1
|
Raja SM, Guptill JT, Mack M, Peterson M, Byard S, Twieg R, Jordan L, Rich N, Castledine R, Bourne S, Wilmshurst M, Oxendine S, Avula SG, Zuleta H, Quigley P, Lawson S, McQuaker SJ, Ahmadkhaniha R, Appelbaum LG, Kowalski K, Barksdale CT, Gufford BT, Awan A, Sancho AR, Moore MC, Berrada K, Cogan GB, DeLaRosa J, Radcliffe J, Pao M, Kennedy M, Lawrence Q, Goldfeder L, Amanfo L, Zanos P, Gilbert JR, Morris PJ, Moaddel R, Gould TD, Zarate CA, Thomas CJ. A Phase 1 Assessment of the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of (2R,6R)-Hydroxynorketamine in Healthy Volunteers. Clin Pharmacol Ther 2024; 116:1314-1324. [PMID: 39054770 PMCID: PMC11479831 DOI: 10.1002/cpt.3391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
(R,S)-Ketamine (ketamine) is a dissociative anesthetic that also possesses analgesic and antidepressant activity. Undesirable dissociative side effects and misuse potential limit expanded use of ketamine in several mental health disorders despite promising clinical activity and intensifying medical need. (2R,6R)-Hydroxynorketamine (RR-HNK) is a metabolite of ketamine that lacks anesthetic and dissociative activity but maintains antidepressant and analgesic activity in multiple preclinical models. To enable future assessments in selected human indications, we report the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of RR-HNK in a Phase 1 study in healthy volunteers (NCT04711005). A six-level single-ascending dose (SAD) (0.1-4 mg/kg) and a two-level multiple ascending dose (MAD) (1 and 2 mg/kg) study was performed using a 40-minute IV administration emulating the common practice for ketamine administration for depression. Safety assessments showed RR-HNK possessed a minimal adverse event profile and no serious adverse events at all doses examined. Evaluations of dissociation and sedation demonstrated that RR-HNK did not possess anesthetic or dissociative characteristics in the doses examined. RR-HNK PK parameters were measured in both the SAD and MAD studies and exhibited dose-proportional increases in exposure. Quantitative electroencephalography (EEG) measurements collected as a PD parameter based on preclinical findings and ketamine's established effect on gamma-power oscillations demonstrated increases of gamma power in some participants at the lower/mid-range doses examined. Cerebrospinal fluid examination confirmed RR-HNK exposure within the central nervous system (CNS). Collectively, these data demonstrate RR-HNK is well tolerated with an acceptable PK profile and promising PD outcomes to support the progression into Phase 2.
Collapse
Affiliation(s)
- Shruti M. Raja
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jeffrey T. Guptill
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
- Argenx BV, 9052 Gent, Belgium
| | - Michelle Mack
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Stephen Byard
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | - Robert Twieg
- Labcorp Bioanalytical Services, Indianapolis, IN, 46214, USA
| | - Lynn Jordan
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | | | - Samuel Bourne
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | | | - Sarah Oxendine
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Helen Zuleta
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Paul Quigley
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | - Sheila Lawson
- Quotient Sciences, Alnwick, Northumberland, NE66 2DH, England
| | | | - Reza Ahmadkhaniha
- National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 22124, USA
| | - Lawrence G. Appelbaum
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kevin Kowalski
- Labcorp Bioanalytical Services, Indianapolis, IN, 46214, USA
| | | | - Brandon T. Gufford
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Asaad Awan
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alfredo R. Sancho
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Max C. Moore
- Drug Discovery and Development Program, Frederick National Laboratory, Fredrick, MD, 21701, USA
| | - Karim Berrada
- Drug Discovery and Development Program, Frederick National Laboratory, Fredrick, MD, 21701, USA
| | - Gregory B. Cogan
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jesse DeLaRosa
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jeanne Radcliffe
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maryland Pao
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | - Lisa Goldfeder
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Leslie Amanfo
- Office of the Director, Intramural Research Program, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia, 2109, Cyprus
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, 20850, USA
| | - Ruin Moaddel
- National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 22124, USA
| | - Todd D. Gould
- Departments of Psychiatry, Pharmacology, and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Baltimore Veterans Affairs Medical Center, Veterans Affairs Maryland Health Care System, Baltimore, MD, 21201, USA
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville MD, 20850, USA
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
2
|
Gomes I, Gupta A, Margolis EB, Fricker LD, Devi LA. Ketamine and Major Ketamine Metabolites Function as Allosteric Modulators of Opioid Receptors. Mol Pharmacol 2024; 106:240-252. [PMID: 39187388 PMCID: PMC11493337 DOI: 10.1124/molpharm.124.000947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
Ketamine is a glutamate receptor antagonist that was developed over 50 years ago as an anesthetic agent. At subanesthetic doses, ketamine and some metabolites are analgesics and fast-acting antidepressants, presumably through targets other than glutamate receptors. We tested ketamine and its metabolites for activity as allosteric modulators of opioid receptors expressed as recombinant receptors in heterologous systems and with native receptors in rodent brain; signaling was examined by measuring GTP binding, β-arrestin recruitment, MAPK activation, and neurotransmitter release. Although micromolar concentrations of ketamine alone had weak agonist activity at μ opioid receptors, the combination of submicromolar concentrations of ketamine with endogenous opioid peptides produced robust synergistic responses with statistically significant increases in efficacies. All three opioid receptors (μ, δ, and κ) showed synergism with submicromolar concentrations of ketamine and either methionine-enkephalin (Met-enk), leucine-enkephalin (Leu-enk), and/or dynorphin A17 (Dyn A17), albeit the extent of synergy was variable between receptors and peptides. S-ketamine exhibited higher modulatory effects compared with R-ketamine or racemic ketamine, with ∼100% increase in efficacy. Importantly, the ketamine metabolite 6-hydroxynorketamine showed robust allosteric modulatory activity at μ opioid receptors; this metabolite is known to have analgesic and antidepressant activity but does not bind to glutamate receptors. Ketamine enhanced potency and efficacy of Met-enkephalin signaling both in mouse midbrain membranes and in rat ventral tegmental area neurons as determined by electrophysiology recordings in brain slices. Taken together, these findings support the hypothesis that some of the therapeutic effects of ketamine and its metabolites are mediated by directly engaging the endogenous opioid system. SIGNIFICANCE STATEMENT: This study found that ketamine and its major biologically active metabolites function as potent allosteric modulators of μ, δ, and κ opioid receptors, with submicromolar concentrations of these compounds synergizing with endogenous opioid peptides, such as enkephalin and dynorphin. This allosteric activity may contribute to ketamine's therapeutic effectiveness for treating acute and chronic pain and as a fast-acting antidepressant drug.
Collapse
Affiliation(s)
- Ivone Gomes
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| | - Achla Gupta
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| | - Elyssa B Margolis
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| | - Lloyd D Fricker
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| | - Lakshmi A Devi
- Departments of Pharmacological Sciences (I.G., A.G., L.A.D.) and Psychiatry (L.A.D.), and Nash Family Department of Neuroscience (L.A.D.), Icahn School of Medicine at Mount Sinai, New York, New York; UCSF Weill Institute for Neurosciences, Department of Neurology, Neuroscience Graduate Program, University of California, San Francisco, California (E.B.M.); and Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York (L.D.F.)
| |
Collapse
|
3
|
Das V, Basovich MB, Thomas CJ, Kroin JS, Buvanendran A, McCarthy RJ. A Pharmacological Evaluation of the Analgesic Effect and Hippocampal Protein Modulation of the Ketamine Metabolite (2R,6R)-Hydroxynorketamine in Murine Pain Models. Anesth Analg 2024; 138:1094-1106. [PMID: 37319016 PMCID: PMC10721716 DOI: 10.1213/ane.0000000000006590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
BACKGROUND The ketamine metabolite (2R,6R)-hydroxynorketamine ([2R,6R]-HNK) has analgesic efficacy in murine models of acute, neuropathic, and chronic pain. The purpose of this study was to evaluate the α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) dependence of (2R,6R)-HNK analgesia and protein changes in the hippocampus in murine pain models administered (2R,6R)-HNK or saline. METHODS All mice were CD-1 IGS outbred mice. Male and female mice underwent plantar incision (PI) (n = 60), spared nerve injury (SNI) (n = 64), or tibial fracture (TF) (n = 40) surgery on the left hind limb. Mechanical allodynia was assessed using calibrated von Frey filaments. Mice were randomized to receive saline, naloxone, or the brain-penetrating AMPA blocker (1,2,3,4-Tetrahydro-6-nitro-2,3-dioxobenzo [f]quinoxaline-7-sulfonamide [NBQX]) before (2R,6R)-HNK 10 mg/kg, and this was repeated for 3 consecutive days. The area under the paw withdrawal threshold by time curve for days 0 to 3 (AUC 0-3d ) was calculated using trapezoidal integration. The AUC 0-3d was converted to percent antiallodynic effect using the baseline and pretreatment values as 0% and 100%. In separate experiments, a single dose of (2R,6R)-HNK 10 mg/kg or saline was administered to naive mice (n = 20) and 2 doses to PI (n = 40), SNI injury (n = 40), or TF (n = 40) mice. Naive mice were tested for ambulation, rearing, and motor strength. Immunoblot studies of the right hippocampal tissue were performed to evaluate the ratios of glutamate ionotropic receptor (AMPA) type subunit 1 (GluA1), glutamate ionotropic receptor (AMPA) type subunit 2 (GluA2), phosphorylated voltage-gated potassium channel 2.1 (p-Kv2.1), phosphorylated-calcium/calmodulin-dependent protein kinase II (p-CaMKII), brain-derived neurotrophic factor (BDNF), phosphorylated protein kinase B (p-AKT), phosphorylated extracellular signal-regulated kinase (p-ERK), CXC chemokine receptor 4 (CXCR4), phosphorylated eukaryotic translation initiation factor 2 subunit 1 (p-EIF2SI), and phosphorylated eukaryotic translation initiation factor 4E (p-EIF4E) to glyceraldehyde 3-phosphate dehydrogenase (GAPDH). RESULTS No model-specific gender difference in antiallodynic responses before (2R,6R)-HNK administration was observed. The antiallodynic AUC 0-3d of (2R,6R)-HNK was decreased by NBQX but not with pretreatment with naloxone or saline. The adjusted mean (95% confidence interval [CI]) antiallodynic effect of (2R,6R)-HNK in the PI, SNI, and TF models was 40.7% (34.1%-47.3%), 55.1% (48.7%-61.5%), and 54.7% (46.5%-63.0%), greater in the SNI, difference 14.3% (95% CI, 3.1-25.6; P = .007) and TF, difference 13.9% (95% CI, 1.9-26.0; P = .019) compared to the PI model. No effect of (2R,6R)-HNK on ambulation, rearing, or motor coordination was observed. Administration of (2R,6R)-HNK was associated with increased GluA1, GluA2, p-Kv2.1, and p-CaMKII and decreased BDNF ratios in the hippocampus, with model-specific variations in proteins involved in other pain pathways. CONCLUSIONS (2R,6R)-HNK analgesia is AMPA-dependent, and (2R,6R)-HNK affected glutamate, potassium, calcium, and BDNF pathways in the hippocampus. At 10 mg/kg, (2R,6R)-HNK demonstrated a greater antiallodynic effect in models of chronic compared with acute pain. Protein analysis in the hippocampus suggests that AMPA-dependent alterations in BDNF-TrkB and Kv2.1 pathways may be involved in the antiallodynic effect of (2R,6R)-HNK.
Collapse
Affiliation(s)
- Vaskar Das
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL 60612
| | - Michael B. Basovich
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL 60612
| | - Craig J. Thomas
- Division of Preclinical Innovation, Chemistry Technologies, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Jeffrey S. Kroin
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL 60612
| | | | - Robert J McCarthy
- Department of Anesthesiology, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|
4
|
Hillhouse TM, Partridge KJ, Garrett PI, Honeycutt SC, Porter JH. Effects of (2R,6R)-hydroxynorketamine in assays of acute pain-stimulated and pain-depressed behaviors in mice. PLoS One 2024; 19:e0301848. [PMID: 38640139 PMCID: PMC11029659 DOI: 10.1371/journal.pone.0301848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/22/2024] [Indexed: 04/21/2024] Open
Abstract
Ketamine has been shown to produce analgesia in various acute and chronic pain states; however, abuse liability concerns have limited its utility. The ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) has been shown to produce antidepressant-like effects similar to ketamine without abuse liability concerns. (2R,6R)-HNK produces sustained analgesia in models of chronic pain, but has yet to be evaluated in models of acute pain. The present study evaluated the efficacy of acute (2R,6R)-HNK administration (one injection) in assays of pain-stimulated (52- and 56-degree hot plate test and acetic acid writhing) and pain-depressed behavior (locomotor activity and rearing) in male and female C57BL/6 mice. In assays of pain-stimulated behaviors, (2R,6R)-HNK (1-32 mg/kg) failed to produce antinociception in the 52- and 56-degree hot plate and acetic acid writhing assays. In assays of pain-depressed behaviors, 0.56% acetic acid produced a robust depression of locomotor activity and rearing that was not blocked by pretreatment of (2R,6R)-HNK (3.2-32 mg/kg). The positive controls morphine (hot plate test) and ketoprofen (acetic acid writhing, locomotor activity, and rearing) blocked pain-stimulated and pain-depressed behaviors. Finally, the effects of intermittent (2R,6R)-HNK administration were evaluated in 52-degree hot plate and pain-depressed locomotor activity and rearing. Intermittent administration of (2R,6R)-HNK also did not produce antinociceptive effects in the hot plate or pain-depressed locomotor activity assays. These results suggest that (2R,6R)-HNK is unlikely to have efficacy in treating acute pain; however, the efficacy of (2R,6R)-HNK in chronic pain states should continue to be evaluated.
Collapse
Affiliation(s)
- Todd M. Hillhouse
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin, United States of America
| | - Kaitlyn J. Partridge
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin, United States of America
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Patrick I. Garrett
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, United States of America
| | - Sarah C. Honeycutt
- Department of Psychology, University at Buffalo, Buffalo, New York, United States of America
| | - Joseph H. Porter
- Department of Psychology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Psychological Sciences, Northern Michigan, Marquette, Michigan, United States of America
| |
Collapse
|
5
|
Drinkuth CR, Lehane MJ, Sartor GC. The effects of (2R,6R)-hydroxynorketamine on oxycodone withdrawal and reinstatement. Drug Alcohol Depend 2023; 253:110987. [PMID: 37864957 PMCID: PMC10842506 DOI: 10.1016/j.drugalcdep.2023.110987] [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: 05/23/2023] [Revised: 09/13/2023] [Accepted: 09/30/2023] [Indexed: 10/23/2023]
Abstract
Despite the thousands of lives lost during the ongoing opioid crisis, a scarcity of new and effective clinical treatments for opioid use disorder (OUD) remains. To address this unmet need, some researchers have turned to dissociative and psychedelic drugs to treat multiple psychiatric conditions. In particular, low doses of ketamine have been shown to attenuate opioid withdrawal and drug use in clinical and preclinical studies. However, ketamine has misuse liability and dissociative side effects that may limit its widespread application as a treatment for OUD. More recently, (2R,6R)-hydroxynorketamine (HNK), a ketamine metabolite that lacks misuse potential, has gained attention for its effectiveness in depression and stress models. To uncover its role in OUD, we tested the time-dependent effects of (2R,6R)-HNK on oxycodone withdrawal and reinstatement of oxycodone conditioned place preference (CPP). In male and female oxycodone-dependent mice, we found that 24h pretreatment with (2R,6R)-HNK (10 or 30mg/kg, s.c.) reduced the frequency of withdrawal-like behaviors and global withdrawal scores during naloxone-precipitated withdrawal, whereas 1h pretreatment with (2R,6R)-HNK only reduced paw tremors and the sum of global withdrawal scores but not GWS Z-scores. In other experiments, both 1h and 24h pretreatment with (2R,6R)-HNK (30mg/kg, s.c.) blocked drug-induced reinstatement of oxycodone CPP. Finally, we found (2R,6R)-HNK (30mg/kg, sc) had no effect on locomotor activity and thigmotaxis. Together, these results indicate that acute (2R,6R)-HNK has efficacy in some preclinical models of OUD without producing locomotor or anxiety-like side effects.
Collapse
Affiliation(s)
- Caryssa R Drinkuth
- Department of Pharmaceutical Sciences, Connecticut Institute for the Brain and Cognitive Sciences (IBACS), University of Connecticut, Storrs, CT 06269, United States
| | - Michael J Lehane
- Department of Pharmaceutical Sciences, Connecticut Institute for the Brain and Cognitive Sciences (IBACS), University of Connecticut, Storrs, CT 06269, United States
| | - Gregory C Sartor
- Department of Pharmaceutical Sciences, Connecticut Institute for the Brain and Cognitive Sciences (IBACS), University of Connecticut, Storrs, CT 06269, United States.
| |
Collapse
|
6
|
Abstract
This paper is the forty-fifth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2022 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, USA.
| |
Collapse
|
7
|
Liu AR, Lin ZJ, Wei M, Tang Y, Zhang H, Peng XG, Li Y, Zheng YF, Tan Z, Zhou LJ, Feng X. The potent analgesia of intrathecal 2R, 6R-HNK via TRPA1 inhibition in LF-PENS-induced chronic primary pain model. J Headache Pain 2023; 24:141. [PMID: 37858040 PMCID: PMC10585932 DOI: 10.1186/s10194-023-01667-1] [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: 04/26/2023] [Accepted: 09/11/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Chronic primary pain (CPP) is an intractable pain of unknown cause with significant emotional distress and/or dysfunction that is a leading factor of disability globally. The lack of a suitable animal model that mimic CPP in humans has frustrated efforts to curb disease progression. 2R, 6R-hydroxynorketamine (2R, 6R-HNK) is the major antidepressant metabolite of ketamine and also exerts antinociceptive action. However, the analgesic mechanism and whether it is effective for CPP are still unknown. METHODS Based on nociplastic pain is evoked by long-term potentiation (LTP)-inducible high- or low-frequency electrical stimulation (HFS/LFS), we wanted to develop a novel CPP mouse model with mood and cognitive comorbidities by noninvasive low-frequency percutaneous electrical nerve stimulation (LF-PENS). Single/repeated 2R, 6R-HNK or other drug was intraperitoneally (i.p.) or intrathecally (i.t.) injected into naïve or CPP mice to investigate their analgesic effect in CPP model. A variety of behavioral tests were used to detect the changes in pain, mood and memory. Immunofluorescent staining, western blot, reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) and calcium imaging of in cultured dorsal root ganglia (DRG) neurons by Fluo-8-AM were used to elucidate the role and mechanisms of 2R, 6R-HNK in vivo or in vitro. RESULTS Intrathecal 2R, 6R-HNK, rather than intraperitoneal 2R, 6R-HNK or intrathecal S-Ketamine, successfully mitigated HFS-induced pain. Importantly, intrathecal 2R, 6R-HNK displayed effective relief of bilateral pain hypersensitivity and depressive and cognitive comorbidities in a dose-dependent manner in LF-PENS-induced CPP model. Mechanically, 2R, 6R-HNK markedly attenuated neuronal hyperexcitability and the upregulation of calcitonin gene-related peptide (CGRP), transient receptor potential ankyrin 1 (TRPA1) or vanilloid-1 (TRPV1), and vesicular glutamate transporter-2 (VGLUT2) in peripheral nociceptive pathway. In addition, 2R, 6R-HNK suppressed calcium responses and CGRP overexpression in cultured DRG neurons elicited by the agonists of TRPA1 or/and TRPV1. Strikingly, the inhibitory effects of 2R, 6R-HNK on these pain-related molecules and mechanical allodynia were substantially occluded by TRPA1 antagonist menthol. CONCLUSIONS In the newly designed CPP model, our findings highlighted the potential utility of intrathecal 2R, 6R-HNK for preventing and therapeutic modality of CPP. TRPA1-mediated uprgulation of CGRP and neuronal hyperexcitability in nociceptive pathways may undertake both unique characteristics and solving process of CPP.
Collapse
Affiliation(s)
- An-Ran Liu
- Department of Anesthesiology and Pain Clinic, First Affiliated Hospital of Sun Yat-Sen University, No.58, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Zhen-Jia Lin
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Ming Wei
- Department of Anesthesiology and Pain Clinic, First Affiliated Hospital of Sun Yat-Sen University, No.58, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Yuan Tang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Hui Zhang
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, No.466, Mid Xingang Road, Haizhu District, Guangzhou, 510317, China
| | - Xiang-Ge Peng
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Ying Li
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Yu-Fan Zheng
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China
| | - Zhi Tan
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China.
| | - Li-Jun Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University, No.74, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China.
| | - Xia Feng
- Department of Anesthesiology and Pain Clinic, First Affiliated Hospital of Sun Yat-Sen University, No.58, 2Nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, China.
| |
Collapse
|
8
|
Inhibition of Microglial GSK3β Activity Is Common to Different Kinds of Antidepressants: A Proposal for an In Vitro Screen to Detect Novel Antidepressant Principles. Biomedicines 2023; 11:biomedicines11030806. [PMID: 36979785 PMCID: PMC10045655 DOI: 10.3390/biomedicines11030806] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/17/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Depression is a major public health concern. Unfortunately, the present antidepressants often are insufficiently effective, whilst the discovery of more effective antidepressants has been extremely sluggish. The objective of this review was to combine the literature on depression with the pharmacology of antidepressant compounds, in order to formulate a conceivable pathophysiological process, allowing proposals how to accelerate the discovery process. Risk factors for depression initiate an infection-like inflammation in the brain that involves activation microglial Toll-like receptors and glycogen synthase kinase-3β (GSK3β). GSK3β activity alters the balance between two competing transcription factors, the pro-inflammatory/pro-oxidative transcription factor NFκB and the neuroprotective, anti-inflammatory and anti-oxidative transcription factor NRF2. The antidepressant activity of tricyclic antidepressants is assumed to involve activation of GS-coupled microglial receptors, raising intracellular cAMP levels and activation of protein kinase A (PKA). PKA and similar kinases inhibit the enzyme activity of GSK3β. Experimental antidepressant principles, including cannabinoid receptor-2 activation, opioid μ receptor agonists, 5HT2 agonists, valproate, ketamine and electrical stimulation of the Vagus nerve, all activate microglial pathways that result in GSK3β-inhibition. An in vitro screen for NRF2-activation in microglial cells with TLR-activated GSK3β activity, might therefore lead to the detection of totally novel antidepressant principles with, hopefully, an improved therapeutic efficacy.
Collapse
|
9
|
Yost JG, Browne CA, Lucki I. (2R,6R)-hydroxynorketamine (HNK) reverses mechanical hypersensitivity in a model of localized inflammatory pain. Neuropharmacology 2022; 221:109276. [PMID: 36198332 DOI: 10.1016/j.neuropharm.2022.109276] [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: 06/07/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 10/07/2022]
Abstract
The ketamine metabolite (2R,6R)-hydroxynorketamine, or (2R,6R)-HNK, was recently reported to evoke antinociception in response to a noxious thermal stimulus in healthy mice and reverse mechanical hypersensitivity in a murine model of neuropathic pain. This study reports the behavioral effects of (2R,6R)-HNK in male and female C57BL/6J mice exposed to a localized inflammatory pain condition and the broad pharmacological mechanism underlying this effect. Hind paw intraplantar injection of λ-carrageenan (CARR) caused inflammation and mechanical hypersensitivity in mice within 2 hours, lasting at least 48 hours. Intraperitoneal administration of (2R,6R)-HNK (10-30 mg/kg i.p.) 2 hours following CARR injection significantly reversed mechanical hypersensitivity within 1 hour in male and female mice, and the effect persisted for 24 hours following a single dose. The magnitude and timing of the analgesic effect of (2R,6R)-HNK were comparable to the non-steroidal anti-inflammatory drug carprofen. The reversal of hypersensitivity by (2R,6R)-HNK was blocked at 4 and 24 hours after administration by pretreatment with the AMPA receptor antagonist NBQX and was not accompanied by changes in locomotor activity. These findings reinforce the growing evidence supporting (2R,6R)-HNK as a novel analgesic in multiple preclinical pain models and further support an AMPAR-dependent mechanism of action. SIGNIFICANCE: The ketamine metabolite (2R,6R)-HNK reversed mechanical hypersensitivity associated with localized inflammation with onset less than one hour and duration greater than 24 hours in an effect comparable to the NSAID carprofen. Reversal of mechanical hypersensitivity by (2R,6R)-HNK is AMPAR-dependent.
Collapse
Affiliation(s)
- Jonathan G Yost
- Neuroscience Graduate Program, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Caroline A Browne
- Neuroscience Graduate Program, Uniformed Services University, Bethesda, MD, 20814, USA; Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Irwin Lucki
- Neuroscience Graduate Program, Uniformed Services University, Bethesda, MD, 20814, USA; Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, Bethesda, MD, 20814, USA; Department of Psychiatry, Uniformed Services University, Bethesda, MD, 20814, USA.
| |
Collapse
|