1
|
Lord B, Simavorian S, Fraser I, Welty N, Wyatt R, Pritchard R, Fletcher L, Van Der Linde H, Bounkhoun D, Libiger O, Maher M, Drevets W, Bischoff F, Bonaventure P, Neff RA. Pharmacological characterisation of JNJ-78911118, a novel, centrally-penetrant, selective GluN2A antagonist. Br J Pharmacol 2025. [PMID: 40361296 DOI: 10.1111/bph.70069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 03/01/2025] [Accepted: 03/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND AND PURPOSE Non-selective NMDA receptor antagonism produces rapid symptom improvement in treatment-resistant depression; however, associated side effects necessitate medical oversight during administration. We hypothesised that selective GluN2A antagonism could provide similar efficacy with an improved side effect profile. Here, we report the pharmacology of JNJ-78911118, a brain-penetrant, GluN2A selective antagonist. EXPERIMENTAL APPROACH JNJ-78911118 pharmacology and mechanism of action was characterised in vitro using fluorescence, voltage clamp and radioligand binding assays. Target engagement was measured using ex vivo receptor autoradiography, and effects on rat prefrontal cortex monoamine levels were measured using microdialysis. Synaptogenesis assays and patch clamp studies were used to demonstrate effects on synaptic plasticity. Cardiovascular safety and neurotoxicity were assessed in rats. KEY RESULTS JNJ-78911118 blocked GluN1/2A receptors with an IC50 of 44 nM and showed selectivity against GluN1/2B, 2C and 2D receptors. Systemic administration produced concentration-dependent receptor occupancy, increased prefrontal cortex monoamine levels in wild type, but not in GluN2A knockout mice, and blocked theta burst induced LTP in the hippocampus. In addition, it produced increases in dendritic complexity and synapse number in vitro, and increased mEPSC frequency in rat cortical neurons in vivo. In rat toxicological studies, no Olney's lesions were observed, but acute increases in heart rate and blood pressure were detected. CONCLUSIONS AND IMPLICATIONS JNJ-78911118 is a potent and selective GluN2A antagonist that reproduces the effect of known rapidly acting antidepressants (RAADs) on neurotransmitter levels and synaptic plasticity. This molecule is a powerful in vivo tool that will enhance understanding of GluN2A biology.
Collapse
Affiliation(s)
- Brian Lord
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Sirak Simavorian
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Ian Fraser
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Natalie Welty
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Ryan Wyatt
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Rory Pritchard
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Lauren Fletcher
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Henk Van Der Linde
- CoE for Cardiovascular Safety Research, Janssen Research and Development, LLC, Beerse, Belgium
| | - Dominic Bounkhoun
- Preclinical Sciences and Translational Safety, Janssen Research and Development, LLC, San Diego, California, USA
| | - Ondrej Libiger
- Statistics and Decision Sciences, Janssen Research and Development, LLC, San Diego, California, USA
| | - Michael Maher
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Wayne Drevets
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - François Bischoff
- Discovery, Product Development and Supply, Janssen Research and Development, LLC, Beerse, Belgium
| | - Pascal Bonaventure
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| | - Robert A Neff
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, California, USA
| |
Collapse
|
2
|
Masaud SM, Nadeem H, Murtaza B, Shamim A. Discovery of a Novel Orally Active Ketamine Derivative with Dual Analgesic and Antidepressant Activities, Lacking Psychomimetic Effects. ACS Chem Neurosci 2025; 16:932-944. [PMID: 39934972 DOI: 10.1021/acschemneuro.4c00887] [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] [Indexed: 02/13/2025] Open
Abstract
This study investigated the synthesis, characterization, and in silico analysis of novel N-acetamide ketamine derivatives aimed at evaluating their analgesic, anesthetic, and antidepressant properties. The synthesis commenced with the preparation of chloroacetylamide derivatives, which were subsequently reacted with ketamine hydrochloride, yielding 16 derivatives k1 to k16. These compounds were characterized through H1 NMR, C13 NMR, mass spectroscopy (EIMS), and elemental analysis, followed by an assessment of their physicochemical properties. The analgesic efficacy of all of the synthesized derivatives was evaluated using the acetic acid-induced writhing test via intraperitoneal administration. The best-performing molecule was further evaluated for analgesic (acetic acid-induced writhing test, tail suspension test (TST), and hot plate test) and anti-inflammatory (carrageenan-induced paw edema) activities. For antidepressant effects, all derivatives were compared with ketamine in a lipopolysaccharide-induced model of depression in mice through the forced swimming test, open field test (OFT), sucrose preference test (SPT), and TST. It was observed that among all the derivatives, molecule k1 demonstrated comparable analgesic activity to ketamine. Further, compound k1 also exhibited the highest antidepressant potential during the forced swimming test, OFT, SPT, and TST. k1 was further compared with ketamine for their activities intraperitoneally and orally where k1 exhibited comparable antidepressant effects to ketamine. Henceforth, the psychomimetic potential of k1 was evaluated through loss of righting reflex and Y-maze tests. Very interestingly, these tests indicated approximately no psychomimetic activity of k1 compared to ketamine intraperitoneally and orally. Finally, molecular docking studies were conducted targeting the NMDA receptor at the JC09 ketamine binding pocket (PDB ID: 7EU7), where all synthesized derivatives exhibited significant binding affinities relative to ketamine. These findings suggest that the newly synthesized N-acetamide ketamine derivative k1 possesses promising pharmacological profiles, warranting further exploration.
Collapse
Affiliation(s)
- Syed Muzzammil Masaud
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Babar Murtaza
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad 44000, Pakistan
| | - Abida Shamim
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
3
|
Wang X, Wu L, Liu J, Ma C, Liu J, Zhang Q. The neuroimmune mechanism of pain induced depression in psoriatic arthritis and future directions. Biomed Pharmacother 2025; 182:117802. [PMID: 39742638 DOI: 10.1016/j.biopha.2024.117802] [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: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Patients suffering from psoriatic arthritis (PsA) often experience depression due to chronic joint pain, which significantly hinders their recovery process. However, the relationship between these two conditions is not well understood. Through a review of existing studies, we revealed that certain neuroendocrine hormones and neurotransmitters are involved in the neuroimmune interactions related to both PsA and depression. These include adrenocorticotropin-releasing hormone (CRH), adrenocorticotropin (ACTH), cortisol, monoamine neurotransmitters, and brain-derived neurotrophic factor (BDNF). Notably, the signalling pathway involving CRH, MCs, and Th17 cells plays a crucial role in linking PsA with depression; thus, this pathway may help clarify their connection. In this review, we outline the inflammatory immune changes associated with PsA and depression. Additionally, we explore how neuroendocrine hormones and neurotransmitters influence inflammatory responses in these two conditions. Finally, our focus will be on potential treatment strategies for patients with PsA and depression through the targeting of the CRH-MC-Th17 pathway. This review aims to provide a theoretical framework as well as new therapeutic targets for managing PsA alongside depression.
Collapse
Affiliation(s)
- Xiaoxu Wang
- Rheumatology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| | - Lingjun Wu
- Shunyi Hospital of Beijing Traditional Chinese Medicine Hospital, Beijing 101300, China
| | - Jing Liu
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing 100010, China
| | - Cong Ma
- Rheumatology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Juan Liu
- Rheumatology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Qin Zhang
- Rheumatology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| |
Collapse
|
4
|
Rosas-Sánchez GU, Germán-Ponciano LJ, Guillen-Ruiz G, Cueto-Escobedo J, Limón-Vázquez AK, Rodríguez-Landa JF, Soria-Fregozo C. Neuroplasticity and Mechanisms of Action of Acute and Chronic Treatment with Antidepressants in Preclinical Studies. Biomedicines 2024; 12:2744. [PMID: 39767650 PMCID: PMC11727250 DOI: 10.3390/biomedicines12122744] [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: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 01/16/2025] Open
Abstract
Pharmacotherapy for depression includes drugs such as monoamine oxidase inhibitors (MAOIs), tricyclic antidepressants (TCAs), selective serotonin reuptake inhibitors (SSRIs), noradrenaline (NA) and serotonin (5-HT) reuptake inhibitors (NaSSAs), and atypical antidepressants; these drugs exert differentially beneficial effects on symptoms of depression after acute and chronic treatment in animal models. Said effects are established through neuroplastic mechanisms involving changes in neurogenesis and synaptogenesis as result of the activation of intracellular signaling pathways associated with neurochemical and behavioral changes. Antidepressants increase the synaptic availability of monoamines (monoaminergic hypothesis) such as 5-HT, NA, and gamma-aminobutyric acid (GABA) by inhibiting their reuptake or degradation and activating intracellular signaling pathways such as the responsive element binding protein (cAMP-CREB) cascade, which regulates the expression of genes related to neuroplasticity and neurogenesis, such as brain-derived neurotrophic factor (BDNF), in various brain structures implicated in depression. The aim of this review is to analyze the mechanisms of action of different antidepressants and to compare the effects of acute and chronic treatment on neuroplasticity in animal models of depression. A thorough search was conducted in PubMed, Scopus, and Web of Science, focusing on studies since 1996 with keywords like antidepressants, acute and chronic treatment, neuroplasticity, and experimental depression. Studies included had to investigate antidepressant effects experimentally, with full-text access, while excluding those that did not. Data extraction focused on study design, findings, and relevance to understanding treatment differences. Only high-quality, peer-reviewed studies were considered to ensure a comprehensive synthesis of current knowledge.
Collapse
Affiliation(s)
| | - León Jesús Germán-Ponciano
- Laboratorio de Neurofarmacología, Instituto de Neuroetología, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico; (L.J.G.-P.); (A.K.L.-V.)
| | - Gabriel Guillen-Ruiz
- Programa Investigadoras e Investigadores por México-CONAHCYT-Instituto de Neuroetología, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico;
| | | | - Ana Karen Limón-Vázquez
- Laboratorio de Neurofarmacología, Instituto de Neuroetología, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico; (L.J.G.-P.); (A.K.L.-V.)
| | - Juan Francisco Rodríguez-Landa
- Laboratorio de Neurofarmacología, Instituto de Neuroetología, Universidad Veracruzana, Xalapa 91190, Veracruz, Mexico; (L.J.G.-P.); (A.K.L.-V.)
| | - César Soria-Fregozo
- Centro Universitario de Los Lagos, Universidad de Guadalajara, Lagos de Moreno 47460, Jalisco, Mexico;
| |
Collapse
|
5
|
Yang P, Chen H, Wang T, Su H, Li J, He Y, Su S. Electroacupuncture promotes synaptic plasticity in rats with chronic inflammatory pain-related depression by upregulating BDNF/TrkB/CREB signaling pathway. Brain Behav 2023; 13:e3310. [PMID: 37948105 PMCID: PMC10726860 DOI: 10.1002/brb3.3310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Chronic inflammatory pain (CIP) frequently coincides with depression among patients. The onset and development of pain and depression are associated with altered neural synaptic plasticity. Electroacupuncture (EA) can effectively relieve CIP and depression. However, the underlying mechanisms have not been fully illustrated. OBJECTIVE To explore whether EA can relieve CIP and depression by regulating hippocampal synaptic plasticity, and the present study offers foundational evidence for the efficacy of EA in treating CIP-related depression (CIPD). METHODS Rats were divided into four groups: 0.9% normal saline group, complete Freund's adjuvant (CFA) group, CFA + duloxetine group, and CFA + EA group. Pain hypersensitivity was detected by mechanical withdrawal threshold and thermal paw withdrawal latency, and the depression level was gauged using the open field test, the sucrose preference test, and the forced swimming test. The morphology of the hippocampal neurons was observed using Nissl staining. The protein expression levels of synuclein (Syn), postsynaptic density protein-95 (PSD-95), brain-derived neurotrophic factors (BDNFs), tyrosine-protein kinase B (TrKB), p-TrkB, cAMP response element binding protein (CREB), and p-CREB were measured by western blotting and immunofluorescence staining. BDNF and TrkB mRNA expression were detected using quantitative real-time polymerase chain reaction (PCR) (qRT-PCR). The content of 5-hydroxytryptamine (5-HT) and γ-aminobutyric acid (GABA) was detected using enzyme-linked immunosorbent assay, and the glutamic acid (Glu) content was determined using the ultraviolet colorimetry method. The hippocampal neuron ultrastructure was observed using transmission electron microscopy. RESULTS EA could alleviate CIP and related depressive behaviors as well as protect the hippocampal neuronal structure from damage and regulate 5-HT/GABA/Glu levels in the hippocampus. Additionally, EA could significantly increase the expression of synapse-associated proteins such as PSD-95 and Syn by activating the BDNF/TrKB/CREB signaling pathway. CONCLUSION EA improves pain and depressive behaviors in CIPD rats, and the mechanism may be related to synaptic plasticity mediated by the BDNF/TrKB/CREB signaling pathway.
Collapse
Affiliation(s)
- Pu Yang
- The First School of Clinical MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Haiyan Chen
- Department of NursingThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningGuangxiChina
| | - Tian Wang
- The First School of Clinical MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Hong Su
- The First School of Clinical MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Jing Li
- The First School of Clinical MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Yujun He
- Faculty of Acupuncture, Moxibustion and TuinaGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Shengyong Su
- Department of Acupuncture and MoxibustionThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningGuangxiChina
- Guangxi Key Laboratory of Molecular Biology of Preventive Medicine of Traditional Chinese MedicineNanningGuangxiChina
| |
Collapse
|
6
|
Yang S, Zhang B, Wang D, Hu S, Wang W, Liu C, Wu Z, Yang C. Role of GABAergic system in the comorbidity of pain and depression. Brain Res Bull 2023:110691. [PMID: 37331640 DOI: 10.1016/j.brainresbull.2023.110691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/23/2023] [Accepted: 06/16/2023] [Indexed: 06/20/2023]
Abstract
Patients with chronic pain often suffer with depressive symptoms, and these two conditions can be aggravated by each other over time, leading to an increase in symptom intensity and duration. The comorbidity of pain and depression poses a significant challenge to human health and quality of life, as it is often difficult to diagnose early and treat effectively. Therefore, exploring the molecular mechanisms underlying the comorbidity of chronic pain and depression is crucial to identifying new therapeutic targets for treatment. However, understanding the pathogenesis of comorbidity requires examining interactions among multiple factors, which calls for an integrative perspective. While several studies have explored the role of the GABAergic system in pain and depression, fewer have examined its interactions with other systems involved in their comorbidity. Here, we review the evidence that the role of GABAergic system in the comorbidity of chronic pain and depression, as well as the interactions between the GABAergic system and other secondary systems involved in pain and depression comorbidity, providing a comprehensive understanding of their intricate interplay.
Collapse
Affiliation(s)
- Siqi Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Bingyuan Zhang
- Department of Anesthesiology, Taizhou People's Hospital Affiliated to Nanjing Medical University, No. 399 Hailing South Road, Taizhou City, 225300, Jiangsu Province, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Suwan Hu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Wenli Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029. China.
| |
Collapse
|
7
|
Lashgari NA, Roudsari NM, Shayan M, Niazi Shahraki F, Hosseini Y, Momtaz S, Abdolghaffari AH. IDO/Kynurenine; novel insight for treatment of inflammatory diseases. Cytokine 2023; 166:156206. [PMID: 37120946 DOI: 10.1016/j.cyto.2023.156206] [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: 01/07/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023]
Abstract
Inflammation and oxidative stress play pivotal roles in pathogenesis of many diseases including cancer, type 2 diabetes, cardiovascular disease, atherosclerosis, neurological diseases, and inflammatory diseases such as inflammatory bowel disease (IBD). Inflammatory mediators such as interleukins (ILs), interferons (INF-s), and tumor necrosis factor (TNF)-α are related to an extended chance of inflammatory diseases initiation or progression due to the over expression of the nuclear factor Kappa B (NF-κB), signal transducer of activators of transcription (STAT), nod-like receptor family protein 3 (NLRP), toll-like receptors (TLR), mitogen-activated protein kinase (MAPK), and mammalian target of rapamycin (mTOR) pathways. These pathways are completely interconnected. Theindoleamine 2,3 dioxygenase (IDO) subset of the kynurenine (KYN) (IDO/KYN), is a metabolic inflammatory pathway involved in production of nicotinamide adenine dinucleotide (NAD + ). It has been shown that IDO/KYN actively participates in inflammatory processes and can increase the secretion of cytokines that provoke inflammatory diseases. Data were extracted from clinical and animal studies published in English between 1990-April 2022, which were collected from PubMed, Google Scholar, Scopus, and Cochrane library. IDO/KYN is completely associated with inflammatory-related pathways, thus leading to the production of cytokines such as TNF-α, IL-1β, and IL-6, and ultimately development and progression of various inflammatory disorders. Inhibition of the IDO/KYN pathway might be a novel therapeutic option for inflammatory diseases. Herein, we gathered data on probable interactions of the IDO/KYN pathway with induction of some inflammatory diseases.
Collapse
Affiliation(s)
- Naser-Aldin Lashgari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Momeni Roudsari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Shayan
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Niazi Shahraki
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin Hosseini
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
8
|
Ma X, Peng J, Chen Y, Wang Z, Zhou Q, Yan J, Jiang H. Esketamine Anesthetizes Mice With a Similar Potency to Racemic Ketamine. Dose Response 2023; 21:15593258231157563. [PMID: 36798635 PMCID: PMC9926386 DOI: 10.1177/15593258231157563] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/13/2023] Open
Abstract
Esketamine, the right-handed optical isomer of racemic ketamine, has recently become widely used for anesthesia and analgesia as a replacement for racemic ketamine. However, there are limited studies comparing the anesthetic and analgesic effects of esketamine and racemic ketamine in mice. This research was conducted to analyze the dose-dependent anesthetic and analgesic efficacy of esketamine in mice and to compare its potency with that of the racemate. We tested the anesthetic effects of different doses of esketamine and compared its potency with that of the racemate using righting reflex tests. Then, the acetic acid-induced pain model and formalin-induced pain model were used to investigate the analgesic effect. Compared with racemic ketamine, an equivalent dose of esketamine at 100 mg/kg was required to induce stable anesthesia. In contrast, 5 mg/kg esketamine was sufficient to provide analgesic effects similar to those of 10 mg/kg ketamine. Together, esketamine had a similar potency to racemic ketamine for anesthesia and a stronger potency for analgesia in mice.
Collapse
Affiliation(s)
- Xiaofan Ma
- Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China
| | - Jiali Peng
- Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China
| | - Yelin Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zeyi Wang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qiang Zhou
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jia Yan
- Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China,Jia Yan, Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, No. 639, Zhizaoju Road, Shanghai 200011, China.
| | - Hong Jiang
- Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People’s Hospital, Shanghai, China,Hong Jiang, Department of Anesthesiology, Shanghai Jiao Tong University School of Medicine Affiliated Ninth People's Hospital, No. 639, Zhizaoju Road, Shanghai, China.
| |
Collapse
|
9
|
Halaris A, Cook J. The Glutamatergic System in Treatment-Resistant Depression and Comparative Effectiveness of Ketamine and Esketamine: Role of Inflammation? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:487-512. [PMID: 36949323 DOI: 10.1007/978-981-19-7376-5_21] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
The glutamatergic system is the primary excitatory pathway within the CNS and is responsible for cognition, memory, learning, emotion, and mood. Because of its significant importance in widespread nervous system function, it is tightly regulated through multiple mechanisms, such as glutamate recycling, microglial interactions, and inflammatory pathways. Imbalance within the glutamatergic system has been implicated in a wide range of pathological conditions including neurodegenerative conditions, neuromuscular conditions, and mood disorders including depression. Major depressive disorder (MDD) is the most common mood disorder worldwide, has a high prevalence rate, and afflicts approximately 280 million people. While there are numerous treatments for the disease, 30-40% of patients are unresponsive to treatment and deemed treatment resistant; approximately another third experience only partial improvement (World Health Organization, Depression fact sheet [Internet], 2020). Esketamine, the S-enantiomer of ketamine, was approved by the Food and Drug Administration for treatment-resistant depression (TRD) in 2019 and has offered new hope to patients. It is the first treatment targeting the glutamatergic system through a complex mechanism. Numerous studies have implicated imbalance in the glutamatergic system in depression and treatment resistance. Esketamine and ketamine principally work through inhibition of the NMDA receptor, though more recent studies have implicated numerous other mechanisms mediating the antidepressant efficacy of these agents. These mechanisms include increase in brain-derived neurotrophic factor (BDNF), activation of mammalian target of the rapamycin complex (mTORC), and reduction in inflammation. Esketamine and ketamine have been shown to decrease inflammation in numerous ways principally through reducing pro-inflammatory cytokines (e.g., TNF-α, IL-6) (Loix et al., Acta Anaesthesiol Belg 62(1):47-58, 2011; Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021). This anti-inflammatory effect has also been shown to be involved in the antidepressive properties of both ketamine and esketamine (Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021).
Collapse
Affiliation(s)
- Angelos Halaris
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA.
| | - John Cook
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
10
|
Soued M, Hamdi L, Ben Rehouma M, Mazoit JX, Benhamou D. Antinociceptive properties of losmapimod in two acute pain models in rats: behavioural analysis, immunohistochemistry, dose response, and comparison with usual analgesic drugs. BJA OPEN 2022; 3:100029. [PMID: 37588580 PMCID: PMC10430813 DOI: 10.1016/j.bjao.2022.100029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/29/2022] [Indexed: 08/18/2023]
Abstract
Background The p38 protein is a ubiquitous mitogen-activated protein kinase involved in the proinflammatory signalling pathway and in the pain response after various noxious stimuli. Many p38 inhibitors have been developed and shown to provide effective analgesia in animal models. They are, however, mainly administered intrathecally or intravenously. Our study aimed to evaluate losmapimod, a novel oral p38 inhibitor, in two murine acute pain models. Methods Losmapimod (12 mg kg-1) was compared with paracetamol, ketamine, and morphine using thermal and mechanical stimulation after carrageenan injection. A dose-effect study was also performed with this model. Behavioural testing was also performed in a plantar incision model to confirm the analgesic effect of losmapimod. Expression of activated p38 in neurones, microglia, and astrocytes was also investigated at 2, 15, and 24 h after carrageenan injection. Results Losmapimod was both antiallodynic and antihyperalgesic in the carrageenan pain model and provided an antinociceptive effect similar to that of morphine. The dose of 12 mg kg-1 was shown to be the ED78 and ED64 after thermal and mechanical stimulation, respectively. After plantar incision, losmapimod provided a significant antinociceptive effect. No life-threatening side-effect was observed in the behavioural study. Losmapimod prevented neurone and microglial activation at 2 and 15 h after carrageenan injection, respectively, but no effect was found on astrocytic activation. Conclusion Losmapimod appears to be a promising drug in severe acute pain conditions. Losmapimod could also be helpful for postoperative pain control, as suggested by its effect after plantar incision.
Collapse
Affiliation(s)
- Mickaël Soued
- Laboratory of Anaesthesia, Inserm U 1195 Neuroprotection et neurorégéneration, Paris-Saclay University, Le Kremlin-Bicêtre, France
- Department of Anaesthesia, Antoine Béclère Hospital, APHP, Paris-Saclay University, Clamart, France
| | - Leila Hamdi
- Laboratory of Anaesthesia, Inserm U 1195 Neuroprotection et neurorégéneration, Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Mouna Ben Rehouma
- Laboratory of Anaesthesia, Inserm U 1195 Neuroprotection et neurorégéneration, Paris-Saclay University, Le Kremlin-Bicêtre, France
- Department of Anaesthesia and Intensive Care Medicine, Bichat Hospital, APHP, Paris Seine Saint Denis, Paris, France
| | - Jean-Xavier Mazoit
- Laboratory of Anaesthesia, Inserm U 1195 Neuroprotection et neurorégéneration, Paris-Saclay University, Le Kremlin-Bicêtre, France
- Department of Anaesthesia and Intensive Care Medicine, Bicêtre Hospital, APHP, Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Dan Benhamou
- Laboratory of Anaesthesia, Inserm U 1195 Neuroprotection et neurorégéneration, Paris-Saclay University, Le Kremlin-Bicêtre, France
- Department of Anaesthesia and Intensive Care Medicine, Bicêtre Hospital, APHP, Paris-Saclay University, Le Kremlin-Bicêtre, France
| |
Collapse
|
11
|
Moraga-Amaro R, Guerrin CGJ, Reali Nazario L, Lima Giacobbo B, J O Dierckx RA, Stehberg J, de Vries EFJ, Doorduin J. A single dose of ketamine cannot prevent protracted stress-induced anhedonia and neuroinflammation in rats. Stress 2022; 25:145-155. [PMID: 35384793 DOI: 10.1080/10253890.2022.2045269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Worldwide, millions of people suffer from treatment-resistant depression. Ketamine, a glutamatergic receptor antagonist, can have a rapid antidepressant effect even in treatment-resistant patients. A proposed mechanism for the antidepressant effect of ketamine is the reduction of neuroinflammation. To further explore this hypothesis, we investigated whether a single dose of ketamine can modulate protracted neuroinflammation in a repeated social defeat (RSD) stress rat model, which resembles features of depression. To this end, male animals exposed to RSD were injected with ketamine (20 mg/kg) or vehicle. A combination of behavioral analyses and PET scans of the inflammatory marker TSPO in the brain were performed. Rats submitted to RSD showed anhedonia-like behavior in the sucrose preference test, decreased weight gain, and increased TSPO levels in the insular and entorhinal cortices, as observed by [11C]-PK11195 PET. Whole brain TSPO levels correlated with corticosterone levels in several brain regions of RSD exposed animals, but not in controls. Ketamine injection 1 day after RSD disrupted the correlation between TSPO levels and serum corticosterone levels, but had no effect on depressive-like symptoms, weight gain or the protracted RSD-induced increase in TSPO expression in male rats. These results suggest that ketamine does not exert its effect on the hypothalamic-pituitary-adrenal axis by modulation of neuroinflammation.
Collapse
Affiliation(s)
- Rodrigo Moraga-Amaro
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Cyprien G J Guerrin
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Luiza Reali Nazario
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Bruno Lima Giacobbo
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Jimmy Stehberg
- Laboratorio de Neurobiología, Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Erik F J de Vries
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Medical Imaging, University Medical Center Groningen, University of Groningen, Groningen, GZ, The Netherlands
| |
Collapse
|
12
|
Ma KH, Cheng CY, Chan WH, Chen SY, Kao LT, Sung CS, Hueng DY, Yeh CC. Pulsed Radiofrequency Upregulates Serotonin Transporters and Alleviates Neuropathic Pain-Induced Depression in a Spared Nerve Injury Rat Model. Biomedicines 2021; 9:biomedicines9101489. [PMID: 34680606 PMCID: PMC8533300 DOI: 10.3390/biomedicines9101489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 02/06/2023] Open
Abstract
Neuropathic pain (NP) is difficult to treat due to complex pathophysiological mechanisms. Pulsed radiofrequency (RRF) has been used widely with neuromodulation effect in refractory chronic pain treatment. A recent study found that PRF treatment may decrease chronic pain-related anxiety-depressant symptoms in patients, even though the mechanisms are unclear. Additionally, accumulated evidence has shown serotonin uptake is correlated with various neuropsychiatric diseases. Therefore, we investigated the effects and underlying mechanisms of PRF on depression-like behaviors, resulting from spared nerve injury (SNI)-induced NP. We examined the indexes of mechanical allodynia, cold allodynia, depression-like behavior, and blood cytokines by dynamic plantar aesthesiometry, acetone spray test, forced swimming test, and ProcartaPlex multiplex immunoassays in male Wistar rats, respectively. Serotonin transporters (SERTs) in rat brains were examined by using 4-[18F]-ADAM/PET imaging. We found that specific uptake ratios (SURs) of SERTs were significantly decreased in the brain regions of the thalamus and striatum in rats with SNI-induced NP and depression-like behaviors. Additionally, the decrease in SERT density was correlated with the development of a depression-like behavior indicated by the forced swimming test results and pronounced IL-6 cytokines. Moreover, we demonstrated that PRF application could modulate the descending serotoninergic pathway to relieve pain and depression behaviors.
Collapse
Affiliation(s)
- Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 115, Taiwan;
| | - Cheng-Yi Cheng
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 115, Taiwan;
| | - Wei-Hung Chan
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 115, Taiwan; (W.-H.C.); (S.-Y.C.)
| | - Shih-Yu Chen
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 115, Taiwan; (W.-H.C.); (S.-Y.C.)
| | - Li-Ting Kao
- Department of Pharmacy Practice, Tri-Service General Hospital, National Defense Medical Center, Taipei 115, Taiwan;
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 115, Taiwan
| | - Chun-Sung Sung
- Department of Anesthesiology, Taipei Veterans General Hospital, Taipei 112, Taiwan;
- School of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112, Taiwan
| | - Dueng-Yuan Hueng
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 115, Taiwan;
| | - Chun-Chang Yeh
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 115, Taiwan; (W.-H.C.); (S.-Y.C.)
- Integrated Pain Management Center, Tri-Service General Hospital, National Defense Medical Center, Taipei 115, Taiwan
- Correspondence:
| |
Collapse
|
13
|
Melanson B, Leri F. Effect of ketamine on the physiological responses to combined hypoglycemic and psychophysical stress. IBRO Neurosci Rep 2021; 11:81-87. [PMID: 34485972 PMCID: PMC8406162 DOI: 10.1016/j.ibneur.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/20/2021] [Indexed: 12/01/2022] Open
Abstract
There is evidence that hypoglycemic stress can interact with other stressors, and that ketamine can mitigate the impact of these stressors on behavior and physiology. The current study in male Sprague-Dawley rats investigated whether pre-treatment with 0, 10, or 20 mg/kg ketamine could modulate the interaction between hypoglycemia induced by 0 or 300 mg/kg 2-deoxy-D-glucose (2-DG) and the psychophysical stress of forced swimming (FSS; 6 sessions, 10 min/session) on serum concentrations of corticosterone (CORT) and the pro-inflammatory cytokine, tumor necrosis factor (TNF)-α. It was found that 2-DG enhanced the CORT response to an initial session of FSS, and this effect dissipated after multiple sessions. More importantly, animals displayed significantly higher levels of CORT and lower levels of TNF-α in response to a drug-free test swim conducted 1 week after exposure to the combined stressors, and these responses were not observed in rats that were pre-treated with ketamine. Overall, these findings indicate that ketamine has the potential to reduce the negative impact of interacting stressors on the biological reactivity of the hypothalamic-pituitary-adrenal axis and the immune system.
Collapse
Affiliation(s)
- Brett Melanson
- Department of Psychology and Collaborative Neuroscience, Program University of Guelph, Guelph, ON, Canada
| | - Francesco Leri
- Department of Psychology and Collaborative Neuroscience, Program University of Guelph, Guelph, ON, Canada
| |
Collapse
|
14
|
Hsu YC, Ma KH, Guo SL, Lin BF, Tsai CS, Yeh CC. The Occurrence of Pain-Induced Depression Is Different between Rat Models of Inflammatory and Neuropathic Pain. J Clin Med 2021; 10:jcm10174016. [PMID: 34501464 PMCID: PMC8432452 DOI: 10.3390/jcm10174016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/18/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022] Open
Abstract
Various pain conditions may be associated with depressed mood. However, the effect of inflammatory or neuropathic pain on depression-like behavior and its associated time frame has not been well established in rat models. This frontward study investigated the differences in pain behavior, depression-like behavior, and serotonin transporter (SERT) distribution in the brain between rats subjected to spared nerve injury (SNI)-induced neuropathic pain or complete Freund’s adjuvant (CFA)-induced inflammatory pain. A dynamic plantar aesthesiometer and an acetone spray test were used to evaluate mechanical and cold allodynia responses, and depression-like behavior was examined using a forced swimming test and sucrose preference test. We also investigated SERT expression by using positron emission tomography. We found that the inflammation-induced pain was less severe than neuropathic pain from days 3 to 28 after induced pain; however, the CFA-injected rats exhibited more noticeable depression-like behavior and had significantly reduced SERT expression in the brain regions (thalamus and striatum) at an early stage (on days 14, 21, and 28 in two groups of CFA-injected rats versus day 28 in SNI rats). We speculated that not only the pain response after initial injury but also the subsequent neuroinflammation may have been the crucial factors influencing depression-like behavior in rats.
Collapse
Affiliation(s)
- Yung-Chi Hsu
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-C.H.); (S.-L.G.); (B.-F.L.)
- Integrated Pain Management Center, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan;
| | - Shu-Lin Guo
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-C.H.); (S.-L.G.); (B.-F.L.)
- Department of Anesthesiology, Cathay General Hospital, Taipei 106, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Bo-Feng Lin
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-C.H.); (S.-L.G.); (B.-F.L.)
- Integrated Pain Management Center, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chien-Sung Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 114, Taiwan
| | - Chun-Chang Yeh
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-C.H.); (S.-L.G.); (B.-F.L.)
- Integrated Pain Management Center, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: ; Tel.: +886-2-8792-3311 (ext. 17051); Fax: +886-2-8792-7127
| |
Collapse
|
15
|
Kopra E, Mondelli V, Pariante C, Nikkheslat N. Ketamine's effect on inflammation and kynurenine pathway in depression: A systematic review. J Psychopharmacol 2021; 35:934-945. [PMID: 34180293 PMCID: PMC8358579 DOI: 10.1177/02698811211026426] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ketamine is a novel rapid-acting antidepressant with high efficacy in treatment-resistant patients. Its exact therapeutic mechanisms of action are unclear; however, in recent years its anti-inflammatory properties and subsequent downstream effects on tryptophan (TRP) metabolism have sparked research interest. AIM This systematic review examined the effect of ketamine on inflammatory markers and TRP-kynurenine (KYN) pathway metabolites in patients with unipolar and bipolar depression and in animal models of depression. METHODS MEDLINE, Embase, and PsycINFO databases were searched on October 2020 (1806 to 2020). RESULTS Out of 807 initial results, nine human studies and 22 animal studies on rodents met the inclusion criteria. Rodent studies provided strong support for ketamine-induced decreases in pro-inflammatory cytokines, namely in interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α and indicated anti-inflammatory effects on TRP metabolism, including decreases in the enzyme indoleamine 2,3-dioxygenase (IDO). Clinical evidence was less robust with high heterogeneity between sample characteristics, but most experiments demonstrated decreases in peripheral inflammation including in IL-1β, IL-6, and TNF-α. Preliminary support was also found for reduced activation of the neurotoxic arm of the KYN pathway. CONCLUSION Ketamine appears to induce anti-inflammatory effects in at least a proportion of depressed patients. Suggestions for future research include investigation of markers in the central nervous system and examination of clinical relevance of inflammatory changes.
Collapse
Affiliation(s)
- Emma Kopra
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Carmine Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Naghmeh Nikkheslat
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| |
Collapse
|
16
|
Melanson B, Lapointe T, Leri F. Impact of impaired glucose metabolism on responses to a psychophysical stressor: modulation by ketamine. Psychopharmacology (Berl) 2021; 238:1005-1015. [PMID: 33404733 DOI: 10.1007/s00213-020-05748-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022]
Abstract
RATIONALE There is evidence that hypoglycemia, a metabolic stressor, can negatively impact mood and motivation, and can interact with other stressors to potentiate their effects on behavior and physiology. OBJECTIVES/METHODS The current study in male Sprague-Dawley rats explored the interaction between impaired glucose metabolism induced by 0, 200, or 300 mg/kg 2-deoxy-D-glucose (2-DG) and a psychophysical stressor induced by forced swimming stress (FSS; 6 sessions, 10 min/session). The endpoints of interest were blood glucose levels, progressive behavioral immobility, and saccharin preference (2-bottle choice test). Furthermore, it was investigated whether pre-treatment with 0, 10, or 20 mg/kg ketamine could modify the interaction between 2-DG and FSS on these endpoints. RESULTS It was found that 2-DG increased blood glucose levels equally in all experimental groups, accelerated the immobile response to FSS, and suppressed saccharin preference 1 week following termination of stress exposure. As well, pre-treatment with ketamine blocked the effects of combined 2-DG and FSS on immobility and saccharin preference without affecting blood glucose levels and produced an anti-immobility effect that was observed during a drug-free test swim 1 week following administration. CONCLUSIONS Overall, these findings demonstrate that impaired glucose metabolism can potentiate the effects of a psychophysical stressor, and that this interaction can be modulated pharmacologically by ketamine.
Collapse
Affiliation(s)
- Brett Melanson
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada
| | - Thomas Lapointe
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada
| | - Francesco Leri
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
17
|
Ji MJ, Yang J, Gao ZQ, Zhang L, Liu C. The Role of the Kappa Opioid System in Comorbid Pain and Psychiatric Disorders: Function and Implications. Front Neurosci 2021; 15:642493. [PMID: 33716658 PMCID: PMC7943636 DOI: 10.3389/fnins.2021.642493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/27/2021] [Indexed: 01/25/2023] Open
Abstract
Both pain and psychiatric disorders, such as anxiety and depression, significantly impact quality of life for the sufferer. The two also share a strong pathological link: chronic pain-induced negative affect drives vulnerability to psychiatric disorders, while patients with comorbid psychiatric disorders tend to experience exacerbated pain. However, the mechanisms responsible for the comorbidity of pain and psychiatric disorders remain unclear. It is well established that the kappa opioid system contributes to depressive and dysphoric states. Emerging studies of chronic pain have revealed the role and mechanisms of the kappa opioid system in pain processing and, in particular, in the associated pathological alteration of affection. Here, we discuss the key findings and summarize compounds acting on the kappa opioid system that are potential candidates for therapeutic strategies against comorbid pain and psychiatric disorders.
Collapse
Affiliation(s)
- Miao-Jin Ji
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Jiao Yang
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Zhi-Qiang Gao
- Jiangsu Province Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Liang Zhang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Liu
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, School of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
18
|
Chronic pain impact on rodents’ behavioral repertoire. Neurosci Biobehav Rev 2020; 119:101-127. [DOI: 10.1016/j.neubiorev.2020.09.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/14/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022]
|
19
|
Campos ACP, Antunes GF, Matsumoto M, Pagano RL, Martinez RCR. Neuroinflammation, Pain and Depression: An Overview of the Main Findings. Front Psychol 2020; 11:1825. [PMID: 32849076 PMCID: PMC7412934 DOI: 10.3389/fpsyg.2020.01825] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic pain is a serious public health problem with a strong affective-motivational component that makes it difficult to treat. Most patients with chronic pain suffer from severe depression; hence, both conditions coexist and exacerbate one another. Brain inflammatory mediators are critical for maintaining depression-pain syndrome and could be substrates for it. The goal of our paper was to review clinical and preclinical findings to identify the neuroinflammatory profile associated with the cooccurrence of pain and depression. In addition, we aimed to explore the regulatory effect of neuronal reorganization on the inflammatory response in pain and depression. We conducted a quantitative review supplemented by manual screening. Our results revealed inflammatory signatures in different preclinical models and clinical articles regarding depression-pain syndrome. We also identified that improvements in depressive symptoms and amelioration of pain can be modulated through direct targeting of inflammatory mediators, such as cytokines and molecular inhibitors of the inflammatory cascade. Additionally, therapeutic targets that improve and regulate the synaptic environment and its neurotransmitters may act as anti-inflammatory compounds, reducing local damage-associated molecular patterns and inhibiting the activation of immune and glial cells. Taken together, our data will help to better elucidate the neuroinflammatory profile in pain and depression and may help to identify pharmacological targets for effective management of depression-pain syndrome.
Collapse
Affiliation(s)
| | | | - Marcio Matsumoto
- Anesthesiology Medical Center, Hospital Sirio-Libanes, São Paulo, Brazil
| | | | - Raquel Chacon Ruiz Martinez
- Division of Neuroscience, Hospital Sirio-Libanes, São Paulo, Brazil.,LIM 23, Institute of Psychiatry, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
20
|
Yang Y, Song Y, Zhang X, Zhao W, Ma T, Liu Y, Ma P, Zhao Y, Zhang H. Ketamine relieves depression-like behaviors induced by chronic postsurgical pain in rats through anti-inflammatory, anti-oxidant effects and regulating BDNF expression. Psychopharmacology (Berl) 2020; 237:1657-1669. [PMID: 32125485 DOI: 10.1007/s00213-020-05490-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 02/14/2020] [Indexed: 12/12/2022]
Abstract
RATIONALE Clinically, chronic postsurgical pain (CPSP) is very common. Many CPSP patients may experience depression. Thus far, little is known about the mechanism of the comorbidity of CPSP and depression. Ketamine has been confirmed to possess analgesic and rapid antidepressant effects, but it is unclear whether ketamine can relieve the comorbidity of CPSP and depression. OBJECTIVES The present study evaluated the effects of ketamine in rats with the comorbidity of CPSP and depression. METHODS We induced CPSP in rats by thoracotomy and screened for rats with or without depression-like phenotype by hierarchical cluster analysis based on the results of depression-related behavioral experiments. Subsequently, rats were intraperitoneally injected with ketamine (20 mg/kg) and were evaluated by mechanical withdrawal threshold, cold hyperalgesia test, sucrose preference test, forced swimming test, and open field test. The inflammatory-related cytokines (IL-1, IL-6, TNF-α, nuclear factor-kappaB), oxidative stress parameters (superoxide dismutase, malondialdehyde, glutathione, catalase), and brain-derived neurotrophic factor (BDNF) in rat hippocampus were detected. RESULTS In the hippocampus of rats with the comorbidity of CPSP and depression, IL-1, IL-6, TNF-α, nuclear factor-kappaB, and malondialdehyde were significantly increased, while superoxide dismutase, glutathione, catalase, and BDNF were significantly decreased. Ketamine relieved depression but did not attenuate hyperalgesia in CPSP rats. Additionally, ketamine reduced proinflammatory cytokines, inhibited oxidative stress, and elevated BDNF levels in rat hippocampus. CONCLUSIONS Ketamine can rapidly relieve CPSP-induced depression in rats, which may be related to the reduction of proinflammatory cytokines, regulating oxidative stress and increasing BDNF in the hippocampus.
Collapse
Affiliation(s)
- Yitian Yang
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China.
| | - Yuxiang Song
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China
| | - Xuan Zhang
- Department of Anesthesiology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, 300060, China
| | - Weixing Zhao
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China
| | - Tao Ma
- Department of Anesthesiology, Rocket Army Characteristic Medical Center, Beijing, 100088, China
| | - Yi Liu
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China
| | - Penglei Ma
- Department of Anesthesiology, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010030, China
| | - Yifan Zhao
- Department of Anesthesiology, The Fourth Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, Beijing, 100037, China
| | - Hong Zhang
- Anesthesia and Operation Center, The First Medical Center of Chinese PLA General Hospital, Medical school of Chinese PLA, No. 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
21
|
Mokhtari T, Tu Y, Hu L. Involvement of the hippocampus in chronic pain and depression. BRAIN SCIENCE ADVANCES 2020. [DOI: 10.26599/bsa.2019.9050025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Increases in depressive behaviors have been reported in patients experiencing chronic pain. In these patients, the symptoms of pain and depression commonly coexist, impairing their lives and challenging effective treatment. The hippocampus may play a role in both chronic pain and depression. A reduction in the volume of the hippocampus is related to reduced neurogenesis and neuroplasticity in cases of chronic pain and depression. Moreover, an increase of proinflammatory factors and a reduction of neurotrophic factors have been reported to modulate the hippocampal neurogenesis and neuroplasticity in chronic pain and depression. This review discusses the mechanisms underlying the depressive-like behavior accompanying chronic pain, emphasizing the structural and functional changes in the hippocampus. We also discuss the hypothesis that pro-inflammatory factors and neurotrophic factors expressed in the hippocampus may serve as a therapeutic target for comorbid chronic pain and depression.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yiheng Tu
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Li Hu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
22
|
Bravo L, Llorca-Torralba M, Suárez-Pereira I, Berrocoso E. Pain in neuropsychiatry: Insights from animal models. Neurosci Biobehav Rev 2020; 115:96-115. [PMID: 32437745 DOI: 10.1016/j.neubiorev.2020.04.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 04/11/2020] [Accepted: 04/23/2020] [Indexed: 02/08/2023]
Abstract
Pain is the most common symptom reported in clinical practice, meaning that it is associated with many pathologies as either the origin or a consequence of other illnesses. Furthermore, pain is a complex emotional and sensorial experience, as the correspondence between pain and body damage varies considerably. While these issues are widely acknowledged in clinical pain research, until recently they have not been extensively considered when exploring animal models, important tools for understanding pain pathophysiology. Interestingly, chronic pain is currently considered a risk factor to suffer psychiatric disorders, mainly stress-related disorders like anxiety and depression. Conversely, pain appears to be altered in many psychiatric disorders, such as depression, anxiety and schizophrenia. Thus, pain and psychiatric disorders have been linked in epidemiological and clinical terms, although the neurobiological mechanisms involved in this pathological bidirectional relationship remain unclear. Here we review the evidence obtained from animal models about the co-morbidity of pain and psychiatric disorders, placing special emphasis on the different dimensions of pain.
Collapse
Affiliation(s)
- Lidia Bravo
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Suárez-Pereira
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Berrocoso
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Neuropsychopharmacology and Psychobiology Research Group, Department of Psychology, University of Cádiz, 11510 Puerto Real, Cádiz, Spain.
| |
Collapse
|
23
|
Kremer M, Becker LJ, Barrot M, Yalcin I. How to study anxiety and depression in rodent models of chronic pain? Eur J Neurosci 2020; 53:236-270. [DOI: 10.1111/ejn.14686] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/06/2020] [Accepted: 01/14/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Mélanie Kremer
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Léa J. Becker
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Michel Barrot
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique Institut des Neurosciences Cellulaires et Intégratives Université de Strasbourg Strasbourg France
| |
Collapse
|
24
|
Polis AJ, Fitzgerald PJ, Hale PJ, Watson BO. Rodent ketamine depression-related research: Finding patterns in a literature of variability. Behav Brain Res 2019; 376:112153. [PMID: 31419519 PMCID: PMC6783386 DOI: 10.1016/j.bbr.2019.112153] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 12/23/2022]
Abstract
Discovering that the anesthetic drug ketamine has rapidly acting antidepressant effects in many individuals with major depression is one of the most important findings in clinical psychopharmacology in recent decades. The initial report of these effects in human subjects was based on a foundation of rodent preclinical studies carried out in the 1990s, and subsequent investigation has included both further studies in individuals with depression, as well as reverse translational experiments in animal models, especially rodents. While there is general agreement in the rodent literature that ketamine has rapidly-acting, and generally sustained, antidepressant-like properties, there are also points of contention across studies, including the precise mechanism of action of this drug. In this review, we briefly summarize prominent yet variable findings regarding the mechanism of action. We also discuss a combination of similarities and variances in the rodent literature in the antidepressant-like effects of ketamine as a function of dose, species and strain, test, stressor, and presumably sex of the experimenter. We then present previously unpublished mouse strain comparison data suggesting that subanesthetic ketamine does not have robust antidepressant-like properties in unstressed animals, and may actually promote depression-like behavior, in contrast to widely reported findings. We conclude that the data best support the notion of ketamine action principally via NMDA receptor antagonism, transiently boosting glutamatergic (and possibly other) signaling in diverse brain circuits. We also suggest that future studies should address in greater detail the extent to which antidepressant-like properties of this drug are stress-sensitive, in an effort to better model major depression present in humans.
Collapse
Affiliation(s)
- Andrew J Polis
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-5720, United States of America
| | - Paul J Fitzgerald
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-5720, United States of America
| | - Pho J Hale
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-5720, United States of America
| | - Brendon O Watson
- University of Michigan, Department of Psychiatry, Ann Arbor, MI 48109-5720, United States of America.
| |
Collapse
|
25
|
Abstract
OBJECTIVE In the context of the current opioid epidemic, there has been a renewed interest in the use of ketamine as an analgesic agent. METHODS We reviewed ketamine analgesia. RESULTS Ketamine is well-known as an antagonist for N-methyl-D-aspartate receptors. In addition, it can regulate the function of opioid receptors and sodium channels. Ketamine also increases signaling through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. These myriad of molecular and cellular mechanisms are responsible for a number of pharmacological functions including pain relief and mood regulation. Clinically, a number of studies have investigated the role of ketamine in the setting of acute and chronic pain, and there is evidence that ketamine can provide analgesia in a variety of pain syndromes. DISCUSSION In this review, we examined basic mechanisms of ketamine and its current clinical use and potential novel use in pain management.
Collapse
|
26
|
Ma JH, Wang SY, Yu HY, Li DY, Luo SC, Zheng SS, Wan LF, Duan KM. Prophylactic use of ketamine reduces postpartum depression in Chinese women undergoing cesarean section ✰. Psychiatry Res 2019; 279:252-258. [PMID: 31147085 DOI: 10.1016/j.psychres.2019.03.026] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/15/2019] [Accepted: 03/15/2019] [Indexed: 01/02/2023]
Abstract
This study aimed to explore the effect of prophylactic ketamine administration on postpartum depression in Chinese woman undergoing cesarean section. This randomized controlled study included 654 Chinese women undergoing cesarean section. At 10 min after child birth, patients in the ketamine group were given 0.5 mg/kg ketamine, whereas patients in the control group received standard postpartum care. At the end of operation, all patients were armed with a patient-controlled intravenous analgesia device. The primary outcome was the prevalence of postpartum depression (PPD), as assessed by the Edinburgh Postnatal Depression Scale (EPDS), and the secondary outcomes included the safety assessment and the Numerical Rating Scale (NRS) of postoperative pain. The prevalence of postpartum blues and postpartum depression were significantly lower in the ketamine group than in the control group. Logistic analysis showed that ketamine administration protected against postpartum depression, and PPD-associated risk factors included stress during pregnancy, antenatal depressive symptom and antenatal suicidal ideation. In addition, the antidepressive effect of prophylactic ketamine was stronger in mothers with a history of moderate stress during pregnancy, antenatal depressive symptom and antenatal suicidal ideation. Our findings suggest that ketamine functions as a prophylactic agent against PPD.
Collapse
Affiliation(s)
- Jia-Hui Ma
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Sai-Ying Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China; Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha 410078, PR China
| | - He-Ya Yu
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Dan-Yang Li
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Shi-Chao Luo
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Shan-Shan Zheng
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | - Li-Fei Wan
- People's Hospital of Ningxiang City, Ningxiang 410600, PR China
| | - Kai-Ming Duan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, PR China.
| |
Collapse
|
27
|
Erdinc M, Uyar E, Kelle I, Akkoc H. Anti-nociceptive effects of low dose ketamine in mice may be mediated by the serotonergic systems. PSYCHIAT CLIN PSYCH 2019. [DOI: 10.1080/24750573.2019.1605665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Meral Erdinc
- Department of Medical Pharmacology, Dicle University, Diyarbakir, Turkey
| | - Emre Uyar
- Department of Medical Pharmacology, Dicle University, Diyarbakir, Turkey
| | - Ilker Kelle
- Department of Medical Pharmacology, Dicle University, Diyarbakir, Turkey
| | - Hasan Akkoc
- Department of Medical Pharmacology, Dicle University, Diyarbakir, Turkey
| |
Collapse
|
28
|
Subanesthetic Dose of Ketamine Improved CFA-induced Inflammatory Pain and Depression-like Behaviors Via Caveolin-1 in Mice. J Neurosurg Anesthesiol 2019; 32:359-366. [DOI: 10.1097/ana.0000000000000610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Abstract
Patients frequently come to the emergency department for pain. For decades, ketamine has been used in the emergency department for procedural sedation but is now receiving attention as a potential alternative to opioids because of its unique analgesic effects. Additionally, ketamine's dissociative properties have made it a popular choice for sedating profoundly agitated patients. In this narrative review, these new roles for ketamine in the emergency department are discussed.
Collapse
Affiliation(s)
- Sophia Sheikh
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, 655 West 8th Street, Jacksonville, FL, 32209, USA.
| | - Phyllis Hendry
- Department of Emergency Medicine, University of Florida College of Medicine-Jacksonville, 655 West 8th Street, Jacksonville, FL, 32209, USA
| |
Collapse
|
30
|
Thompson SJ, Pitcher MH, Stone LS, Tarum F, Niu G, Chen X, Kiesewetter DO, Schweinhardt P, Bushnell MC. Chronic neuropathic pain reduces opioid receptor availability with associated anhedonia in rat. Pain 2019; 159:1856-1866. [PMID: 29794614 PMCID: PMC6095806 DOI: 10.1097/j.pain.0000000000001282] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Supplemental Digital Content is Available in the Text. Chronic pain reduces opioid receptor expression in the rat striatum, where the correlation between receptor expression and anhedonia may represent a molecular substrate for comorbid depression. The opioid system plays a critical role in both the experience and management of pain. Although acute activation of the opioid system can lead to pain relief, the effects of chronic pain on the opioid system remain opaque. Cross-sectional positron emission tomography (PET) studies show reduced availability of brain opioid receptors in patients with chronic pain but are unable to (1) determine whether these changes are due to the chronic pain itself or due to preexisting or medication-induced differences in the endogenous opioid system, and (2) identify the neurobiological substrate of reduced opioid receptor availability. We investigated these possibilities using a well-controlled longitudinal study design in rat. Using [18F]-FDPN-PET in either sham rats (n = 17) or spared nerve injury rats (n = 17), we confirmed reduced opioid receptor availability in the insula, caudate–putamen, and motor cortex of nerve injured rats 3 months after surgery, indicating that painful neuropathy altered the endogenous opioid system. Immunohistochemistry showed reduced expression of the mu-opioid receptor, MOR1, in the caudate–putamen and insula. Neither the opioid peptide enkephalin nor the neuronal marker NeuN differed between groups. In nerve-injured animals, sucrose preference, a measure of anhedonia/depression-like behavior, positively correlated with PET opioid receptor availability and MOR1-immunoreactivity in the caudate–putamen. These findings provide new evidence that the altered supraspinal opioid receptor availability observed in human patients with chronic pain may be a direct result of chronic pain. Moreover, reduced opioid receptor availability seems to reflect decreased receptor expression, which may contribute to pain-induced depression.
Collapse
Affiliation(s)
- Scott J Thompson
- Division of Intramural Research, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States.,Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Mark H Pitcher
- Division of Intramural Research, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Laura S Stone
- Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Farid Tarum
- Division of Intramural Research, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Gang Niu
- Division of Intramural Research, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, United States
| | - Xiaoyuan Chen
- Division of Intramural Research, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, United States
| | - Dale O Kiesewetter
- Division of Intramural Research, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, United States
| | | | - M Catherine Bushnell
- Division of Intramural Research, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
31
|
Humo M, Lu H, Yalcin I. The molecular neurobiology of chronic pain-induced depression. Cell Tissue Res 2019; 377:21-43. [PMID: 30778732 DOI: 10.1007/s00441-019-03003-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/01/2019] [Indexed: 12/18/2022]
Abstract
The increasing number of individuals with comorbidities poses an urgent need to improve the management of patients with multiple co-existing diseases. Among these comorbidities, chronic pain and mood disorders, two long-lasting disabling conditions that significantly reduce the quality of life, could be cited first. The recent development of animal models accelerated the studies focusing on the underlying mechanisms of the chronic pain and depression/anxiety comorbidity. This review provides an overview of clinical and pre-clinical studies performed over the past two decades addressing the molecular aspects of the comorbid relationship of chronic pain and depression. We thus focused on the studies that investigated the molecular characteristics of the comorbid relationship between chronic pain and mood disorders, especially major depressive disorders, from the genetic and epigenetic point of view to key neuromodulators which have been shown to play an important role in this comorbidity.
Collapse
Affiliation(s)
- Muris Humo
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France
| | - Han Lu
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France.,Faculty of Biology and Bernstein Center Freiburg, University of Freiburg, D-79104, Freiburg, Germany
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France.
| |
Collapse
|
32
|
Zhou Y, Zheng W, Liu W, Wang C, Zhan Y, Li H, Chen L, Li M, Ning Y. Antidepressant effect of repeated ketamine administration on kynurenine pathway metabolites in patients with unipolar and bipolar depression. Brain Behav Immun 2018; 74:205-212. [PMID: 30213652 DOI: 10.1016/j.bbi.2018.09.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/16/2018] [Accepted: 09/09/2018] [Indexed: 01/23/2023] Open
Abstract
Ketamine has rapid antidepressant effects on treatment-resistant depression, but the biological mechanism underpinning this effect is less clear. Our aims were to examine whether kynurenine pathway metabolites were altered by six infusions of ketamine and whether these biological factors could act as potential biomarkers to predict ketamine's antidepressant effects. Six intravenous infusions of ketamine (0.5 mg/kg) were administered to 84 patients with unipolar and bipolar depression over a 12-d period. Symptom severity and response were assessed using the Montgomery-Asberg Scale (MADRS), and blood samples were collected at baseline and 24 h following the first infusion and at 24 h and 14 d after the sixth infusion (24 h, 13 d and 26 d). Blood samples from sixty healthy controls were collected for comparison with samples from the patients. Serum concentrations of tryptophan (TRP), kynurenine (KYN) and kynurenic acid (KYNA) were measured by the liquid chromatography-tandem mass spectrometry method. At baseline, serum levels of TRP and KYNA and the KYNA/KYN ratio were lower and the KYN/TRP ratio was greater in depressed patients than in healthy controls. Overall, fifty (59.5%) patients responded to ketamine at 13 d. Ketamine responders had a greater KYNA level and KYNA/KYN ratio than nonresponders at 24 h and 13 d (all P < 0.05). Elevations in the KYNA levels and KYNA/KYN ratio at 24 h were significantly associated with reductions in MADRS scores at 24 h, 13 d and 26 d in the linear regression analysis (all P < 0.05). Our results showed a possible involvement of the kynurenine pathway in the rapid antidepressant effect of ketamine. Early changes in serum KYNA levels and the KYNA/KYN ratio could be potential predictors of antidepressanteffects of repeated ketamine administration.
Collapse
Affiliation(s)
- Yanling Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Wei Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Weijian Liu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Chengyu Wang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Yanni Zhan
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Hanqiu Li
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Lijian Chen
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Mingding Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China.
| |
Collapse
|
33
|
Pitcher MH, Tarum F, Lehmann M, Bushnell MC. Persistent inflammatory pain alters sexually-motivated behavior in male rats. Behav Brain Res 2018; 356:380-389. [PMID: 30205121 PMCID: PMC7485009 DOI: 10.1016/j.bbr.2018.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 02/06/2023]
Abstract
Urine from pro-œstrus female rodents evokes increased levels of sexually-motivated behaviors in males, including sniffing and scent marking of the urine spot as well as activation of brain reward regions. Stressors such as social defeat can adversely impact urine scent marking behavior in male rodents, an effect that can be mitigated with anti-depressant drugs. Persistent pain is also known to be a potent stressor, producing elevated levels of plasma corticosterone as well as reduced sucrose preference and reduced social interaction. However, the effect of persistent pain on sexually-motivated behavior is unknown. Here, we compared urine scent marking behavior in male rats for up to 3 weeks following intra-articular injection of Complete Freund’s Adjuvant (CFA) or sham injection. CFA-injected rats exhibited profound and ongoing deficits in static weight bearing capacity. CFA-induced persistent inflammatory pain increased plasma corticosterone levels and reduced urine scent marking behavior in male rats. Moreover, while the vast majority of injured rats showed decreased urine scent marking preference for the pro-œstrus female urine spot, male rats with higher baseline scent marking preference also exhibited higher post-injury scent marking preference, more sniffing behavior and lower levels of plasma corticosterone, compared to those with lower baseline scent marking preference. Overall, scent marking behavior may be an ethologically relevant behavioral predictor of persistent pain-induced stress in rats, representing a novel translational approach to understanding chronic pain comorbidities.
Collapse
Affiliation(s)
- Mark Henry Pitcher
- Pain and Integrative Neuroscience Laboratory, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, 20892, United States.
| | - Farid Tarum
- Pain and Integrative Neuroscience Laboratory, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Michael Lehmann
- Section on Functional Neuroanatomy, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, United States
| | - M Catherine Bushnell
- Pain and Integrative Neuroscience Laboratory, National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, 20892, United States
| |
Collapse
|
34
|
Michelet D, Brasher C, Horlin AL, Bellon M, Julien-Marsollier F, Vacher T, Pontone S, Dahmani S. Ketamine for chronic non-cancer pain: A meta-analysis and trial sequential analysis of randomized controlled trials. Eur J Pain 2017; 22:632-646. [DOI: 10.1002/ejp.1153] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2017] [Indexed: 01/17/2023]
Affiliation(s)
- D. Michelet
- Department of Anaesthesia and Intensive Care; Robert Debre University Hospital; Paris France
- Paris Diderot University; France
| | - C. Brasher
- Department of Anaesthesia and Pain Management; Royal Children's Hospital; Melbourne Australia
| | - A.-L. Horlin
- Department of Anaesthesia and Intensive Care; Robert Debre University Hospital; Paris France
- Paris Diderot University; France
| | - M. Bellon
- Department of Anaesthesia and Intensive Care; Robert Debre University Hospital; Paris France
- Paris Diderot University; France
| | - F. Julien-Marsollier
- Department of Anaesthesia and Intensive Care; Robert Debre University Hospital; Paris France
- Paris Diderot University; France
| | - T. Vacher
- Department of Anaesthesia and Intensive Care; Robert Debre University Hospital; Paris France
- Paris Diderot University; France
| | - S. Pontone
- Paris Diderot University; France
- Department of Palliative Care and Pain Management; Robert Debre University Hospital; Paris France
| | - S. Dahmani
- Department of Anaesthesia and Intensive Care; Robert Debre University Hospital; Paris France
- Paris Diderot University; France
- DHU PROTECT; INSERM U1141; Robert Debre University Hospital; Paris France
| |
Collapse
|
35
|
Xie ZM, Wang XM, Xu N, Wang J, Pan W, Tang XH, Zhou ZQ, Hashimoto K, Yang JJ. Alterations in the inflammatory cytokines and brain-derived neurotrophic factor contribute to depression-like phenotype after spared nerve injury: improvement by ketamine. Sci Rep 2017; 7:3124. [PMID: 28600519 PMCID: PMC5466642 DOI: 10.1038/s41598-017-03590-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/02/2017] [Indexed: 12/17/2022] Open
Abstract
Although pain is frequently accompanied with depression, little is known about the risk factors contributing to individual differences to the comorbidity of pain and depression. In this study, we examined whether cytokines and brain-derived neurotrophic factor (BDNF) might contribute to the individual differences in the development of neuropathic pain-induced depression. Rats were randomly subjected to spared nerved ligation (SNI) or sham surgery. The SNI rats were divided into two groups by the data from depression-related behavioral tests. Rats with depression-like phenotype displayed higher levels of pro-inflammatory cytokines (e.g., interleukin (IL)-1β, IL-6) as well as imbalance of pro/anti-inflammatory cytokines compared with rats without depression-like phenotype and sham-operated rats. Levels of BDNF in the prefrontal cortex of rats with depression-like phenotype were lower than those of rats without depression-like phenotype and sham-operated rats. A single dose of ketamine ameliorated depression-like behaviors in the rats with depression-like phenotype. Interestingly, higher serum levels of IL-1β and IL-6 in the rat with depression-like phenotype were normalized after a single dose of ketamine. These findings suggest that alterations in the inflammatory cytokines and BDNF might contribute to neuropathic pain-induced depression, and that serum cytokines may be predictable biomarkers for ketamine's antidepressant actions.
Collapse
Affiliation(s)
- Ze-Min Xie
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Laboratory of Anesthetic and Analgesia Application Technology, Xuzhou Medicine University, Xuzhou, China
| | - Xing-Ming Wang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Ning Xu
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Laboratory of Anesthetic and Analgesia Application Technology, Xuzhou Medicine University, Xuzhou, China
| | - Jing Wang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Wei Pan
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xiao-Hui Tang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Zhi-Qiang Zhou
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan.
| | - Jian-Jun Yang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Laboratory of Anesthetic and Analgesia Application Technology, Xuzhou Medicine University, Xuzhou, China.
| |
Collapse
|