliation(s)
- Juha R Savinainen
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Emilia Kansanen
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tatu Pantsar
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Dina Navia-Paldanius
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Teija Parkkari
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Marko Lehtonen
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tuomo Laitinen
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tapio Nevalainen
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Antti Poso
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anna-Liisa Levonen
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jarmo T Laitinen
- School of Medicine, Institute of Biomedicine (J.R.S., D.N-P., Te.P., J.T.L.), A.I. Virtanen Institute for Molecular Sciences (E.K., A-L.L.), School of Pharmacy (Ta.P., Te.P., M.L., T.L., T.N., A.P.), Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
13
|
Nithipatikom K, Endsley MP, Pfeiffer AW, Falck JR, Campbell WB. A novel activity of microsomal epoxide hydrolase: metabolism of the endocannabinoid 2-arachidonoylglycerol. J Lipid Res 2014; 55:2093-102. [PMID: 24958911 DOI: 10.1194/jlr.m051284] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Microsomal epoxide hydrolase (EPHX1, EC 3.3.2.9) is a highly abundant α/β-hydrolase enzyme that is known for its catalytical epoxide hydrolase activity. A wide range of EPHX1 functions have been demonstrated including xenobiotic metabolism; however, characterization of its endogenous substrates is limited. In this study, we present evidence that EPHX1 metabolizes the abundant endocannabinoid 2-arachidonoylglycerol (2-AG) to free arachidonic acid (AA) and glycerol. The EPHX1 metabolism of 2-AG was demonstrated using commercially available EPHX1 microsomes as well as PC-3 cells overexpressing EPHX1. Conversely, EPHX1 siRNA markedly reduced the EPHX1 expression and 2-AG metabolism in HepG2 cells and LNCaP cells. A selective EPHX1 inhibitor, 10-hydroxystearamide, inhibited 2-AG metabolism and hydrolysis of a well-known EPHX1 substrate, cis-stilbene oxide. Among the inhibitors studied, a serine hydrolase inhibitor, methoxy-arachidonyl fluorophosphate, was the most potent inhibitor of 2-AG metabolism by EPHX1 microsomes. These results demonstrate that 2-AG is an endogenous substrate for EPHX1, a potential role of EPHX1 in the endocannabinoid signaling and a new AA biosynthetic pathway.
Collapse
Affiliation(s)
- Kasem Nithipatikom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Michael P Endsley
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Adam W Pfeiffer
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - John R Falck
- Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - William B Campbell
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
14
|
Wilhelmsen K, Khakpour S, Tran A, Sheehan K, Schumacher M, Xu F, Hellman J. The endocannabinoid/endovanilloid N-arachidonoyl dopamine (NADA) and synthetic cannabinoid WIN55,212-2 abate the inflammatory activation of human endothelial cells. J Biol Chem 2014; 289:13079-100. [PMID: 24644287 DOI: 10.1074/jbc.m113.536953] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Although cannabinoids, such as Δ(9)-tetrahydrocannabinol, have been studied extensively for their psychoactive effects, it has become apparent that certain cannabinoids possess immunomodulatory activity. Endothelial cells (ECs) are centrally involved in the pathogenesis of organ injury in acute inflammatory disorders, such as sepsis, because they express cytokines and chemokines, which facilitate the trafficking of leukocytes to organs, and they modulate vascular barrier function. In this study, we find that primary human ECs from multiple organs express the cannabinoid receptors CB1R, GPR18, and GPR55, as well as the ion channel transient receptor potential cation channel vanilloid type 1. In contrast to leukocytes, CB2R is only minimally expressed in some EC populations. Furthermore, we show that ECs express all of the known endocannabinoid (eCB) metabolic enzymes. Examining a panel of cannabinoids, we demonstrate that the synthetic cannabinoid WIN55,212-2 and the eCB N-arachidonoyl dopamine (NADA), but neither anandamide nor 2-arachidonoylglycerol, reduce EC inflammatory responses induced by bacterial lipopeptide, LPS, and TNFα. We find that endothelial CB1R/CB2R are necessary for the effects of NADA, but not those of WIN55,212-2. Furthermore, transient receptor potential cation channel vanilloid type 1 appears to counter the anti-inflammatory properties of WIN55,212-2 and NADA, but conversely, in the absence of these cannabinoids, its inhibition exacerbates the inflammatory response in ECs activated with LPS. These data indicate that the eCB system can modulate inflammatory activation of the endothelium and may have important implications for a variety of acute inflammatory disorders that are characterized by EC activation.
Collapse
|
15
|
Woodhams SG, Wong A, Barrett DA, Bennett AJ, Chapman V, Alexander SPH. Spinal administration of the monoacylglycerol lipase inhibitor JZL184 produces robust inhibitory effects on nociceptive processing and the development of central sensitization in the rat. Br J Pharmacol 2013; 167:1609-19. [PMID: 22924700 DOI: 10.1111/j.1476-5381.2012.02179.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 08/07/2012] [Accepted: 08/13/2012] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The cannabinoid receptor-mediated analgesic effects of 2-arachidonoylglycerol (2-AG) are limited by monoacylglycerol lipase (MAGL). 4-nitrophenyl 4-[bis (1,3-benzodioxol-5-yl) (hydroxy) methyl] piperidine-1-carboxylate (JZL184) is a potent inhibitor of MAGL in the mouse, though potency is reportedly reduced in the rat. Here we have assessed the effects of spinal inhibition of MAGL with JZL184 on nociceptive processing in rats. EXPERIMENTAL APPROACH In vivo spinal electrophysiological assays in anaesthetized rats were used to determine the effects of spinal administration of JZL184 on spinal nociceptive processing in the presence and absence of hindpaw inflammation. Contributions of CB(1) receptors to these effects was assessed with AM251. Inhibition of 2-oleoylglycerol hydrolytic activity and alterations of 2-AG in the spinal cord after JZL 184 were also assessed. KEY RESULTS Spinal JZL184 dose-dependently inhibited mechanically evoked responses of wide dynamic range (WDR) neurones in naïve anaesthetized rats, in part via the CB(1) receptor. A single spinal administration of JZL184 abolished inflammation-induced expansion of the receptive fields of spinal WDR neurones. However, neither spinal nor systemic JZL184 altered levels of 2-AG, or 2-oleoylglycerol hydrolytic activity in the spinal cord, although JZL184 displayed robust inhibition of MAGL when incubated with spinal cord tissue in vitro. CONCLUSIONS AND IMPLICATIONS JZL184 exerted robust anti-nociceptive effects at the level of the spinal cord in vivo and inhibited rat spinal cord MAGL activity in vitro. The discordance between in vivo and in vitro assays suggests that localized sites of action of JZL184 produce these profound functional inhibitory effects. LINKED ARTICLES This article is part of a themed section on Cannabinoids. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.167.issue-8.
Collapse
Affiliation(s)
- S G Woodhams
- School of Biomedical Sciences, University of Nottingham Medical School, Nottingham, UK
| | | | | | | | | | | |
Collapse
|
16
|
Chicca A, Marazzi J, Gertsch J. The antinociceptive triterpene β-amyrin inhibits 2-arachidonoylglycerol (2-AG) hydrolysis without directly targeting cannabinoid receptors. Br J Pharmacol 2013; 167:1596-608. [PMID: 22646533 DOI: 10.1111/j.1476-5381.2012.02059.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Pharmacological activation of cannabinoid CB(1) and CB(2) receptors is a therapeutic strategy to treat chronic and inflammatory pain. It was recently reported that a mixture of natural triterpenes α- and β-amyrin bound selectively to CB(1) receptors with a subnanomolar K(i) value (133 pM). Orally administered α/β-amyrin inhibited inflammatory and persistent neuropathic pain in mice through both CB(1) and CB(2) receptors. Here, we investigated effects of amyrins on the major components of the endocannabinoid system. EXPERIMENTAL APPROACH We measured CB receptor binding interactions of α- and β-amyrin in validated binding assays using hCB(1) and hCB(2) transfected CHO-K1 cells. Effects on endocannabinoid transport in U937 cells and breakdown using homogenates of BV2 cells and pig brain, as well as purified enzymes, were also studied. KEY RESULTS There was no binding of either α- or β-amyrin to hCB receptors in our assays (K(i) > 10 µM). The triterpene β-amyrin potently inhibited 2-arachidonoyl glycerol (2-AG) hydrolysis in pig brain homogenates, but not that of anandamide. Although β-amyrin only weakly inhibited purified human monoacylglycerol lipase (MAGL), it also inhibited α,β-hydrolases and more potently inhibited 2-AG breakdown than α-amyrin and the MAGL inhibitor pristimerin in BV2 cell and pig brain homogenates. CONCLUSIONS AND IMPLICATIONS We propose that β-amyrin exerts its analgesic and anti-inflammatory pharmacological effects via indirect cannabimimetic mechanisms by inhibiting the degradation of the endocannabinoid 2-AG without interacting directly with CB receptors. Triterpenoids appear to offer a very broad and largely unexplored scaffold for inhibitors of the enzymic degradation of 2-AG. LINKED ARTICLES This article is part of a themed section on Cannabinoids. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.167.issue-8.
Collapse
Affiliation(s)
- A Chicca
- Institute of Biochemistry and Molecular Medicine, National Centre of Competence in Research NCCR TransCure, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
17
|
Abstract
The endocannabinoid (eCB) system is involved in processes as diverse as control of appetite, perception of pain and the limitation of cancer cell growth and invasion. The enzymes responsible for eCB breakdown are attractive pharmacological targets, and fatty acid amide hydrolase inhibitors, which potentiate the levels of the eCB anandamide, are now undergoing pharmaceutical development. 'Drugable' selective inhibitors of monoacylglycerol lipase, a key enzyme regulating the levels of the other main eCB, 2-arachidonoylglycerol, were however not identified until very recently. Their availability has resulted in a large expansion of our knowledge concerning the pharmacological consequences of monoacylglycerol lipase inhibition and hence the role(s) played by the enzyme in the body. In this review, the pharmacology of monoacylglycerol lipase will be discussed, together with an analysis of the therapeutic potential of monoacylglycerol lipase inhibitors as analgesics and anticancer agents.
Collapse
Affiliation(s)
- C J Fowler
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Sweden.
| |
Collapse
|
18
|
Chicca A, Marazzi J, Nicolussi S, Gertsch J. Evidence for bidirectional endocannabinoid transport across cell membranes. J Biol Chem 2012; 287:34660-82. [PMID: 22879589 DOI: 10.1074/jbc.m112.373241] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Despite extensive research on the trafficking of anandamide (AEA) across cell membranes, little is known about the membrane transport of other endocannabinoids, such as 2-arachidonoylglycerol (2-AG). Previous studies have provided data both in favor and against a cell membrane carrier-mediated transport of endocannabinoids, using different methodological approaches. Because AEA and 2-AG undergo rapid and almost complete intracellular hydrolysis, we employed a combination of radioligand assays and absolute quantification of cellular and extracellular endocannabinoid levels. In human U937 leukemia cells, 100 nm AEA and 1 μm 2-AG were taken up through a fast and saturable process, reaching a plateau after 5 min. Employing differential pharmacological blockage of endocannabinoid uptake, breakdown, and interaction with intracellular binding proteins, we show that eicosanoid endocannabinoids harboring an arachidonoyl chain compete for a common membrane target that regulates their transport, whereas other N-acylethanolamines did not interfere with AEA and 2-AG uptake. By combining fatty acid amide hydrolase or monoacyl glycerol lipase inhibitors with hydrolase-inactive concentrations of the AEA transport inhibitors UCM707 (1 μm) and OMDM-2 (5 μm), a functional synergism on cellular AEA and 2-AG uptake was observed. Intriguingly, structurally unrelated AEA uptake inhibitors also blocked the cellular release of AEA and 2-AG. We show, for the first time, that UCM707 and OMDM-2 inhibit the bidirectional movement of AEA and 2-AG across cell membranes. Our findings suggest that a putative endocannabinoid cell membrane transporter controls the cellular AEA and 2-AG trafficking and metabolism.
Collapse
Affiliation(s)
- Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research TransCure, University of Bern, CH-3012 Bern, Switzerland
| | | | | | | |
Collapse
|
19
|
Cisneros JA, Björklund E, González-Gil I, Hu Y, Canales A, Medrano FJ, Romero A, Ortega-Gutiérrez S, Fowler CJ, López-Rodríguez ML. Structure-activity relationship of a new series of reversible dual monoacylglycerol lipase/fatty acid amide hydrolase inhibitors. J Med Chem 2012; 55:824-36. [PMID: 22185522 DOI: 10.1021/jm201327p] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The two endocannabinoids, anandamide (AEA) and 2-arachidonoylglycerol (2-AG), play independent and nonredundant roles in the body. This makes the development of both selective and dual inhibitors of their inactivation an important priority. In this work we report a new series of inhibitors of monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH). Among them, (±)-oxiran-2-ylmethyl 6-(1,1'-biphenyl-4-yl)hexanoate (8) and (2R)-(-)-oxiran-2-ylmethyl(4-benzylphenyl)acetate (30) stand out as potent inhibitors of human recombinant MAGL (IC(50) (8) = 4.1 μM; IC(50) (30) = 2.4 μM), rat brain monoacylglycerol hydrolysis (IC(50) (8) = 1.8 μM; IC(50) (30) = 0.68 μM), and rat brain FAAH (IC(50) (8) = 5.1 μM; IC(50) (30) = 0.29 μM). Importantly, and in contrast to the other previously described MAGL inhibitors, these compounds behave as reversible inhibitors either of competitive (8) or noncompetitive nature (30). Hence, they could be useful to explore the therapeutic potential of reversible MAGL inhibitors.
Collapse
Affiliation(s)
- José A Cisneros
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, E-28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Grabiec U, Koch M, Kallendrusch S, Kraft R, Hill K, Merkwitz C, Ghadban C, Lutz B, Straiker A, Dehghani F. The endocannabinoid N-arachidonoyldopamine (NADA) exerts neuroprotective effects after excitotoxic neuronal damage via cannabinoid receptor 1 (CB(1)). Neuropharmacology 2011; 62:1797-807. [PMID: 22186081 DOI: 10.1016/j.neuropharm.2011.11.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 11/29/2011] [Accepted: 11/30/2011] [Indexed: 12/16/2022]
Abstract
Endocannabinoids exert numerous effects in the CNS under physiological and pathological conditions. The aim of the present study was to examine whether the endocannabinoid N-arachidonoyldopamine (NADA) may protect neurons in excitotoxically lesioned organotypic hippocampal slice cultures (OHSC). OHSC were excitotoxically lesioned by application of N-methyl-d-aspartate (NMDA, 50 μM) for 4 h and subsequently treated with different NADA concentrations (0.1 pM-50 μM) alone or in combination with cannabinoid receptor antagonists. NADA protected dentate gyrus granule cells and caused a slight reduction in the number of microglial cells. The number of degenerated neurons significantly decreased between 100 pM and 10 μM NADA (p < 0.05). To identify the responsive receptor type of NADA mediated neuroprotection, we applied the cannabinoid (CB) receptor 1 (CB(1)) inverse agonist/antagonist AM251, CB(2) inverse agonist/antagonist AM630, abnormal-cannabidiol (abn-CBD)-sensitive receptor antagonist O-1918, transient receptor potential channel V1 (TRPV1) antagonist 6-iodonordihydrocapsaicin and A1 (TRPA1) antagonist HC-030031. Neuroprotective properties of low (1 nM) but not high (10 μM) NADA concentrations were solely blocked by AM251 and were absent in CB(1)(-/-) mice. AM630, O-1918, 6-iodonordihydrocapsaicin and HC-030031 showed no effects at all NADA concentrations applied. Our findings demonstrate that NADA protects dentate gyrus granule cells by acting via CB(1). NADA reduced the number of microglial cells at distinct concentrations. TRPV1 and TRPA1 were not involved in NADA mediated neuroprotection. Thus, our data implicate that NADA mediated activation of neuronal CB(1) may serve as a novel pharmacological target to mitigate symptoms of neuronal damage.
Collapse
Affiliation(s)
- Urszula Grabiec
- Institute of Anatomy, Leipzig University, Liebigstr. 13, 04103 Leipzig, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|