1
|
Wilson PR, Bridges KH, Scofield M, Wilson SH. Perioperative N-acetylcysteine: evidence and indications. Pain Manag 2024; 14:385-396. [PMID: 39166871 PMCID: PMC11486111 DOI: 10.1080/17581869.2024.2388504] [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: 05/10/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Nonopioid analgesics serve to improve analgesia and limit side effects and risks of perioperative opioids. N-acetylcysteine (NAC), the primary treatment of acetaminophen toxicity, may have perioperative indications, including analgesia. NAC impacts glutathione synthesis, oxidant scavenging, glutamate receptor modulation and neuroinflammation. Potential perioperative benefits include arrhythmia prevention after cardiac surgery, decreased contrast-induced nephropathy, improved post-transplant liver function and superior pulmonary outcomes with general anesthesia. NAC may improve perioperative analgesia, with some studies displaying a reduction in postoperative opioid use. NAC is generally well tolerated with an established safety profile. NAC administration may predispose to gastrointestinal effects, while parenteral administration may carry a risk of anaphylactoid reactions, including bronchospasm. Larger randomized trials may clarify the impact of NAC on perioperative analgesic outcomes.
Collapse
Affiliation(s)
- Phillip Ryan Wilson
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kathryn H Bridges
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Michael Scofield
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Sylvia H Wilson
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
2
|
Busceti CL, Di Menna L, Castaldi S, D'Errico G, Taddeucci A, Bruno V, Fornai F, Pittaluga A, Battaglia G, Nicoletti F. Adaptive Changes in Group 2 Metabotropic Glutamate Receptors Underlie the Deficit in Recognition Memory Induced by Methamphetamine in Mice. eNeuro 2024; 11:ENEURO.0523-23.2024. [PMID: 38969501 PMCID: PMC11298959 DOI: 10.1523/eneuro.0523-23.2024] [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: 12/11/2023] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 07/07/2024] Open
Abstract
Cognitive dysfunction is associated with methamphetamine use disorder (MUD). Here, we used genetic and pharmacological approaches to examine the involvement of either Group 2 metabotropic glutamate (mGlu2) or mGlu3 receptors in memory deficit induced by methamphetamine in mice. Methamphetamine treatment (1 mg/kg, i.p., once a day for 5 d followed by 7 d of withdrawal) caused an impaired performance in the novel object recognition test in wild-type mice, but not in mGlu2-/- or mGlu3-/- mice. Memory deficit in wild-type mice challenged with methamphetamine was corrected by systemic treatment with selectively negative allosteric modulators of mGlu2 or mGlu3 receptors (compounds VU6001966 and VU0650786, respectively). Methamphetamine treatment in wild-type mice caused large increases in levels of mGlu2/3 receptors, the Type 3 activator of G-protein signaling (AGS3), Rab3A, and the vesicular glutamate transporter, vGlut1, in the prefrontal cortex (PFC). Methamphetamine did not alter mGlu2/3-mediated inhibition of cAMP formation but abolished the ability of postsynaptic mGlu3 receptors to boost mGlu5 receptor-mediated inositol phospholipid hydrolysis in PFC slices. Remarkably, activation of presynaptic mGlu2/3 receptors did not inhibit but rather amplified depolarization-induced [3H]-D-aspartate release in synaptosomes prepared from the PFC of methamphetamine-treated mice. These findings demonstrate that exposure to methamphetamine causes changes in the expression and function of mGlu2 and mGlu3 receptors, which might alter excitatory synaptic transmission in the PFC and raise the attractive possibility that selective inhibitors of mGlu2 or mGlu3 receptors (or both) may be used to improve cognitive dysfunction in individuals affected by MUD.
Collapse
Affiliation(s)
| | - Luisa Di Menna
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
| | - Sonia Castaldi
- Department of Physiology and Pharmacology, University Sapienza, Roma 00185, Italy
| | - Giovanna D'Errico
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
| | - Alice Taddeucci
- Department of Pharmacy, University of Genova, Genova 16148, Italy
| | - Valeria Bruno
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
- Department of Physiology and Pharmacology, University Sapienza, Roma 00185, Italy
| | - Francesco Fornai
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56126, Italy
| | - Anna Pittaluga
- Department of Pharmacy, University of Genova, Genova 16148, Italy
- IRCCS Ospedale Policlinico San Martino, Genova 16145, Italy
| | - Giuseppe Battaglia
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
- Department of Physiology and Pharmacology, University Sapienza, Roma 00185, Italy
| | - Ferdinando Nicoletti
- Department of Molecular Pathology, IRCCS Neuromed, Pozzilli 86077, Italy
- Department of Physiology and Pharmacology, University Sapienza, Roma 00185, Italy
| |
Collapse
|
3
|
Wilson SH, Sirianni JM, Bridges KH, Wolf BJ, Valente IE, Scofield MD. The impact of intraoperative N-acetylcysteine on opioid consumption following spine surgery: a randomized pilot trial. Pain Manag 2023; 13:593-602. [PMID: 37877260 PMCID: PMC10694787 DOI: 10.2217/pmt-2023-0061] [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: 04/02/2023] [Accepted: 08/22/2023] [Indexed: 10/26/2023] Open
Abstract
Aim: N-acetylcysteine (NAC) decreases inflammation and could augment perioperative analgesia. Materials & methods: This prospective pilot trial examined postoperative opioid consumption at 12 h following intraoperative NAC. In phase I, 20 adults scheduled for posterior spine surgery were randomized to NAC (0, 50, 100 and 150 mg/kg) to determine the optimal dose. In phase II, 30 patients were randomized to placebo or NAC (150 mg/kg). Opioid consumption, pain ratings and time to opioid rescue were recorded. Results: Postoperative opioid consumption was reduced in the NAC group 19.3% at 12 h and 20% at 18 and 36 h. Opioid consumption was reduced 22-24% in the NAC group at all times after adjusting for intraoperative opioid administration. NAC subjects reported lower pain scores relative to placebo. Conclusion: Subjects randomized to NAC consumed less postoperative opioids and reported less pain versus placebo. Larger randomized controlled trials are needed to further evaluate NAC for analgesia. Clinical Trial Registration: NCT04562597 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Sylvia H Wilson
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Joel M Sirianni
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kathryn H Bridges
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bethany J Wolf
- Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Isabella E Valente
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael D Scofield
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
4
|
Bernatoniene J, Sciupokas A, Kopustinskiene DM, Petrikonis K. Novel Drug Targets and Emerging Pharmacotherapies in Neuropathic Pain. Pharmaceutics 2023; 15:1799. [PMID: 37513986 PMCID: PMC10384314 DOI: 10.3390/pharmaceutics15071799] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Neuropathic pain is a debilitating condition characterized by abnormal signaling within the nervous system, resulting in persistent and often intense sensations of pain. It can arise from various causes, including traumatic nerve injury, neuropathy, and certain diseases. We present an overview of current and emerging pharmacotherapies for neuropathic pain, focusing on novel drug targets and potential therapeutic agents. Current pharmacotherapies, including tricyclic antidepressants, gabapentinoids, and serotonin norepinephrine re-uptake inhibitors, are discussed, as are emerging treatments, such as ambroxol, cannabidiol, and N-acetyl-L-cysteine. Additionally, the article highlights the need for further research in this field to identify new targets and develop more effective and targeted therapies for neuropathic pain management.
Collapse
Affiliation(s)
- Jurga Bernatoniene
- Department of Drug Technology and Social Pharmacy, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania
| | - Arunas Sciupokas
- Pain Clinic, Lithuanian University of Health Sciences Hospital Kauno Klinikos, Eivenių Str. 2, LT-50009 Kaunas, Lithuania
- Department of Neurology, Lithuanian University of Health Sciences, Eivenių Str. 2, LT-50009 Kaunas, Lithuania
| | - Dalia Marija Kopustinskiene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania
| | - Kestutis Petrikonis
- Department of Neurology, Lithuanian University of Health Sciences, Eivenių Str. 2, LT-50009 Kaunas, Lithuania
| |
Collapse
|
5
|
Group II metabotropic glutamate receptor activation attenuates acid-sensing ion channel currents in rat primary sensory neurons. J Biol Chem 2023; 299:102953. [PMID: 36731795 PMCID: PMC9976456 DOI: 10.1016/j.jbc.2023.102953] [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: 09/21/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Acid-sensing ion channels (ASICs) play an important role in pain associated with tissue acidification. Peripheral inhibitory group II metabotropic glutamate receptors (mGluRs) have analgesic effects in a variety of pain conditions. Whether there is a link between ASICs and mGluRs in pain processes is still unclear. Herein, we show that the group II mGluR agonist LY354740 inhibited acid-evoked ASIC currents and action potentials in rat dorsal root ganglia neurons. LY354740 reduced the maximum current response to protons, but it did not change the sensitivity of ASICs to protons. LY354740 inhibited ASIC currents by activating group II mGluRs. We found that the inhibitory effect of LY354740 was blocked by intracellular application of the Gi/o protein inhibitor pertussis toxin and the cAMP analogue 8-Br-cAMP and mimicked by the protein kinase A (PKA) inhibitor H-89. LY354740 also inhibited ASIC3 currents in CHO cells coexpressing mGluR2 and ASIC3 but not in cells expressing ASIC3 alone. In addition, intraplantar injection of LY354740 dose-dependently alleviated acid-induced nociceptive behavior in rats through local group II mGluRs. Together, these results suggested that activation of peripheral group II mGluRs inhibited the functional activity of ASICs through a mechanism that depended on Gi/o proteins and the intracellular cAMP/PKA signaling pathway in rat dorsal root ganglia neurons. We propose that peripheral group II mGluRs are an important therapeutic target for ASIC-mediated pain.
Collapse
|
6
|
Mazzitelli M, Presto P, Antenucci N, Meltan S, Neugebauer V. Recent Advances in the Modulation of Pain by the Metabotropic Glutamate Receptors. Cells 2022; 11:2608. [PMID: 36010684 PMCID: PMC9406805 DOI: 10.3390/cells11162608] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 01/22/2023] Open
Abstract
Metabotropic glutamate receptors (mGluR or mGlu) are G-protein coupled receptors activated by the binding of glutamate, the main classical neurotransmitter of the nervous system. Eight different mGluR subtypes (mGluR1-8) have been cloned and are classified in three groups based on their molecular, pharmacological and signaling properties. mGluRs mediate several physiological functions such as neuronal excitability and synaptic plasticity, but they have also been implicated in numerous pathological conditions including pain. The availability of new and more selective allosteric modulators together with the canonical orthosteric ligands and transgenic technologies has led to significant advances in our knowledge about the role of the specific mGluR subtypes in the pathophysiological mechanisms of various diseases. Although development of successful compounds acting on mGluRs for clinical use has been scarce, the subtype-specific-pharmacological manipulation might be a compelling approach for the treatment of several disorders in humans, including pain; this review aims to summarize and update on preclinical evidence for the roles of different mGluRs in the pain system and discusses knowledge gaps regarding mGluR-related sex differences and neuroimmune signaling in pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Nico Antenucci
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Shakira Meltan
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
7
|
CXCR4/CX43 Regulate Diabetic Neuropathic Pain via Intercellular Interactions between Activated Neurons and Dysfunctional Astrocytes during Late Phase of Diabetes in Rats and the Effects of Antioxidant N-Acetyl-L-Cysteine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8547563. [PMID: 35799894 PMCID: PMC9256426 DOI: 10.1155/2022/8547563] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/15/2022] [Indexed: 12/12/2022]
Abstract
Growing evidence suggests that the interactions between astrocytes and neurons exert important functions in the central sensitization of the spinal cord dorsal horn in rodents with diabetes and neuropathic pain (DNP). However, it still remains unclear how signal transmission occurs in the spinal cord dorsal horn between astrocytes and neurons, especially in subjects with DNP. Chemokine CXC receptor 4 (CXCR4) plays critical roles in DNP, and connexin 43 (CX43), which is also primarily expressed by astrocytes, contributes to the development of neuropathy. We thus postulated that astrocytic and neuronal CXCR4 induces and produces inflammatory factors under persistent peripheral noxious stimulation in DNP, while intercellular CX43 can transmit inflammatory stimulation signals. The results showed that streptozotocin-induced type 1 diabetic rats developed heat hyperalgesia and mechanical allodynia. Diabetes led to persistent neuropathic pain. Diabetic rats developed peripheral sensitization at the early phase (2 weeks) and central sensitization at the late phase (5 weeks) after diabetes induction. Both CXCR4 and CX43, which are localized and coexpressed in neurons and astrocytes, were enhanced significantly in the dorsal horn of spinal cord in rats undergoing DNP during late phase of diabetes, and the CXCR4 antagonist AMD3100 reduced the expression of CX43. The nociceptive behavior was reversed, respectively, by AMD3100 at the early phase and by the antioxidant N-acetyl-L-cysteine (NAC) at the late phase. Furthermore, rats with DNP demonstrated downregulation of glial fibrillary acidic protein (GFAP) as well as upregulation of c-fos in the spinal cord dorsal horn at the late phase compared to the controls, and upregulation of GFAP and downregulation of c-fos were observed upon treatment with NAC. Given that GFAP and c-fos are, respectively, makers of astrocyte and neuronal activation, our findings suggest that CXCR4 as an inflammatory stimulation protein and CX43 as an intercellular signal transmission protein both may induce neurons excitability and astrocytes dysfunction in developing DNP.
Collapse
|
8
|
Yaryari AM, Mousavibahar SH, Amirhassani S, Bagheri M, Mohammadi Y, Mehrpooya M. Men suffering from category III chronic prostatitis may benefit from N-acetylcysteine as an adjunct to alpha-blockers. Low Urin Tract Symptoms 2022; 14:199-207. [PMID: 35068061 DOI: 10.1111/luts.12425] [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: 10/22/2021] [Revised: 12/17/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE We designed this study to investigate the potential use of N-acetylcysteine (NAC) as an adjunct to alpha-blockers in the treatment of category III chronic prostatitis (CP). METHODS Sixty-three men with category III CP with a National Institutes of Health Chronic Prostatitis Symptom Index (NIH-CPSI) total score of 15 or more were randomized to either the NAC treatment group or the placebo treatment group. Besides tamsulosin at a dose of 0.4 mg once daily, participants based on their allocation group received NAC or placebo at a dose of 600 mg twice daily for 12 weeks. The efficacy of the medications was assessed by measuring changes in the NIH-CPSI total score and its subscales, including pain, urinary symptoms, and quality of life. RESULTS Based on the general linear model analysis of the data, over the 12-week treatment, NAC+tamsulosin was statistically superior to placebo+tamsulosin in reducing the total NIH-CPSI score, pain subscore, and quality-of-life subscore (P value <.001). Further, after 12 weeks, more patients in the NAC+tamsulosin group than in the placebo+tamsulosin group met the responder criterion, defined as a decrease of at least 6 points in the NIH-CPSI total score (65.6% vs 29.0%). A more favorable outcome was also noted in the NAC+tamsulosin group regarding the number of patients reporting moderate or marked improvement in symptoms (62.5% vs 25.80%). No significant difference was seen between the groups concerning changes in urinary symptoms. CONCLUSIONS Our study provided clinical evidence that men with category III CP might benefit from NAC treatment. Further studies are needed for the validation of these findings.
Collapse
Affiliation(s)
- Amir-Mohammad Yaryari
- Department of Clinical Pharmacy, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Shahriar Amirhassani
- Urology and Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Bagheri
- Department of Clinical Pharmacy, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Younes Mohammadi
- Modeling of Noncommunicable Diseases Research Center, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Mehrpooya
- Department of Clinical Pharmacy, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Urology and Nephrology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
9
|
Bradlow RCJ, Berk M, Kalivas PW, Back SE, Kanaan RA. The Potential of N-Acetyl-L-Cysteine (NAC) in the Treatment of Psychiatric Disorders. CNS Drugs 2022; 36:451-482. [PMID: 35316513 PMCID: PMC9095537 DOI: 10.1007/s40263-022-00907-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2022] [Indexed: 12/22/2022]
Abstract
N-acetyl-L-cysteine (NAC) is a compound of increasing interest in the treatment of psychiatric disorders. Primarily through its antioxidant, anti-inflammatory, and glutamate modulation activity, NAC has been investigated in the treatment of neurodevelopmental disorders, schizophrenia spectrum disorders, bipolar-related disorders, depressive disorders, anxiety disorders, obsessive compulsive-related disorders, substance-use disorders, neurocognitive disorders, and chronic pain. Whilst there is ample preclinical evidence and theoretical justification for the use of NAC in the treatment of multiple psychiatric disorders, clinical trials in most disorders have yielded mixed results. However, most studies have been underpowered and perhaps too brief, with some evidence of benefit only after months of treatment with NAC. Currently NAC has the most evidence of having a beneficial effect as an adjuvant agent in the negative symptoms of schizophrenia, severe autism, depression, and obsessive compulsive and related disorders. Future research with well-powered studies that are of sufficient length will be critical to better understand the utility of NAC in the treatment of psychiatric disorders.
Collapse
Affiliation(s)
| | - Michael Berk
- IMPACT-The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, VIC Australia ,Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Melbourne, VIC Australia ,Florey Institute of Neuroscience and Mental Health, Melbourne, VIC Australia ,Department of Psychiatry, University of Melbourne, Parkville, VIC Australia
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC USA ,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC USA
| | - Sudie E. Back
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC USA ,Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC USA
| | - Richard A. Kanaan
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC Australia ,Department of Psychiatry, University of Melbourne, Parkville, VIC Australia
| |
Collapse
|
10
|
Mulkens CE, Staatsen M, van Genugten L, Snoeker BAM, Vissers KCP, Bruhn J, Bucx MJL. Postoperative pain reduction by pre-emptive N-acetylcysteine: an exploratory randomized controlled clinical trial. Reg Anesth Pain Med 2021; 46:960-964. [PMID: 34446544 DOI: 10.1136/rapm-2021-102884] [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: 05/25/2021] [Accepted: 08/19/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND A new potential target for multimodal pain management is the group-II metabotropic glutamate receptor subtypes, which can be activated by N-acetylcysteine. We investigated whether pre-emptive administration of N-acetylcysteine leads to a reduction in postoperative pain after laparoscopic inguinal hernia repair. METHODS Sixty American Society of Anesthesiologists I-II patients scheduled for elective inguinal hernia repair were randomized to receive either N-acetylcysteine (150 mg/kg) or placebo intravenously 1 hour before surgery. The primary outcome was the visual analogue score during movement in the morning (approximately 24 hours) after surgery. Among secondary outcomes were postoperative opioid consumption and safety of intravenous N-acetylcysteine. RESULTS In total, 23 patients were analyzed per group. Pain scores were similar at all timepoints with a 24 hours median score of 34 (IQR of 19.0 to 42.5) in the N-acetylcysteine group and a median score of 26 (16.0 to 50.0) in the placebo group. The percentage of patients using opioids after surgery was 22% versus 39% day 1 (p=0.63); 9% versus 26% day 2 (p=0.14); 9% versus 17% day 3 (p=0.35) in the N-acetylcysteine group compared with placebo group. Side effects resembling anaphylactoid reactions in response to the administration of N-acetylcysteine were present in more than half of the patients. CONCLUSIONS Without finding important differences between N-acetylcysteine and placebo group in pain scores postoperatively, but with a high percentage of bothersome side effects for the N-acetylcysteine group, we would not recommend the use of pre-emptive intravenous N-acetylcysteine to reduce postoperative pain in laparoscopic inguinal hernia repair patients based on this study. TRIAL REGISTRATION NUMBER NCT03354572.
Collapse
Affiliation(s)
- Chantal Elise Mulkens
- Department of Anesthesiology, Pain and Palliative Medicine, Radboudumc, Nijmegen, Gelderland, The Netherlands
| | - Marieke Staatsen
- Department of Anesthesiology, Máxima Medical Centre, Eindhoven, Noord-Brabant, The Netherlands
| | - Lucie van Genugten
- Department of Anesthesiology, Slingeland Hospital, Doetinchem, The Netherlands
| | - Barbara A M Snoeker
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, The Netherlands
| | - Kris C P Vissers
- Department of Anesthesiology, Pain and Palliative Medicine, Radboudumc, Nijmegen, Gelderland, The Netherlands
| | - Jörgen Bruhn
- Department of Anesthesiology, Pain and Palliative Medicine, Radboudumc, Nijmegen, Gelderland, The Netherlands
| | - Martin J L Bucx
- Department of Anesthesiology, Pain and Palliative Medicine, Radboudumc, Nijmegen, Gelderland, The Netherlands
| |
Collapse
|
11
|
Lofthouse EM, Manousopoulou A, Cleal JK, O'Kelly IM, Poore KR, Garbis SD, Lewis RM. N-acetylcysteine, xCT and suppression of Maxi-chloride channel activity in human placenta. Placenta 2021; 110:46-55. [PMID: 34120018 DOI: 10.1016/j.placenta.2021.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/04/2021] [Accepted: 05/26/2021] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Placental oxidative stress features in pregnancy pathologies but in clinical trials antioxidant supplementation has not improved outcomes. N-acetylcysteine (NAC) stimulates glutathione production and is proposed as a therapeutic agent in pregnancy. However, key elements of N-acetylcysteine biology, including its cellular uptake mechanism, remains unclear. This study explores how the cystine/glutamate transporter xCT may mediate N-acetylcysteine uptake and how N-acetylcysteine alters placental redox status. METHODS The involvement of xCT in NAC uptake by the human placenta was studied in perfused placenta and Xenopus oocytes. The effect of short-term N-acetylcysteine exposure on the placental villous proteome was determined using LC-MS. The effect of N-acetylcysteine on Maxi-chloride channel activity was investigated in perfused placenta, villous fragments and cell culture. RESULTS Maternoplacental N-acetylcysteine administration stimulated intracellular glutamate efflux suggesting a role of the exchange transporter xCT, which was localised to the microvillous membrane of the placental syncytiotrophoblast. Placental exposure to a bolus of N-acetylcysteine inhibited subsequent activation of the redox sensitive Maxi-chloride channel independently of glutathione synthesis. Stable isotope quantitative proteomics of placental villi treated with N-acetylcysteine demonstrated changes in pathways associated with oxidative stress, apoptosis and the acute phase response. DISCUSSION This study suggests that xCT mediates N-acetylcysteine uptake into the placenta and that N-acetylcysteine treatment of placental tissue alters the placental proteome while regulating the redox sensitive Maxi-chloride channel. Interestingly N-acetylcysteine had antioxidant effects independent of the glutathione pathway. Effective placental antioxidant therapy in pregnancy may require maintaining the balance between normalising redox status without inhibiting physiological redox signalling.
Collapse
Affiliation(s)
- Emma M Lofthouse
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK.
| | - Antigoni Manousopoulou
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK; Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Jane K Cleal
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| | | | | | - Spiros D Garbis
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK; Proteome Exploration Laboratory, Beckman Institute, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rohan M Lewis
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| |
Collapse
|
12
|
Notartomaso S, Scarselli P, Mascio G, Liberatore F, Mazzon E, Mammana S, Gugliandolo A, Cruccu G, Bruno V, Nicoletti F, Battaglia G. N-Acetylcysteine causes analgesia in a mouse model of painful diabetic neuropathy. Mol Pain 2021; 16:1744806920904292. [PMID: 32009537 PMCID: PMC6997966 DOI: 10.1177/1744806920904292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
N-Acetylcysteine, one of the most prescribed antioxidant drugs, enhances pain
threshold in rodents and humans by activating mGlu2 metabotropic glutamate
receptors. Here, we assessed the analgesic activity of N-acetylcysteine in the
streptozotocin model of painful diabetic neuropathy and examined the effect of
N-acetylcysteine on proteins that are involved in mechanisms of nociceptive
sensitization. Mice with blood glucose levels ≥250 mg/dl in response to a single
intraperitoneal (i.p.) injection of streptozotocin (200 mg/kg) were used for the
assessment of mechanical pain thresholds. Systemic treatment with
N-acetylcysteine (100 mg/kg, i.p., either single injection or daily injections
for seven days) caused analgesia in diabetic mice. N-acetylcysteine-induced
analgesia was abrogated by the Sxc− inhibitors, sulfasalazine (8 mg/kg, i.p.), erastin (30 mg/kg,
i.p.), and sorafenib (10 mg/kg, i.p.), or by the mGlu2/3 receptor antagonist,
LY341495 (1 mg/kg, i.p.). Repeated administrations of N-acetylcysteine in
diabetic mice reduced ERK1/2 phosphorylation in the dorsal region of the lumbar
spinal cord. The analgesic activity of N-acetylcysteine was occluded by the MEK
inhibitor, PD0325901 (25 mg/kg, i.p.), the TRPV1 channel blocker, capsazepine
(40 mg/kg, i.p.), or by a cocktail of NMDA and mGlu5 metabotropic glutamate
receptor antagonists (memantine, 25 mg/kg, plus MTEP, 5 mg/kg,
both i.p.). These findings offer the first demonstration that N-acetylcysteine
relieves pain associated with diabetic neuropathy and holds promise for the use
of N-acetylcysteine as an add-on drug in diabetic patients.
Collapse
Affiliation(s)
| | - Pamela Scarselli
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Giada Mascio
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | | | | | - Santa Mammana
- IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | | | - Giorgio Cruccu
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Valeria Bruno
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Ferdinando Nicoletti
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Giuseppe Battaglia
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| |
Collapse
|
13
|
Quintanilla ME, Morales P, Ezquer F, Ezquer M, Herrera-Marschitz M, Israel Y. Administration of N-acetylcysteine Plus Acetylsalicylic Acid Markedly Inhibits Nicotine Reinstatement Following Chronic Oral Nicotine Intake in Female Rats. Front Behav Neurosci 2021; 14:617418. [PMID: 33633548 PMCID: PMC7902020 DOI: 10.3389/fnbeh.2020.617418] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Background Nicotine is the major addictive component of cigarette smoke and the prime culprit of the failure to quit smoking. Common elements perpetuating the use of addictive drugs are (i) cues associated with the setting in which drug was used and (ii) relapse/reinstatement mediated by an increased glutamatergic tone (iii) associated with drug-induced neuroinflammation and oxidative stress. Aims The present study assessed the effect of the coadministration of the antioxidant N-acetylcysteine (NAC) plus the anti-inflammatory acetylsalicylic acid (ASA) on oral nicotine reinstatement intake following a post-deprivation re-access in female rats that had chronically and voluntarily consumed a nicotine solution orally. The nicotine-induced oxidative stress and neuroinflammation in the hippocampus and its effects on the glutamate transporters GLT-1 and XCT mRNA levels in prefrontal cortex were also analyzed. Results The oral coadministration of NAC (40 mg/kg/day) and ASA (15 mg/kg/day) inhibited by 85% of the oral nicotine reinstatement intake compared to control (vehicle), showing an additive effect of both drugs. Acetylsalicylic acid and N-acetylcysteine normalized hippocampal oxidative stress and blunted the hippocampal neuroinflammation observed upon oral nicotine reinstatement. Nicotine downregulated GLT-1 and xCT gene expression in the prefrontal cortex, an effect reversed by N-acetylcysteine, while acetylsalicylic acid reversed the nicotine-induced downregulation of GLT-1 gene expression. The inhibitory effect of N-acetylcysteine on chronic nicotine intake was blocked by the administration of sulfasalazine, an inhibitor of the xCT transporter. Conclusion Nicotine reinstatement, following post-deprivation of chronic oral nicotine intake, downregulates the mRNA levels of GLT-1 and xCT transporters, an effect reversed by the coadministration of N-acetylcysteine and acetylsalicylic acid, leading to a marked inhibition of nicotine intake. The combination of these drugs may constitute a valuable adjunct in the treatment of nicotine-dependent behaviors.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
14
|
Nakhaee S, Dastjerdi M, Roumi H, Mehrpour O, Farrokhfall K. N-acetylcysteine dose-dependently improves the analgesic effect of acetaminophen on the rat hot plate test. BMC Pharmacol Toxicol 2021; 22:4. [PMID: 33413696 PMCID: PMC7791802 DOI: 10.1186/s40360-020-00469-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Acetaminophen (APAP) induced hepatotoxicity is a clinically important problem. Up to now, interventive therapy with n-acetylcysteine (NAC) has been considered as a gold-standard treatment for APAP overdose. However, no study has focused on the efficacy of these drugs' concurrent administration on probable enhancing therapeutic outcomes. Thus, this study was aimed to investigate the analgesic effect of co-administration of NAC and acetaminophen in male rats. The NAC-APAP drug formulation may demonstrate the stranger antinociceptive effect. METHODS Forty-eight male Sprague-Dawley rats (12-14 weeks) randomly divided into six equal groups; control, APAP (received 300 mg/kg APAP), NAC (received 600 mg/kg NAC) and APAP+ NAC groups that received simultaneously 300 mg/kg APAP with 200-600 mg/kg NAC (AN200, AN400, AN600). All administrations were done orally for once. The antinociceptive effect was recorded by measurement of latency period on a hot plate in 30, 60, and 90 min after administrations. RESULTS The results showed that NAC's concurrent administration with APAP, dose-dependently increased APAP analgesic effects (p< 0.0001). Moreover, NAC treatment exhibited an antinociceptive effect in 60 and 90 min, per se. The treatments had no adverse effect on liver enzymes and oxidative stress. CONCLUSION Co-administration of NAC with APAP can improve the antinociceptive effect of APAP. It is suggested that this compound can enhance analgesic effects of APAP and eventually lead to a reduction in acetaminophen dose. Further studies are needed to evaluate the molecular mechanism of this hyper analgesic effect.
Collapse
Affiliation(s)
- Samaneh Nakhaee
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), Birjand, Iran
| | - Mohammad Dastjerdi
- Cardiovascular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Hesam Roumi
- Cardiovascular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Omid Mehrpour
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), Birjand, Iran
- Mel and Enid Zuckerman, College of Public Health, University of Arizona, Tucson, Arizona, USA
| | - Khadijeh Farrokhfall
- Cardiovascular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
15
|
Raghu G, Berk M, Campochiaro PA, Jaeschke H, Marenzi G, Richeldi L, Wen FQ, Nicoletti F, Calverley PMA. The Multifaceted Therapeutic Role of N-Acetylcysteine (NAC) in Disorders Characterized by Oxidative Stress. Curr Neuropharmacol 2021; 19:1202-1224. [PMID: 33380301 PMCID: PMC8719286 DOI: 10.2174/1570159x19666201230144109] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/27/2020] [Accepted: 12/13/2020] [Indexed: 02/08/2023] Open
Abstract
Oxidative stress, which results in the damage of diverse biological molecules, is a ubiquitous cellular process implicated in the etiology of many illnesses. The sulfhydryl-containing tripeptide glutathione (GSH), which is synthesized and maintained at high concentrations in all cells, is one of the mechanisms by which cells protect themselves from oxidative stress. N-acetylcysteine (NAC), a synthetic derivative of the endogenous amino acid L-cysteine and a precursor of GSH, has been used for several decades as a mucolytic and as an antidote to acetaminophen (paracetamol) poisoning. As a mucolytic, NAC breaks the disulfide bonds of heavily cross-linked mucins, thereby reducing mucus viscosity. In vitro, NAC has antifibrotic effects on lung fibroblasts. As an antidote to acetaminophen poisoning, NAC restores the hepatic GSH pool depleted in the drug detoxification process. More recently, improved knowledge of the mechanisms by which NAC acts has expanded its clinical applications. In particular, the discovery that NAC can modulate the homeostasis of glutamate has prompted studies of NAC in neuropsychiatric diseases characterized by impaired glutamate homeostasis. This narrative review provides an overview of the most relevant and recent evidence on the clinical application of NAC, with a focus on respiratory diseases, acetaminophen poisoning, disorders of the central nervous system (chronic neuropathic pain, depression, schizophrenia, bipolar disorder, and addiction), cardiovascular disease, contrast-induced nephropathy, and ophthalmology (retinitis pigmentosa).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peter M. A. Calverley
- Address correspondence to this author at Clinical Science Centre, University Hospital Aintree, Longmoor Lane, Liverpool UK L9 7AL; Tel: +44 151 529 5886, Fax: +44 151 529 5888; E-mail:
| |
Collapse
|
16
|
Cisani F, Roggeri A, Olivero G, Garrone B, Tongiani S, Di Giorgio FP, Pittaluga A. Acute Low Dose of Trazodone Recovers Glutamate Release Efficiency and mGlu2/3 Autoreceptor Impairments in the Spinal Cord of Rats Suffering From Chronic Sciatic Ligation. Front Pharmacol 2020; 11:1108. [PMID: 32765286 PMCID: PMC7379891 DOI: 10.3389/fphar.2020.01108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/08/2020] [Indexed: 11/16/2022] Open
Abstract
We investigated whether chronic sciatic ligation modifies the glutamate release in spinal cord nerve endings (synaptosomes) as well as the expression and the function of presynaptic release-regulating mGlu2/3 autoreceptors and 5-HT2A heteroreceptors in these particles. Synaptosomes were from the spinal cord of animals suffering from the sciatic ligation that developed on day 6 post-surgery a significant decrease of the force inducing paw-withdrawal in the lesioned paw. The exocytosis of glutamate (quantified as release of preloaded [3H]D-aspartate, [3H]D-Asp) elicited by a mild depolarizing stimulus (15 mM KCl) was significantly increased in synaptosomes from injured rats when compared to controls (uninjured rats). The mGlu2/3 agonist LY379268 (1000 pM) significantly inhibited the 15 mM KCl-evoked [3H]D-Asp overflow from control synaptosomes, but not in terminals isolated from injured animals. Differently, a low concentration (10 nM) of (±) DOI, unable to modify the 15 mM KCl-evoked [3H]D-Asp overflow in control spinal cord synaptosomes, significantly reduced the glutamate exocytosis in nerve endings isolated from the injured rats. Acute oral trazodone (TZD, 0.3 mg/kg on day 7 post-surgery) efficiently recovered glutamate exocytosis as well as the efficiency of LY379268 in inhibiting this event in spinal cord synaptosomes from injured animals. The sciatic ligation significantly reduced the expression of mGlu2/3, but not of 5-HT2A, receptor proteins in spinal cord synaptosomal lysates. Acute TZD recovered this parameter. Our results support the use of 5-HT2A antagonists for restoring altered spinal cord glutamate plasticity in rats suffering from sciatic ligation.
Collapse
Affiliation(s)
- Francesca Cisani
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Alessandra Roggeri
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Guendalina Olivero
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Beatrice Garrone
- Angelini RR&D (Research, Regulatory & Development), Angelini Pharma S.p.A., Rome, Italy
| | - Serena Tongiani
- Angelini RR&D (Research, Regulatory & Development), Angelini Pharma S.p.A., Rome, Italy
| | | | - Anna Pittaluga
- Department of Pharmacy, DIFAR, Pharmacology and Toxicology Section and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
17
|
Quintanilla ME, Ezquer F, Morales P, Ezquer M, Olivares B, Santapau D, Herrera-Marschitz M, Israel Y. N-Acetylcysteine and Acetylsalicylic Acid Inhibit Alcohol Consumption by Different Mechanisms: Combined Protection. Front Behav Neurosci 2020; 14:122. [PMID: 32848653 PMCID: PMC7412547 DOI: 10.3389/fnbeh.2020.00122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic ethanol intake results in brain oxidative stress and neuroinflammation, which have been postulated to perpetuate alcohol intake and to induce alcohol relapse. The present study assessed the mechanisms involved in the inhibition of: (i) oxidative stress; (ii) neuroinflammation; and (iii) ethanol intake that follow the administration of the antioxidant N-acetylcysteine (NAC) and the anti-inflammatory acetylsalicylic acid (ASA) to animals that had consumed ethanol chronically. At doses used clinically, NAC [40 mg/kg per day orally (p.o.)] and ASA (15 mg/kg per day p.o.) significantly inhibited chronic alcohol intake and relapse intake in alcohol-preferring rats. The coadministration of both drugs reduced ethanol intake by 65% to 70%. N-acetylcysteine administration: (a) induced the Nrf2-ARE system, lowering the hippocampal oxidative stress assessed as the ratio of oxidized glutathione (GSSG)/reduced glutathione (GSH); (b) reduced the neuroinflammation assessed by astrocyte and microglial activation by immunofluorescence; and (c) inhibited chronic and relapse ethanol intake. These effects were blocked by sulfasalazine, an inhibitor of the xCT transporter, which incorporates cystine (precursor of GSH) and extrudes extracellular glutamate, an agonist of the inhibitory mGlu2/3 receptor, which lowers the synaptic glutamatergic tone. The inhibitor of mGlu2/3 receptor (LY341495) blocked the NAC-induced inhibition of both relapse ethanol intake and neuroinflammation without affecting the GSSG/GSH ratio. Unlike N-acetylcysteine, ASA inhibited chronic alcohol intake and relapse via lipoxin A4, a strong anti-inflammatory metabolite of arachidonic acid generated following the ASA acetylation of cyclooxygenases. Accordingly, the lipoxin A4 receptor inhibitor, WRW4, blocked the ASA-induced reduction of ethanol intake. Overall, via different mechanisms, NAC and ASA administered in clinically relevant doses combine their effects inhibiting ethanol intake.
Collapse
Affiliation(s)
- María Elena Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Belen Olivares
- Centro de Química Médica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Daniela Santapau
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
18
|
Heidari N, Sajedi F, Mohammadi Y, Mirjalili M, Mehrpooya M. Ameliorative Effects Of N-Acetylcysteine As Adjunct Therapy On Symptoms Of Painful Diabetic Neuropathy. J Pain Res 2019; 12:3147-3159. [PMID: 31819599 PMCID: PMC6875491 DOI: 10.2147/jpr.s228255] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/01/2019] [Indexed: 01/08/2023] Open
Abstract
Purpose Painful diabetic neuropathy (PDN) is a variant of diabetic peripheral neuropathy which is highly prevalent and distressing in diabetic patients. Despite its high burden, the optimal treatment of PDN has remained a clinical challenge. To explain the emergence and maintenance of PDN, increasing attention has been focused on dimensions of inflammation and oxidative toxic stress (OTS). Accordingly, the aim of this study was to investigate the effects of oral N-acetylcysteine (NAC), an agent with known anti-oxidant and anti-inflammatory effects, as an adjunct therapy in patients suffering from PDN. Patients and methods 113 eligible patients with type 2 diabetes suffering from PDN were randomly assigned to either the pregabalin + placebo or pregabalin + NAC group for 8 weeks (pregabalin at a dose of 150 mg per day, NAC and matched placebo at doses of 600 mg twice a day). Mean pain score was evaluated at baseline, week 1, 2, 4, 6, and 8 of the study based on the mean 24 hr average pain score, using an 11-point numeric rating scale (NRS). As secondary efficacy measures, mean sleep interference score (SIS) resulting from PDN, responder rates, Patient Global Impression of Change (PGIC), Clinical Global Impression of Change (CGIC), and safety were also assessed. Additionally, serum levels of total antioxidant capacity (TAC), total thiol groups (TTG), catalase activity (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD), nitric oxide (NO), and malondialdehyde (MDA) were assessed at baseline and at the end of the study. Results Ninety patients completed the eight-week course of the study. The decrease in mean pain scores and mean sleep interference score in pregabalin + NAC group was greater in comparison with pregabalin + placebo group (p value<0.001 in both conditions). Moreover, more responders (defined as ≥50% reduction in mean pain score from baseline to end-point) were observed in the pregabalin + NAC group, in comparison with pregabalin + placebo group (72.1% vs 46.8%). More improvement in PGIC and CGIC from baseline to the end of the study was reported in pregabalin + NAC group. Oral NAC had minimal adverse effects and was well tolerated in almost all patients. Furthermore, in respect to OTS biomarkers, adjuvant NAC significantly decreased serum level of MDA and significantly increased serum levels of SOD, GPx, TAC, and TTG. Conclusion The pattern of results suggests that compared to placebo and over a time period of 8 weeks, adjuvant NAC is more efficacious in improving neuropathic pain associated with diabetic neuropathy than placebo. Ameliorative effects of NAC on OTS biomarkers demonstrated that NAC may alleviate painful symptoms of diabetic neuropathy, at least in part by its antioxidant effects.
Collapse
Affiliation(s)
- Narges Heidari
- Department of Clinical Pharmacy, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Firozeh Sajedi
- Department of Internal Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Younes Mohammadi
- Modeling of Noncommunicable Diseases Research Center, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahtabalsadat Mirjalili
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Mehrpooya
- Department of Clinical Pharmacy, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
19
|
Mazzitelli M, Neugebauer V. Amygdala group II mGluRs mediate the inhibitory effects of systemic group II mGluR activation on behavior and spinal neurons in a rat model of arthritis pain. Neuropharmacology 2019; 158:107706. [PMID: 31306647 DOI: 10.1016/j.neuropharm.2019.107706] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022]
Abstract
The amygdala plays a critical role in emotional-affective aspects of behaviors and pain modulation. The central nucleus of amygdala (CeA) serves major output functions, and neuroplasticity in the CeA is linked to pain-related behaviors in different models. Activation of Gi/o-coupled group II metabotropic glutamate receptors (mGluRs), which consist of mGluR2 and mGluR3, can decrease neurotransmitter release and regulate synaptic plasticity. Group II mGluRs have emerged as targets for neuropsychiatric disorders and can inhibit pain-related processing and behaviors. Surprisingly, site and mechanism of antinociceptive actions of systemically applied group II mGluR agonists are not clear. Our previous work showed that group II mGluR activation in the amygdala inhibits pain-related CeA activity, but behavioral and spinal consequences remain to be determined. Here we studied the contribution of group II mGluRs in the amygdala to the antinociceptive effects of a systemically applied group II mGluR agonist (LY379268) on behavior and spinal dorsal horn neuronal activity, using the kaolin/carrageenan-induced knee joint arthritis pain model. Audible and ultrasonic vocalizations (emotional responses) and mechanical reflex thresholds were measured in adult rats with and without arthritis (5-6 h postinduction). Extracellular single-unit recordings were made from spinal dorsal horn wide dynamic range neurons of anesthetized (isoflurane) rats with and without arthritis (5-6 h postinduction). Systemic (intraperitoneal) application of a group II mGluR agonist (LY379268) decreased behaviors and activity of spinal neurons in the arthritis pain model but not under normal conditions. Stereotaxic administration of LY379268 into the CeA mimicked the effects of systemic application. Conversely, stereotaxic administration of a group II mGluR antagonist (LY341495) into the CeA reversed the effects of systemic application of LY379268 on behaviors and dorsal horn neuronal activity in arthritic rats. The data show for the first time that the amygdala is the critical site of action for the antinociceptive behavioral and spinal neuronal effects of systemically applied group II mGluR agonists.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA.
| |
Collapse
|
20
|
Tardiolo G, Bramanti P, Mazzon E. Overview on the Effects of N-Acetylcysteine in Neurodegenerative Diseases. Molecules 2018; 23:molecules23123305. [PMID: 30551603 PMCID: PMC6320789 DOI: 10.3390/molecules23123305] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023] Open
Abstract
N-acetylcysteine (NAC), which is an acetylated cysteine compound, has aroused scientific interest for decades due to its important medical applications. It also represents a nutritional supplement in the human diet. NAC is a glutathione precursor and shows antioxidant and anti-inflammatory activities. In addition to the uses quoted in the literature, NAC may be considered helpful in therapies to counteract neurodegenerative and mental health diseases. Furthermore, this compound has been evaluated for its neuroprotective potential in the prevention of cognitive aging dementia. NAC is inexpensive, commercially available and no relevant side effects were observed after its administration. The purpose of this paper is to give an overview on the effects and applications of NAC in Parkinson's and Alzheimer's disorders and in neuropathic pain and stroke.
Collapse
Affiliation(s)
- Giuseppe Tardiolo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
21
|
Mazzitelli M, Palazzo E, Maione S, Neugebauer V. Group II Metabotropic Glutamate Receptors: Role in Pain Mechanisms and Pain Modulation. Front Mol Neurosci 2018; 11:383. [PMID: 30356691 PMCID: PMC6189308 DOI: 10.3389/fnmol.2018.00383] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter in the nervous system and plays a critical role in nociceptive processing and pain modulation. G-protein coupled metabotropic glutamate receptors (mGluRs) are widely expressed in the central and peripheral nervous system, and they mediate neuronal excitability and synaptic transmission. Eight different mGluR subtypes have been identified so far, and are classified into Groups I-III. Group II mGluR2 and mGluR3 couple negatively to adenylyl cyclase through Gi/Go proteins, are mainly expressed presynaptically, and typically inhibit the release of neurotransmitters, including glutamate and GABA. Group II mGluRs have consistently been linked to pain modulation; they are expressed in peripheral, spinal and supraspinal elements of pain-related neural processing. Pharmacological studies have shown anti-nociceptive/analgesic effects of group II mGluR agonists in preclinical models of acute and chronic pain, although much less is known about mechanisms and sites of action for mGluR2 and mGluR3 compared to other mGluRs. The availability of orthosteric and new selective allosteric modulators acting on mGluR2 and mGluR3 has provided valuable tools for elucidating (subtype) specific contributions of these receptors to the pathophysiological mechanisms of pain and other disorders and their potential as therapeutic targets. This review focuses on the important role of group II mGluRs in the neurobiology of pain mechanisms and behavioral modulation, and discusses evidence for their therapeutic potential in pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Enza Palazzo
- Section of Pharmacology L. Donatelli, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Section of Pharmacology L. Donatelli, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
22
|
Sałat K, Gryzło B, Kulig K. Experimental Drugs for Neuropathic Pain. Curr Neuropharmacol 2018; 16:1193-1209. [PMID: 29745335 PMCID: PMC6187752 DOI: 10.2174/1570159x16666180510151241] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 02/02/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Neuropathic pain (NP) is an important public health problem and despite recent progress in the understanding, diagnosis, pathophysiological mechanisms and the treatment of NP, many patients remain refractory to pharmacotherapy. OBJECTIVE Currently used drugs have limited efficacy and dose-limiting adverse effects, and thus there is a substantial need for further development of novel medications for its treatment. Alternatively, drugs approved for use in diseases other than NP can be applied as experimental for NP conditions. This paper covers advances in the field of NP treatment. RESULTS The prime focus of this paper is on drugs with well-established pharmacological activity whose current therapeutic applications are distinct from NP. These drugs could be a potential novel treatment of NP. Data from preclinical studies and clinical trials on these experimental drugs are presented. The development of advanced methods of genomics enabled to propose new targets for drugs which could be effective in the NP treatment. CONCLUSION Experimental drugs for NP can be a treatment option which should be tailor-made for each individual on the basis of pain features, previous therapies, associated clinical conditions, recurrence of pain, adverse effects, contraindications and patients' preferences. At present, there are only some agents which may have potential as novel treatments. Increasing knowledge about mechanisms underlying NP, mechanisms of drug action, as well as available data from preclinical and clinical studies make botulinum toxin A, minocycline, ambroxol, statins and PPAR agonists (ATx086001) promising potential future treatment options.
Collapse
Affiliation(s)
- Kinga Sałat
- Address correspondence to this author at the Faculty of Pharmacy,
Jagiellonian University, 9 Medyczna St., 30-688 Kraków, Poland; Tel: + 48 12 6205 555; Fax: + 48 12 6205 554; E-mail:
| | | | | |
Collapse
|
23
|
Nasca C, Bigio B, Zelli D, de Angelis P, Lau T, Okamoto M, Soya H, Ni J, Brichta L, Greengard P, Neve RL, Lee FS, McEwen BS. Role of the Astroglial Glutamate Exchanger xCT in Ventral Hippocampus in Resilience to Stress. Neuron 2017; 96:402-413.e5. [PMID: 29024663 DOI: 10.1016/j.neuron.2017.09.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 08/10/2017] [Accepted: 09/13/2017] [Indexed: 12/23/2022]
Abstract
We demonstrate that stress differentially regulates glutamate homeostasis in the dorsal and ventral hippocampus and identify a role for the astroglial xCT in ventral dentate gyrus (vDG) in stress and antidepressant responses. We provide an RNA-seq roadmap for the stress-sensitive vDG. The transcription factor REST binds to xCT promoter in co-occupancy with the epigenetic marker H3K27ac to regulate expression of xCT, which is also reduced in a genetic mouse model of inherent susceptibility to depressive-like behavior. Pharmacologically, modulating histone acetylation with acetyl-L-carnitine (LAC) or acetyl-N-cysteine (NAC) rapidly increases xCT and activates a network with mGlu2 receptors to prime an enhanced glutamate homeostasis that promotes both pro-resilient and antidepressant-like responses. Pharmacological xCT blockage counteracts NAC prophylactic effects. GFAP+-Cre-dependent overexpression of xCT in vDG mimics pharmacological actions in promoting resilience. This work establishes a mechanism by which vDG protection leads to stress resilience and antidepressant responses via epigenetic programming of an xCT-mGlu2 network.
Collapse
Affiliation(s)
- Carla Nasca
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| | - Benedetta Bigio
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Danielle Zelli
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Paolo de Angelis
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Timothy Lau
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Masahiro Okamoto
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA; Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki 305-8574, Japan
| | - Hideyo Soya
- Department of Sports Neuroscience, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki 305-8574, Japan; Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki 305-8574, Japan
| | - Jason Ni
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Lars Brichta
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Francis S Lee
- Sackler Institute for Developmental Psychobiology, Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
24
|
Horst A, de Souza JA, Santos MCQ, Riffel APK, Kolberg C, Partata WA. Effects of N-acetylcysteine on spinal cord oxidative stress biomarkers in rats with neuropathic pain. ACTA ACUST UNITED AC 2017; 50:e6533. [PMID: 29069230 PMCID: PMC5649872 DOI: 10.1590/1414-431x20176533] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/28/2017] [Indexed: 01/06/2023]
Abstract
N-acetylcysteine (NAC) inhibits nociceptive transmission. This effect has been associated partly with its antioxidant properties. However, the effect of NAC on the levels of lipid hydroperoxides (a pro-oxidant marker), content of ascorbic acid (a key antioxidant molecule of nervous tissue) and total antioxidant capacity (TAC) is unknown. Thus, our study assessed these parameters in the lumbosacral spinal cord of rats with chronic constriction injury (CCI) of the sciatic nerve, one of the most commonly employed animal models of neuropathic pain. Thirty-six male Wistar rats weighing 200–300 g were equally divided into the following groups: Naive (rats did not undergo surgical manipulation); Sham (rats in which all surgical procedures involved in CCI were used except the ligature), and CCI (rats in which four ligatures were tied loosely around the right common sciatic nerve). All rats received intraperitoneal injections of NAC (150 mg·kg−1·day−1) or saline for 1, 3, or 7 days. Rats were killed 1, 3, and 7 days after surgery. NAC treatment prevented the CCI-induced increase in lipid hydroperoxide levels only at day 1, although the amount was higher than that found in naive rats. NAC treatment also prevented the CCI-induced increase in ascorbic acid content, which occurred at days 1, 3, and 7. No significant change was found in TAC with NAC treatment. The changes observed here may be related to the antinociceptive effect of NAC because modulation of oxidative-stress parameters seemed to help normalize the spinal cord oxidative status altered by pain.
Collapse
Affiliation(s)
- A Horst
- Laboratório de Neurobiologia Comparada, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil.,Univates, Lajeado, RS, Brasil
| | - J A de Souza
- Laboratório de Neurobiologia Comparada, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - M C Q Santos
- Laboratório de Neurobiologia Comparada, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - A P K Riffel
- Laboratório de Neurobiologia Comparada, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - C Kolberg
- Laboratório de Neurobiologia Comparada, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - W A Partata
- Laboratório de Neurobiologia Comparada, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| |
Collapse
|
25
|
Notartomaso S, Mascio G, Bernabucci M, Zappulla C, Scarselli P, Cannella M, Imbriglio T, Gradini R, Battaglia G, Bruno V, Nicoletti F. Analgesia induced by the epigenetic drug, L-acetylcarnitine, outlasts the end of treatment in mouse models of chronic inflammatory and neuropathic pain. Mol Pain 2017; 13:1744806917697009. [PMID: 28326943 PMCID: PMC5407675 DOI: 10.1177/1744806917697009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background L-acetylcarnitine, a drug marketed for the treatment of chronic pain, causes analgesia by epigenetically up-regulating type-2 metabotropic glutamate (mGlu2) receptors in the spinal cord. Because the epigenetic mechanisms are typically long-lasting, we hypothesized that analgesia could outlast the duration of L-acetylcarnitine treatment in models of inflammatory and neuropathic pain. Results A seven-day treatment with L-acetylcarnitine (100 mg/kg, once a day, i.p.) produced an antiallodynic effect in the complete Freund adjuvant mouse model of chronic inflammatory pain. L-Acetylcarnitine-induced analgesia persisted for at least 14 days after drug withdrawal. In contrast, the analgesic effect of pregabalin, amitryptiline, ceftriaxone, and N-acetylcysteine disappeared seven days after drug withdrawal. L-acetylcarnitine treatment enhanced mGlu2/3 receptor protein levels in the dorsal region of the spinal cord. This effect also persisted for two weeks after drug withdrawal and was associated with increased levels of acetylated histone H3 bound to the Grm2 gene promoter in the dorsal root ganglia. A long-lasting analgesic effect of L-acetylcarnitine was also observed in mice subjected to chronic constriction injury of the sciatic nerve. In these animals, a 14-day treatment with pregabalin, amitryptiline, tramadol, or L-acetylcarnitine produced a significant antiallodynic effect, with pregabalin displaying the greatest efficacy. In mice treated with pregabalin, tramadol or L-acetylcarnitine the analgesic effect was still visible 15 days after the end of drug treatment. However, only in mice treated with L-acetylcarnitine analgesia persisted 37 days after drug withdrawal. This effect was associated with an increase in mGlu2/3 receptor protein levels in the dorsal horns of the spinal cord. Conclusions Our findings suggest that L-acetylcarnitine has the unique property to cause a long-lasting analgesic effect that might reduce relapses in patients suffering from chronic pain.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Roberto Gradini
- 1 I.R.C.C.S. Neuromed, Pozzilli, Italy.,2 Department of Experimental Medicine, Sapienza University, Rome, Italy
| | | | - Valeria Bruno
- 1 I.R.C.C.S. Neuromed, Pozzilli, Italy.,3 Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Ferdinando Nicoletti
- 1 I.R.C.C.S. Neuromed, Pozzilli, Italy.,3 Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| |
Collapse
|
26
|
Gleixner AM, Hutchison DF, Sannino S, Bhatia TN, Leak LC, Flaherty PT, Wipf P, Brodsky JL, Leak RK. N-Acetyl-l-Cysteine Protects Astrocytes against Proteotoxicity without Recourse to Glutathione. Mol Pharmacol 2017; 92:564-575. [PMID: 28830914 DOI: 10.1124/mol.117.109926] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-l-cysteine (NAC) exhibits protective properties in brain injury models and has undergone a number of clinical trials. Most studies of NAC have focused on neurons. However, neuroprotection may be complemented by the protection of astrocytes because healthier astrocytes can better support the viability of neurons. Here, we show that NAC can protect astrocytes against protein misfolding stress (proteotoxicity), the hallmark of neurodegenerative disorders. Although NAC is thought to be a glutathione precursor, NAC protected primary astrocytes from the toxicity of the proteasome inhibitor MG132 without eliciting any increase in glutathione. Furthermore, glutathione depletion failed to attenuate the protective effects of NAC. MG132 elicited a robust increase in the folding chaperone heat shock protein 70 (Hsp70), and NAC mitigated this effect. Nevertheless, three independent inhibitors of Hsp70 function ablated the protective effects of NAC, suggesting that NAC may help preserve Hsp70 chaperone activity and improve protein quality control without need for Hsp70 induction. Consistent with this view, NAC abolished an increase in ubiquitinated proteins in MG132-treated astrocytes. However, NAC did not affect the loss of proteasome activity in response to MG132, demonstrating that it boosted protein homeostasis and cell viability without directly interfering with the efficacy of this proteasome inhibitor. The thiol-containing molecules l-cysteine and d-cysteine both mimicked the protective effects of NAC, whereas the thiol-lacking molecule N-acetyl-S-methyl-l-cysteine failed to exert protection or blunt the rise in ubiquitinated proteins. Collectively, these findings suggest that the thiol group in NAC is required for its effects on glial viability and protein quality control.
Collapse
Affiliation(s)
- Amanda M Gleixner
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Daniel F Hutchison
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Sara Sannino
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Tarun N Bhatia
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Lillian C Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Patrick T Flaherty
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Peter Wipf
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Jeffrey L Brodsky
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania (A.M.G., D.F.H., T.N.B., L.C.L., P.T.F., R.K.L.); and Departments of Biological Sciences (S.S., J.L.B.) and Chemistry and Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania (P.W.)
| |
Collapse
|
27
|
Liu Y, Ni Y, Zhang W, Sun YE, Ma Z, Gu X. N-acetyl-cysteine attenuates remifentanil-induced postoperative hyperalgesia via inhibiting matrix metalloproteinase-9 in dorsal root ganglia. Oncotarget 2017; 8:16988-17001. [PMID: 28199982 PMCID: PMC5370016 DOI: 10.18632/oncotarget.15217] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/27/2017] [Indexed: 11/25/2022] Open
Abstract
Treatment of remifentanil-induced postoperative hyperalgesia (RIH) remains a clinical challenge because the mechanisms are not fully understood. Matrix metalloproteinase-9 (MMP-9) is a key component in neuroinflammation because of its facilitation of pro-inflammatory cytokine maturation. Therefore, inhibition of MMP-9 may represent a novel therapeutic approach to the treatment of RIH. Sprague-Dawley rats were randomly divided into three groups: Control, Incision and Remifentanil. A right plantar surgical incision was performed in Group Incision, and intraoperative remifentanil (0.04 mg/kg, 0.4 ml) was infused subcutaneously for 30 min in Group Remifentanil. The results indicated that intraoperative remifentanil induced an up-regulation and activation of MMP-9 in DRGs but not spinal cords. MMP-9 was expressed primarily in DRG neurons co-expressing mu opioid receptors (MOR), and elicited interleukin-1β (IL-1β) cleavage in DRG neurons and satellite glial cells (SGCs). Intraperitoneal injection of N-acetyl-cysteine (NAC), a broadly used safe drug, significantly attenuated RIH via suppressing the activation of MMP-9 in DRGs. NAC inhibited the cleavage of IL-1β in DRGs, which is a critical substrate of MMP-9, and markedly suppressed glial activation and neuron excitability in spinal dorsal horn induced by remifentanil. These results demonstrated that NAC can effectively alleviate RIH via powerfully inhibiting MMP-9 activation in DRGs.
Collapse
Affiliation(s)
- Yue Liu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yuan Ni
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Wei Zhang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yu-E Sun
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
28
|
Neuronal P2X7 receptor-induced reactive oxygen species production contributes to nociceptive behavior in mice. Sci Rep 2017; 7:3539. [PMID: 28615626 PMCID: PMC5471238 DOI: 10.1038/s41598-017-03813-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/03/2017] [Indexed: 11/27/2022] Open
Abstract
ATP can activate a variety of pathways through P2 purinoreceptors, leading to neuroprotection and pathology in the CNS. Among all P2X receptors, the P2X7 receptor (P2X7R) is a well-defined therapeutic target for inflammatory and neuropathic pain. Activation of P2X7R can generate reactive oxygen species (ROS) in macrophages and microglia. However, the role of ROS in P2X7R–induced pain remains unexplored. Here, we investigated the downstream effects of neuronal P2X7R activation in the spinal cord. We found that ATP induces ROS production in spinal cord dorsal horn neurons, an effect eliminated by ROS scavenger N-tert-butyl-α-phenylnitrone (PBN) and P2X7R antagonist A438079. A similar effect was observed with a P2X7R agonist, BzATP, and was attenuated by a NADPH oxidase inhibitor apocynin. Intrathecal administration of BzATP resulted in ROS production in the spinal cord and oxidative DNA damage in dorsal horn neurons. BzATP also induced robust biphasic spontaneous nociceptive behavior. Pre-treatment with A438079 abolished all BzATP-induced nociceptive behaviors, while ROS scavengers dose-dependently attenuated the secondary response. Here, we provide evidence that neuronal P2X7R activation leads to ROS production and subsequent nociceptive pain in mice. Together, the data indicate that P2X7R-induced ROS play a critical role in the P2X7R signaling pathway of the CNS.
Collapse
|
29
|
Abstract
The treatment of neuropathic pain remains a clinical challenge because of its unclear mechanisms and broad clinical morbidity. Matrix metalloproteinase (MMP)-9 and MMP-2 have previously been described as key components in neuropathic pain because of their facilitation of inflammatory cytokine maturation and induction of neural inflammation. Therefore, the inhibition of MMPs may represent a novel therapeutic approach to the treatment of neuropathic pain. In this study, we report that N-acetyl-cysteine (NAC), which is a broadly used respiratory drug, significantly attenuates neuropathic pain through a unique mechanism of MMP inhibition. Both the in vitro (0.1 mM) and in vivo application of NAC significantly suppressed the activity of MMP-9/2. Orally administered NAC (50, 100, and 200 mg/kg) not only postponed the occurrence but also inhibited the maintenance of chronic constrictive injury (CCI)-induced neuropathic pain in rats. The administration of NAC blocked the maturation of interleukin-1β, which is a critical substrate of MMPs, and markedly suppressed the neuronal activation induced by CCI, including inhibiting the phosphorylation of protein kinase Cγ, NMDAR1, and mitogen-activated protein kinases. Finally, NAC significantly inhibited CCI-induced microglia activation but elicited no notable effects on astrocytes. These results demonstrate an effective and safe approach that has been used clinically to alleviate neuropathic pain through the powerful inhibition of the activation of MMPs.
Collapse
|
30
|
Johnson MP, Muhlhauser MA, Nisenbaum ES, Simmons RMA, Forster BM, Knopp KL, Yang L, Morrow D, Li DL, Kennedy JD, Swanson S, Monn JA. Broad spectrum efficacy with LY2969822, an oral prodrug of metabotropic glutamate 2/3 receptor agonist LY2934747, in rodent pain models. Br J Pharmacol 2017; 174:822-835. [PMID: 28177520 DOI: 10.1111/bph.13740] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 01/27/2017] [Accepted: 01/31/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE A body of evidence suggests activation of metabotropic glutamate 2/3 (mGlu2/3 ) receptors would be an effective analgesic in chronic pain conditions. Thus, the analgesic properties of a novel mGlu2/3 receptor agonist prodrug were investigated. EXPERIMENTAL APPROACH After oral absorption, the prodrug LY2969822 rapidly converts to the brain penetrant, potent and subtype-selective mGlu2/3 receptor agonist LY2934747. Behavioural assessments of allodynia, hyperalgesia and nocifensive behaviours were determined in preclinical pain models after administration of LY2969822 0.3-10 mg·kg-1 . In addition, the ability of i.v. LY2934747 to modulate dorsal horn spinal cord wide dynamic range (WDR) neurons in spinal nerve ligated (SNL) rats was assessed. KEY RESULTS Following treatment with LY2934747, the spontaneous activity and electrically-evoked wind-up of WDR neurons in rats that had undergone spinal nerve ligation and developed mechanical allodynia were suppressed. In a model of sensitization, orally administered LY2969822 prevented the nociceptive behaviours induced by an intraplantar injection of formalin. The on-target nature of this effect was confirmed by blockade with an mGlu2/3 receptor antagonist. LY2969822 prevented capsaicin-induced tactile hypersensitivity, reversed the SNL-induced tactile hypersensitivity and reversed complete Freund's adjuvant - induced mechanical hyperalgesia. The mGlu2/3 receptor agonist prodrug demonstrated efficacy in visceral pain models, including a colorectal distension model and partially prevented the nocifensive behaviours in the mouse acetic acid writhing model. CONCLUSIONS AND IMPLICATIONS Following oral administration of the prodrug LY2969822, the mGlu2/3 receptor agonist LY2934747 was formed and this attenuated pain behaviours across a broad range of preclinical pain models.
Collapse
Affiliation(s)
- Michael P Johnson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mark A Muhlhauser
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Eric S Nisenbaum
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Rosa M A Simmons
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Beth M Forster
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Kelly L Knopp
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Lijuan Yang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Denise Morrow
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Dominic L Li
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jeffrey D Kennedy
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Steven Swanson
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - James A Monn
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
31
|
Palazzo E, Marabese I, Luongo L, Guida F, de Novellis V, Maione S. Nociception modulation by supraspinal group III metabotropic glutamate receptors. J Neurochem 2017; 141:507-519. [DOI: 10.1111/jnc.13725] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 02/02/2023]
Affiliation(s)
- Enza Palazzo
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Ida Marabese
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Livio Luongo
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Francesca Guida
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Vito de Novellis
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| | - Sabatino Maione
- Department of Experimental Medicine; The Second University of Naples; Naples Italy
| |
Collapse
|
32
|
Davidson S, Golden JP, Copits BA, Ray PR, Vogt SK, Valtcheva MV, Schmidt RE, Ghetti A, Price T, Gereau RW. Group II mGluRs suppress hyperexcitability in mouse and human nociceptors. Pain 2016; 157:2081-2088. [PMID: 27218869 PMCID: PMC4988887 DOI: 10.1097/j.pain.0000000000000621] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We introduce a strategy for preclinical research wherein promising targets for analgesia are tested in rodent and subsequently validated in human sensory neurons. We evaluate group II metabotropic glutamate receptors, the activation of which is efficacious in rodent models of pain. Immunohistochemical analysis showed positive immunoreactivity for mGlu2 in rodent dorsal root ganglia (DRG), peripheral fibers in skin, and central labeling in the spinal dorsal horn. We also found mGlu2-positive immunoreactivity in human neonatal and adult DRG. RNA-seq analysis of mouse and human DRG revealed a comparative expression profile between species for group II mGluRs and for opioid receptors. In rodent sensory neurons under basal conditions, activation of group II mGluRs with a selective group II agonist produced no changes to membrane excitability. However, membrane hyperexcitability in sensory neurons exposed to the inflammatory mediator prostaglandin E2 (PGE2) was prevented by (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate (APDC). In human sensory neurons from donors without a history of chronic pain, we show that PGE2 produced hyperexcitability that was similarly blocked by group II mGluR activation. These results reveal a mechanism for peripheral analgesia likely shared by mice and humans and demonstrate a translational research strategy to improve preclinical validation of novel analgesics using cultured human sensory neurons.
Collapse
Affiliation(s)
- Steve Davidson
- Washington University in St. Louis, School of Medicine, Pain Center and Department of Anesthesiology. St. Louis, MO. 63110
| | - Judith P. Golden
- Washington University in St. Louis, School of Medicine, Pain Center and Department of Anesthesiology. St. Louis, MO. 63110
| | - Bryan A. Copits
- Washington University in St. Louis, School of Medicine, Pain Center and Department of Anesthesiology. St. Louis, MO. 63110
| | - Pradipta R. Ray
- School of Brain and Behavioral Sciences, University of Texas at Dallas. 75080
| | - Sherri K. Vogt
- Washington University in St. Louis, School of Medicine, Pain Center and Department of Anesthesiology. St. Louis, MO. 63110
| | - Manouela V. Valtcheva
- Washington University in St. Louis, School of Medicine, Pain Center and Department of Anesthesiology. St. Louis, MO. 63110
| | - Robert E. Schmidt
- Washington University in St. Louis, School of Medicine Department of Neuropathology, St. Louis, MO. 63110
| | | | - Theodore Price
- School of Brain and Behavioral Sciences, University of Texas at Dallas. 75080
| | - Robert W. Gereau
- Washington University in St. Louis, School of Medicine, Pain Center and Department of Anesthesiology. St. Louis, MO. 63110
| |
Collapse
|
33
|
Chiechio S. Modulation of Chronic Pain by Metabotropic Glutamate Receptors. PHARMACOLOGICAL MECHANISMS AND THE MODULATION OF PAIN 2016; 75:63-89. [DOI: 10.1016/bs.apha.2015.11.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
34
|
Wang Q, Mazur A, Guerrero F, Lambrechts K, Buzzacott P, Belhomme M, Theron M. Antioxidants, endothelial dysfunction, and DCS: in vitro and in vivo study. J Appl Physiol (1985) 2015; 119:1355-62. [PMID: 26472863 DOI: 10.1152/japplphysiol.00167.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 10/08/2015] [Indexed: 11/22/2022] Open
Abstract
Reactive oxygen species (ROS) production is a well-known effect in individuals after an undersea dive. This study aimed to delineate the links between ROS, endothelial dysfunction, and decompression sickness (DCS) through the use of antioxidants in vitro and in vivo. The effect of N-acetylcysteine (NAC) on superoxide and peroxynitrite, nitric oxide (NO) generation, and cell viability during in vitro diving simulation were analyzed. Also analyzed was the effect of vitamin C and NAC on plasma glutathione thiol and thiobarbituric acid reactive substances (TBARS), plasma angiotensin-converting enzyme (ACE) activity, and angiotensin-II and DCS morbidity during in vivo diving simulation. During an in vitro diving simulation, vascular endothelial cells showed overproduction of superoxide and peroxynitrite, obvious attenuation of NO generation, and promotion of cell death, all of which were reversed by NAC treatment. After in vivo diving simulation, plasma ACE activity and angiotensin-II level were not affected. The plasma level of glutathione thiol was downregulated after the dive, which was attenuated partially by NAC treatment. Plasma TBARS level was upregulated; however, either NAC or vitamin C treatment failed to prevent DCS morbidity. During in vitro simulation, endothelial superoxide and peroxynitrite-mediated oxidative stress were involved in the attenuation of NO availability and cell death. This study is the first attempt to link oxidative stress and DCS occurrence, and the link could not be confirmed in vivo. Even in the presence of antioxidants, ROS and bubbles generated during diving and/or decompression might lead to embolic or biochemical stress and DCS. Diving-induced oxidative stress might not be the only trigger of DCS morbidity.
Collapse
Affiliation(s)
- Qiong Wang
- Laboratory ORPHY, Department of UFR Sciences and Technologies, European University of Bretagne, University of Brest, Brest, France
| | - Aleksandra Mazur
- Laboratory ORPHY, Department of UFR Sciences and Technologies, European University of Bretagne, University of Brest, Brest, France
| | - François Guerrero
- Laboratory ORPHY, Department of UFR Sciences and Technologies, European University of Bretagne, University of Brest, Brest, France
| | - Kate Lambrechts
- Laboratory ORPHY, Department of UFR Sciences and Technologies, European University of Bretagne, University of Brest, Brest, France
| | - Peter Buzzacott
- Laboratory ORPHY, Department of UFR Sciences and Technologies, European University of Bretagne, University of Brest, Brest, France
| | - Marc Belhomme
- Laboratory ORPHY, Department of UFR Sciences and Technologies, European University of Bretagne, University of Brest, Brest, France
| | - Michaël Theron
- Laboratory ORPHY, Department of UFR Sciences and Technologies, European University of Bretagne, University of Brest, Brest, France
| |
Collapse
|
35
|
Truini A, Piroso S, Pasquale E, Notartomaso S, Di Stefano G, Lattanzi R, Battaglia G, Nicoletti F, Cruccu G. N-acetyl-cysteine, a drug that enhances the endogenous activation of group-II metabotropic glutamate receptors, inhibits nociceptive transmission in humans. Mol Pain 2015; 11:14. [PMID: 25889381 PMCID: PMC4379592 DOI: 10.1186/s12990-015-0009-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/20/2015] [Indexed: 01/21/2023] Open
Abstract
Background Emerging research seeking novel analgesic drugs focuses on agents targeting group-II metabotropic glutamate receptors (mGlu2 and mGlu3 receptors). N-Acetylcysteine (NAC) enhances the endogenous activation of mGlu2/3 receptors by activating the glial glutamate:cystine membrane exchanger. Here, we examined whether NAC inhibits nociceptive responses in humans and animals. We tested the effect of oral NAC (1.2 g) on thermal-pain thresholds and laser-evoked potentials in 10 healthy volunteers, according to a crossover, double-blind, placebo-controlled design, and the effect of NAC (100 mg/kg, i.p.) on the tail-flick response evoked by radiant heat stimulation in mice. Results In healthy subjects, NAC treatment left thermal-pain thresholds unchanged, but significantly reduced pain ratings to laser stimuli and amplitudes of laser-evoked potentials. NAC induced significantly greater changes in these measures than placebo. In the tail-flick test, NAC strongly reduced the nocifensive reflex response to radiant heat. The action of NAC was abolished by the preferential mGlu2/3 receptor antagonist, LY341495 (1 mg/kg, i.p.). Conclusions Our findings show for the first time that NAC inhibits nociceptive transmission in humans, and does the same in mice by activating mGlu2/3 receptors. These data lay the groundwork for investigating the therapeutic potential of NAC in patients with chronic pain.
Collapse
Affiliation(s)
- Andrea Truini
- Department of Neurology and Psychiatry, University Sapienza, Rome, Italy.
| | - Serena Piroso
- Department of Neurology and Psychiatry, University Sapienza, Rome, Italy.
| | - Erica Pasquale
- Department of Neurology and Psychiatry, University Sapienza, Rome, Italy.
| | - Serena Notartomaso
- Department of Neurology and Psychiatry, University Sapienza, I.R.C.C.S. Neuromed, Pozzilli, Italy.
| | - Giulia Di Stefano
- Department of Neurology and Psychiatry, University Sapienza, Rome, Italy.
| | - Roberta Lattanzi
- Department of Physiology and Pharmacology, University Sapienza, Piazzale Aldo Moro, Rome, 500185, Italy.
| | - Giuseppe Battaglia
- Department of Molecular Pathology, I.R.C.C.S. Neuromed, Pozzilli, Italy.
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza, Piazzale Aldo Moro, Rome, 500185, Italy. .,I.N.M. Neuromed, Pozzilli, Italy. .,Department of Molecular Pathology, I.R.C.C.S. Neuromed, Pozzilli, Italy.
| | - Giorgio Cruccu
- Department of Neurology and Psychiatry, University Sapienza, Rome, Italy.
| |
Collapse
|
36
|
Lutgen V, Resch J, Qualmann K, Raddatz NJ, Panhans C, Olander EM, Kong L, Choi S, Mantsch JR, Baker DA. Behavioral assessment of acute inhibition of system xc (-) in rats. Psychopharmacology (Berl) 2014; 231:4637-47. [PMID: 24828877 PMCID: PMC4474164 DOI: 10.1007/s00213-014-3612-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 05/01/2014] [Indexed: 12/20/2022]
Abstract
RATIONALE Gaps in our understanding of glutamatergic signaling may be key obstacles in accurately modeling complex CNS diseases. System xc (-) is an example of a poorly understood component of glutamate homeostasis that has the potential to contribute to CNS diseases. OBJECTIVES This study aims to determine whether system xc (-) contributes to behaviors used to model features of CNS disease states. METHODS In situ hybridization was used to map mRNA expression of xCT throughout the brain. Microdialysis in the prefrontal cortex was used to sample extracellular glutamate levels; HPLC was used to measure extracellular glutamate and tissue glutathione concentrations. Acute administration of sulfasalazine (8-16 mg/kg, IP) was used to decrease system xc (-) activity. Behavior was measured using attentional set shifting, elevated plus maze, open-field maze, Porsolt swim test, and social interaction paradigm. RESULTS The expression of xCT mRNA was detected throughout the brain, with high expression in several structures including the basolateral amygdala and prefrontal cortex. Doses of sulfasalazine that produced a reduction in extracellular glutamate levels were identified and subsequently used in the behavioral experiments. Sulfasalazine impaired performance in attentional set shifting and reduced the amount of time spent in an open arm of an elevated plus maze and the center of an open-field maze without altering behavior in a Porsolt swim test, total distance moved in an open-field maze, or social interaction. CONCLUSIONS The widespread distribution of system xc (-) and involvement in a growing list of behaviors suggests that this form of nonvesicular glutamate release is a key component of excitatory signaling.
Collapse
Affiliation(s)
- Victoria Lutgen
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - Jon Resch
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - Krista Qualmann
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - Nicholas J. Raddatz
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - Cristina Panhans
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - Ellen M. Olander
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - Linghai Kong
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - John R. Mantsch
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| | - David A. Baker
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15 St, Milwaukee, WI 53233
| |
Collapse
|
37
|
Montagna C, Di Giacomo G, Rizza S, Cardaci S, Ferraro E, Grumati P, De Zio D, Maiani E, Muscoli C, Lauro F, Ilari S, Bernardini S, Cannata S, Gargioli C, Ciriolo MR, Cecconi F, Bonaldo P, Filomeni G. S-nitrosoglutathione reductase deficiency-induced S-nitrosylation results in neuromuscular dysfunction. Antioxid Redox Signal 2014; 21:570-87. [PMID: 24684653 PMCID: PMC4086474 DOI: 10.1089/ars.2013.5696] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
AIMS Nitric oxide (NO) production is implicated in muscle contraction, growth and atrophy, and in the onset of neuropathy. However, many aspects of the mechanism of action of NO are not yet clarified, mainly regarding its role in muscle wasting. Notably, whether NO production-associated neuromuscular atrophy depends on tyrosine nitration or S-nitrosothiols (SNOs) formation is still a matter of debate. Here, we aim at assessing this issue by characterizing the neuromuscular phenotype of S-nitrosoglutathione reductase-null (GSNOR-KO) mice that maintain the capability to produce NO, but are unable to reduce SNOs. RESULTS We demonstrate that, without any sign of protein nitration, young GSNOR-KO mice show neuromuscular atrophy due to loss of muscle mass, reduced fiber size, and neuropathic behavior. In particular, GSNOR-KO mice show a significant decrease in nerve axon number, with the myelin sheath appearing disorganized and reduced, leading to a dramatic development of a neuropathic phenotype. Mitochondria appear fragmented and depolarized in GSNOR-KO myofibers and myotubes, conditions that are reverted by N-acetylcysteine treatment. Nevertheless, although atrogene transcription is induced, and bulk autophagy activated, no removal of damaged mitochondria is observed. These events, alongside basal increase of apoptotic markers, contribute to persistence of a neuropathic and myopathic state. INNOVATION Our study provides the first evidence that GSNOR deficiency, which affects exclusively SNOs reduction without altering nitrotyrosine levels, results in a clinically relevant neuromuscular phenotype. CONCLUSION These findings provide novel insights into the involvement of GSNOR and S-nitrosylation in neuromuscular atrophy and neuropathic pain that are associated with pathological states; for example, diabetes and cancer.
Collapse
|
38
|
Horst A, Kolberg C, Moraes MS, Riffel APK, Finamor IA, Belló-Klein A, Pavanato MA, Partata WA. Effect of N-acetylcysteine on the spinal-cord glutathione system and nitric-oxide metabolites in rats with neuropathic pain. Neurosci Lett 2014; 569:163-8. [PMID: 24704379 DOI: 10.1016/j.neulet.2014.03.063] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/21/2014] [Accepted: 03/07/2014] [Indexed: 11/19/2022]
Abstract
Since N-acetylcysteine (NAC) is a donor of cysteine, we studied the relationship between NAC and concentration of oxidized and reduced glutathione (GSH/GSSG ratio), and glutathione peroxidase (GPx) and glutathione-S-transferase (GST) activities in the lumbosacral spinal cord of rats with chronic constriction injury (CCI) of the sciatic nerve that received NAC (150mg/kg/day, i.p.) or 0.9% saline solution for 3 or 10 days. Hydrogen peroxide (H2O2) and nitric-oxide (NO) metabolites were also measured. Von Frey hair and hot-plate tests showed hyperalgesia at day 1 in CCI rats. Hyperalgesia persisted at all other times in saline-treated CCI rats, but returned to pre-injury values in NAC-treated CCI rats after 3 postoperative days. GST activity and the GSH/GSSG ratio increased in saline-treated CCI rats, while the NAC treatment increased GST and GPx activities at day 10, with no significant change in the GSH/GSSG ratio. NAC treatment did not affect H2O2 levels, but it reduced NO metabolites in CCI rats 3 days after the surgery. Thus, the anti-hyperalgesic effect of NAC appears not to involve its action as a cysteine precursor for GSH synthesis, but involves a decrease in NO.
Collapse
Affiliation(s)
- Andréa Horst
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Carolina Kolberg
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maira S Moraes
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ana Paula K Riffel
- Departamento de Farmacologia e Fisiologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Isabela A Finamor
- Departamento de Farmacologia e Fisiologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Adriane Belló-Klein
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Amália Pavanato
- Departamento de Farmacologia e Fisiologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Wania A Partata
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
39
|
Coburn C, Gems D. The mysterious case of the C. elegans gut granule: death fluorescence, anthranilic acid and the kynurenine pathway. Front Genet 2013; 4:151. [PMID: 23967012 PMCID: PMC3735983 DOI: 10.3389/fgene.2013.00151] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 07/21/2013] [Indexed: 12/31/2022] Open
Abstract
Gut granules are lysosome-like organelles with acidic interiors that are found in large numbers within the intestine of the nematode Caenorhabditis elegans. They are particularly prominent when viewed under ultraviolet light, which causes them to emit intense blue fluorescence. Yet the function of these large and abundant organelles in this heavily-studied model organism remains unclear. One possibility is that they serve as storage organelles, for example of zinc. A new clue to gut granule function is the identification of the blue fluorescent material that they contain as a glycosylated form of anthranilic acid, which is derived from tryptophan by action of the kynurenine pathway. This compound can also serve a surprising role as a natural, endogenous marker of organismal death.
Collapse
Affiliation(s)
| | - David Gems
- Institute of Healthy Ageing, and Department of Genetics, Evolution and Environment, University College LondonLondon, UK
| |
Collapse
|
40
|
Evidence for glutamate as a neuroglial transmitter within sensory ganglia. PLoS One 2013; 8:e68312. [PMID: 23844184 PMCID: PMC3699553 DOI: 10.1371/journal.pone.0068312] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 05/28/2013] [Indexed: 11/19/2022] Open
Abstract
This study examines key elements of glutamatergic transmission within sensory ganglia of the rat. We show that the soma of primary sensory neurons release glutamate when depolarized. Using acute dissociated mixed neuronal/glia cultures of dorsal root ganglia (DRG) or trigeminal ganglia and a colorimetric assay, we show that when glutamate uptake by satellite glial cells (SGCs) is inhibited, KCl stimulation leads to simultaneous increase of glutamate in the culture medium. With calcium imaging we see that the soma of primary sensory neurons and SGCs respond to AMPA, NMDA, kainate and mGluR agonists, and selective antagonists block this response. Using whole cell patch-clamp technique, inward currents were recorded from small diameter (<30 µm) DRG neurons from intact DRGs (ex-vivo whole ganglion preparation) in response to local application of the above glutamate receptor agonists. Following a chronic constriction injury (CCI) of either the inferior orbital nerve or the sciatic nerve, glutamate expression increases in the trigeminal ganglia and DRG respectively. This increase occurs in neurons of all diameters and is present in the somata of neurons with injured axons as well as in somata of neighboring uninjured neurons. These data provides additional evidence that glutamate can be released within the sensory ganglion, and that the somata of primary sensory neurons as well as SGCs express functional glutamate receptors at their surface. These findings, together with our previous gene knockdown data, suggest that glutamatergic transmission within the ganglion could impact nociceptive threshold.
Collapse
|
41
|
P2X7 Cell Death Receptor Activation and Mitochondrial Impairment in Oxaliplatin-Induced Apoptosis and Neuronal Injury: Cellular Mechanisms and In Vivo Approach. PLoS One 2013; 8:e66830. [PMID: 23826152 PMCID: PMC3695015 DOI: 10.1371/journal.pone.0066830] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 05/10/2013] [Indexed: 12/25/2022] Open
Abstract
Limited information is available regarding the cellular mechanisms of oxaliplatin-induced painful neuropathy during exposure of patients to this drug. We therefore determined oxidative stress in cultured cells and evaluated its occurrence in C57BL/6 mice. Using both cultured neuroblastoma (SH-SY5Y) and macrophage (RAW 264.7) cell lines and also brain tissues of oxaliplatin-treated mice, we investigated whether oxaliplatin (OXA) induces oxidative stress and apoptosis. Cultured cells were treated with 2–200 µM OXA for 24 h. The effects of pharmacological inhibitors of oxidative stress or inflammation (N-acetyl cysteine, ibuprofen, acetaminophen) were also tested. Inhibitors were added 30 min before OXA treatment and then in combination with OXA for 24 h. In SH-SY5Y cells, OXA caused a significant dose-dependent decrease in viability, a large increase in ROS and NO production, lipid peroxidation and mitochondrial impairment as assessed by a drop in mitochondrial membrane potential, which are deleterious for the cell. An increase in levels of negatively charged phospholipids such as cardiolipin but also phosphatidylserine and phosphatidylinositol, was also observed. Additionally, OXA caused concentration-dependent P2X7 receptor activation, increased chromatin condensation and caspase-3 activation associated with TNF-α and IL-6 release. The majority of these toxic effects were equally observed in Raw 264.7 which also presented high levels of PGE2. Pretreatment of SH-SY5Y cells with pharmacological inhibitors significantly reduced or blocked all the neurotoxic OXA effects. In OXA-treated mice (28 mg/kg cumulated dose) significant cold hyperalgesia and oxidative stress in the tested brain areas were shown. Our study suggests that targeting P2X7 receptor activation and mitochondrial impairment might be a potential therapeutic strategy against OXA-induced neuropathic pain.
Collapse
|