1
|
Flores CM, Carson JR, Codd EE, Dax SL, Kuffner EK, Stahle PL, Neff RA, Eichenbaum GE. The discovery and preclinical pharmacology of JNJ-10450232 (NTM-006), a centrally penetrant, non-opioid structural analog of acetaminophen with comparable analgesic and anti-pyretic properties but no evidence of hepatotoxicity. Regul Toxicol Pharmacol 2025:105830. [PMID: 40222476 DOI: 10.1016/j.yrtph.2025.105830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 03/08/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
A mainstay of the analgesic pharmacopeia for nearly seven decades and alone in its class, acetaminophen relieves mild-to-moderate pain and fever, without similar adverse gastrointestinal and cardiovascular effects associated with non-steroidal anti-inflammatory drugs. While safe and effective when used as directed, acetaminophen overdose may produce liver injury. This report describes discovery and pharmacological characterization of JNJ-10450232/NTM-006, an acetaminophen structural analog designed to retain the efficacy and overall safety profile of acetaminophen without risk of hepatotoxicity following overdose. In the carrageenan and complete Freund's adjuvant models of inflammatory pain and yeast model of fever in rats, JNJ-10450232/NTM-006 exhibited statistically significant effects comparable to acetaminophen in both maximal efficacy and potency. In rat pharmacokinetic studies, JNJ-10450232/NTM-006 exhibited a comparable maximal plasma concentration but higher volume of distribution and longer half-life than acetaminophen, potentially conferring an extended duration of action. In a mouse model of liver injury, acetaminophen produced elevations in aspartate and alanine transaminase activities and signs of hepatic necrosis, whereas JNJ-10450232/NTM-006 did not. Finally, following systemic administration, JNJ-10450232/NTM-006 and acetaminophen produced comparable peripheral levels of para-aminophenol and brain levels of pharmacologically active metabolite N-arachidonoyl-phenolamine (AM404), consistent with the hypothesis that both parent molecules are prodrugs and share the same central mechanism of analgesic action. Taken together, these results suggest JNJ-10450232/NTM-006 as a potentially clinically useful analgesic/antipyretic with improved benefit-to-risk ratio compared with current standards of care.
Collapse
Affiliation(s)
| | - John R Carson
- Janssen Research & Development, LLC, Springhouse, PA, USA
| | - Ellen E Codd
- Janssen Research & Development, LLC, Springhouse, PA, USA.
| | - Scott L Dax
- Janssen Research & Development, LLC, Springhouse, PA, USA.
| | | | - Paul L Stahle
- Janssen Research & Development, LLC, Springhouse, PA, USA.
| | - Robert A Neff
- Janssen Research & Development, LLC, San Diego, CA, USA.
| | | |
Collapse
|
2
|
Bunsick DA, Baghaie L, Li Y, Yaish AM, Aucoin EB, Skapinker E, Aldbai R, Szewczuk MR. Synthetic CB1 Cannabinoids Promote Tunneling Nanotube Communication, Cellular Migration, and Epithelial-Mesenchymal Transition in Pancreatic PANC-1 and Colorectal SW-620 Cancer Cell Lines. Cells 2025; 14:71. [PMID: 39851499 PMCID: PMC11763365 DOI: 10.3390/cells14020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
Metastasizing cancer cells surreptitiously can adapt to metabolic activity during their invasion. By initiating their communications for invasion, cancer cells can reprogram their cellular activities to initiate their proliferation and migration and uniquely counteract metabolic stress during their progression. During this reprogramming process, cancer cells' metabolism and other cellular activities are integrated and mutually regulated by tunneling nanotube communications to alter their specific metabolic functional drivers of tumor growth and progression. Here, we investigated the in vitro effects of the synthetic CB1 cannabinoids AM-404, arvanil, and olvanil on human pancreatic PANC-1 and colorectal SW-620 cancer cell lines to understand further cellular behaviors and the potential risks of their use in cancer therapy. For the first time, the synthetic CB1 cannabinoids AM-404, arvanil, and olvanil significantly altered cancer cells in forming missile-like shapes to induce tunneling nanotube (TNT) communications in PANC-1 cells. Oseltamivir phosphate (OP) significantly prevented TNT formation. To assess the key survival pathways critical for pancreatic cancer progression, we used the AlamarBlue assay to determine synthetic CB1 cannabinoids to induce the cell's metabolic viability drivers to stage migratory intercellular communication. The synthetic CB1 cannabinoids significantly increased cell viability compared to the untreated control for PANC-1 and SW-620 cells, and this response was significantly reduced with the NMBR inhibitor BIM-23127, neuraminidase-1 inhibitor OP, and MMP-9 inhibitor (MMP-9i). CB1 cannabinoids also significantly increased N-cadherin and decreased E-cadherin EMT markers compared to the untreated controls, inducing the process of metastatic phenotype for invasion. BIM-23127, MMP9i, and OP significantly inhibited CB1 agonist-induced NFκB-dependent secretory alkaline phosphatase (SEAP) activity. To confirm this concept, we investigated the migratory invasiveness of PANC-1 and SW-620 cancer cells treated with the synthetic CB1 cannabinoids AM-404, arvanil, and olvanil in a scratch wound assay. CB1 cannabinoids significantly induced the rate of migration and invasiveness of PANC-1 cancer cells, whereas they had minimal effect on the rate of migration of already metastatic SW-620 cancer cells. Interestingly, olvanil-treated SW-620 cells significantly enhanced the migration rate and invasiveness of these cells. The data support the cellular and molecular mechanisms of the synthetic CB1 cannabinoids, orchestrating intercellular conduits to enhance metabolic drivers to stage migratory intercellular communication in pancreatic cancer cells.
Collapse
Affiliation(s)
- David A. Bunsick
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (L.B.); (R.A.)
| | - Leili Baghaie
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (L.B.); (R.A.)
| | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (Y.L.); (E.S.)
| | | | - Emilyn B. Aucoin
- Faculty of Science, Biology (Biomedical Science), York University, Toronto, ON M3J 1P3, Canada;
| | - Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (Y.L.); (E.S.)
| | - Rashelle Aldbai
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (L.B.); (R.A.)
| | - Myron R. Szewczuk
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (L.B.); (R.A.)
| |
Collapse
|
3
|
Yun M, Regen ND, Anchondo Y, Eddinger K, Malkmus S, Roberts SW, Donati E, Leonardi A, Yaksh TL. Acetaminophen effects upon formalin-evoked flinching, postformalin, and postincisional allodynia and conditioned place preference. Pain Rep 2024; 9:e1168. [PMID: 39139364 PMCID: PMC11321755 DOI: 10.1097/pr9.0000000000001168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/11/2024] [Accepted: 05/09/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction We explored in mice, the analgesic, tolerance, dependency, and rewarding effects of systemic acetaminophen (APAP). Methods Studies employed adult mice (C57Bl6). (1) Intraplantar formalin flinching + post formalin allodynia. Mice were given intraperitoneal APAP in a DMSO (5%)/Tween 80 (5%) or a water-based formulation before formalin flinching on day 1 and tactile thresholds assessed before and after APAP at day 12. (2) Paw incision. At 24 hours and 8 days after hind paw incision in male mice, effects of intraperitoneal APAP on tactile allodynia were assessed. (3) Repeated delivery. Mice received daily (4 days) analgesic doses of APAP or vehicle and tested upon formalin flinching on day 5. (4) Conditioned place preference. For 3 consecutive days, vehicle was given in the morning in either of 2 chambers and in each afternoon, an analgesic dose of morphine or APAP in the other chamber. On days 5 and 10, animals were allowed to select a "preferred" chamber. Results Formalin in male mice resulted in biphasic flinching and an enduring postformalin tactile allodynia. Acetaminophen dose dependently decreased phase 2 flinching, and reversed allodynia was observed postflinching. At a comparable APAP dose, female mice showed similarly reduced phase 2 flinching. Incision allodynia was transiently reversed by APAP. Repeated APAP delivery showed no loss of effect after sequential injections or signs of withdrawal. Morphine, but not APAP or vehicle, resulted in robust place preference. Conclusions APAP decreased flinching and allodynia observed following formalin and paw incision and an absence of tolerance, dependence, or rewarding properties.
Collapse
Affiliation(s)
- Mijung Yun
- Department of Anesthesiology, University of California, San Diego, CA, USA
- Pain Clinic, Department of Anesthesiology and Pain Medicine, National Medical Center, Jung-gu, Seoul, Korea
| | | | - Yuvicza Anchondo
- Department of Anesthesiology, University of California, San Diego, CA, USA
| | - Kelly Eddinger
- Department of Anesthesiology, University of California, San Diego, CA, USA
| | - Shelle Malkmus
- Department of Anesthesiology, University of California, San Diego, CA, USA
| | - Steven W. Roberts
- Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA, USA
| | | | | | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA, USA
| |
Collapse
|
4
|
Simon CG, Bozenhardt EH, Celluzzi CM, Dobnik D, Grant ML, Lakshmipathy U, Nebel T, Peltier L, Ratcliffe A, Sherley JL, Stacey GN, Taghizadeh RR, Tan EHP, Vessillier S. Mechanism of action, potency and efficacy: considerations for cell therapies. J Transl Med 2024; 22:416. [PMID: 38698408 PMCID: PMC11067168 DOI: 10.1186/s12967-024-05179-7] [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: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
One of the most challenging aspects of developing advanced cell therapy products (CTPs) is defining the mechanism of action (MOA), potency and efficacy of the product. This perspective examines these concepts and presents helpful ways to think about them through the lens of metrology. A logical framework for thinking about MOA, potency and efficacy is presented that is consistent with the existing regulatory guidelines, but also accommodates what has been learned from the 27 US FDA-approved CTPs. Available information regarding MOA, potency and efficacy for the 27 FDA-approved CTPs is reviewed to provide background and perspective. Potency process and efficacy process charts are introduced to clarify and illustrate the relationships between six key concepts: MOA, potency, potency test, efficacy, efficacy endpoint and efficacy endpoint test. Careful consideration of the meaning of these terms makes it easier to discuss the challenges of correlating potency test results with clinical outcomes and to understand how the relationships between the concepts can be misunderstood during development and clinical trials. Examples of how a product can be "potent but not efficacious" or "not potent but efficacious" are presented. Two example applications of the framework compare how MOA is assessed in cell cultures, animal models and human clinical trials and reveals the challenge of establishing MOA in humans. Lastly, important considerations for the development of potency tests for a CTP are discussed. These perspectives can help product developers set appropriate expectations for understanding a product's MOA and potency, avoid unrealistic assumptions and improve communication among team members during the development of CTPs.
Collapse
Affiliation(s)
- Carl G Simon
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD, USA.
| | - Erich H Bozenhardt
- United Therapeutics Corporation, Regenerative Medicine Operations, Research Triangle Park, NC, USA
| | - Christina M Celluzzi
- Association for the Advancement of Blood and Biotherapies (AABB), Bethesda, MD, USA
| | - David Dobnik
- Niba Labs, Ljubljana, Slovenia
- National Institute of Biology, Ljubljana, Slovenia
| | - Melanie L Grant
- Department of Pediatrics, Children's Healthcare of Atlanta, Marcus Center for Cellular and Gene Therapies, Correlative Studies Laboratory, Emory University School of Medicine, Atlanta, GA, USA
| | - Uma Lakshmipathy
- Pharma Services, Science and Technology, Thermo Fisher Scientific, San Diego, CA, USA
| | - Thiana Nebel
- Medical Education, Sports Medicine and Orthobiologics, Medical Sales Institute, San Diego, CA, USA
| | - Linda Peltier
- Cellular Therapy Lab, Research Institute of McGill University Health Center, Montreal, QC, Canada
| | | | | | - Glyn N Stacey
- International Stem Cell Banking Initiative, Barley, Herts, UK
- National Stem Cell Resource Centre, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cells and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
| | | | - Eddie H P Tan
- Cell and Gene Therapy Facility, Health Sciences Authority, Singapore, Singapore
| | - Sandrine Vessillier
- Science, Research and Innovation Group, Biotherapeutics and Advanced Therapies Division, Medicines and Healthcare Products Regulatory Agency, South Mimms, Hertfordshire, UK
| |
Collapse
|
5
|
Kouznetsov VV. Exploring acetaminophen prodrugs and hybrids: a review. RSC Adv 2024; 14:9691-9715. [PMID: 38525062 PMCID: PMC10958773 DOI: 10.1039/d4ra00365a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/17/2024] [Indexed: 03/26/2024] Open
Abstract
This critical review highlights the advances in developing new molecules for treating pain syndrome, an important issue for human health. Acetaminophen (APAP, known as paracetamol) and nonsteroidal anti-inflammatory drugs (NSAIDs) are commonly used in clinical practice despite their adverse effects. Research is being conducted to develop innovative drugs with improved pharmaceutical properties to mitigate these effects. A more practical way to achieve that is to study well-known and time-tested drugs in their molecular combinations. Accordingly, the present work explores APAP and their combined chemical entities, i.e., prodrugs (soft drugs), codrugs (mutual prodrugs), and hybrids. Due to their molecular structure, APAP prodrugs or codrugs could be considered merged or conjugated hybrids; all these names are very fluid terms. This article proposed a structural classification of these entities to better analyze their advances. So, the following: carrier-linked O-modified APAP, -linked N-modified APAP derivatives (prodrugs), and direct- and spacer-N,O-linked APAP hybrids (codrugs) are the central parts of this review and are examined, especially ester and amide NSAID-APAP molecules. The C-linked APAP and nitric oxide (NO)-releasing APAP hybrids were also briefly discussed. Prime examples of APAP-based drugs such as propacetamol, benorylate, acetaminosalol, nitroparacetamol, and agent JNJ-10450232 weave well into this classification. The proposed classification is the first and original, giving a better understanding of the SAR studies for new pain relievers research and the design development for the analgesic APAP-(or NSAID)-based compounds.
Collapse
Affiliation(s)
- Vladimir V Kouznetsov
- Laboratorio de Química Orgánica y Biomolecular, Escuela de Química, Universidad Industrial de Santander Cl. 9 # Cra 27 A.A. 680006 Bucaramanga Colombia
| |
Collapse
|
6
|
Bunsick DA, Matsukubo J, Aldbai R, Baghaie L, Szewczuk MR. Functional Selectivity of Cannabinoid Type 1 G Protein-Coupled Receptor Agonists in Transactivating Glycosylated Receptors on Cancer Cells to Induce Epithelial-Mesenchymal Transition Metastatic Phenotype. Cells 2024; 13:480. [PMID: 38534324 PMCID: PMC10969603 DOI: 10.3390/cells13060480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Understanding the role of biased G protein-coupled receptor (GPCR) agonism in receptor signaling may provide novel insights into the opposing effects mediated by cannabinoids, particularly in cancer and cancer metastasis. GPCRs can have more than one active state, a phenomenon called either 'biased agonism', 'functional selectivity', or 'ligand-directed signaling'. However, there are increasing arrays of cannabinoid allosteric ligands with different degrees of modulation, called 'biased modulation', that can vary dramatically in a probe- and pathway-specific manner, not from simple differences in orthosteric ligand efficacy or stimulus-response coupling. Here, emerging evidence proposes the involvement of CB1 GPCRs in a novel biased GPCR signaling paradigm involving the crosstalk between neuraminidase-1 (Neu-1) and matrix metalloproteinase-9 (MMP-9) in the activation of glycosylated receptors through the modification of the receptor glycosylation state. The study findings highlighted the role of CB1 agonists AM-404, Aravnil, and Olvanil in significantly inducing Neu-1 sialidase activity in a dose-dependent fashion in RAW-Blue, PANC-1, and SW-620 cells. This approach was further substantiated by findings that the neuromedin B receptor inhibitor, BIM-23127, MMP-9 inhibitor, MMP9i, and Neu-1 inhibitor, oseltamivir phosphate, could specifically block CB1 agonist-induced Neu-1 sialidase activity. Additionally, we found that CB1 receptors exist in a multimeric receptor complex with Neu-1 in naïve, unstimulated RAW-Blue, PANC-1, and SW-620 cells. This complex implies a molecular link that regulates the interaction and signaling mechanism among these molecules present on the cell surface. Moreover, the study results demonstrate that CB1 agonists induce NFκB-dependent secretory alkaline phosphatase (SEAP) activity in influencing the expression of epithelial-mesenchymal markers, E-cadherin, and vimentin in SW-620 cells, albeit the impact on E-cadherin expression is less pronounced compared to vimentin. In essence, this innovative research begins to elucidate an entirely new molecular mechanism involving a GPCR signaling paradigm in which cannabinoids, as epigenetic stimuli, may traverse to influence gene expression and contribute to cancer and cancer metastasis.
Collapse
Affiliation(s)
- David A. Bunsick
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (J.M.); (R.A.); (L.B.)
| | - Jenna Matsukubo
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (J.M.); (R.A.); (L.B.)
- Faculty of Medicine, University of Ottawa, Roger Guindon Hall, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada
| | - Rashelle Aldbai
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (J.M.); (R.A.); (L.B.)
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (J.M.); (R.A.); (L.B.)
| | - Myron R. Szewczuk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (D.A.B.); (J.M.); (R.A.); (L.B.)
| |
Collapse
|
7
|
Suárez J, de Ceglia M, Rodríguez-Pozo M, Vargas A, Santos I, Melgar-Locatelli S, Castro-Zavala A, Castilla-Ortega E, Rodríguez de Fonseca F, Decara J, Rivera P. Inhibition of Adult Neurogenesis in Male Mice after Repeated Exposure to Paracetamol Overdose. Int J Mol Sci 2024; 25:1964. [PMID: 38396643 PMCID: PMC10888347 DOI: 10.3390/ijms25041964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/24/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Paracetamol, or acetaminophen (N-acetyl-para-aminophenol, APAP), is an analgesic and antipyretic drug that is commonly used worldwide, implicated in numerous intoxications due to overdose, and causes serious liver damage. APAP can cross the blood-brain barrier and affects brain function in numerous ways, including pain signals, temperature regulation, neuroimmune response, and emotional behavior; however, its effect on adult neurogenesis has not been thoroughly investigated. We analyze, in a mouse model of hepatotoxicity, the effect of APAP overdose (750 mg/kg/day) for 3 and 4 consecutive days and after the cessation of APAP administration for 6 and 15 days on cell proliferation and survival in two relevant neurogenic zones: the subgranular zone of the dentate gyrus and the hypothalamus. The involvement of liver damage (plasma transaminases), neuronal activity (c-Fos), and astroglia (glial fibrillar acidic protein, GFAP) were also evaluated. Our results indicated that repeated APAP overdoses are associated with the inhibition of adult neurogenesis in the context of elevated liver transaminase levels, neuronal hyperactivity, and astrogliosis. These effects were partially reversed after the cessation of APAP administration for 6 and 15 days. In conclusion, these results suggest that APAP overdose impairs adult neurogenesis in the hippocampus and hypothalamus, a fact that may contribute to the effects of APAP on brain function.
Collapse
Affiliation(s)
- Juan Suárez
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain; (J.S.); (M.R.-P.); (I.S.)
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
| | - Marialuisa de Ceglia
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
| | - Miguel Rodríguez-Pozo
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain; (J.S.); (M.R.-P.); (I.S.)
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
| | - Antonio Vargas
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
| | - Ignacio Santos
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain; (J.S.); (M.R.-P.); (I.S.)
| | - Sonia Melgar-Locatelli
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain
| | - Adriana Castro-Zavala
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain
| | - Estela Castilla-Ortega
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
- Unidad Clínica de Neurología, Hospital Regional Universitario de Málaga, Instituto IBMA-Plataforma BIONAND, 29010 Málaga, Spain
| | - Juan Decara
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
| | - Patricia Rivera
- Grupo de Neuropsicofarmacología, Instituto IBIMA-Plataforma BIONAND, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, Av. de Carlos Haya, 29010 Málaga, Spain; (M.d.C.); (A.V.); (S.M.-L.); (A.C.-Z.); (E.C.-O.); (F.R.d.F.)
| |
Collapse
|
8
|
Tsuzawa K, Onimaru H, Inagaki K, Izumizaki M. Involvement of cannabinoid receptors in depression of the putative nociceptive response in spinal cord preparations isolated from neonatal rats. J Physiol Sci 2023; 73:23. [PMID: 37803279 PMCID: PMC10717773 DOI: 10.1186/s12576-023-00881-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/30/2023] [Indexed: 10/08/2023]
Abstract
A metabolite of acetaminophen, AM404, which is an anandamide transporter inhibitor, induces analgesia mainly via activation of transient receptor potential channel 1 in the spinal cord, although the role of cannabinoid receptors remains to be studied. The ventral root reflex response induced by stimulation of the dorsal root in in vitro preparations of rat spinal cord is useful to assess the effect of analgesics. We analyzed the effects of AM404 and cannabinoid receptor antagonist AM251 on reflex responses in lumbar spinal cord preparations from newborn rats and found that the amplitude of the slow ventral root potential after administration of 10 µM AM404 was not significantly changed, whereas 10 µM AM251 significantly increased the amplitude. Administration of the cannabinoid receptor 1 agonist WIN55,212-2 (10 µM) did not significantly affect the reflex response. We suggest that endogenous cannabinoids in the spinal cord are involved in the antinociceptive mechanism through suppressive effects.
Collapse
Affiliation(s)
- Kayo Tsuzawa
- Department of Orthopedic Surgery, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8555, Japan
| | - Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8555, Japan.
| | - Katsunori Inagaki
- Department of Orthopedic Surgery, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8555, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8555, Japan
| |
Collapse
|
9
|
Klein RM, Motomura VN, Debiasi JD, Moreira EG. Gestational paracetamol exposure induces core behaviors of neurodevelopmental disorders in infant rats and modifies response to a cannabinoid agonist in females. Neurotoxicol Teratol 2023; 99:107279. [PMID: 37391024 DOI: 10.1016/j.ntt.2023.107279] [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: 12/16/2022] [Revised: 05/23/2023] [Accepted: 06/20/2023] [Indexed: 07/02/2023]
Abstract
Paracetamol (PAR) is an over-the-counter analgesic/antipyretic used during pregnancy worldwide. Epidemiological studies have been associating gestational PAR exposure with neurobehavioral alterations in the progeny resembling autism spectrum disorders and attention-deficit hyperactivity disorder symptoms. The endocannabinoid (eCB) dysfunction was previously hypothesized as one of the modes of action by which PAR may harm the developing nervous system. We aimed to evaluate possible effects of gestational exposure to PAR on male and female rat's offspring behavior and if an acute injection of WIN 55,212-2 (WIN, 0.3 mg/kg), a non-specific cannabinoid agonist, prior to behavioral tests, would induce different effects in PAR exposed and non-exposed animals. Pregnant Wistar rats were gavaged with PAR (350 mg/kg/day) or water from gestational day 6 until delivery. Nest-seeking, open field, apomorphine-induced stereotypy, marble burying and three-chamber tests were conducted in 10-, 24-, 25- or 30-days-old rats, respectively. PAR exposure resulted in increased apomorphine-induced stereotyped behavior and time spent in the central area of the open field in exposed female pups. Additionally, it induced hyperactivity in the open field and increased marble burying behavior in both male and female pups. WIN injection modified the behavioral response only in the nest seeking test, and opposite effects were observed in control and PAR-exposed neonate females. Reported alterations are relevant for the neurodevelopmental disorders that have been associated with maternal PAR exposure and suggest that eCB dysfunction may play a role in the action by which PAR may harm the developing brain.
Collapse
Affiliation(s)
- Rodrigo Moreno Klein
- Graduation Program in Health Sciences, State University of Londrina, Londrina, PR 86047-610, Brazil
| | | | - Juliana Diosti Debiasi
- Department of Physiological Sciences, State University of Londrina, Londrina, PR 86047-610, Brazil
| | - Estefânia Gastaldello Moreira
- Graduation Program in Health Sciences, State University of Londrina, Londrina, PR 86047-610, Brazil; Department of Physiological Sciences, State University of Londrina, Londrina, PR 86047-610, Brazil.
| |
Collapse
|
10
|
Spillers NJ, Luther PM, Talbot NC, Ly GH, Downs EM, Lavespere G, Pavlickova D, Ahmadzadeh S, Viswanath O, Varrassi G, Shekoohi S, Kaye AD. Association of Acetaminophen With Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis: Pharmacologic Considerations and Treatment Options. Cureus 2023; 15:e41116. [PMID: 37519510 PMCID: PMC10382713 DOI: 10.7759/cureus.41116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Acetaminophen is an extremely common drug with many implications for its analgesic and antipyretic properties. It has a unique mechanism of action and downstream effects that separate it categorically from non-steroidal anti-inflammatory drugs. These differences come with potential adverse effects that range from mild drug reactions to severe life-threatening emergencies. While acetaminophen's toxic liver effects are well known, a lesser-known adverse effect of this drug is its association with the development of Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN). These dermatological emergencies involve similar pathological processes, including apoptosis of the epidermis and sloughing of the dermis and mucosa from the underlying layers with a positive Nikolsky sign. Currently, SJS and TEN are considered immune-mediated type IV hypersensitivity reactions predominantly involving CD8+ T lymphocytes. Other immune mediators, including regulatory T cells, natural killer cells, interleukins, and drug metabolites are speculated to be involved, but their mechanisms have not been entirely determined. These conditions are differentially diagnosed by the percentage of body area affected with SJS and TENS, involving <10% and >30%, respectively. Genomic variations in human leukocyte antigens (HLA) genes have been implicated in the susceptibility and severity of acetaminophen-induced SJS/TENS, however, details of these interactions remain unclear. Acetaminophen's widespread use and the morbidity of its associated skin pathologies SJS and TENS warrant an in-depth examination of the causative processes involved in their pathogenesis. It is critical that both physicians and patients be made aware that while acetaminophen is widely tolerated by most individuals, severe and potentially fatal interactions do occur, and further investigation is necessary to reduce these adverse effects.
Collapse
Affiliation(s)
- Noah J Spillers
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Patrick M Luther
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Norris C Talbot
- Radiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Gianni H Ly
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Evan M Downs
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Gabriel Lavespere
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Denisa Pavlickova
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Shahab Ahmadzadeh
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Omar Viswanath
- Pain Management, Valley Pain Consultants - Envision Physician Services, Phoenix, USA
| | | | - Sahar Shekoohi
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alan D Kaye
- Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| |
Collapse
|
11
|
Myers AL, Jeske AH. Provider-directed analgesia for dental pain. Expert Rev Clin Pharmacol 2023; 16:435-451. [PMID: 37083548 DOI: 10.1080/17512433.2023.2206118] [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: 04/22/2023]
Abstract
INTRODUCTION Extraction of impacted molar teeth is a common procedure performed by oral surgeons and general dentists, with postoperative pain being a significant adverse event post-surgery. If mismanaged, pain can lead to complications that impact oral and systemic health. The current scourge of the opioid epidemic has ushered in a new era of provider-directed analgesic (PDA) therapy in dentistry. AREAS COVERED This article provides an in-depth review on the major pharmacological and therapeutic properties of established and alternative analgesics used to manage dental pain. EXPERT OPINION Substantial evidence-based literature shows combination of a non-steroidal anti-inflammatory drug (NSAID; e.g. ibuprofen) and acetaminophen provides superior pain relief than single-agent or combination opioid regimens. However, there are clinical scenarios (e.g. severe pain) when short-course opioid prescription is appropriate in select patients, in which a 2-3-day treatment duration is typically sufficient. Alternative agents (e.g. caffeine, gabapentin, phytotherapies), typically in combination with established agents, can mitigate postoperative dental pain. Some evidence suggests preemptive therapies (e.g. corticosteroids, NSAIDs) reduce amounts of postsurgical analgesic consumption and might lessen opioid prescription burden. In summary, this comprehensive review provides an opportune update on the evolving landscape of pharmacotherapy for acute postsurgical dental pain, informing best practices for PDA in the dental setting.
Collapse
Affiliation(s)
- Alan L Myers
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Arthur H Jeske
- Office of the Dean, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
12
|
Hines III JE, Agu OA, Deere CJ, Fronczek FR, Uppu RM. N-(4-Meth-oxy-3-nitro-phen-yl)acetamide. IUCRDATA 2023; 8:x230298. [PMID: 37151207 PMCID: PMC10162034 DOI: 10.1107/s2414314623002985] [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: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
The title compound, C9H10N2O4, crystallizes with a disordered nitro group in twinned crystals. Both the meth-oxy group and the acetamide groups are nearly coplanar with the phenyl ring, and the C-N-C-O torsion angle [0.2 (4)°] is also insignificantly different from zero. Overall, the 12-atom meth-oxy-phenyl-acetamide group is nearly planar, with an r.m.s. deviation of 0.042 Å. The nitro group is twisted out of this plane by about 30°, disordered into two orientations with opposite senses of twist. In the crystal, the N-H group donates a hydrogen bond to a nitro oxygen atom, generating chains propagating in the [101] direction. The amide carbonyl oxygen atom is not involved in the hydrogen bonding.
Collapse
Affiliation(s)
- James E. Hines III
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA 70813, USA
| | - Ogad A. Agu
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA 70813, USA
| | - Curtistine J. Deere
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA 70813, USA
| | - Frank R. Fronczek
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Rao M. Uppu
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA 70813, USA
- Correspondence e-mail:
| |
Collapse
|
13
|
Mallet C, Desmeules J, Pegahi R, Eschalier A. An Updated Review on the Metabolite (AM404)-Mediated Central Mechanism of Action of Paracetamol (Acetaminophen): Experimental Evidence and Potential Clinical Impact. J Pain Res 2023; 16:1081-1094. [PMID: 37016715 PMCID: PMC10066900 DOI: 10.2147/jpr.s393809] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Paracetamol remains the recommended first-line option for mild-to-moderate acute pain in general population and particularly in vulnerable populations. Despite its wide use, debate exists regarding the analgesic mechanism of action (MoA) of paracetamol. A growing body of evidence challenged the notion that paracetamol exerts its analgesic effect through cyclooxygenase (COX)-dependent inhibitory effect. It is now more evident that paracetamol analgesia has multiple pathways and is mediated by the formation of the bioactive AM404 metabolite in the central nervous system (CNS). AM404 is a potent activator of TRPV1, a major contributor to neuronal response to pain in the brain and dorsal horn. In the periaqueductal grey, the bioactive metabolite AM404 activated the TRPV1 channel-mGlu5 receptor-PLC-DAGL-CB1 receptor signaling cascade. The present article provides a comprehensive literature review of the centrally located, COX-independent, analgesic MoA of paracetamol and relates how the current experimental evidence can be translated into clinical practice. The evidence discussed in this review established paracetamol as a central, COX-independent, antinociceptive medication that has a distinct MoA from non-steroidal anti-inflammatory drugs (NSAIDs) and a more tolerable safety profile. With the establishment of the central MoA of paracetamol, we believe that paracetamol remains the preferred first-line option for mild-to-moderate acute pain for healthy adults, children, and patients with health concerns. However, safety concerns remain with the high dose of paracetamol due to the NAPQI-mediated liver necrosis. Centrally acting paracetamol/p-aminophenol derivatives could potentiate the analgesic effect of paracetamol without increasing the risk of hepatoxicity. Moreover, the specific central MoA of paracetamol allows its combination with other analgesics, including NSAIDs, with a different MoA. Future experiments to better explain the central actions of paracetamol could pave the way for discovering new central analgesics with a better benefit-to-risk ratio.
Collapse
Affiliation(s)
- Christophe Mallet
- Université Clermont Auvergne, INSERM, NEURO-DOL Basics & Clinical Pharmacology of Pain, Clermont-Ferrand, France
| | - Jules Desmeules
- Faculty of Medicine and The School of Pharmaceutical Sciences, Faculty of Sciences, Geneva University, Geneva, Switzerland
| | | | - Alain Eschalier
- Université Clermont Auvergne, INSERM, NEURO-DOL Basics & Clinical Pharmacology of Pain, Clermont-Ferrand, France
- Correspondence: Alain Eschalier, Faculté de Médecine, UMR Neuro-Dol, 49 Bd François Mitterrand, Clermont-Ferrand, 63000, France, Email
| |
Collapse
|
14
|
Schmiedhofer P, Vogel FD, Koniuszewski F, Ernst M. Cys-loop receptors on cannabinoids: All high? Front Physiol 2022; 13:1044575. [PMID: 36439263 PMCID: PMC9682269 DOI: 10.3389/fphys.2022.1044575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/24/2022] [Indexed: 11/10/2022] Open
Abstract
Endocannabinoids (eCBS) are endogenously derived lipid signaling molecules that serve as tissue hormones and interact with multiple targets, mostly within the endocannabinoid system (ECS). The ECS is a highly conserved regulatory system involved in homeostatic regulation, organ formation, and immunomodulation of chordates. The term “cannabinoid” evolved from the distinctive class of plant compounds found in Cannabis sativa, an ancient herb, due to their action on CB1 and CB2 receptors. CB1/2 receptors are the primary targets for eCBs, but their effects are not limited to the ECS. Due to the high interest and extensive research on the ECS, knowledge on its constituents and physiological role is substantial and still growing. Crosstalk and multiple targeting of molecules are common features of endogenous and plant compounds. Cannabimimetic molecules can be divided according to their origin, natural or synthetic, including phytocannabinoids (pCB’s) or synthetic cannabinoids (sCB’s). The endocannabinoid system (ECS) consists of receptors, transporters, enzymes, and signaling molecules. In this review, we focus on the effects of cannabinoids on Cys-loop receptors. Cys-loop receptors belong to the class of membrane-bound pentameric ligand gated ion channels, each family comprising multiple subunits. Mammalians possess GABA type A receptors (GABAAR), glycine receptors (GlyR), serotonin receptors type 3 (5-HT3R), and nicotinic acetylcholine receptors (nAChR). Several studies have shown different modulatory effects of CBs on multiple members of the Cys-loop receptor family. We highlight the existing knowledge, especially on subunits and protein domains with conserved binding sites for CBs and their possible pharmacological and physiological role in epilepsy and in chronic pain. We further discuss the potential for cannabinoids as first line treatments in epilepsy, chronic pain and other neuropsychiatric conditions, indicated by their polypharmacology and therapeutic profile.
Collapse
Affiliation(s)
- Philip Schmiedhofer
- SBR Development Holding, Vienna, Austria
- *Correspondence: Philip Schmiedhofer, ; Margot Ernst,
| | - Florian Daniel Vogel
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Filip Koniuszewski
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Margot Ernst
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University Vienna, Vienna, Austria
- *Correspondence: Philip Schmiedhofer, ; Margot Ernst,
| |
Collapse
|
15
|
Scienza-Martin K, Lotz FN, Zanona QK, Santana-Kragelund F, Crestani AP, Boos FZ, Calcagnotto ME, Quillfeldt JA. Memory consolidation depends on endogenous hippocampal levels of anandamide: CB1 and M4, but possibly not TRPV1 receptors mediate AM404 effects. Neuroscience 2022; 497:53-72. [DOI: 10.1016/j.neuroscience.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/14/2022] [Accepted: 04/08/2022] [Indexed: 11/15/2022]
|
16
|
Why Do Marijuana and Synthetic Cannabimimetics Induce Acute Myocardial Infarction in Healthy Young People? Cells 2022; 11:cells11071142. [PMID: 35406706 PMCID: PMC8997492 DOI: 10.3390/cells11071142] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 12/19/2022] Open
Abstract
The use of cannabis preparations has steadily increased. Although cannabis was traditionally assumed to only have mild vegetative side effects, it has become evident in recent years that severe cardiovascular complications can occur. Cannabis use has recently even been added to the risk factors for myocardial infarction. This review is dedicated to pathogenetic factors contributing to cannabis-related myocardial infarction. Tachycardia is highly important in this respect, and we provide evidence that activation of CB1 receptors in brain regions important for cardiovascular regulation and of presynaptic CB1 receptors on sympathetic and/or parasympathetic nerve fibers are involved. The prototypical factors for myocardial infarction, i.e., thrombus formation and coronary constriction, have also been considered, but there is little evidence that they play a decisive role. On the other hand, an increase in the formation of carboxyhemoglobin, impaired mitochondrial respiration, cardiotoxic reactions and tachyarrhythmias associated with the increased sympathetic tone are factors possibly intensifying myocardial infarction. A particularly important factor is that cannabis use is frequently accompanied by tobacco smoking. In conclusion, additional research is warranted to decipher the mechanisms involved, since cannabis use is being legalized increasingly and Δ9-tetrahydrocannabinol and its synthetic analogue nabilone are indicated for the treatment of various disease states.
Collapse
|
17
|
Apweiler M, Streyczek J, Saliba SW, Ditrich J, Muñoz E, Fiebich BL. Anti-Inflammatory and Anti-Oxidative Effects of AM404 in IL-1β-Stimulated SK-N-SH Neuroblastoma Cells. Front Pharmacol 2021; 12:789074. [PMID: 34867421 PMCID: PMC8635764 DOI: 10.3389/fphar.2021.789074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022] Open
Abstract
An emerging number of studies address the involvement of neuroinflammation and oxidative stress in the pathophysiology of central nervous system (CNS) disorders such as depression, schizophrenia, anxiety, and neurodegenerative diseases. Different cytokines and molecules, such as prostaglandin (PG) E2, are associated with neuroinflammatory processes. The active acetaminophen metabolite AM404 has been shown to prevent inflammation and neuroinflammation in primary microglia and organotypic hippocampal slice cultures. However, its effects on pathophysiological conditions in the CNS and especially on neurons are still poorly understood. In this study, we therefore evaluated the effects of AM404 and acetaminophen on the arachidonic acid cascade and oxidative stress induced by interleukin (IL)-1β in human SK-N-SH neuronal cells. We observed that AM404 and acetaminophen significantly and concentration-dependent inhibited IL-1β-induced release of PGE2, independent of cyclooxygenases (COX)-1 and COX-2 enzymatic activity as well as COX-2 mRNA and protein levels in SK-N-SH-cells. The reduction of IL-1β-induced PGE2-release by AM404 and acetaminophen treatment might be mediated by the 8-iso-PGF2α pathway since IL-1β-induced synthesis of this free radical marker is dose-dependently reduced by both compounds, respectively. Therefore, understanding of the potential therapeutic properties of AM404 in neuroinflammation and oxidative stress might lead to future treatment options of different neurological disorders.
Collapse
Affiliation(s)
- Matthias Apweiler
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jana Streyczek
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Soraya Wilke Saliba
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Eduardo Muñoz
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain.,Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain.,Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Bernd L Fiebich
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
18
|
Bloukh S, Wazaify M, Matheson C. Paracetamol: unconventional uses of a well-known drug. INTERNATIONAL JOURNAL OF PHARMACY PRACTICE 2021; 29:527-540. [PMID: 34455434 DOI: 10.1093/ijpp/riab058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/04/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To describe and map what is known about unconventional uses of paracetamol through a scoping review of published literature by adopting adopted a PRISMA systematic approach methodology. KEY FINDINGS Four themes for unconventional uses of paracetamol emerged: (a) use of paracetamol in sleep (a-1) positive effect of paracetamol on sleep (n = 9) or (a-2) neutral or negative effect of paracetamol on sleep (n = 9); (b) use of paracetamol in sport (n = 13); (c) mixing paracetamol with drinks, waterpipe and illicit drugs (n = 5); and (d) miscellaneous uses (n = 4). Forty records were reviewed and charted. Available literature supports concern around the potential of harmful or non-medical use of paracetamol, especially among patients with a history of substance use, parents of young children or athletes. SUMMARY Paracetamol (acetaminophen) is one of the most popular and widely used drugs for the treatment of pain and fever. It is considered remarkably safe if used within instructions. However, there is growing evidence that paracetamol, is sometimes used outside approved indications or abused (i.e. used for non-medical reasons). This review highlights the need for enhanced pharmacovigilance and surveillance of non-medical paracetamol use and raising general public awareness of its potential dangers especially in higher than recommended doses.
Collapse
Affiliation(s)
- Sarah Bloukh
- School of Pharmacy, The University of Jordan (UJ), Amman, Jordan
| | - Mayyada Wazaify
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan (UJ), Amman, Jordan
| | | |
Collapse
|
19
|
Hoshijima H, Hunt M, Nagasaka H, Yaksh T. Systematic Review of Systemic and Neuraxial Effects of Acetaminophen in Preclinical Models of Nociceptive Processing. J Pain Res 2021; 14:3521-3552. [PMID: 34795520 PMCID: PMC8594782 DOI: 10.2147/jpr.s308028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/11/2021] [Indexed: 12/29/2022] Open
Abstract
Acetaminophen (APAP) in humans has robust effects with a high therapeutic index in altering postoperative and inflammatory pain states in clinical and experimental pain paradigms with no known abuse potential. This review considers the literature reflecting the preclinical actions of acetaminophen in a variety of pain models. Significant observations arising from this review are as follows: 1) acetaminophen has little effect upon acute nociceptive thresholds; 2) acetaminophen robustly reduces facilitated states as generated by mechanical and thermal hyperalgesic end points in mouse and rat models of carrageenan and complete Freund’s adjuvant evoked inflammation; 3) an antihyperalgesic effect is observed in models of facilitated processing with minimal inflammation (eg, phase II intraplantar formalin); and 4) potent anti-hyperpathic effects on the thermal hyperalgesia, mechanical and cold allodynia, allodynic thresholds in rat and mouse models of polyneuropathy and mononeuropathies and bone cancer pain. These results reflect a surprisingly robust drug effect upon a variety of facilitated states that clearly translate into a wide range of efficacy in preclinical models and to important end points in human therapy. The specific systems upon which acetaminophen may act based on targeted delivery suggest both a spinal and a supraspinal action. Review of current targets for this molecule excludes a role of cyclooxygenase inhibitor but includes effects that may be mediated through metabolites acting on the TRPV1 channel, or by effect upon cannabinoid and serotonin signaling. These findings suggest that the mode of action of acetaminophen, a drug with a long therapeutic history of utilization, has surprisingly robust effects on a variety of pain states in clinical patients and in preclinical models with a good therapeutic index, but in spite of its extensive use, its mechanisms of action are yet poorly understood.
Collapse
Affiliation(s)
- Hiroshi Hoshijima
- Department of Anesthesiology, Saitama Medical University Hospital, Saitama, Japan
| | - Matthew Hunt
- Departments of Anesthesiology and Pharmacology, University of California, San Diego Anesthesia Research Laboratory, La Jolla, CA, USA
| | - Hiroshi Nagasaka
- Department of Anesthesiology, Saitama Medical University Hospital, Saitama, Japan
| | - Tony Yaksh
- Departments of Anesthesiology and Pharmacology, University of California, San Diego Anesthesia Research Laboratory, La Jolla, CA, USA
| |
Collapse
|
20
|
|
21
|
Winters BL, Vaughan CW. Mechanisms of endocannabinoid control of synaptic plasticity. Neuropharmacology 2021; 197:108736. [PMID: 34343612 DOI: 10.1016/j.neuropharm.2021.108736] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/13/2023]
Abstract
The endogenous cannabinoid transmitter system regulates synaptic transmission throughout the nervous system. Unlike conventional transmitters, specific stimuli induce synthesis of endocannabinoids (eCBs) in the postsynaptic neuron, and these travel backwards to modulate presynaptic inputs. In doing so, eCBs can induce short-term changes in synaptic strength and longer-term plasticity. While this eCB regulation is near ubiquitous, it displays major regional and synapse specific variations with different synapse specific forms of short-versus long-term plasticity throughout the brain. These differences are due to the plethora of pre- and postsynaptic mechanisms which have been implicated in eCB signalling, the intricacies of which are only just being realised. In this review, we shall describe the current understanding and highlight new advances in this area, with a focus on the retrograde action of eCBs at CB1 receptors (CB1Rs).
Collapse
Affiliation(s)
- Bryony Laura Winters
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney at Royal North Shore Hospital, NSW, Australia.
| | - Christopher Walter Vaughan
- Pain Management Research Institute, Kolling Institute of Medical Research, Northern Clinical School, University of Sydney at Royal North Shore Hospital, NSW, Australia
| |
Collapse
|
22
|
Cannabidiol has therapeutic potential for myofascial pain in female and male parkinsonian rats. Neuropharmacology 2021; 196:108700. [PMID: 34246682 DOI: 10.1016/j.neuropharm.2021.108700] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/21/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022]
Abstract
The musculoskeletal orofacial pain is a complex symptom of Parkinson's disease (PD) resulting in stomatognathic system dysfunctions aggravated by the disease rigidity and postural instability. We tested the effect of cannabidiol (CBD), a non-psychotomimetic constituent of Cannabis sativa, in PD-related myofascial pain. Wistar adult female and male rats orofacial allodynic and hyperalgesic responses were tested by Von Frey and formalin tests, before and 21 days past 6-OHDA lesion. Algesic response was tested after masseter muscle injection of CBD (10, 50, 100 μg in 10 μL) or vehicle. Males compared to females in all estrous cycles' phases presented reduced orofacial allodynia and hyperalgesia. According to the estrous cycle's phases, females presented distinct orofacial nociceptive responses, being the estrus phase well-chosen for nociceptive analysis after 6-OHDA lesion (phase with fewer hormone alterations and adequate length). Dopaminergic neuron lesion decreased mechanical and inflammatory nociceptive thresholds in females and males in a higher proportion in females. CBD local treatment reduced the increased orofacial allodynia and hyperalgesia, in males and females. The female rats were more sensitive to CBD effect considering allodynia, responding to the lowest dose. Although females and males respond to the effect of three doses of CBD in the formalin test, males showed a superior reduction in the hyperalgesic response. These results indicate that hemiparkinsonian female in the estrus phase and male answer differently to the different doses of CBD therapy and nociceptive tests. CBD therapy is effective for parkinsonism-induced orofacial nociception.
Collapse
|
23
|
Garrone B, Durando L, Prenderville J, Sokolowska E, Milanese C, Di Giorgio FP, Callaghan C, Bianchi M. Paracetamol (acetaminophen) rescues cognitive decline, neuroinflammation and cytoskeletal alterations in a model of post-operative cognitive decline (POCD) in middle-aged rats. Sci Rep 2021; 11:10139. [PMID: 33980934 PMCID: PMC8115335 DOI: 10.1038/s41598-021-89629-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/22/2021] [Indexed: 11/09/2022] Open
Abstract
Post-operative cognitive dysfunction (POCD) is a debilitating clinical phenomenon in elderly patients. Management of pain in elderly is complicated because analgesic opiates elicit major side effects. In contrast, paracetamol (acetaminophen) has shown analgesic efficacy, no impact on cognition, and its side effects are well tolerated. We investigated the efficacy of paracetamol, compared to the opioid analgesic buprenorphine, in a model of POCD by investigating cognitive decline, allodynia, peripheral and hippocampal cytokines levels, and hippocampal microtubule dynamics as a key modulator of synaptic plasticity. A POCD model was developed in middle-aged (MA) rats by inducing a tibia fracture via orthopaedic surgery. Control MA rats did not undergo any surgery and only received isoflurane anaesthesia. We demonstrated that cognitive decline and increased allodynia following surgery was prevented in paracetamol-treated animals, but not in animals which were exposed to anesthesia alone or underwent the surgery and received buprenorphine. Behavioral alterations were associated with different peripheral cytokine changes between buprenorphine and paracetamol treated animals. Buprenorphine showed no central effects, while paracetamol showed modulatory effects on hippocampal cytokines and markers of microtubule dynamics which were suggestive of neuroprotection. Our data provide the first experimental evidence corroborating the use of paracetamol as first-choice analgesic in POCD.
Collapse
Affiliation(s)
- B Garrone
- Angelini Pharma S.p.A., Viale Amelia, 70, 00181, Rome, Italy
| | - L Durando
- Angelini Pharma S.p.A., Viale Amelia, 70, 00181, Rome, Italy
| | - J Prenderville
- Transpharmation Ireland Ltd., Trinity College Dublin-Institute of Neuroscience (TCIN), Lloyd Institute, Trinity College, Dublin 2, Ireland
| | - E Sokolowska
- Transpharmation Ireland Ltd., Trinity College Dublin-Institute of Neuroscience (TCIN), Lloyd Institute, Trinity College, Dublin 2, Ireland
| | - C Milanese
- Angelini Pharma S.p.A., Viale Amelia, 70, 00181, Rome, Italy
| | - F P Di Giorgio
- Angelini Pharma S.p.A., Viale Amelia, 70, 00181, Rome, Italy
| | - C Callaghan
- Ulysses Neuroscience Ltd, Room 3.57B, Trinity College Dublin-Institute of Neuroscience (TCIN), Lloyd Institute, Trinity College, Dublin 2, Ireland
| | - M Bianchi
- Ulysses Neuroscience Ltd, Room 3.57B, Trinity College Dublin-Institute of Neuroscience (TCIN), Lloyd Institute, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
24
|
Ayoub SS. Paracetamol (acetaminophen): A familiar drug with an unexplained mechanism of action. Temperature (Austin) 2021; 8:351-371. [PMID: 34901318 PMCID: PMC8654482 DOI: 10.1080/23328940.2021.1886392] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 02/02/2023] Open
Abstract
Paracetamol (acetaminophen) is undoubtedly one of the most widely used drugs worldwide. As an over-the-counter medication, paracetamol is the standard and first-line treatment for fever and acute pain and is believed to remain so for many years to come. Despite being in clinical use for over a century, the precise mechanism of action of this familiar drug remains a mystery. The oldest and most prevailing theory on the mechanism of analgesic and antipyretic actions of paracetamol relates to the inhibition of CNS cyclooxygenase (COX) enzyme activities, with conflicting views on the COX isoenzyme/variant targeted by paracetamol and on the nature of the molecular interactions with these enzymes. Paracetamol has been proposed to selectively inhibit COX-2 by working as a reducing agent, despite the fact that in vitro screens demonstrate low potency on the inhibition of COX-1 and COX-2. In vivo data from COX-1 transgenic mice suggest that paracetamol works through inhibition of a COX-1 variant enzyme to mediate its analgesic and particularly thermoregulatory actions (antipyresis and hypothermia). A separate line of research provides evidence on potentiation of the descending inhibitory serotonergic pathway to mediate the analgesic action of paracetamol, but with no evidence of binding to serotonergic molecules. AM404 as a metabolite for paracetamol has been proposed to activate the endocannabinoid and the transient receptor potential vanilloid-1 (TRPV1) systems. The current review gives an update and in some cases challenges the different theories on the pharmacology of paracetamol and raises questions on some of the inadequately explored actions of paracetamol. List of Abbreviations: AM404, N-(4-hydroxyphenyl)-arachidonamide; CB1R, Cannabinoid receptor-1; Cmax, Maximum concentration; CNS, Central nervous system; COX, Cyclooxygenase; CSF, Cerebrospinal fluid; ED50, 50% of maximal effective dose; FAAH, Fatty acid amidohydrolase; IC50, 50% of the maximal inhibitor concentration; LPS, Lipopolysaccharide; NSAIDs, Non-steroidal anti-inflammatory drugs; PGE2, Prostaglandin E2; TRPV1, Transient receptor potential vanilloid-1.
Collapse
Affiliation(s)
- Samir S Ayoub
- School of Health, Sport and Bioscience, Medicines Research Group, University of East London, London, UK
| |
Collapse
|
25
|
Å Nilsson JL, Mallet C, Shionoya K, Blomgren A, Sundin AP, Grundemar L, Boudieu L, Blomqvist A, Eschalier A, Nilsson UJ, Zygmunt PM. Paracetamol analogues conjugated by FAAH induce TRPV1-mediated antinociception without causing acute liver toxicity. Eur J Med Chem 2021; 213:113042. [PMID: 33257173 DOI: 10.1016/j.ejmech.2020.113042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022]
Abstract
Paracetamol, one of the most widely used pain-relieving drugs, is deacetylated to 4-aminophenol (4-AP) that undergoes fatty acid amide hydrolase (FAAH)-dependent biotransformation into N-arachidonoylphenolamine (AM404), which mediates TRPV1-dependent antinociception in the brain of rodents. However, paracetamol is also converted to the liver-toxic metabolite N-acetyl-p-benzoquinone imine already at therapeutic doses, urging for safer paracetamol analogues. Primary amine analogues with chemical structures similar to paracetamol were evaluated for their propensity to undergo FAAH-dependent N-arachidonoyl conjugation into TRPV1 activators both in vitro and in vivo in rodents. The antinociceptive and antipyretic activity of paracetamol and primary amine analogues was examined with regard to FAAH and TRPV1 as well as if these analogues produced acute liver toxicity. 5-Amino-2-methoxyphenol (2) and 5-aminoindazole (3) displayed efficient target protein interactions with a dose-dependent antinociceptive effect in the mice formalin test, which in the second phase was dependent on FAAH and TRPV1. No hepatotoxicity of the FAAH substrates transformed into TRPV1 activators was observed. While paracetamol attenuates pyrexia via inhibition of brain cyclooxygenase, its antinociceptive FAAH substrate 4-AP was not antipyretic, suggesting separate mechanisms for the antipyretic and antinociceptive effect of paracetamol. Furthermore, compound 3 reduced fever without a brain cyclooxygenase inhibitory action. The data support our view that analgesics and antipyretics without liver toxicity can be derived from paracetamol. Thus, research into the molecular actions of paracetamol could pave the way for the discovery of analgesics and antipyretics with a better benefit-to-risk ratio.
Collapse
Affiliation(s)
- Johan L Å Nilsson
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, Box 117, SE-221 00, Lund, Sweden
| | - Christophe Mallet
- Université Clermont Auvergne, INSERM, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, F-63000, Clermont-Ferrand, France
| | - Kiseko Shionoya
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85, Linköping, Sweden
| | - Anders Blomgren
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, Box 117, SE-221 00, Lund, Sweden
| | - Anders P Sundin
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, 221 00, Lund, Sweden
| | - Lars Grundemar
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, Box 117, SE-221 00, Lund, Sweden
| | - Ludivine Boudieu
- Université Clermont Auvergne, INSERM, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, F-63000, Clermont-Ferrand, France
| | - Anders Blomqvist
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, SE-581 85, Linköping, Sweden
| | - Alain Eschalier
- Université Clermont Auvergne, INSERM, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000, Clermont-Ferrand, France; ANALGESIA Institute, Faculty of Medicine, F-63000, Clermont-Ferrand, France
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, 221 00, Lund, Sweden
| | - Peter M Zygmunt
- Department of Clinical Sciences Malmö, Lund University, SE-214 28, Malmö, Sweden.
| |
Collapse
|
26
|
Morena M, Nastase AS, Santori A, Cravatt BF, Shansky RM, Hill MN. Sex-dependent effects of endocannabinoid modulation of conditioned fear extinction in rats. Br J Pharmacol 2021; 178:983-996. [PMID: 33314038 PMCID: PMC8311789 DOI: 10.1111/bph.15341] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 10/05/2020] [Accepted: 12/07/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Women are twice as likely as men to develop post-traumatic stress disorder (PTSD) making the search for biological mechanisms underlying these gender disparities especially crucial. One of the hallmark symptoms of PTSD is an alteration in the ability to extinguish fear responses to trauma-associated cues. In male rodents, the endocannabinoid system can modulate fear extinction and has been suggested as a therapeutic target for PTSD. However, whether and how the endocannabinoid system may modulate fear expression and extinction in females remains unknown. EXPERIMENTAL APPROACH To answer this question, we pharmacologically manipulated endocannabinoid signalling in male and female rats prior to extinction of auditory conditioned fear and measured both passive (freezing) and active (darting) conditioned responses. KEY RESULTS Surprisingly, we found that acute systemic inhibition of the endocannabinoid anandamide (AEA) or 2-arachidonoyl glycerol (2-AG) hydrolysis did not significantly alter fear expression or extinction in males. However, the same manipulations in females produced diverging effects. Increased AEA signalling at vanilloid TRPV1 receptors impaired fear memory extinction. In contrast, inhibition of 2-AG hydrolysis promoted active over passive fear responses acutely via activation of cannabinoid1 (CB1 ) receptors. Measurement of AEA and 2-AG levels after extinction training revealed sex- and brain region-specific changes. CONCLUSION AND IMPLICATIONS We provide the first evidence that AEA and 2-AG signalling affect fear expression and extinction in females in opposite directions. These findings are relevant to future research on sex differences in mechanisms of fear extinction and may help develop sex-specific therapeutics to treat trauma-related disorders.
Collapse
Affiliation(s)
- Maria Morena
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
- Mathison Centre for Mental Health Research, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
- Departments of Cell Biology and Anatomy & Psychiatry, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
| | - Andrei S. Nastase
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
- Mathison Centre for Mental Health Research, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
- Neuroscience Program, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
| | - Alessia Santori
- Department of Physiology and Pharmacology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Rome, Italy
| | - Benjamin F. Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road La Jolla, CA 92037, USA
| | - Rebecca M. Shansky
- Department of Psychology, Northeastern University, 360 Huntington Ave, 125 NI, Boston, MA 02115, USA
| | - Matthew N. Hill
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
- Mathison Centre for Mental Health Research, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
- Departments of Cell Biology and Anatomy & Psychiatry, University of Calgary, 3330 Hospital Dr. NW, T2N 4N1 Calgary, AB, Canada
| |
Collapse
|
27
|
Ishitsuka Y, Kondo Y, Kadowaki D. Toxicological Property of Acetaminophen: The Dark Side of a Safe Antipyretic/Analgesic Drug? Biol Pharm Bull 2020; 43:195-206. [PMID: 32009106 DOI: 10.1248/bpb.b19-00722] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acetaminophen (paracetamol, N-acetyl-p-aminophenol; APAP) is the most popular analgesic/antipyretic agent in the world. APAP has been regarded as a safer drug compared with non-steroidal anti-inflammatory drugs (NSAIDs) particularly in terms of lower risks of renal dysfunction, gastrointestinal injury, and asthma/bronchospasm induction, even in high-risk patients such as the elderly, children, and pregnant women. On the other hand, the recent increasing use of APAP has raised concerns about its toxicity. In this article, we review recent pharmacological and toxicological findings about APAP from basic, clinical, and epidemiological studies, including spontaneous drug adverse events reporting system, especially focusing on drug-induced asthma and pre-and post-natal closure of ductus arteriosus. Hepatotoxicity is the greatest fault of APAP and the most frequent cause of drug-induced acute liver failure in Western countries. However, its precise mechanism remains unclear and no effective cure beyond N-acetylcysteine has been developed. Recent animal and cellular studies have demonstrated that some cellular events, such as c-jun N-terminal kinase (JNK) pathway activation, endoplasmic reticulum (ER) stress, and mitochondrial oxidative stress may play important roles in the development of hepatitis. Herein, the molecular mechanisms of APAP hepatotoxicity are summarized. We also discuss the not-so-familiar "dark side" of APAP as an otherwise safe analgesic/antipyretic drug.
Collapse
Affiliation(s)
- Yoichi Ishitsuka
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Kondo
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Daisuke Kadowaki
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Sojo University
| |
Collapse
|
28
|
Ohashi N, Kohno T. Analgesic Effect of Acetaminophen: A Review of Known and Novel Mechanisms of Action. Front Pharmacol 2020; 11:580289. [PMID: 33328986 PMCID: PMC7734311 DOI: 10.3389/fphar.2020.580289] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/22/2020] [Indexed: 11/13/2022] Open
Abstract
Acetaminophen is one of the most commonly used analgesic agents for treating acute and chronic pain. However, its metabolism is complex, and its analgesic mechanisms have not been completely understood. Previously, it was believed that acetaminophen induces analgesia by inhibiting cyclooxygenase enzymes; however, it has been considered recently that the main analgesic mechanism of acetaminophen is its metabolization to N-acylphenolamine (AM404), which then acts on the transient receptor potential vanilloid 1 (TRPV1) and cannabinoid 1 receptors in the brain. We also recently revealed that the acetaminophen metabolite AM404 directly induces analgesia via TRPV1 receptors on terminals of C-fibers in the spinal dorsal horn. It is known that, similar to the brain, the spinal dorsal horn is critical to pain pathways and modulates nociceptive transmission. Therefore, acetaminophen induces analgesia by acting not only on the brain but also the spinal cord. In addition, acetaminophen is not considered to possess any anti-inflammatory activity because of its weak inhibition of cyclooxygenase (COX). However, we also revealed that AM404 induces analgesia via TRPV1 receptors on the spinal dorsal horn in an inflammatory pain rat model, and these analgesic effects were stronger in the model than in naïve rats. The purpose of this review was to summarize the previous and new issues related to the analgesic mechanisms of acetaminophen. We believe that it will allow clinicians to consider new pain management techniques involving acetaminophen.
Collapse
Affiliation(s)
- Nobuko Ohashi
- Division of Anesthesiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tatsuro Kohno
- Department of Anesthesiology and Intensive Care Medicine, International University of Health and Welfare School of Medicine, Narita, Japan
| |
Collapse
|
29
|
Bamps D, Vriens J, de Hoon J, Voets T. TRP Channel Cooperation for Nociception: Therapeutic Opportunities. Annu Rev Pharmacol Toxicol 2020; 61:655-677. [PMID: 32976736 DOI: 10.1146/annurev-pharmtox-010919-023238] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chronic pain treatment remains a sore challenge, and in our aging society, the number of patients reporting inadequate pain relief continues to grow. Current treatment options all have their drawbacks, including limited efficacy and the propensity of abuse and addiction; the latter is exemplified by the ongoing opioid crisis. Extensive research in the last few decades has focused on mechanisms underlying chronic pain states, thereby producing attractive opportunities for novel, effective and safe pharmaceutical interventions. Members of the transient receptor potential (TRP) ion channel family represent innovative targets to tackle pain sensation at the root. Three TRP channels, TRPV1, TRPM3, and TRPA1, are of particular interest, as they were identified as sensors of chemical- and heat-induced pain in nociceptor neurons. This review summarizes the knowledge regarding TRP channel-based pain therapies, including the bumpy road of the clinical development of TRPV1 antagonists, the current status of TRPA1 antagonists, and the future potential of targeting TRPM3.
Collapse
Affiliation(s)
- Dorien Bamps
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Jan de Hoon
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium; .,Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
30
|
Abstract
Non-steroidal anti-inflammatory drugs produce antinociceptive effects mainly through peripheral cyclooxygenase inhibition. In opposition to the classical non-steroidal anti-inflammatory drugs, paracetamol and dipyrone exert weak anti-inflammatory activity, their antinociceptive effects appearing to be mostly due to mechanisms other than peripheral cyclooxygenase inhibition. In this review, we classify classical non-steroidal anti-inflammatory drugs, paracetamol and dipyrone as “non-opioid analgesics” and discuss the mechanisms mediating participation of the endocannabinoid system in their antinociceptive effects. Non-opioid analgesics and their metabolites may activate cannabinoid receptors, as well as elevate endocannabinoid levels through different mechanisms: reduction of endocannabinoid degradation via fatty acid amide hydrolase and/or cyclooxygenase-2 inhibition, mobilization of arachidonic acid for the biosynthesis of endocannabinoids due to cyclooxygenase inhibition, inhibition of endocannabinoid cellular uptake directly or through the inhibition of nitric oxide synthase production, and induction of endocannabinoid release.
Collapse
Affiliation(s)
- Ruhan Deniz Topuz
- Department of Medical Pharmacology, Trakya University School of Medicine, Edirne, Turkey
| | - Özgur Gündüz
- Department of Medical Pharmacology, Trakya University School of Medicine, Edirne, Turkey
| | - Çetin Hakan Karadağ
- Department of Medical Pharmacology, Trakya University School of Medicine, Edirne, Turkey
| | - Ahmet Ulugöl
- Department of Medical Pharmacology, Trakya University School of Medicine, Edirne, Turkey
| |
Collapse
|
31
|
Nakagawa F, Higashi S, Ando E, Ohsumi T, Watanabe S, Takeuchi H. Modification of TRPV4 activity by acetaminophen. Heliyon 2020; 6:e03301. [PMID: 32051870 PMCID: PMC7002858 DOI: 10.1016/j.heliyon.2020.e03301] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 12/15/2019] [Accepted: 01/22/2020] [Indexed: 11/25/2022] Open
Abstract
N-Acetyl-p-aminophenol (APAP/acetaminophen) is a widely used analgesic/antipyretic with weaker inhibitory effects on cyclooxygenase compared to those of non-steroidal anti-inflammatory drugs. The effect of APAP is mediated by its metabolites, N-arachidonoyl-phenolamine and N-acetyl-p-benzoquinone imine, which activate transient receptor potential (TRP) channels, including TRP vanilloid 1 (TRPV1) and TRP ankyrin 1 (TRPA1) or cannabinoid receptor type 1. However, the exact molecular mechanism underlying the cellular actions of APAP remains unclear. Recently, we observed that APAP promotes cell migration through TRPV4; in this study, we examined the effect of APAP on Ca2+-channel activity of TRPV4. In the rat cell line PC12 expressing TRPV4, GSK1016790A (GSK), a TRPV4 agonist, stimulated an increase in [Ca2+]i; these effects were abrogated by HC-067047 treatment. This GSK-induced Ca2+ entry through TRPV4 was inhibited by APAP in a dose-dependent manner, whereas APAP alone did not affect [Ca2+]i. The specificity of the effect of APAP on TRPV4 was further confirmed using HeLa cells, which lack endogenous TRPV4 but stably express exogenous TRPV4 (HeLa-mTRPV4). GSK-induced [Ca2+]i elevation was only observed in HeLa-mTRPV4 cells compared to that in the control HeLa cells, indicating the specific action of GSK on TRPV4. APAP dose-dependently suppressed this GSK-induced Ca2+ entry in HeLa-mTRPV4. However, it is unlikely that the metabolites of APAP were involved in these effects as the reaction in this study was rapid. The results suggest that APAP suppresses the newly identified target TRPV4 without being metabolized and exerts antipyretic/analgesic and/or other effects on TRPV4-related phenomena in the body. The effect of APAP on TRPV4 was opposite to that on TRPV1 or TRPA1, as the latter is activated by APAP.
Collapse
Affiliation(s)
- Fumio Nakagawa
- Division of Dental Anesthesiology, Department of Control of Physical Functions, Kyushu Dental University, Kitakyushu, 803-8580, Japan
- Division of Applied Pharmacology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, 803-8580, Japan
| | - Sen Higashi
- Division of Applied Pharmacology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, 803-8580, Japan
| | - Eika Ando
- Division of Dental Anesthesiology, Department of Control of Physical Functions, Kyushu Dental University, Kitakyushu, 803-8580, Japan
| | - Tomoko Ohsumi
- Division of Applied Pharmacology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, 803-8580, Japan
| | - Seiji Watanabe
- Division of Dental Anesthesiology, Department of Control of Physical Functions, Kyushu Dental University, Kitakyushu, 803-8580, Japan
| | - Hiroshi Takeuchi
- Division of Applied Pharmacology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, 803-8580, Japan
- Corresponding author.
| |
Collapse
|
32
|
Saliba SW, Bonifacino T, Serchov T, Bonanno G, de Oliveira ACP, Fiebich BL. Neuroprotective Effect of AM404 Against NMDA-Induced Hippocampal Excitotoxicity. Front Cell Neurosci 2019; 13:566. [PMID: 31920563 PMCID: PMC6932953 DOI: 10.3389/fncel.2019.00566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/06/2019] [Indexed: 11/13/2022] Open
Abstract
Different studies have demonstrated that inflammation and alterations in glutamate neurotransmission are two events contributing to the pathophysiology of neurodegenerative or neurological disorders. There are evidences that N-arachidonoylphenolamine (AM404), a cannabinoid system modulator and paracetamol metabolite, modulates inflammation and exerts neuroprotective effects on Huntington's (HD) and Parkinson's diseases (PD), and ischemia. However, the effects of AM404 on the production of inflammatory mediators and excitotoxicity in brain tissue stimulated with N-methyl-D-aspartic acid (NMDA) are not elucidated. In this present study, we investigated the effects of AM404 on the production of inflammatory mediators and neuronal cell death induced by NMDA in organotypic hippocampal slices cultures (OHSC) using qPCR, western blot (WB), and immunohistochemistry. Moreover, to comprehend the mechanism of excitotoxicity, we evaluated the effects of AM404 on glutamate release in hippocampal synaptosomes and the NMDA-induced calcium responses in acute hippocampal slices. Our results showed that AM404 led to a significant decrease in cell death induced by NMDA, through a mechanism possibly involving the reduction of glutamate release and the calcium ions responses. Furthermore, it decreased the expression of the interleukin (IL)-1β. This study provides new significant insights about the anti-inflammatory and neuroprotection effects of AM404 on NMDA-induced excitotoxicity. To understand the effects of AM404 in these processes might contribute to the therapeutic potential of AM404 in diseases with involvement of neuroinflammation and neurodegeneration and might lead to a possible future treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Soraya Wilke Saliba
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tiziana Bonifacino
- Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Tsvetan Serchov
- Laboratory of Stereotaxy and Interventional Neuroscience, Department of Stereotactic and Functional Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Giambattista Bonanno
- Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Bernd L Fiebich
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Vigo MB, Pérez MJ, De Fino F, Gómez G, Martínez SA, Bisagno V, Di Carlo MB, Scazziota A, Manautou JE, Ghanem CI. Acute acetaminophen intoxication induces direct neurotoxicity in rats manifested as astrogliosis and decreased dopaminergic markers in brain areas associated with locomotor regulation. Biochem Pharmacol 2019; 170:113662. [DOI: 10.1016/j.bcp.2019.113662] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/08/2019] [Indexed: 01/13/2023]
|
34
|
Horton JS, Shiraishi T, Alfulaij N, Small-Howard AL, Turner HC, Kurokawa T, Mori Y, Stokes AJ. "TRPV1 is a component of the atrial natriuretic signaling complex, and using orally delivered antagonists, presents a valid therapeutic target in the longitudinal reversal and treatment of cardiac hypertrophy and heart failure". Channels (Austin) 2019; 13:1-16. [PMID: 30424709 PMCID: PMC6298697 DOI: 10.1080/19336950.2018.1547611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Activation of the atrial natriuretic signaling pathway is intrinsic to the pathological responses associated with a range of cardiovascular diseases that stress the heart, especially those involved in sustained cardiac pressure overload which induces hypertrophy and the pathological remodeling that frequently leads to heart failure. We identify transient receptor potential cation channel, subfamily V, member 1, as a regulated molecular component, and therapeutic target of this signaling system. Data show that TRPV1 is a physical component of the natriuretic peptide A, cGMP, PKG signaling complex, interacting with the Natriuretic Peptide Receptor 1 (NPR1), and upon binding its ligand, Natriuretic Peptide A (NPPA, ANP) TRPV1 activation is subsequently suppressed through production of cGMP and PKG mediated phosphorylation of the TRPV1 channel. Further, inhibition of TRPV1, with orally delivered drugs, suppresses chamber and myocyte hypertrophy, and can longitudinally improve in vivo heart function in mice exposed to chronic pressure overload induced by transverse aortic constriction, reversing pre-established hypertrophy induced by pressure load while restoring chamber function. TRPV1 is a physical and regulated component of the natriuretic peptide signaling system, and TRPV1 inhibition may provide a new treatment strategy for treating, and reversing the loss of function associated with cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Jaime S Horton
- a Laboratory of Experimental Medicine, John A. Burns School of Medicine , University of Hawaii , Honolulu , HI 96813 USA
| | - Takuya Shiraishi
- i Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Naghum Alfulaij
- a Laboratory of Experimental Medicine, John A. Burns School of Medicine , University of Hawaii , Honolulu , HI 96813 USA
| | | | - Helen C Turner
- b Department of Cell and Molecular Biology, John A. Burns School of Medicine , University of Hawaii , Honolulu , HI 96813 USA.,c Queen's Medical Center, Punchbowl Street , Honolulu, HI, USA.,d Division of Natural Sciences and Mathematics, Chaminade University , Honolulu, HI USA
| | - Tatsuki Kurokawa
- h Department of Pathophysiology Faculty of Medicine, Oita University 1-1 Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan.,i Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Yasuo Mori
- i Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Alexander J Stokes
- a Laboratory of Experimental Medicine, John A. Burns School of Medicine , University of Hawaii , Honolulu , HI 96813 USA.,b Department of Cell and Molecular Biology, John A. Burns School of Medicine , University of Hawaii , Honolulu , HI 96813 USA.,c Queen's Medical Center, Punchbowl Street , Honolulu, HI, USA.,d Division of Natural Sciences and Mathematics, Chaminade University , Honolulu, HI USA.,e Department of Molecular Biosciences and Bioengineering, University of Hawaii , Honolulu, HI 96822 USA.,f Diabetes Research Center, John A. Burns School of Medicine, University of Hawaii , Honolulu, HI 96813 USA
| |
Collapse
|
35
|
Malinowska B, Toczek M, Pędzińska‐Betiuk A, Schlicker E. Cannabinoids in arterial, pulmonary and portal hypertension - mechanisms of action and potential therapeutic significance. Br J Pharmacol 2019; 176:1395-1411. [PMID: 29455452 PMCID: PMC6487561 DOI: 10.1111/bph.14168] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/24/2018] [Accepted: 02/05/2018] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system is overactivated in arterial, pulmonary and portal hypertension. In this paper, we present limited clinical data concerning the role of cannabinoids in human hypertension including polymorphism of endocannabinoid system components. We underline differences between the acute cannabinoid administration and their potential hypotensive effect after chronic application in experimental hypertension. We discuss pleiotropic effects of cannabinoids on the cardiovascular system mediated via numerous neuronal and non‐neuronal mechanisms both in normotension and in hypertension. The final results are dependent on the model of hypertension, age, sex, the cannabinoid ligands used or the action via endocannabinoid metabolites. More experimental and clinical studies are needed to clarify the role of endocannabinoids in hypertension, not only in the search for new therapeutic strategies but also in the context of cardiovascular effects of cannabinoids and the steadily increasing legalization of cannabis use for recreational and medical purposes.Linked ArticlesThis article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc
Collapse
Affiliation(s)
- Barbara Malinowska
- Department of Experimental Physiology and PathophysiologyMedical University of BiałystokBiałystokPoland
| | - Marek Toczek
- Department of Experimental Physiology and PathophysiologyMedical University of BiałystokBiałystokPoland
| | - Anna Pędzińska‐Betiuk
- Department of Experimental Physiology and PathophysiologyMedical University of BiałystokBiałystokPoland
| | | |
Collapse
|
36
|
Aitken P, Stanescu I, Playne R, Zhang J, Frampton CMA, Atkinson HC. An integrated safety analysis of combined acetaminophen and ibuprofen (Maxigesic ® /Combogesic ®) in adults. J Pain Res 2019; 12:621-634. [PMID: 30804681 PMCID: PMC6371943 DOI: 10.2147/jpr.s189605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Acetaminophen (APAP) and ibuprofen (IBP) are two analgesic compounds with a long history of use. Both are considered safe at recommended over-the-counter daily doses. Chronic use, high doses, or concomitant medication can produce safety risks for both drugs. APAP is associated with increased risk of hepatic injury, while IBP can produce gastric bleeding and thromboembolic events. Using a combination of APAP and IBP provides superior analgesia without transgressing daily dose limits of each individual drug. METHODS The present study aimed to determine if treatment with a fixed-dose combination (FDC) containing APAP and IBP results in any unexpected adverse events (AEs) and/or changes in the safety profiles of its two ingredients compared to monotherapy. The analysis will examine clinical safety data obtained from either single dose trials, multiple dose trials, a long-term exposure trial, and post-marketing surveillance data of APAP/IBP FDC tablets (Maxigesic®/Combogesic®, AFT Pharmaceuticals Ltd). The largest dataset was obtained by pooling the four randomized-controlled, multiple-dose clinical studies with either APAP 325 mg + IBP 97.5 mg (FDC 325/97.5, three tablets per dose) or APAP 500 mg + IBP 150 mg (FDC 500/150, two tablets per dose). At maximum doses, the two FDCs are bioequivalent, permitting the pooling of data for the analysis of safety. RESULTS A safety population of 922 patients who received full doses of either FDC, APAP alone, IBP alone, or placebo was compiled from the four studies. A total of 521 AEs were experienced with the incidence of FDC AEs similar to or below either monotherapy group or placebo. The FDC did not alter the incidence and percentage of the most common AEs, including gastrointestinal events and postoperative bleeding. CONCLUSION Overall, the FDC is well tolerated and has a strong safety profile at single and multiple doses with improved efficacy over monotherapy.
Collapse
Affiliation(s)
- Phillip Aitken
- Drug Development, AFT Pharmaceuticals Ltd, Auckland, New Zealand,
| | - Ioana Stanescu
- Drug Development, AFT Pharmaceuticals Ltd, Auckland, New Zealand,
| | - Rebecca Playne
- Drug Development, AFT Pharmaceuticals Ltd, Auckland, New Zealand,
| | - Jennifer Zhang
- Drug Development, AFT Pharmaceuticals Ltd, Auckland, New Zealand,
| | | | | |
Collapse
|
37
|
Gianessi CA, Groman SM, Taylor JR. Bi-directional modulation of food habit expression by the endocannabinoid system. Eur J Neurosci 2019; 49:1610-1622. [PMID: 30589475 DOI: 10.1111/ejn.14330] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/03/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022]
Abstract
The compulsive, habitual behaviors that have been observed in individuals diagnosed with substance use disorders may be due to disruptions in the neural circuits that mediate goal-directed actions. The endocannabinoid system has been shown to play a critical role in habit learning, but the role of this neuromodulatory system in habit expression is unclear. Here, we investigated the role of the endocannabinoid system in established habitual actions using contingency degradation in male C57BL/6 mice. We found that administration of the endocannabinoid transport inhibitor AM404 reduced habitual responding for food and that antagonism of cannabinoid receptor type 1 (CB1), but not transient receptor potential cation subfamily V (TRPV1), receptors produced a similar reduction in habitual responding. Moreover, pharmacological stimulation of CB1 receptors increased habitual responding for food. Co-administration of an enzyme inhibitor that selectively increases the endocannabinoid 2-arachidonoyl glycerol (2-AG) with AM404 partially restored habitual responding for food. Together, these findings demonstrate an important role for the endocannabinoid system in the expression of habits and provide novel insights into potential pharmacological strategies for reducing habitual behaviors in mental disorders.
Collapse
Affiliation(s)
- Carol A Gianessi
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Interdepartmental Neuroscience Program, Yale University Graduate School of Arts and Sciences, New Haven, Connecticut
| | - Stephanie M Groman
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Jane R Taylor
- Department of Psychiatry, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, Connecticut.,Interdepartmental Neuroscience Program, Yale University Graduate School of Arts and Sciences, New Haven, Connecticut.,Department of Psychology, Yale University, New Haven, Connecticut
| |
Collapse
|
38
|
TRPV1 mediates the anticonvulsant effects of acetaminophen in mice. Epilepsy Res 2018; 145:153-159. [DOI: 10.1016/j.eplepsyres.2018.06.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 12/16/2022]
|
39
|
Nazıroğlu M, Taner AN, Balbay E, Çiğ B. Inhibitions of anandamide transport and FAAH synthesis decrease apoptosis and oxidative stress through inhibition of TRPV1 channel in an in vitro seizure model. Mol Cell Biochem 2018; 453:143-155. [PMID: 30159798 DOI: 10.1007/s11010-018-3439-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/24/2018] [Indexed: 02/03/2023]
Abstract
The expression level of TRPV1 is high in hippocampus which is a main epileptic area in the brain. In addition to the actions of capsaicin (CAP) and reactive oxygen species (ROS), the TRPV1 channel is activated in neurons by endogenous cannabinoid, anandamide (AEA). In the current study, we investigated the role of inhibitors of TRPV1 (capsazepine, CPZ), AEA transport (AM404), and FAAH (URB597) on the modulation of Ca2+ entry, apoptosis, and oxidative stress in in vitro seizure-induced rat hippocampus and human glioblastoma (DBTRG) cell line. The seizure was induced in the hippocampal and DBTRG neurons using in vitro 4-aminopyridine (4-AP) to trigger a seizure-like activity model. CPZ and AM404 were fully effective in reversing 4-AP-induced intracellular free Ca2+ concentration of the hippocampus and TRPV1 current density of DBTRG. However, AEA and CAP did not activate TRPV1 in the URB597-treated neurons. Hence, we observed TRPV1 blocker effects of URB597 in the DBTRG neurons. In addition, the AM404 and CPZ treatments decreased intracellular ROS production, mitochondrial membrane depolarization, apoptosis, caspases 3 and 9 values in the hippocampus. In conclusion, the results indicate that inhibition of AEA transport, FAAH synthesis, and TRPV1 activity can result in remarkable neuroprotective effects in the epileptic neurons. Possible molecular pathways of involvement of capsazepine (CPZ) and AM4040 in anandamide and capsaicin (CAP)-induced apoptosis, oxidative stress, and Ca2+ accumulation through TRPV1 channel in the seizure-induced rat hippocampus and human glioblastoma neurons. The TRPV1 channel is activated by different stimuli including reactive oxygen species (ROS), anandamide (AEA), and CAP and it is blocked by capsazepine (CPZ). Cannabinoid receptor type 1 (CB1) is also activated by AEA. The AEA levels in cytosol are decreased by fatty acid amide hydrolase (FAAH) enzyme. Inhibition of FAAH through URB597 induces stimulation of CB1 receptor through accumulation AEA. URB597 acts antiepileptic effects through inhibition of TRPV1. Overloaded Ca2+ concentration of mitochondria can induce an apoptotic program by stimulating the release of apoptosis-promoting factors such as caspases 3 and caspase 9 by generating ROS due to respiratory chain damage. AM404 and CPZ reduce TRPV1 channel activation and Ca2+ entry in the in vitro 4-AP seizure model-induced hippocampal and glioblastoma neurons.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, 32260, Isparta, Turkey. .,Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.
| | - Afife Nur Taner
- Medicine Faculty, Suleyman Demirel University, Isparta, Turkey
| | - Esra Balbay
- Medicine Faculty, Suleyman Demirel University, Isparta, Turkey
| | - Bilal Çiğ
- Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
40
|
Di Marzo V. New approaches and challenges to targeting the endocannabinoid system. Nat Rev Drug Discov 2018; 17:623-639. [DOI: 10.1038/nrd.2018.115] [Citation(s) in RCA: 256] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Mirrasekhian E, Nilsson JLÅ, Shionoya K, Blomgren A, Zygmunt PM, Engblom D, Högestätt ED, Blomqvist A. The antipyretic effect of paracetamol occurs independent of transient receptor potential ankyrin 1-mediated hypothermia and is associated with prostaglandin inhibition in the brain. FASEB J 2018; 32:5751-5759. [PMID: 29738273 DOI: 10.1096/fj.201800272r] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The mode of action of paracetamol (acetaminophen), which is widely used for treating pain and fever, has remained obscure, but may involve several distinct mechanisms, including cyclooxygenase inhibition and transient receptor potential ankyrin 1 (TRPA1) channel activation, the latter being recently associated with paracetamol's propensity to elicit hypothermia at higher doses. Here, we examined whether the antipyretic effect of paracetamol was due to TRPA1 activation or cyclooxygenase inhibition. Treatment of wild-type and TRPA1 knockout mice rendered febrile by immune challenge with LPS with a dose of paracetamol that did not produce hypothermia (150 mg/kg) but is known to be analgetic, abolished fever in both genotypes. Paracetamol completely suppressed the LPS-induced elevation of prostaglandin E2 in the brain and also reduced the levels of several other prostanoids. The hypothermia induced by paracetamol was abolished in mice treated with the electrophile-scavenger N-acetyl cysteine. We conclude that paracetamol's antipyretic effect in mice is dependent on inhibition of cyclooxygenase activity, including the formation of pyrogenic prostaglandin E2, whereas paracetamol-induced hypothermia likely is mediated by the activation of TRPA1 by electrophilic metabolites of paracetamol, similar to its analgesic effect in some experimental paradigms.-Mirrasekhian, E., Nilsson, J. L. Å., Shionoya, K., Blomgren, A., Zygmunt, P. M., Engblom, D., Högestätt, E. D., Blomqvist, A. The antipyretic effect of paracetamol occurs independent of transient receptor potential ankyrin 1-mediated hypothermia and is associated with prostaglandin inhibition in the brain.
Collapse
Affiliation(s)
- Elahe Mirrasekhian
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; and
| | - Johan L Å Nilsson
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kiseko Shionoya
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; and
| | - Anders Blomgren
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Peter M Zygmunt
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - David Engblom
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; and
| | - Edward D Högestätt
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anders Blomqvist
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; and
| |
Collapse
|
42
|
Stueber T, Meyer S, Jangra A, Hage A, Eberhardt M, Leffler A. Activation of the capsaicin-receptor TRPV1 by the acetaminophen metabolite N-arachidonoylaminophenol results in cytotoxicity. Life Sci 2017; 194:67-74. [PMID: 29273526 DOI: 10.1016/j.lfs.2017.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 12/11/2022]
Abstract
AIMS The anandamide reuptake inhibitor N-arachidonoylaminophenol (AM404) and the reactive substance N-acetyl-p-benzoquinone imine (NAPQI) are both metabolites of acetaminophen and may contribute to acetaminophen-induced analgesia by acting at TRPV1 expressed in the peripheral or central nervous system. While NAPQI slowly sensitizes and activates TRPV1 by interacting with distinct intracellular cysteine residues, detailed properties of AM404 as an agonist of TRPV1 have not yet been reported on. We explored the effects of AM404 on recombinant human TRPV1 and in rodent dorsal root ganglion (DRG) neurons. MATERIALS AND METHODS HEK 293 cells expressing different isoforms of recombinant TRPV1 and rodent DRG neurons were employed for patch clamp and calcium imaging experiments. Cytotoxicity was assessed by propidium iodide and Annexin V staining on TRPV1-HEK 293 cells and with trypan blue staining on DRG neurons. KEY FINDINGS AM404 activates hTRPV1 at concentrations >1μM and in a concentration-dependent manner. AM404 also potentiates TRPV1-mediated currents evoked by heat and anandamide. Moreover, AM404-evoked currents are potentiated by NAPQI. While the partly capsaicin-insensitive rabbit (o) TRPV1 fails to respond to AM404, AM404-sensitivity is restored by insertion of the capsaicin binding-domain of rat TRPV1 into oTRPV1. In DRG neurons, AM404-evoked calcium influx as well as cell death is mediated by TRPV1. SIGNIFICANCE AM404 gates TRPV1 by interacting with the vanilloid-binding site, and TRPV1 is the main receptor for AM404 in DRG neurons. While direct activation of TRPV1 requires high concentrations of AM404, it is possible that synergistic effects of AM404 with further TRPV1-agonists may occur at clinically relevant concentrations.
Collapse
Affiliation(s)
- Thomas Stueber
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Susanne Meyer
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Annette Jangra
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Axel Hage
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Mirjam Eberhardt
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Andreas Leffler
- Department of Anaesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
43
|
Saliba SW, Marcotegui AR, Fortwängler E, Ditrich J, Perazzo JC, Muñoz E, de Oliveira ACP, Fiebich BL. AM404, paracetamol metabolite, prevents prostaglandin synthesis in activated microglia by inhibiting COX activity. J Neuroinflammation 2017; 14:246. [PMID: 29237478 PMCID: PMC5729401 DOI: 10.1186/s12974-017-1014-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/27/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND N-arachidonoylphenolamine (AM404), a paracetamol metabolite, is a potent agonist of the transient receptor potential vanilloid type 1 (TRPV1) and low-affinity ligand of the cannabinoid receptor type 1 (CB1). There is evidence that AM404 exerts its pharmacological effects in immune cells. However, the effect of AM404 on the production of inflammatory mediators of the arachidonic acid pathway in activated microglia is still not fully elucidated. METHOD In the present study, we investigated the effects of AM404 on the eicosanoid production induced by lipopolysaccharide (LPS) in organotypic hippocampal slices culture (OHSC) and primary microglia cultures using Western blot, immunohistochemistry, and ELISA. RESULTS Our results show that AM404 inhibited LPS-mediated prostaglandin E2 (PGE2) production in OHSC, and LPS-stimulated PGE2 release was totally abolished in OHSC if microglial cells were removed. In primary microglia cultures, AM404 led to a significant dose-dependent decrease in the release of PGE2, independent of TRPV1 or CB1 receptors. Moreover, AM404 also inhibited the production of PGD2 and the formation of reactive oxygen species (8-iso-PGF2 alpha) with a reversible reduction of COX-1- and COX-2 activity. Also, it slightly decreased the levels of LPS-induced COX-2 protein, although no effect was observed on LPS-induced mPGES-1 protein synthesis. CONCLUSIONS This study provides new significant insights about the potential anti-inflammatory role of AM404 and new mechanisms of action of paracetamol on the modulation of prostaglandin production by activated microglia.
Collapse
Affiliation(s)
- Soraya Wilke Saliba
- Department of Psychiatry and Psychotherapy, Laboratory of Translational Psychiatry, Faculty of Medicine, Medical Center – University of Freiburg, Hauptstr. 5, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ariel R. Marcotegui
- Laboratory of Hepatic Encephalopathy and Portal Hypertension, Center of Applied and Experimental Pathology, University of Buenos Aires, Buenos Aires, Argentina
| | - Ellen Fortwängler
- Department of Psychiatry and Psychotherapy, Laboratory of Translational Psychiatry, Faculty of Medicine, Medical Center – University of Freiburg, Hauptstr. 5, 79104 Freiburg, Germany
| | - Johannes Ditrich
- Department of Psychiatry and Psychotherapy, Laboratory of Translational Psychiatry, Faculty of Medicine, Medical Center – University of Freiburg, Hauptstr. 5, 79104 Freiburg, Germany
| | - Juan Carlos Perazzo
- Laboratory of Hepatic Encephalopathy and Portal Hypertension, Center of Applied and Experimental Pathology, University of Buenos Aires, Buenos Aires, Argentina
| | - Eduardo Muñoz
- Departamento de Biología Celular, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain
| | | | - Bernd L. Fiebich
- Department of Psychiatry and Psychotherapy, Laboratory of Translational Psychiatry, Faculty of Medicine, Medical Center – University of Freiburg, Hauptstr. 5, 79104 Freiburg, Germany
| |
Collapse
|
44
|
Sharma CV, Long JH, Shah S, Rahman J, Perrett D, Ayoub SS, Mehta V. First evidence of the conversion of paracetamol to AM404 in human cerebrospinal fluid. J Pain Res 2017; 10:2703-2709. [PMID: 29238213 PMCID: PMC5716395 DOI: 10.2147/jpr.s143500] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Paracetamol is arguably the most commonly used analgesic and antipyretic drug worldwide, however its mechanism of action is still not fully established. It has been shown to exert effects through multiple pathways, some actions suggested to be mediated via N-arachidonoylphenolamine (AM404). AM404, formed through conjugation of paracetamol-derived p-aminophenol with arachidonic acid in the brain, is an activator of the capsaicin receptor, TRPV1, and inhibits the reuptake of the endocannabinoid, anandamide, into postsynaptic neurons, as well as inhibiting synthesis of PGE2 by COX-2. However, the presence of AM404 in the central nervous system following administration of paracetamol has not yet been demonstrated in humans. Cerebrospinal fluid (CSF) and blood were collected from 26 adult male patients between 10 and 211 minutes following intravenous administration of 1 g of paracetamol. Paracetamol was measured by high-performance liquid chromatography with UV detection. AM404 was measured by liquid chromatography-tandem mass spectrometry. AM404 was detected in 17 of the 26 evaluable CSF samples at 5–40 nmol⋅L−1. Paracetamol was measurable in CSF within 10 minutes, with a maximum measured concentration of 60 μmol⋅L−1 at 206 minutes. This study is the first to report on the presence of AM404 in human CSF following paracetamol administration. This may represent an important finding in our understanding of paracetamol’s mechanism of action, although measured concentrations were far below the previously documented IC50 for this metabolite.
Collapse
Affiliation(s)
- Chhaya V Sharma
- Pain & Anaesthesia Research Centre, St Bartholomew's and The Royal London Hospitals, Barts Health NHS Trust, London, UK
| | - Jamie H Long
- Barts & The London School of Medicine, Queen Mary University of London, London, UK
| | - Seema Shah
- Pain & Anaesthesia Research Centre, St Bartholomew's and The Royal London Hospitals, Barts Health NHS Trust, London, UK
| | - Junia Rahman
- Pain & Anaesthesia Research Centre, St Bartholomew's and The Royal London Hospitals, Barts Health NHS Trust, London, UK
| | - David Perrett
- BioAnalytical Science, Barts & The London School of Medicine, Queen Mary University of London, London, UK
| | - Samir S Ayoub
- School of Health, Sport and Bioscience, Medicines Research Group, University of East London, London, UK
| | - Vivek Mehta
- Pain & Anaesthesia Research Centre, St Bartholomew's and The Royal London Hospitals, Barts Health NHS Trust, London, UK
| |
Collapse
|
45
|
Reactive metabolites of acetaminophen activate and sensitize the capsaicin receptor TRPV1. Sci Rep 2017; 7:12775. [PMID: 28986540 PMCID: PMC5630573 DOI: 10.1038/s41598-017-13054-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 09/13/2017] [Indexed: 02/07/2023] Open
Abstract
The irritant receptor TRPA1 was suggested to mediate analgesic, antipyretic but also pro-inflammatory effects of the non-opioid analgesic acetaminophen, presumably due to channel activation by the reactive metabolites parabenzoquinone (pBQ) and N-acetyl-parabenzoquinonimine (NAPQI). Here we explored the effects of these metabolites on the capsaicin receptor TRPV1, another redox-sensitive ion channel expressed in sensory neurons. Both pBQ and NAPQI, but not acetaminophen irreversibly activated and sensitized recombinant human and rodent TRPV1 channels expressed in HEK 293 cells. The reducing agents dithiothreitol and N-acetylcysteine abolished these effects when co-applied with the metabolites, and both pBQ and NAPQI failed to gate TRPV1 following substitution of the intracellular cysteines 158, 391 and 767. NAPQI evoked a TRPV1-dependent increase in intracellular calcium and a potentiation of heat-evoked currents in mouse spinal sensory neurons. Although TRPV1 is expressed in mouse hepatocytes, inhibition of TRPV1 did not alleviate acetaminophen-induced hepatotoxicity. Finally, intracutaneously applied NAPQI evoked burning pain and neurogenic inflammation in human volunteers. Our data demonstrate that pBQ and NAQPI activate and sensitize TRPV1 by interacting with intracellular cysteines. While TRPV1 does not seem to mediate acetaminophen-induced hepatotoxicity, our data identify TRPV1 as a target of acetaminophen with a potential relevance for acetaminophen-induced analgesia, antipyresia and inflammation.
Collapse
|
46
|
Acetaminophen Metabolite N-Acylphenolamine Induces Analgesia via Transient Receptor Potential Vanilloid 1 Receptors Expressed on the Primary Afferent Terminals of C-fibers in the Spinal Dorsal Horn. Anesthesiology 2017; 127:355-371. [PMID: 28542001 DOI: 10.1097/aln.0000000000001700] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The widely used analgesic acetaminophen is metabolized to N-acylphenolamine, which induces analgesia by acting directly on transient receptor potential vanilloid 1 or cannabinoid 1 receptors in the brain. Although these receptors are also abundant in the spinal cord, no previous studies have reported analgesic effects of acetaminophen or N-acylphenolamine mediated by the spinal cord dorsal horn. We hypothesized that clinical doses of acetaminophen induce analgesia via these spinal mechanisms. METHODS We assessed our hypothesis in a rat model using behavioral measures. We also used in vivo and in vitro whole cell patch-clamp recordings of dorsal horn neurons to assess excitatory synaptic transmission. RESULTS Intravenous acetaminophen decreased peripheral pinch-induced excitatory responses in the dorsal horn (53.1 ± 20.7% of control; n = 10; P < 0.01), while direct application of acetaminophen to the dorsal horn did not reduce these responses. Direct application of N-acylphenolamine decreased the amplitudes of monosynaptic excitatory postsynaptic currents evoked by C-fiber stimulation (control, 462.5 ± 197.5 pA; N-acylphenolamine, 272.5 ± 134.5 pA; n = 10; P = 0.022) but not those evoked by stimulation of Aδ-fibers. These phenomena were mediated by transient receptor potential vanilloid 1 receptors, but not cannabinoid 1 receptors. The analgesic effects of acetaminophen and N-acylphenolamine were stronger in rats experiencing an inflammatory pain model compared to naïve rats. CONCLUSIONS Our results suggest that the acetaminophen metabolite N-acylphenolamine induces analgesia directly via transient receptor potential vanilloid 1 receptors expressed on central terminals of C-fibers in the spinal dorsal horn and leads to conduction block, shunt currents, and desensitization of these fibers.
Collapse
|
47
|
Extinction of avoidance behavior by safety learning depends on endocannabinoid signaling in the hippocampus. J Psychiatr Res 2017; 90:46-59. [PMID: 28222356 DOI: 10.1016/j.jpsychires.2017.02.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/24/2017] [Accepted: 02/02/2017] [Indexed: 12/29/2022]
Abstract
The development of exaggerated avoidance behavior is largely responsible for the decreased quality of life in patients suffering from anxiety disorders. Studies using animal models have contributed to the understanding of the neural mechanisms underlying the acquisition of avoidance responses. However, much less is known about its extinction. Here we provide evidence in mice that learning about the safety of an environment (i.e., safety learning) rather than repeated execution of the avoided response in absence of negative consequences (i.e., response extinction) allowed the animals to overcome their avoidance behavior in a step-down avoidance task. This process was context-dependent and could be blocked by pharmacological (3 mg/kg, s.c.; SR141716) or genetic (lack of cannabinoid CB1 receptors in neurons expressing dopamine D1 receptors) inactivation of CB1 receptors. In turn, the endocannabinoid reuptake inhibitor AM404 (3 mg/kg, i.p.) facilitated safety learning in a CB1-dependent manner and attenuated the relapse of avoidance behavior 28 days after conditioning. Safety learning crucially depended on endocannabinoid signaling at level of the hippocampus, since intrahippocampal SR141716 treatment impaired, whereas AM404 facilitated safety learning. Other than AM404, treatment with diazepam (1 mg/kg, i.p.) impaired safety learning. Drug effects on behavior were directly mirrored by drug effects on evoked activity propagation through the hippocampal trisynaptic circuit in brain slices: As revealed by voltage-sensitive dye imaging, diazepam impaired whereas AM404 facilitated activity propagation to CA1 in a CB1-dependent manner. In line with this, systemic AM404 enhanced safety learning-induced expression of Egr1 at level of CA1. Together, our data render it likely that AM404 promotes safety learning by enhancing information flow through the trisynaptic circuit to CA1.
Collapse
|
48
|
Nazıroğlu M, Çiğ B, Blum W, Vizler C, Buhala A, Marton A, Katona R, Jósvay K, Schwaller B, Oláh Z, Pecze L. Targeting breast cancer cells by MRS1477, a positive allosteric modulator of TRPV1 channels. PLoS One 2017. [PMID: 28640864 PMCID: PMC5481018 DOI: 10.1371/journal.pone.0179950] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
There is convincing epidemiological and experimental evidence that capsaicin, a potent natural transient receptor potential cation channel vanilloid member 1 (TRPV1) agonist, has anticancer activity. However, capsaicin cannot be given systemically in large doses, because of its induction of acute pain and neurological inflammation. MRS1477, a dihydropyridine derivative acts as a positive allosteric modulator of TRPV1, if added together with capsaicin, but is ineffective, if given alone. Addition of MRS1477 evoked Ca2+ signals in MCF7 breast cancer cells, but not in primary breast epithelial cells. This indicates that MCF7 cells not only express functional TRPV1 channels, but also produce endogenous TRPV1 agonists. We investigated the effects of MRS1477 and capsaicin on cell viability, caspase-3 and -9 activities and reactive oxygen species production in MCF7 cells. The fraction of apoptotic cells was increased after 3 days incubation with capsaicin (10 μM) paralleled by increased reactive oxygen species production and caspase activity. These effects were even more pronounced, when cells were incubated with MRS1477 (2 μM) either alone or together with CAPS (10 μM). Capsazepine, a TRPV1 blocker, inhibited both the effect of capsaicin and MRS1477. Whole-cell patch clamp recordings revealed that capsaicin-evoked TRPV1-mediated current density levels were increased after 3 days incubation with MRS1477 (2 μM). However, the tumor growth in MCF7 tumor-bearing immunodeficient mice was not significantly decreased after treatment with MRS1477 (10 mg/ kg body weight, i.p., injection twice a week). In conclusion, in view of a putative in vivo treatment with MRS1477 or similar compounds further optimization is required.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey
- Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey
| | - Bilal Çiğ
- Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey
| | - Walter Blum
- Unit of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Csaba Vizler
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Andrea Buhala
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Annamária Marton
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Róbert Katona
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Katalin Jósvay
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Beat Schwaller
- Unit of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Zoltán Oláh
- Institute of Chemistry, Faculty of Materials Science and Engineering, University of Miskolc, Miskolc-Egyetemváros, Hungary
- Acheuron Ltd., Szeged, Hungary
| | - László Pecze
- Unit of Anatomy, Department of Medicine, University of Fribourg, Fribourg, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Kress HG, Untersteiner G. Clinical update on benefit versus risks of oral paracetamol alone or with codeine: still a good option? Curr Med Res Opin 2017; 33:289-304. [PMID: 27842443 DOI: 10.1080/03007995.2016.1254606] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND After decades of worldwide use of paracetamol/acetaminophen as a popular and apparently safe prescription and over-the-counter medicine, the future role of this poorly understood analgesic has been seriously questioned by recent concerns about prenatal, cardiovascular (CV) and hepatic safety, and also about its analgesic efficacy. At the same time the usefulness of codeine in combination products has come under debate. METHODS Based on a PubMed database literature search on the terms efficacy, safety, paracetamol, acetaminophen, codeine and their combinations up to and including June 2016, this clinical update reviews the current evidence of the benefit and risks of oral paracetamol alone and with codeine for mild-to-moderate pain in adults, and compares the respective efficacy and safety profiles with those of nonsteroidal anti-inflammatory drugs (NSAIDs). RESULTS Whereas there is a clear strong association of NSAID use and gastrointestinal (GI) and CV morbidity and mortality, evidence for paracetamol with and without codeine supports the recommended use even in most vulnerable individuals, such as the elderly, pregnant women, alcoholics, and compromised GI and CV patients. The controversies and widespread misconceptions about the complex hepatic metabolism and potential hepatotoxicity have been corrected by recent reviews, and paracetamol remains the first-line nonopioid analgesic in patients with liver diseases if notes of caution are applied. CONCLUSION Due to its safety and tolerability profile paracetamol remained a first-line treatment in many international guidelines. Alone and with codeine it is a safe and effective option in adults, whilst NSAIDs are obviously less safe as alternatives, given the risk of potentially fatal GI and CV adverse effects.
Collapse
|
50
|
Manteghi F, Nasehi M, Zarrindast MR. Precondition of right frontal region with anodal tDCS can restore the fear memory impairment induced by ACPA in male mice. EXCLI JOURNAL 2017; 16:1-13. [PMID: 28337114 PMCID: PMC5318674 DOI: 10.17179/excli2016-693] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/05/2016] [Indexed: 01/12/2023]
Abstract
Fear memory and learning cause behavioural patterns such as fight or flight responses, which increase survival probability, but unfit processing of fear memory and learning can lead to maladaptive behaviours and maladies such as phobias, Post-Traumatic Stress Disorder (PTSD) and anxiety disorders. The growing prevalence of these maladies shows the need to quest novel methods for their treatment. We used anodal transcranial direct current stimulation (tDCS) on the right frontal region as a precondition neuromodulator and arachidonylcyclopropylamide (ACPA), a selective CB1 cannabinoid receptor agonist, as a fear memory impairing agent to assess their effects on contextual and auditory fear conditioning (reliable model for fear studies). Right frontal anodal tDCS (0.2 mA for. 20 minutes) 24 hours before the train did not alter contextual and auditory learning and memory in short-term (24 hrs after the training phase). Moreover, intraperitoneal pre-train injection of ACPA (0.1 mg/kg) alone, decreased both contextual and auditory learning and memory in short- but not long-term. Right frontal anodal tDCS improved short-term contextual fear memory in subthreshold doses of ACPA. On the other hand, right frontal anodal tDCS in long-term improved (lower doses of ACPA) and restored (higher doses of ACPA) both fear memories. These findings showed that, aforementioned approach could cause durable learning and memory improvements. Also this combined modality could be useful for fear extinction training and maladies which inflict amnesia.
Collapse
Affiliation(s)
| | - Mohammad Nasehi
- Institute for Cognitive Science Studies (ICSS), Tehran, Iran; Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Institute for Cognitive Science Studies (ICSS), Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|