1
|
Chen J, Liang S, Li C, Li B, He M, Li K, Fu W, Li S, Mi H. Mitochondrial damage causes inflammation via cGAS-STING signaling in ketamine-induced cystitis. Inflamm Res 2025; 74:6. [PMID: 39762437 PMCID: PMC11703929 DOI: 10.1007/s00011-024-01973-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Mitochondrial dysfunction and damage can result in the release of mitochondrial DNA (mtDNA) into the cytoplasm, which subsequently activates the cGAS-STING pathway, promoting the onset of inflammatory diseases. Various factors, such as oxidative stress, viral infection, and drug toxicity, have been identified as inducers of mitochondrial damage. This study aims to investigate the role of mtDNA as a critical inflammatory mediator in the pathogenesis of ketamine (KET)-induced cystitis (KC) through the cGAS-STING pathway. METHODS To investigate the role of the cGAS-STING pathway in KET-induced cystitis, we assessed the expression of cGAS and STING in rats with KET cystitis. Additionally, we evaluated STING expression in conditionally deficient Simian Virus-transformed Human Uroepithelial Cell Line 1 (SV-HUC-1) cells in vitro. Morphological changes in mitochondria were examined using transmission electron microscopy. We measured intracellular reactive oxygen species (ROS) production through flow cytometry and immunofluorescence techniques. Furthermore, alterations in associated inflammatory factors and cytokines were quantified using real-time quantitative PCR with fluorescence detection. RESULTS We observed up-regulation of cGAS and STING expressions in the bladder tissue of rats in the KET group, stimulation with KET also led to increased cGAS and STING levels in SV-HUC-1 cells. Notably, the knockdown of STING inhibited the nuclear translocation of NF-κB p65 and IRF3, resulting in a decrease in the expression of inflammatory cytokines, including IL-6, IL-8, and CXCL10. Additionally, KET induced damage to the mitochondria of SV-HUC-1 cells, facilitating the release of mtDNA into the cytoplasm. This significant depletion of mtDNA inhibited the activation of cGAS-STING pathway, subsequently affecting the expression of NF-κB p65 and IRF3. Importantly, the reintroduction of mtDNA after STING knockdown partially restored the inflammatory response. CONCLUSION Our findings confirmed the activation of the cGAS-STING pathway in KC rats and revealed mitochondrial damage in vitro. These results highlight the involvement of the cGAS-STING pathway in the pathogenesis of KC, suggesting its potential as a therapeutic target for intervention.
Collapse
Affiliation(s)
- Jinji Chen
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Shengsheng Liang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Cheng Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Bowen Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Mingdong He
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Kezhen Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Weijin Fu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Shenghua Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Hua Mi
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| |
Collapse
|
2
|
Yin YY, Yan JZ, Wei QQ, Sun SR, Ding YQ, Zhang LM, Li YF. Serotonergic transmission plays differentiated roles in the rapid and sustained antidepressant-like effects of ketamine. Br J Pharmacol 2024; 181:4874-4889. [PMID: 39238235 DOI: 10.1111/bph.17324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND AND PURPOSE The emerging antidepressant effects of ketamine have inspired tremendous interest in its underlying neurobiological mechanisms, although the involvement of 5-HT in the antidepressant effects of ketamine remains unclear. EXPERIMENTAL APPROACH The chronic restraint stress procedure was performed to induce depression-like behaviours in mice. OFT, FST, TST, and NSFT tests were used to evaluate the antidepressant-like effects of ketamine. Tph2 knockout or depletion of 5-HT by PCPA and 5,7-DHT were used to manipulate the brain 5-HT system. ELISA and fibre photometry recordings were used to measure extracellular 5-HT levels in the brain. KEY RESULTS 60 min after injection, ketamine (10 mg·kg-1, i.p.) produced rapid antidepressant-like effects and increased brain 5-HT levels. After 24 h, ketamine significantly reduced immobility time in TST and FST tests and increased brain 5-HT levels, as measured by ELISA and fibre photometry recordings. The sustained (24 h) but not rapid (60 min) antidepressant-like effects of ketamine were abrogated by PCPA and 5,7-DHT, or by Tph2 knockout. Importantly, NBQX (10 mg·kg-1, i.p.), an AMPA receptor antagonist, significantly inhibited the effect of ketamine on brain 5-HT levels and abolished the sustained antidepressant-like effects of ketamine in naïve or CRS-treated mice. CONCLUSION AND IMPLICATIONS This study confirms the requirement of serotonergic neurotransmission for the sustained antidepressant-like effects of ketamine, which appears to involve AMPA receptors, and provides avenues to search for antidepressant pharmacological targets.
Collapse
Affiliation(s)
- Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Jiao-Zhao Yan
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Qian-Qian Wei
- School of Medicine, Nantong University, Nantong, China
| | - Si-Rui Sun
- Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yu-Qiang Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
- Beijing Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Chen H, Chen J, Lan J. Acute manipulation of Drd1 neurons in the prefrontal cortex bidirectionally regulates anxiety and depression-like behaviors. Neurosci Lett 2024; 832:137805. [PMID: 38705453 DOI: 10.1016/j.neulet.2024.137805] [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: 02/07/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND CONTEXT The medial prefrontal cortex (mPFC) has been implicated in modulating anxiety and depression. Manipulation of Drd1 neurons in the mPFC resulted in variable neuronal activity and, consequently, strikingly different behaviors. The acute regulation of anxiety- and depression-like behaviors by Drd1 neurons, a major neuronal subtype in the mPFC, has not yet been investigated. PURPOSE The purpose of this study was to investigate whether acute manipulation of Drd1 neurons in the mPFC affects anxiety- and depression-like behaviors. STUDY DESIGN Male Drd1-Cre mice were injected with an adeno-associated virus (AAV) expressing hM3DGq or hM4DGi. Clozapine-n-oxide (CNO, 1 mg/kg, i.p.) was injected 30 min before the behavioral tests. METHODS Male Drd1-Cre mice were injected with AAV-Ef1α-DIO-hM4DGi-mCherry-WPRE-pA, AAV-Ef1α-DIO-hM3DGq-mCherry-WPRE-pA or AAV-Ef1α-DIO-mCherry-WPRE-pA. Three weeks later, whole-cell recordings after CNO (5 μM) were applied to the bath were used to validate the functional expression of hM4DGi and hM3DGq. Four groups of mice underwent all the behavioral tests, and after each of the tests, the mice were allowed to rest for 3-4 days. CNO (1 mg/kg) was injected intraperitoneally 30 min before the behavior test. Anxiety-like behaviors were evaluated by the open field test (OFT), the elevated plus maze test (EPMT), and the novelty-suppressed feeding test (NSFT). Depression-like behaviors were evaluated by the sucrose preference test (SPT) and force swimming test (FST). For all experiments, coronal sections of the targeted brain area were used to confirm virus expression. RESULTS Whole-cell recordings from brain slices demonstrated that infusions of CNO (5 µM) into mPFC slices dramatically increased the firing activity of hM3DGq-mCherry+ neurons and abolished the firing activity of hM4DGi-mCherry+ neurons. Acute chemogenetic activation of Drd1 neurons in the mPFC increased the time spent in the central area in the OFT, increased the time spent in the open arms in the EMPT, decreased the latency to bite the food in the NSFT, increased the sucrose preference in the SPT, and decreased the immobility time in the FST. Acute chemogenetic inhibition of Drd1 neurons in the mPFC decreased the time spent in the central area in the OFT, decreased the time spent in the open arms in the EMPT, increased the latency to bite the food in the NSFT, decreased the sucrose preference in the SPT, and increased the immobility time in the FST. CONCLUSIONS The present study showed that acute activation of Drd1 neurons in the mPFC produced rapid anxiolytic- and antidepressant-like effects, and acute inhibition had the opposite effect, revealing that Drd1 neurons in the mPFC bidirectionally regulate anxiety- and depression-like behaviors. CLINICAL SIGNIFICANCE The findings of the present study regarding the acute effects of stimulating Drd1 neurons in the mPFC on anxiety and depression suggest that Drd1 neurons in the mPFC are a focus for the treatment of anxiety disorders and depression.
Collapse
Affiliation(s)
- He Chen
- Department of Neurosurgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Jun Chen
- Department of Neurosurgery, Nanning First People's Hospital, Nanning 530000, China; Phase I Clinical Trial Laboratory, Nanning First People's Hospital, Nanning 530000, China
| | - Jie Lan
- Department of Neurosurgery, Nanning First People's Hospital, Nanning 530000, China.
| |
Collapse
|
4
|
Cuttoli RDD, Sweis BM. Ketamine reverses stress-induced hypersensitivity to sunk costs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593597. [PMID: 38798536 PMCID: PMC11118454 DOI: 10.1101/2024.05.12.593597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
How mood interacts with information processing in the brain is thought to mediate the maladaptive behaviors observed in depressed individuals. However, the neural mechanisms underlying impairments in emotion-cognition interactions are poorly understood. This includes influencing the balance between how past-sensitive vs. future-looking one is during decision-making. Recent insights from the field of neuroeconomics offer novel approaches to study changes in such valuation processes in a manner that is biologically tractable and readily translatable across species. We recently discovered that rodents are sensitive to "sunk costs" - a feature of higher cognition previously thought to be unique to humans. The sunk costs bias describes the phenomenon in which an individual overvalues and escalates commitment to continuing an ongoing endeavor, even if suboptimal, as a function of irrecoverable past (sunk) losses - information that, according to classic economic theory, should be ignored. In the present study, mice were exposed to chronic social defeat stress paradigm, a well-established animal model used for the study of depression. Mice were then tested on our longitudinal neuroeconomic foraging task, Restaurant Row. We found mice exposed to this severe stressor displayed an increased sensitivity to sunk costs, without altering overall willingness to wait. Mice were then randomly assigned to receive a single intraperitoneal injection of either saline or ketamine (20 mg/kg). We discovered that stress-induced hypersensitivity to sunk costs was renormalized following a single dose of ketamine. Interestingly, in non-defeated mice, ketamine treatment completely abolished sunk cost sensitivity, causing mice to no longer value irrecoverable losses during re-evaluation decisions who instead based choices solely on the future investment required to obtain a goal. These findings suggest that the antidepressant effects of ketamine may be mediated in part through changes in the processing of past-sensitive information during on-going decision-making, reducing its weight as a potential source of cognitive dissonance that could modulate behavior and instead promoting more future-thinking behavior.
Collapse
|
5
|
Mitsi V, Ruiz A, Polizu C, Farzinpour Z, Ramakrishnan A, Serafini RA, Parise EM, Floodstrand M, Sial OK, Gaspari S, Tang CY, Nestler EJ, Schmidt EF, Shen L, Zachariou V. RGS4 Actions in Mouse Prefrontal Cortex Modulate Behavioral and Transcriptomic Responses to Chronic Stress and Ketamine. Mol Pharmacol 2024; 105:272-285. [PMID: 38351270 PMCID: PMC10949159 DOI: 10.1124/molpharm.123.000753] [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: 07/06/2023] [Accepted: 01/16/2024] [Indexed: 03/16/2024] Open
Abstract
The signal transduction protein, regulator of G protein signaling 4 (RGS4), plays a prominent role in physiologic and pharmacological responses by controlling multiple intracellular pathways. Our earlier work identified the dynamic but distinct roles of RGS4 in the efficacy of monoamine-targeting versus fast-acting antidepressants. Using a modified chronic variable stress (CVS) paradigm in mice, we demonstrate that stress-induced behavioral abnormalities are associated with the downregulation of RGS4 in the medial prefrontal cortex (mPFC). Knockout of RGS4 (RGS4KO) increases susceptibility to CVS, as mutant mice develop behavioral abnormalities as early as 2 weeks after CVS resting-state functional magnetic resonance imaging I (rs-fMRI) experiments indicate that stress susceptibility in RGS4KO mice is associated with changes in connectivity between the mediodorsal thalamus (MD-THL) and the mPFC. Notably, RGS4KO also paradoxically enhances the antidepressant efficacy of ketamine in the CVS paradigm. RNA-sequencing analysis of naive and CVS samples obtained from mPFC reveals that RGS4KO triggers unique gene expression signatures and affects several intracellular pathways associated with human major depressive disorder. Our analysis suggests that ketamine treatment in the RGS4KO group triggers changes in pathways implicated in synaptic activity and responses to stress, including pathways associated with axonal guidance and myelination. Overall, we show that reducing RGS4 activity triggers unique gene expression adaptations that contribute to chronic stress disorders and that RGS4 is a negative modulator of ketamine actions. SIGNIFICANCE STATEMENT: Chronic stress promotes robust maladaptation in the brain, but the exact intracellular pathways contributing to stress vulnerability and mood disorders have not been thoroughly investigated. In this study, the authors used murine models of chronic stress and multiple methodologies to demonstrate the critical role of the signal transduction modulator regulator of G protein signaling 4 in the medial prefrontal cortex in vulnerability to chronic stress and the efficacy of the fast-acting antidepressant ketamine.
Collapse
Affiliation(s)
- Vasiliki Mitsi
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Anne Ruiz
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Claire Polizu
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Zahra Farzinpour
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Randal A Serafini
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Eric M Parise
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Madeline Floodstrand
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Omar K Sial
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Sevasti Gaspari
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Cheuk Y Tang
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Eric F Schmidt
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| | - Venetia Zachariou
- Nash Family Department of Neuroscience and Friedman Brain Institute (V.M., A.Ru., C.P., A.Ra., R.A.S., E.M.P. M.F., S.G., E.J.N., L.S.) and BioMedical Engineering and Imaging Institute (C.Y.T.), Icahn School of Medicine at Mount Sinai, New York, New York; University of Crete, Department of Basic Sciences, Crete, Greece (V.M.); Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts (Z.F., R.A.S., V.Z.); Department of Psychology, Texas A&M University, College Station, Texas (O.K.S.); and Laboratory of Molecular Biology, Rockefeller University, New York, New York (E.F.S.)
| |
Collapse
|
6
|
Lineham A, Avila-Quintero VJ, Bloch MH, Dwyer J. Exploring Predictors of Ketamine Response in Adolescent Treatment-Resistant Depression. J Child Adolesc Psychopharmacol 2024; 34:73-79. [PMID: 38170185 PMCID: PMC11262580 DOI: 10.1089/cap.2023.0047] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Objective: Ketamine has proved effective as a rapid-acting antidepressant agent, but treatment is not effective for everyone (approximately a quarter to a half of patients). Some adult studies have begun to investigate predictors of ketamine's antidepressant response, but no studies have examined this in adolescents with depression. Methods: We conducted a secondary data analysis of adolescents who participated in a randomized, single-dose, midazolam-controlled crossover trial of ketamine for adolescents with treatment-resistant depression. We examined the relationship between 19 exploratory demographic and clinical variables and depression symptom improvement (using the Montgomery-Åsberg Depression Rating Scale [MADRS]) at 1 and 7 days postinfusion. Results: Subjects who had fewer medication trials of both antidepressant medications and augmentation treatments were more likely to experience depression symptom improvement with ketamine. Subjects with shorter duration of their current depressive episode were more likely to experience depression symptom improvement with ketamine. Subjects currently being treated with selective serotonin reuptake inhibitor medications, and not being treated with serotonin-norepinephrine reuptake inhibitor medications, also experienced greater symptom improvement with ketamine. When receiving the midazolam control, less severe depressive symptoms, as measured by the Children's Depression Rating Scale (CDRS) (but not MADRS), and a comorbid attention-deficit/hyperactivity disorder diagnosis were associated with increased response. Conclusions: Findings should be viewed as preliminary and exploratory given the small sample size and multiple secondary analyses. Identifying meaningful predictors of ketamine response is important to inform future therapeutic use of this compound, however, considerably more research is warranted before such clinical guidance is established. The trial was registered in clinicaltrials.gov with the identifier NCT02579928.
Collapse
Affiliation(s)
- Alice Lineham
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Michael H. Bloch
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Psychiatry and Yale School of Medicine, New Haven, Connecticut, USA
| | - Jennifer Dwyer
- Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
7
|
Ledesma-Corvi S, Jornet-Plaza J, Gálvez-Melero L, García-Fuster MJ. Novel rapid treatment options for adolescent depression. Pharmacol Res 2024; 201:107085. [PMID: 38309382 DOI: 10.1016/j.phrs.2024.107085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/13/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
There is an urgent need for novel fast-acting antidepressants for adolescent treatment-resistant depression and/or suicidal risk, since the selective serotonin reuptake inhibitors that are clinically approved for that age (i.e., fluoxetine or escitalopram) take weeks to work. In this context, one of the main research lines of our group is to characterize at the preclinical level novel approaches for rapid-acting antidepressants for adolescence. The present review summarizes the potential use in adolescence of non-pharmacological options, such as neuromodulators (electroconvulsive therapy and other innovative types of brain stimulation), as well as pharmacological options, including consciousness-altering drugs (mainly ketamine but also classical psychedelics) and cannabinoids (i.e., cannabidiol), with promising fast-acting responses. Following a brief analytical explanation of adolescent depression, we present a general introduction for each therapeutical approach together with the clinical evidence supporting its potential beneficial use in adolescence (mainly extrapolated from prior successful examples for adults), to then report recent and/or ongoing preclinical studies that will aid in improving the inclusion of these therapies in the clinic, by considering potential sex-, age-, and dose-related differences, and/or other factors that might affect efficacy or long-term safety. Finally, we conclude the review by providing future avenues to maximize treatment response, including the need for more clinical studies and the importance of designing and/or testing novel treatment options that are safe and fast-acting for adolescent depression.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Jordi Jornet-Plaza
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Laura Gálvez-Melero
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- Neuropharmacology Research Group, IUNICS, University of the Balearic Islands, Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain; Department of Medicine, University of the Balearic Islands, Palma, Spain.
| |
Collapse
|
8
|
Rengasamy M, Mathew S, Howland R, Griffo A, Panny B, Price R. Neural connectivity moderators and mechanisms of ketamine treatment among treatment-resistant depressed patients: a randomized controlled trial. EBioMedicine 2024; 99:104902. [PMID: 38141395 PMCID: PMC10788398 DOI: 10.1016/j.ebiom.2023.104902] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND Intravenous (IV) ketamine has emerged as a rapid and effective treatment for TRD. However, the specific neural mechanisms of ketamine's effects in humans remains unclear. Although neuroplasticity is implicated as a mechanism of action in animal models, relatively few randomized controlled trials (RCTs) in TRD patients have examined ketamine's impact on functional connectivity, a posited functional marker of neuroplasticity-particularly in the context of a mood-induction paradigm (termed miFC). METHODS 152 adults with TRD (63% female; 37% male) were randomly allocated to receive a single infusion of ketamine or saline in a 2:1 ratio. We examined changes in connectivity (from baseline to 24-h post-infusion) that differed by treatment, and whether clinical treatment response at 24-h post-infusion was uniquely related (among patients allocated to ketamine relative to saline) to (1) pre-treatment connectivity and (2) changes in connectivity. We examined both miFC and rsFC, using prefrontal cortex and limbic seed regions. We also conducted a multiverse analysis to examine findings most robust against analytic decisions. FINDINGS Across both miFC and rsFC, ketamine was associated with greater in prefrontal/limbic connectivity compared to saline, and lower baseline connectivity of limbic and prefrontal regions predicted greater treatment response in patients receiving ketamine. Greater connectivity increases in participants receiving ketamine was uniquely related to greater treatment response. In addition, certain findings were identified as being reproducible against different analytic decisions in multiverse analyses. INTERPRETATION Our findings identify specific neural connectivity patterns impacted by ketamine and were uniquely related to outcomes following ketamine (relative to saline). These findings generally support prominent neuroplasticity models of ketamine's therapeutic efficacy. These findings lay new groundwork for understanding how to enhance and optimize ketamine treatments and develop novel rapid-acting treatments for depression. FUNDING This research was supported by NIH grant R01MH113857 and by the Clinical and Translational Sciences Institute at the University of Pittsburgh (UL1-TR-001857).
Collapse
Affiliation(s)
- Manivel Rengasamy
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Sanjay Mathew
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA; Michael E. Debakey VA Medical Center, Houston, TX, USA; The Menninger Clinic, Houston, TX, USA
| | - Robert Howland
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Angela Griffo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin Panny
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebecca Price
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Awasthi N, Yadav R, Kumar D. Revealing metabolic path of Ketamine catalyzed by CYP450 via quantum mechanical approach. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
10
|
Kang MJY, Vazquez GH. Association between peripheral biomarkers and clinical response to IV ketamine for unipolar treatment-resistant depression: An open label study. J Affect Disord 2022; 318:331-337. [PMID: 36070831 DOI: 10.1016/j.jad.2022.08.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 08/01/2022] [Accepted: 08/20/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Major Depression is the leading cause of disability worldwide. A cohort of patients do not respond adequately to available antidepressants, leading to treatment-resistant depression (TRD). We evaluated the antidepressant efficacy of an acute intravenous ketamine treatment (0.5 mg/kg) for patients with unipolar TRD, and measured peripheral blood-based biomarkers associated with response to treatment. METHODS Fifteen adults diagnosed with TRD completed an open label study of ten infusions of subanesthetic ketamine over four weeks. Out of fifteen patients, blood was collected from eleven patients at three timepoints to analyze peripheral biomarkers in isolated plasma, including IL-6, IL-10, TNF-α, BDNF, and irisin. Irisin analysis was completed using an ELISA assay, and the remaining biomarkers were analyzed together simultaneously using a multiplex immunoassay. RESULTS Repeated ketamine infusions produced a significant decrease in total average depressive symptoms (MADRS) at all timepoints. Improvements in depressive symptoms were significant at one week, and continued to significantly decrease until two weeks, where it was maintained. Ketamine was generally well tolerated, and we observed improvements in functional impairment, anhedonia, and psychiatric symptoms, with no increases in manic symptoms. Levels of BDNF throughout treatment inversely correlated to decreases in MADRS scores, and higher levels of baseline BDNF predicted mood responses at one- and four weeks. LIMITATIONS The study was observational and uncontrolled, with a sample size of 15. Outpatients remained on their course of medications, unless they were pharmacological agents that have previously been identified to block ketamine's effects. CONCLUSIONS Ketamine may be an efficacious and safe pharmacological option for the acute treatment of patients suffering from severe TRD. BDNF has the potential to function as a prognostic biomarker for predicting response to ketamine treatments.
Collapse
Affiliation(s)
- Melody J Y Kang
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada; Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Gustavo H Vazquez
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada; Department of Psychiatry, Queen's University Medical School, Kingston, Ontario, Canada; International Consortium for Research on Mood & Psychotic Disorders, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
11
|
Guran E, Hu J, Wefel JS, Chung C, Cata JP. Perioperative considerations in patients with chemotherapy-induced cognitive impairment: a narrative review. Br J Anaesth 2022; 129:909-922. [PMID: 36270848 DOI: 10.1016/j.bja.2022.08.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/01/2022] [Accepted: 08/23/2022] [Indexed: 11/02/2022] Open
Abstract
Patients with cancer may suffer from a decline in their cognitive function after various cancer therapies, including surgery, radiation, and chemotherapy, and in some cases, this decline in cognitive function persists even years after completion of treatment. Chemobrain or chemotherapy-induced cognitive impairment, a well-established clinical syndrome, has become an increasing concern as the number of successfully treated cancer patients has increased significantly. Chemotherapy-induced cognitive impairment can originate from direct neurotoxicity, neuroinflammation, and oxidative stress, resulting in alterations in grey matter volume, white matter integrity, and brain connectivity. Surgery has been associated with exacerbating the inflammatory response associated with chemotherapy and predisposes patients to develop postoperative cognitive dysfunction. As the proportion of patients living longer after these therapies increases, the magnitude of impact and growing concern of post-treatment cognitive dysfunction in these patients has also come to the fore. We review the clinical presentation, potential mechanisms, predisposing factors, diagnostic methods, neuropsychological testing, and imaging findings of chemotherapy-induced cognitive impairment and its intersection with postoperative cognitive dysfunction.
Collapse
Affiliation(s)
- Ekin Guran
- Department of Anaesthesiology and Reanimation, University of Health Sciences, Ankara Oncology Training and Research Hospital, Ankara, Turkey; Anaesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Jian Hu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey S Wefel
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Juan P Cata
- Anaesthesiology and Surgical Oncology Research Group, Houston, TX, USA; Department of Anaesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
Leung E, Lau EW, Liang A, de Dios C, Suchting R, Östlundh L, Masdeu JC, Fujita M, Sanches M, Soares JC, Selvaraj S. Alterations in brain synaptic proteins and mRNAs in mood disorders: a systematic review and meta-analysis of postmortem brain studies. Mol Psychiatry 2022; 27:1362-1372. [PMID: 35022529 DOI: 10.1038/s41380-021-01410-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 11/09/2022]
Abstract
The pathophysiological mechanisms underlying bipolar (BD) and major depressive disorders (MDD) are multifactorial but likely involve synaptic dysfunction and dysregulation. There are multiple synaptic proteins but three synaptic proteins, namely SNAP-25, PSD-95, and synaptophysin, have been widely studied for their role in synaptic function in human brain postmortem studies in BD and MDD. These studies have yielded contradictory results, possibly due to the small sample size and sourcing material from different cortical regions of the brain. We performed a systematic review and meta-analysis to understand the role of these three synaptic proteins and other synaptic proteins, messenger RNA (mRNA) and their regional localizations in BD and MDD. A systematic literature search was conducted and the review is reported in accordance with the MOOSE Guidelines. Meta-analysis was performed to compare synaptic marker levels between BD/MDD groups and controls separately. 1811 papers were identified in the literature search and screened against the preset inclusion and exclusion criteria. A total of 72 studies were screened in the full text, of which 47 were identified as eligible to be included in the systematic review. 24 of these 47 papers were included in the meta-analysis. The meta-analysis indicated that SNAP-25 protein levels were significantly lower in BD. On average, PSD-95 mRNA levels were lower in BD, and protein levels of SNAP-25, PSD-95, and syntaxin were lower in MDD. Localization analysis showed decreased levels of PSD-95 protein in the frontal cortex. We found specific alterations in synaptic proteins and RNAs in both BD and MDD. The review was prospectively registered online in PROSPERO international prospective register of systematic reviews, registration no. CRD42020196932.
Collapse
Affiliation(s)
- Edison Leung
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Depression Research Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ethan W Lau
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Andi Liang
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Constanza de Dios
- Depression Research Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Robert Suchting
- Depression Research Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Linda Östlundh
- The National Medical Library, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Joseph C Masdeu
- Houston Methodist Neurological Institute, Houston, TX, USA.,Weill Cornell Medicine, New York, NY, USA
| | - Masahiro Fujita
- Weill Cornell Medicine, New York, NY, USA.,PET Core Facility, Houston Methodist Research Insitute, Houston, TX, USA
| | - Marsal Sanches
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Depression Research Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jair C Soares
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.,Depression Research Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sudhakar Selvaraj
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA. .,Depression Research Program, Faillace Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
13
|
Luo YF, Ye XX, Fang YZ, Li MD, Xia ZX, Liu JM, Lin XS, Huang Z, Zhu XQ, Huang JJ, Tan DL, Zhang YF, Liu HP, Zhou J, Shen ZC. mTORC1 Signaling Pathway Mediates Chronic Stress-Induced Synapse Loss in the Hippocampus. Front Pharmacol 2022; 12:801234. [PMID: 34987410 PMCID: PMC8722735 DOI: 10.3389/fphar.2021.801234] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Background: The mechanistic target of rapamycin complex 1 (mTORC1) signaling has served as a promising target for therapeutic intervention of major depressive disorder (MDD), but the mTORC1 signaling underlying MDD has not been well elucidated. In the present study, we investigated whether mTORC1 signaling pathway mediates synapse loss induced by chronic stress in the hippocampus. Methods: Chronic restraint stress-induced depression-like behaviors were tested by behavior tests (sucrose preference test, forced swim test and tail suspension test). Synaptic proteins and alternations of phosphorylation levels of mTORC1 signaling-associated molecules were measured using Western blotting. In addition, mRNA changes of immediate early genes (IEGs) and glutamate receptors were measured by RT-PCR. Rapamycin was used to explore the role of mTORC1 signaling in the antidepressant effects of fluoxetine. Results: After successfully establishing the chronic restraint stress paradigm, we observed that the mRNA levels of some IEGs were significantly changed, indicating the activation of neurons and protein synthesis alterations. Then, there was a significant downregulation of glutamate receptors and postsynaptic density protein 95 at protein and mRNA levels. Additionally, synaptic fractionation assay revealed that chronic stress induced synapse loss in the dorsal and ventral hippocampus. Furthermore, these effects were associated with the mTORC1 signaling pathway-mediated protein synthesis, and subsequently the phosphorylation of associated downstream signaling targets was reduced after chronic stress. Finally, we found that intracerebroventricular infusion of rapamycin simulated depression-like behavior and also blocked the antidepressant effects of fluoxetine. Conclusion: Overall, our study suggests that mTORC1 signaling pathway plays a critical role in mediating synapse loss induced by chronic stress, and has part in the behavioral effects of antidepressant treatment.
Collapse
Affiliation(s)
- Yu-Fei Luo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Clinical Medical Research Center, Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, China
| | - Xiao-Xia Ye
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Ying-Zhao Fang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Meng-Die Li
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Zhi-Xuan Xia
- Department of Pharmacology, School of Basic Medicine and Life Science, Hainan Medical University, Haikou, China
| | - Jian-Min Liu
- Department of Pharmacy, Wuhan No. 1 Hospital, Wuhan, China
| | - Xiao-Shan Lin
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Zhen Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xiao-Qian Zhu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jun-Jie Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Dong-Lin Tan
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Fei Zhang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Hai-Ping Liu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jun Zhou
- Translational Medicine Center, Xi'an Chest Hospital, Medical College of Xi'an Jiaotong University, Xi'an, China
| | - Zu-Cheng Shen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
14
|
Ren Z, Wang M, Aldhabi M, Zhang R, Liu Y, Liu S, Tang R, Chen Z. Low-dose S-ketamine exerts antidepressant-like effects via enhanced hippocampal synaptic plasticity in postpartum depression rats. Neurobiol Stress 2022; 16:100422. [PMID: 34977283 PMCID: PMC8686162 DOI: 10.1016/j.ynstr.2021.100422] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 11/05/2022] Open
Abstract
Rapid antidepressant effects of S-ketamine have repeatedly been confirmed in patients with depression, as well as in chronic unpredictable mild stress (CUMS) animal models. However, the pharmacological study of S-ketamine for anti-postpartum depression has not been considered. In this study, the classical method of reproductive hormone withdrawal was used to construct a rat model of postpartum depression (PPD). Subsequently, the study evaluated the effects of low-dose S-ketamine on behavior and synaptic plasticity, which is related to depression, in the hippocampus of PPD rats. Multiple behavioral tests were used to evaluate depression-like behaviors in PPD models. Synaptic plasticity of the hippocampus can be demonstrated by Western blot, Golgi staining, transmission electron microscopy, and electrophysiological recording. Our study provides insight into the role of low-dose S-ketamine in antidepressant as well as antianxiety and indicates that maintaining synaptic plasticity is a key target for S-ketamine therapy for postpartum depression induced by reproductive hormone withdrawal.
Collapse
Affiliation(s)
- Zhuoyu Ren
- Department of Anesthesiology, Qingdao Women and Children's Hospital of Qingdao University, Qingdao, Shandong, China
| | - Mingling Wang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Mokhtar Aldhabi
- Department of Urology of the Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Rui Zhang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Yongxin Liu
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Shaoyan Liu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rundong Tang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zuolei Chen
- Department of Anesthesiology, Qingdao Women and Children's Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Anesthesiology of the Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| |
Collapse
|
15
|
Shen Y, Lv F, Min S, Hao X, Yu J. Ketamine alleviating depressive-like behaviors is associated with regulation of nNOS-CAPON-Dexras1 complex in chronic unpredictable mild stress rats. Transl Neurosci 2022; 13:309-319. [PMID: 36212606 PMCID: PMC9508647 DOI: 10.1515/tnsci-2022-0245] [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: 06/29/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND A growing number of studies have demonstrated that ketamine induces rapid and sustained antidepressant action. Neuronal nitric oxide synthase (nNOS) signaling has been explored for the treatment of neuropsychiatric disorders for decades. But the effect of ketamine on nNOS signaling is poorly understood. The aim of the present study was to investigate the effect of ketamine on nNOS signaling in a chronic unpredictable mild stress (CUMS) model of depression. METHODS Forty-eight rats were randomly divided into four groups: the control group of healthy rats (group C), the healthy rats treated with ketamine 10 mg/kg for 3 days (group CK), the rats model of stress-induced depression group (group D), and the depressed group treated with ketamine 10 mg/kg for 3 days (group DK). The sucrose preference test and open field test were used to assess behavioral changes. Immunohistochemistry, immunofluorescence, and real-time PCR analysis were carried out to measure the expression of nNOS, CAPON, and Dexras1 in the prefrontal cortex (PFC) of the CUMS rats. RESULTS Compared with healthy rats, the total distance traveled, the rearing counts, the sucrose preference percentage (SPP), and CAPON and Dexras1 expression in the PFC significantly decreased, while nNOS expression increased in CUMS rats. After treating with ketamine, the total distance traveled, the rearing counts, the SPP, and CAPON and Dexras1 expression significantly increased, while nNOS expression significantly decreased. CONCLUSION The results indicated that ketamine improved the depressive behavior of rats, which may be related to the reduced nNOS expression and enhanced CAPON and Dexras1 expression.
Collapse
Affiliation(s)
- Yiwei Shen
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| | - Feng Lv
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| | - Su Min
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| | - Xuechao Hao
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| | - Jian Yu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| |
Collapse
|
16
|
Kang MJY, Hawken E, Vazquez GH. The Mechanisms Behind Rapid Antidepressant Effects of Ketamine: A Systematic Review With a Focus on Molecular Neuroplasticity. Front Psychiatry 2022; 13:860882. [PMID: 35546951 PMCID: PMC9082546 DOI: 10.3389/fpsyt.2022.860882] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
The mechanism of action underlying ketamine's rapid antidepressant effects in patients with depression, both suffering from major depressive disorder (MDD) and bipolar disorder (BD), including treatment resistant depression (TRD), remains unclear. Of the many speculated routes that ketamine may act through, restoring deficits in neuroplasticity may be the most parsimonious mechanism in both human patients and preclinical models of depression. Here, we conducted a literature search using PubMed for any reports of ketamine inducing neuroplasticity relevant to depression, to identify cellular and molecular events, relevant to neuroplasticity, immediately observed with rapid mood improvements in humans or antidepressant-like effects in animals. After screening reports using our inclusion/exclusion criteria, 139 publications with data from cell cultures, animal models, and patients with BD or MDD were included (registered on PROSPERO, ID: CRD42019123346). We found accumulating evidence to support that ketamine induces an increase in molecules involved in modulating neuroplasticity, and that these changes are paired with rapid antidepressant effects. Molecules or complexes of high interest include glutamate, AMPA receptors (AMPAR), mTOR, BDNF/TrkB, VGF, eEF2K, p70S6K, GSK-3, IGF2, Erk, and microRNAs. In summary, these studies suggest a robust relationship between improvements in mood, and ketamine-induced increases in molecular neuroplasticity, particularly regarding intracellular signaling molecules.
Collapse
Affiliation(s)
- Melody J Y Kang
- Center of Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada
| | - Emily Hawken
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada.,Providence Care Hospital, Kingston, ON, Canada
| | - Gustavo Hector Vazquez
- Center of Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada.,Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada.,Providence Care Hospital, Kingston, ON, Canada
| |
Collapse
|
17
|
Kowalczyk M, Kowalczyk E, Kwiatkowski P, Łopusiewicz Ł, Sienkiewicz M, Talarowska M. Ketamine-New Possibilities in the Treatment of Depression: A Narrative Review. Life (Basel) 2021; 11:1186. [PMID: 34833062 PMCID: PMC8619908 DOI: 10.3390/life11111186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 12/11/2022] Open
Abstract
The SARS-CoV-2 coronavirus epidemic has led to an increase in the number of people with depression. Symptoms related to the mental sphere (mainly depression and anxiety) may be experienced by one third of the worldwide population. This entails the need for the effective and rapid treatment of depressive episodes. An effective drug seems to be s-ketamine, which was accepted in March 2019 by the Food and Drug Administration (FDA) for the treatment of drug-resistant depression. This drug provides a quick antidepressant effect with maximum effectiveness achieved after 24 h. It also appears to reduce the occurrence of suicidal thoughts. However, research into undesirable effects, especially in groups of people susceptible to psychotic episodes or those who use alcohol or psychoactive substances, is necessary.
Collapse
Affiliation(s)
- Mateusz Kowalczyk
- Babinski Memorial Hospital, Aleksandrowska St. 159, 91-229 Lodz, Poland;
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego St. 7/9, 90-752 Lodz, Poland;
| | - Paweł Kwiatkowski
- Department of Diagnostic Immunology, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Łukasz Łopusiewicz
- Center of Bioimmobilisation and Innovative Packaging Materials, Faculty of Food Sciences and Fisheries, West Pomeranian University of Technology Szczecin, Janickiego 35, 71-270 Szczecin, Poland;
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostic, Medical University of Lodz, Muszyńskiego St. 1, 90-151 Lodz, Poland
| | - Monika Talarowska
- Department of Clinical Psychology and Psychopathology, Institute of Psychology, University of Lodz, Smugowa St. 10/12, 91-433 Lodz, Poland;
| |
Collapse
|
18
|
Takahashi N, Yamada A, Shiraishi A, Shimizu H, Goto R, Tominaga Y. Efficacy and safety of fixed doses of intranasal Esketamine as an add-on therapy to Oral antidepressants in Japanese patients with treatment-resistant depression: a phase 2b randomized clinical study. BMC Psychiatry 2021; 21:526. [PMID: 34696742 PMCID: PMC8547110 DOI: 10.1186/s12888-021-03538-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 10/07/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Esketamine nasal spray (Spravato) in conjunction with oral antidepressants (ADs) is approved in the European Union, United States, and other markets for treatment-resistant depression (TRD). Efficacy, safety, and tolerability of esketamine nasal spray in Japanese patients with TRD needs to be assessed. METHODS This Phase 2b, randomized, double-blind (DB), placebo-controlled study was conducted in adult Japanese patients with TRD meeting the Diagnostic and Statistical Manual of Mental Disorders (fifth edition) criteria of major depressive disorder with nonresponse to ≥ 1 but < 5 different ADs in the current episode at screening. Patients were treated with a new oral AD for 6 weeks (prospective lead-in phase); nonresponders were randomized (2:1:1:1) to placebo or esketamine (28-, 56-, or 84-mg) nasal spray along with the continued use of AD for 4 weeks (DB induction phase). Responders (≥50% reduction from baseline in the Montgomery-Asberg Depression Rating Scale [MADRS] total score) from the DB induction phase continued into the 24-week posttreatment phase and patients who relapsed could participate in a 4-week open-label (OL) second induction (flexibly-dosed esketamine). The primary efficacy endpoint, change from baseline in the MADRS total score at Day 28 in the DB induction phase, was based on mixed-effects model using repeated measures pairwise comparisons using a Dunnett adjustment. RESULTS Of the 202 patients randomized in the DB induction phase (esketamine [n = 122] or placebo [n = 80]), the MADRS total scores decreased from baseline to Day 28 of the DB induction phase (- 15.2, - 14.5, - 15.1, and - 15.3 for esketamine 28 mg, 56 mg, 84 mg, and placebo groups, respectively), indicating an improvement in depressive symptoms; however, the difference between the esketamine and placebo groups was not statistically significant. The most common treatment-emergent adverse events during the DB induction phase in the combined esketamine group (incidences ranging from 12.3 to 41.0%) were blood pressure increased, dissociation, dizziness, somnolence, nausea, hypoaesthesia, vertigo, and headache; the incidence of each of these events was > 2-fold higher than the corresponding incidence in the placebo group. CONCLUSIONS Efficacy of esketamine plus oral AD in Japanese TRD patients was not established; further investigation is warranted. All esketamine doses were safe and tolerated. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02918318 . Registered: 28 September 2016.
Collapse
Affiliation(s)
- Nagahide Takahashi
- grid.27476.300000 0001 0943 978XDepartment of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Aya Yamada
- Janssen Pharmaceutical K.K, 5-2, Nishikanda 3-chome, Chiyoda-ku, Tokyo, 101-0065 Japan
| | - Ayako Shiraishi
- Janssen Pharmaceutical K.K, 5-2, Nishikanda 3-chome, Chiyoda-ku, Tokyo, 101-0065 Japan
| | - Hiroko Shimizu
- Janssen Pharmaceutical K.K, 5-2, Nishikanda 3-chome, Chiyoda-ku, Tokyo, 101-0065 Japan
| | - Ryosuke Goto
- Janssen Pharmaceutical K.K, 5-2, Nishikanda 3-chome, Chiyoda-ku, Tokyo, 101-0065 Japan
| | - Yushin Tominaga
- Janssen Pharmaceutical K.K, 5-2, Nishikanda 3-chome, Chiyoda-ku, Tokyo, 101-0065, Japan.
| |
Collapse
|
19
|
Parise EM, Parise LF, Sial OK, Cardona-Acosta AM, Gyles TM, Juarez B, Chaudhury D, Han MH, Nestler EJ, Bolaños-Guzmán CA. The Resilient Phenotype Induced by Prophylactic Ketamine Exposure During Adolescence Is Mediated by the Ventral Tegmental Area-Nucleus Accumbens Pathway. Biol Psychiatry 2021; 90:482-493. [PMID: 34247781 PMCID: PMC8761260 DOI: 10.1016/j.biopsych.2021.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 04/12/2021] [Accepted: 05/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Major depressive disorder is prevalent in children and adolescents and is associated with a high degree of morbidity throughout life, with potentially devastating personal consequences and public health impact. The efficacy of ketamine (KET) as an antidepressant has been demonstrated in adolescent rodents; however, the neurobiological mechanisms underlying these effects are unknown. Recent evidence showed that KET reverses stress-induced (i.e., depressive-like) deficits within major mesocorticolimbic regions, such as the prefrontal cortex, nucleus accumbens (NAc), and hippocampus, in adult rodents. However, little is known about KET's effect in the ventral tegmental area (VTA), which provides the majority of dopaminergic input to these brain regions. METHODS We characterized behavioral, biochemical, and electrophysiological effects produced by KET treatment in C57BL/6J male mice during adolescence (n = 7-10 per condition) within the VTA and its major projection regions, namely, the NAc and prefrontal cortex. Subsequently, molecular targets within the VTA-NAc projection were identified for viral gene transfer manipulations to recapitulate the effects of stress or KET treatment. RESULTS Repeated KET treatment produced a robust proresilient response to chronic social defeat stress. This effect was largely driven by Akt signaling activity within the VTA and NAc, and it could be blocked or recapitulated through direct Akt-viral-mediated manipulation. Additionally, we found that the KET-induced resilient phenotype is dependent on VTA-NAc, but not VTA-prefrontal cortex, pathway activity. CONCLUSIONS These findings indicate that KET exposure during adolescence produces a proresilient phenotype mediated by changes in Akt intracellular signaling and altered neuronal activity within the VTA-NAc pathway.
Collapse
Affiliation(s)
- Eric M Parise
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lyonna F Parise
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Omar K Sial
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas; Institute for Neuroscience, Texas A&M University, College Station, Texas
| | - Astrid M Cardona-Acosta
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas
| | - Trevonn M Gyles
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Barbara Juarez
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacology, University of Washington, Seattle, Washington
| | - Dipesh Chaudhury
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York; Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Ming-Hu Han
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Carlos A Bolaños-Guzmán
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas; Institute for Neuroscience, Texas A&M University, College Station, Texas.
| |
Collapse
|
20
|
Murphy N, Lijffijt M, Ramakrishnan N, Vo-Le B, Vo-Le B, Iqbal S, Iqbal T, O'Brien B, Smith MA, Swann AC, Mathew SJ. Does mismatch negativity have utility for NMDA receptor drug development in depression? ACTA ACUST UNITED AC 2021; 44:61-73. [PMID: 33825765 PMCID: PMC8827377 DOI: 10.1590/1516-4446-2020-1685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/13/2021] [Indexed: 11/22/2022]
Abstract
CLINICAL TRIAL REGISTRATION Rapid antidepressant effects associated with ketamine have shifted the landscape for the development of therapeutics to treat major depressive disorder (MDD) from a monoaminergic to glutamatergic model. Treatment with ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, may be effective, but has many non-glutamatergic targets, and clinical and logistical problems are potential challenges. These factors underscore the importance of manipulations of binding mechanics to produce antidepressant effects without concomitant clinical side effects. This will require identification of efficient biomarkers to monitor target engagement. The mismatch negativity (MMN) is a widely used electrophysiological signature linked to the activity of NMDA receptors (NMDAR) in humans and animals and validated in pre-clinical and clinical studies of ketamine. In this review, we explore the flexibility of the MMN and its capabilities for reliable use in drug development for NMDAR antagonists in MDD. We supplement this with findings from our own research with three distinct NMDAR antagonists. The research described illustrates that there are important distinctions between the mechanisms of NMDAR antagonism, which are further crystallized when considering the paradigm used to study the MMN. We conclude that the lack of standardized methodology currently prevents MMN from being ready for common use in drug discovery. This manuscript describes data collected from the following National Institutes of Health (NIH) and Veterans Affairs (VA) studies: AV-101, NCT03583554; lanicemine, NCT03166501; ketamine, NCT02556606.
Collapse
Affiliation(s)
- Nicholas Murphy
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA.,The Menninger Clinic, Houston, TX, USA
| | - Marijn Lijffijt
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Nithya Ramakrishnan
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Bylinda Vo-Le
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Brittany Vo-Le
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Sidra Iqbal
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Tabish Iqbal
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Brittany O'Brien
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Mark A Smith
- VistaGen Therapeutics, Inc., South San Francisco, CA, USA.,Medical College of Georgia, Augusta, GA, USA
| | - Alan C Swann
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA
| | - Sanjay J Mathew
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey VA Medical Center, Houston, TX, USA.,The Menninger Clinic, Houston, TX, USA
| |
Collapse
|
21
|
Zhang M, Zhang Y, Sun H, Ni H, Sun J, Yang X, Chen W, Zhao W, Zhong X, He C, Ao H, He S. Sinisan Protects Primary Hippocampal Neurons Against Corticosterone by Inhibiting Autophagy via the PI3K/Akt/mTOR Pathway. Front Psychiatry 2021; 12:627056. [PMID: 34122166 PMCID: PMC8192823 DOI: 10.3389/fpsyt.2021.627056] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Objective: Corticosterone causes significant neurotoxicity in primary hippocampal neurons which is associated with depression. Dysfunctional autophagy is implicated in cognitive impairment and depressive-like behavior. The traditional Chinese medicine Sinisan (SNS) is highly effective in clinical treatment of depression. However, the molecular mechanisms underlying therapeutic effects of SNS are unknown. Purpose: The aim of this study was to elucidate the protective effect of SNS and the underlying mechanisms against corticosterone-induced neuronal damage. Study Design: The effects of serum derived from rats containing SNS (or untreated controls) on the expression of autophagy-related molecules in primary rat hippocampal neurons exposed to different concentrations of corticosterone for different intervals were explored. Methods: CCK-8 assay, LDH assay were used to analyze cell viability and LDH activity. Western blot, qRT-PCR, and immunofluorescence assays were used to determine protein and mRNA expression levels of molecules such as LC3, p62, Beclin1, ULK1, PI3K, p-PI3K, Akt p-Akt, mTOR, p-mTOR, p70S6, p-p70S6, 4ebp1 and p-4ebp1. Results: Corticosterone induced a dose- and time-dependent reduction in cellular viability. Moreover, corticosterone (100-400 μM) treatment for 24 h increased LC3-II/LC3-I protein ratio, increased Beclin1 and ULK1 protein expression levels, and decreased p62, PI3K, p-PI3K, p-Akt, p-mTOR, p-p70S6, and p-4ebp1 protein expression levels. Notably, SNS-containing serum reversed corticosterone-induced reduction of neuronal viability, and increased p62, PI3K, p-Akt, p-mTOR, p-p70S6, and p-4ebp1 protein and mRNA expression levels. In addition, SNS-containing serum decreased LC3-II/LC3-I protein ratio, and downregulated Beclin1, and ULK1 protein and mRNA expression in primary hippocampal neurons. Conclusion: SNS protects primary hippocampal neurons against corticosterone-induced neurotoxicity by preventing excessive autophagy through activation of PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Mingjia Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yi Zhang
- Department of Psychology, School of Economics and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haitao Sun
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hui Ni
- Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Jialing Sun
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xuemei Yang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Weicong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wenting Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaodan Zhong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chunyu He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Haiqing Ao
- Department of Psychology, School of Economics and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Songqi He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Crisanti C, Enrico P, Fiorentini A, Delvecchio G, Brambilla P. Neurocognitive impact of ketamine treatment in major depressive disorder: A review on human and animal studies. J Affect Disord 2020; 276:1109-1118. [PMID: 32777649 DOI: 10.1016/j.jad.2020.07.119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/29/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Most recent evidence support a rapid and sustained antidepressant effect of subanesthetic dose of intravenous ketamine in patients with major depressive disorder (MDD). However, clinical and animal studies investigating the effects of intravenous ketamine on specific functional domains disrupted by depression reported conflicting results. Therefore, the aim of this review is to provide an overview of the recent findings exploring the cognitive effects of ketamine in depression. METHODS After a bibliographic search on PubMed, Medline and PsycInfo, we retrieved 11 original studies meeting our research criteria, 7 in humans with MDD or Treatment Resistant Disorder and 4 using rats models for depression. RESULTS Overall the results showed that a) ketamine reduced activation and normalized connectivity measures of several brain regions related to depressive behaviors and reversed deficits in cognitive flexibility and coping response strategy in rats with depressive features, and b) ketamine leads to a no significant impairment on neurocognitive functions in most of the studies, with only three studies observing improvements in speed of processing, verbal learning, sustained attention and response control, verbal and working memory. LIMITATIONS The methodological heterogeneity, in terms of neuropsychological tests used and cognitive domain explored, of the studies included. CONCLUSIONS Most of the studies included showed no significant cognitive impairments in MDD patients after ketamine treatment. Furthermore, the results of the fMRI studies considered suggest that ketamine may have a normalizing effect on brain functions during attentional and emotional processing in MDD patients. However, further studies are needed to confirm these preliminary evidences.
Collapse
Affiliation(s)
- Camilla Crisanti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Paolo Enrico
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Alessio Fiorentini
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giuseppe Delvecchio
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| | - Paolo Brambilla
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy; Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
23
|
Pang L, Cui M, Dai W, Kong J, Chen H, Wu S. Can Intraoperative Low-Dose R, S-Ketamine Prevent Depressive Symptoms After Surgery? The First Meta-Analysis of Clinical Trials. Front Pharmacol 2020; 11:586104. [PMID: 33192527 PMCID: PMC7604489 DOI: 10.3389/fphar.2020.586104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Postoperative depression is a common complication after surgery that profoundly affects recovery and prognosis. New research indicates that (R,S)-ketamine is a potent antidepressant that exerts a rapid and sustained antidepressive effect. However, there is no consensus on whether intraoperative low-dose (R,S)-ketamine prevents postoperative depression. Objectives: This study aimed to investigate the safety, feasibility, and short-term complications of intraoperative low-dose (R,S)-ketamine in preventing postoperative depressive symptoms. Methods: The Web of Science, Cochrane, PubMed, and CNKI databases were systematically searched (last search February 28, 2020) to identify studies involving ketamine. Sensitivity and metaregression analyses were performed to identify potential confounders. The meta-analysis was performed using Review Manager 5.3. Results: A total of 13 studies (seven in Chinese and six in English) representing 1,148 cases of patients who were treated with (R,S)-ketamine and 874 cases of patients who received other treatments were included in the meta-analysis. Anesthesia duration and blood loss did not significantly differ between the two groups, demonstrating that (R,S)-ketamine was safe (odds ratio,OR: 0.27; 95% CI: -1.14 to 1.68; P = 0.71) for prophylactic treatment of postoperative depression. Blood loss (OR: -1.83; 95% CI: -8.34 to 4.68; P = 0.58), the number of postoperative depressive patients (95% CI: 0.8-1.07; P = 0.08; (R,S)-ketamine: control = 12.9%:15.8%), and postoperative complications (OR: 0.83, 95% CI: 0.44-1.58; P = 0.57; (R,S)-ketamine: control = 19.3%:19.3%) were all similar across groups. Intra-operative low-dose (R,S)-ketamine reduced extubation time (OR: -2.84; 95% CI: -5.48 to -0.21; P = 0.03). Conclusions: The prophylactic anti-depressant effect of (R,S)-ketamine did not significantly differ between the (R,S)-ketamine and control groups in patients undergoing general or spinal anesthesia. However, (R,S)-ketamine use led to a higher incidence of adverse reactions in patients under 40 years of age who underwent a Cesarean section under spinal anesthesia.
Collapse
Affiliation(s)
- Liwei Pang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Meiying Cui
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wanling Dai
- Innovation Institute of China Medical University, Shenyang, China
| | - Jing Kong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongzhi Chen
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Shuodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
24
|
Fukumoto K, Fogaça MV, Liu RJ, Duman CH, Li XY, Chaki S, Duman RS. Medial PFC AMPA receptor and BDNF signaling are required for the rapid and sustained antidepressant-like effects of 5-HT 1A receptor stimulation. Neuropsychopharmacology 2020; 45:1725-1734. [PMID: 32396921 PMCID: PMC7419563 DOI: 10.1038/s41386-020-0705-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 04/26/2020] [Accepted: 05/05/2020] [Indexed: 12/28/2022]
Abstract
We previously reported that the serotonergic system is important for the antidepressant-like effects of ketamine, a non-competitive N-methyl-D-aspartate receptor antagonist, which produces rapid and long-lasting antidepressant effects in patients with major depressive disorder (MDD). In particular, selective stimulation of the 5-HT1A receptor in the medial prefrontal cortex (mPFC), as opposed to the somatic 5-HT1A autoreceptor, has been shown to play a critical role in the antidepressant-like actions of ketamine. However, the detailed mechanisms underlying mPFC 5-HT1A receptor-mediated antidepressant-like effects are not fully understood. Here we examined the involvement of the glutamate AMPA receptor and brain-derived neurotrophic factor (BDNF) in the antidepressant-like effects of 5-HT1A receptor activation in the mPFC. The results show that intra-mPFC infusion of the 5-HT1A receptor agonist 8-OH-DPAT induces rapid and long-lasting antidepressant-like effects in the forced swim, novelty-suppressed feeding, female urine sniffing, and chronic unpredictable stress tests. In addition, the results demonstrate that the antidepressant-like effects of intra-mPFC infusion of 8-OH-DPAT are blocked by co-infusion of an AMPA receptor antagonist or an anti-BDNF neutralizing antibody. In addition, mPFC infusion of 8-OH-DPAT increased the phosphorylation of signaling proteins downstream of BDNF, including mTOR, ERK, 4EBP1, and p70S6K. Finally, selective stimulation of the 5-HT1A receptor increased levels of synaptic proteins and synaptic function in the mPFC. Collectively, these results indicate that selective stimulation of 5-HT1A receptor in the mPFC exerts rapid and sustained antidepressant-like effects via activation of AMPA receptor/BDNF/mTOR signaling in mice, which subsequently increase synaptic function in the mPFC, and provide evidence for the 5-HT1A receptor as a target for the treatment of MDD.
Collapse
Affiliation(s)
- Kenichi Fukumoto
- Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06520, USA. .,Research Headquarters, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama, 331-9530, Japan.
| | - Manoela V. Fogaça
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| | - Rong-Jian Liu
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| | - Catharine H. Duman
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| | - Xiao-Yuan Li
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| | - Shigeyuki Chaki
- grid.419836.10000 0001 2162 3360Research Headquarters, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama 331-9530 Japan
| | - Ronald S. Duman
- grid.47100.320000000419368710Departments of Psychiatry and Neurosciences, Yale University School of Medicine, 34 Park Street, New Haven, CT 06520 USA
| |
Collapse
|
25
|
Garcia-Carachure I, Flores-Ramirez FJ, Castillo SA, Themann A, Arenivar MA, Preciado-Piña J, Zavala AR, Lobo MK, Iñiguez SD. Enduring effects of adolescent ketamine exposure on cocaine- and sucrose-induced reward in male and female C57BL/6 mice. Neuropsychopharmacology 2020; 45:1536-1544. [PMID: 32165718 PMCID: PMC7360558 DOI: 10.1038/s41386-020-0654-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022]
Abstract
Ketamine has shown promising antidepressant efficacy for adolescent treatment-resistant depression. However, the potential enduring consequences of ketamine exposure have not been thoroughly evaluated. Thus, we examined if juvenile ketamine treatment results in long-lasting changes for the rewarding properties of sucrose and cocaine in adulthood, across three separate experiments. In Experiment 1, adolescent male and female C57BL/6 mice received ketamine (20 mg/kg) for 15 consecutive days (Postnatal Day [PD] 35-49). Twenty-one days later (PD70; adulthood) we examined their behavioral responsivity to sucrose (1%) on a two-bottle choice design, or cocaine (0, 5, 10 mg/kg) using the conditioned place preference (CPP) test. We found that juvenile ketamine-pretreatment increased preference for sucrose and environments paired with cocaine in male, but not female, adult mice. This long-term outcome was not observed when male and female mice received ketamine as adults (PD70-84) and tested for sucrose and cocaine preference 21-days later (Experiment 2). Similarly, in Experiment 3, no long-lasting differences in these measures were observed when adolescent male mice were exposed to concomitant ketamine and social stressors (PD35-44), namely the social defeat or vicarious defeat stress paradigms-procedures that mediated a depression-related phenotype (along with a ketamine antidepressant-like response). Collectively, we demonstrate that in the absence of physical or psychological stress, adolescent ketamine exposure increases later life preference for the rewarding properties of sucrose and cocaine in a sex- and age-specific manner. As such, this preclinical work provides awareness for the potential long-term behavioral consequences associated with juvenile ketamine exposure.
Collapse
Affiliation(s)
- Israel Garcia-Carachure
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Francisco J. Flores-Ramirez
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Samuel A. Castillo
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Anapaula Themann
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Miguel A. Arenivar
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Joshua Preciado-Piña
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Arturo R. Zavala
- 0000 0000 9093 6830grid.213902.bDepartment of Psychology, California State University, Long Beach, CA USA
| | - Mary Kay Lobo
- 0000 0001 2175 4264grid.411024.2Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Sergio D. Iñiguez
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| |
Collapse
|
26
|
Chaves TV, Wilffert B, Sanchez ZM. The use of ketamine to cope with depression and post-traumatic stress disorder: A qualitative analysis of the discourses posted on a popular online forum. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2020; 46:613-624. [DOI: 10.1080/00952990.2020.1769118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Tharcila V. Chaves
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bob Wilffert
- Groningen Research Institute of Pharmacy, Pharmacotherapy, Epidemiology and Economics, Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Zila M. Sanchez
- Department of Preventive Medicine, Universidade Federal De São Paulo, São Paulo, Brazil
| |
Collapse
|
27
|
Efficacy and safety of ketamine in the management of anxiety and anxiety spectrum disorders: a review of the literature. CNS Spectr 2020; 25:331-342. [PMID: 31339086 DOI: 10.1017/s1092852919001238] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Anxiety disorders are among the most prevalent psychiatric conditions. Despite many proven pharmacological and non-pharmacological treatments available, high rates of partial response and low rates of long-term remission remain. Ketamine has been receiving increasing attention as an interventional treatment modality in psychiatry, especially among refractory conditions, including major depressive disorder. There is limited yet growing evidence to support the use of ketamine in anxiety disorders. In this review of the literature, we present case reports, case series, and controlled trials demonstrating proof-of-concept for its potential role in the treatment of anxiety and anxiety spectrum disorders. Its unique mechanism of action, rapid onset, and high rate of response have driven its use in clinical practice. Ketamine is generally well tolerated by patients and has a limited side effect profile; however, the effects of long-term use are unknown. While there is a growing body of research and increasing clinical experience to suggest ketamine may have clinical applications in the treatment of refractory anxiety disorders, further research to determine long-term safety and tolerability is indicated.
Collapse
|
28
|
|
29
|
Hao X, Ou M, Zhang D, Zhao W, Yang Y, Liu J, Yang H, Zhu T, Li Y, Zhou C. The Effects of General Anesthetics on Synaptic Transmission. Curr Neuropharmacol 2020; 18:936-965. [PMID: 32106800 PMCID: PMC7709148 DOI: 10.2174/1570159x18666200227125854] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
General anesthetics are a class of drugs that target the central nervous system and are widely used for various medical procedures. General anesthetics produce many behavioral changes required for clinical intervention, including amnesia, hypnosis, analgesia, and immobility; while they may also induce side effects like respiration and cardiovascular depressions. Understanding the mechanism of general anesthesia is essential for the development of selective general anesthetics which can preserve wanted pharmacological actions and exclude the side effects and underlying neural toxicities. However, the exact mechanism of how general anesthetics work is still elusive. Various molecular targets have been identified as specific targets for general anesthetics. Among these molecular targets, ion channels are the most principal category, including ligand-gated ionotropic receptors like γ-aminobutyric acid, glutamate and acetylcholine receptors, voltage-gated ion channels like voltage-gated sodium channel, calcium channel and potassium channels, and some second massager coupled channels. For neural functions of the central nervous system, synaptic transmission is the main procedure for which information is transmitted between neurons through brain regions, and intact synaptic function is fundamentally important for almost all the nervous functions, including consciousness, memory, and cognition. Therefore, it is important to understand the effects of general anesthetics on synaptic transmission via modulations of specific ion channels and relevant molecular targets, which can lead to the development of safer general anesthetics with selective actions. The present review will summarize the effects of various general anesthetics on synaptic transmissions and plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yu Li
- Address correspondence to these authors at the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; E-mail: and Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China; E-mail:
| | - Cheng Zhou
- Address correspondence to these authors at the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; E-mail: and Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China; E-mail:
| |
Collapse
|
30
|
Amidfar M, Woelfer M, Réus GZ, Quevedo J, Walter M, Kim YK. The role of NMDA receptor in neurobiology and treatment of major depressive disorder: Evidence from translational research. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109668. [PMID: 31207274 DOI: 10.1016/j.pnpbp.2019.109668] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/24/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022]
Abstract
There is accumulating evidence demonstrating that dysfunction of glutamatergic neurotransmission, particularly via N-methyl-d-aspartate (NMDA) receptors, is involved in the pathophysiology of major depressive disorder (MDD). Several studies have revealed an altered expression of NMDA receptor subtypes and impaired NMDA receptor-mediated intracellular signaling pathways in brain circuits of patients with MDD. Clinical studies have demonstrated that NMDA receptor antagonists, particularly ketamine, have rapid antidepressant effects in treatment-resistant depression, however, neurobiological mechanisms are not completely understood. Growing body of evidence suggest that signal transduction pathways involved in synaptic plasticity play critical role in molecular mechanisms underlying rapidly acting antidepressant properties of ketamine and other NMDAR antagonists in MDD. Discovering the molecular mechanisms underlying the unique antidepressant actions of ketamine will facilitate the development of novel fast acting antidepressants which lack undesirable effects of ketamine. This review provides a critical examination of the NMDA receptor involvement in the neurobiology of MDD including analyses of alterations in NMDA receptor subtypes and their interactive signaling cascades revealed by postmortem studies. Furthermore, to elucidate mechanisms underlying rapid-acting antidepressant properties of NMDA receptor antagonists we discussed their effects on the neuroplasticity, mostly based on signaling systems involved in synaptic plasticity of mood-related neurocircuitries.
Collapse
Affiliation(s)
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory, University Magdeburg, Germany; New Jersey Institute of Technology, Newark, NJ, USA
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Martin Walter
- Clinical Affective Neuroimaging Laboratory, University Magdeburg, Germany; Department of Psychiatry, University Tuebingen, Germany
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
31
|
Sayson LV, Botanas CJ, Custodio RJP, Abiero A, Kim M, Lee HJ, Kim HJ, Yoo SY, Lee KW, Ryu HW, Acharya S, Kim KM, Lee YS, Cheong JH. The novel methoxetamine analogs N-ethylnorketamine hydrochloride (NENK), 2-MeO-N-ethylketamine hydrochloride (2-MeO-NEK), and 4-MeO-N-ethylketamine hydrochloride (4-MeO-NEK) elicit rapid antidepressant effects via activation of AMPA and 5-HT 2 receptors. Psychopharmacology (Berl) 2019; 236:2201-2210. [PMID: 30891619 DOI: 10.1007/s00213-019-05219-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/01/2019] [Indexed: 12/28/2022]
Abstract
RATIONALE Depressive syndrome or depression is a debilitating brain disorder affecting numerous people worldwide. Although readily available, current antidepressants have low remission rates and late onset times. Recently, N-methyl-D-aspartate (NMDA) receptor antagonists, like ketamine and methoxetamine (MXE), were found to elicit rapid antidepressant effects. As the search for glutamatergic-based antidepressants is increasing, we synthesized three novel MXE analogs, N-ethylnorketamine hydrochloride (NENK), 2-MeO-N-ethylketamine hydrochloride (2-MeO-NEK), and 4-MeO-N-ethylketamine hydrochloride (4-MeO-NEK). OBJECTIVES To determine whether the three novel MXE analogs induce antidepressant effects and explore their mechanistic correlation. METHODS We examined their affinity for NMDA receptors through a radioligand binding assay. Mice were treated with each drug (2.5, 5, and 10 mg/kg), and their behavior was assessed 30 min later in the forced swimming test (FST), tail suspension test (TST), elevated plus-maze (EPM) test, and open-field test (OFT). Another group of mice were pretreated with 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo(f)quinoxaline-2,3-dione (NBQX), an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist, or ketanserin (KS), a 5-HT2 receptor antagonist, during the FST. We also measured mRNA levels of the AMPA receptor subunits GluA1 and GluA2, brain-derived neurotrophic factor (BDNF), and mammalian target of rapamycin (mTOR) in the hippocampus and prefrontal cortex. RESULTS The MXE analogs showed affinity to NMDA receptors and decreased immobility time during the FST and TST. NBQX and KS blocked their effects in the FST. The compounds did not induce behavioral alteration during the EPM and OFT. The compounds altered GluA1, GluA2, and BDNF mRNA levels. CONCLUSION These results suggest that the novel MXE analogs induce antidepressant effects, which is likely via AMPA and 5-HT2 receptor activation.
Collapse
Affiliation(s)
- Leandro Val Sayson
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Chrislean Jun Botanas
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Raly James Perez Custodio
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Arvie Abiero
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Hyun Jun Lee
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea
| | - Sung Yeun Yoo
- Medicinal Chemistry Laboratory, Department of Pharmacy & Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Kun Won Lee
- Medicinal Chemistry Laboratory, Department of Pharmacy & Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Hye Won Ryu
- Medicinal Chemistry Laboratory, Department of Pharmacy & Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Srijan Acharya
- College of Pharmacy, Chonnam National University, 77 Yongbong-ro, Yongbong-dong, Buk-gu, Gwangju, Republic of Korea
| | - Kyeong-Man Kim
- College of Pharmacy, Chonnam National University, 77 Yongbong-ro, Yongbong-dong, Buk-gu, Gwangju, Republic of Korea
| | - Yong Sup Lee
- Medicinal Chemistry Laboratory, Department of Pharmacy & Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea.
| | - Jae Hoon Cheong
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, 815 Hwarangro, Nowon-gu, Seoul, 01795, Republic of Korea.
| |
Collapse
|
32
|
Brakel K, Hook MA. SCI and depression: Does inflammation commandeer the brain? Exp Neurol 2019; 320:112977. [PMID: 31203113 DOI: 10.1016/j.expneurol.2019.112977] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/29/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022]
Abstract
The incidence of depression is almost twice as high in the spinally injured population compared to the general population. While this incidence has long been attributed to the psychological, economic, and social burdens that accompany spinal cord injury (SCI), data from animal studies indicate that the biology of SCI may play an important role in the development of depression. Inflammation has been shown to impact stress response in rodents and humans, and inflammatory cytokines have been associated with depression for decades. The inflammation inherent to SCI may disrupt necessary mechanisms of mental homeostasis, such as serotonin production, dopamine production, and the hypothalamic pituitary adrenal axis. Additionally, gut dysbiosis that occurs after SCI can exacerbate inflammation and may cause further mood and behavior changes. These mediators combined may significantly contribute to the rise in depression seen after SCI. Currently, there are no therapies specific to depression after SCI. Elucidation of the molecular pathways that contribute to SCI-specific depression is crucial for the understanding of this disease and its potential treatments.
Collapse
Affiliation(s)
- Kiralyn Brakel
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Medical Research and Education Building, Ste. 1005, 8447 Riverside Pkwy, Bryan, TX 77807, United States; Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474 College Station, TAMU, TX, United States.
| | - Michelle A Hook
- School of Medicine, Department of Neuroscience and Experimental Therapeutics, Texas A&M University, Medical Research and Education Building, Ste. 1005, 8447 Riverside Pkwy, Bryan, TX 77807, United States; Texas A&M Institute of Neuroscience, Texas A&M University, Interdisciplinary Life Sciences Building, Rm 3148, 3474 College Station, TAMU, TX, United States
| |
Collapse
|
33
|
Brain penetration of ketamine: Intranasal delivery VS parenteral routes of administraion. J Psychiatr Res 2019; 112:7-11. [PMID: 30818171 DOI: 10.1016/j.jpsychires.2019.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 02/08/2023]
Abstract
Ketamine is approved by the FDA to be used as an anesthetic however, recent reports have exhibited its success in the treatment of major depressive disorder (MDD). Studies have suggested that a sub-anesthetic dose produces rapid antidepressant activity providing significant symptomatic relief particularly in patients with a history of treatment resistant depression (TRD). Many reports have been published on the intranasal (IN) efficacy of ketamine in the treatment of depression, however studies that have investigated the effects of the route of administration on drug delivery to the brain appear to be absent in literature. Therefore, in this study, a single dose (15 mg/kg body weight) was administered via different routes of administration [oral (PO), intranasal (IN) and intraperitoneal (IP)] to healthy male Sprague-Dawley rats in order to determine the brain tissue pharmacokinetics of ketamine. A novel validated liquid chromatography-mass spectrometry (LC-MS) method was developed using a fused core column for the determination of ketamine in plasma and brain homogenates. While IP administration resulted in favorable concentrations in the brain and plasma; IN administration, which is supposed to favour drug delivery to the brain, exhibited moderately low drug levels post administration. PO administration produced significantly lower levels due to extensive first-pass metabolism in the liver and intestines. These results have implications for future studies exploring the use of ketamine for the treatment of MDD in order to optimize treatment regimens and suggest that parenteral administration of ketamine should be used in the treatment of depression.
Collapse
|
34
|
Zheng W, Zhou YL, Liu WJ, Wang CY, Zhan YN, Li HQ, Chen LJ, Li MD, Ning YP. Investigation of medical effect of multiple ketamine infusions on patients with major depressive disorder. J Psychopharmacol 2019; 33:494-501. [PMID: 30789302 DOI: 10.1177/0269881119827811] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Single-dose intravenous ketamine has rapid but time-limited antidepressant effects. We aimed to examine the sustained effects of six consecutive ketamine infusions in Chinese patients with major depressive disorder. METHODS Seventy-seven patients with major depressive disorder were eligible to receive augmentation with six ketamine infusions (0.5 mg/kg over 40 min) administered over the course of 12 days (Monday-Wednesday-Friday). The coprimary outcome measures were the rates of response and remission as measured on the 10-item Montgomery-Asberg Depression Rating Scale. Psychotomimetic and dissociative symptoms were measured with the Brief Psychiatric Rating Scale-positive symptoms and the Clinician Administered Dissociative States Scale, respectively. RESULTS After the first ketamine infusion, only 10 (13.0%) and 6 (7.8%) patients responded and remitted, respectively; after six ketamine infusions, 52 (67.5%) patients responded and 37 (48.1%) remitted. There was a significant mean decrease in Montgomery-Asberg Depression Rating Scale score at four hours after the first ketamine infusion (7.0±7.5, p<0.001), and this decrease was maintained for the duration of the infusion period. The response to ketamine treatment was positively associated with no history of psychiatric hospitalization (odds ratio=3.56, p=0.009). Suicidal ideation rapidly decreased across the entire study sample, even among the nonresponder group. No significant differences were found regarding Brief Psychiatric Rating Scale and Clinician Administered Dissociative States Scale scores from the first infusion at baseline to four hours post-infusion. CONCLUSION Six ketamine infusions increased rates of response and remission when compared to a single-dose ketamine infusion in patients with major depressive disorder. Future controlled studies are warranted to confirm and expand these findings.
Collapse
Affiliation(s)
- Wei Zheng
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Yan-Ling Zhou
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Wei-Jian Liu
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Cheng-Yu Wang
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Yan-Ni Zhan
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Han-Qiu Li
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Li-Jian Chen
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| | - Ming D Li
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China.,2 State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu-Ping Ning
- 1 The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China
| |
Collapse
|
35
|
Khakpai F, Ebrahimi-Ghiri M, Alijanpour S, Zarrindast MR. Ketamine-induced antidepressant like effects in mice: A possible involvement of cannabinoid system. Biomed Pharmacother 2019; 112:108717. [PMID: 30970516 DOI: 10.1016/j.biopha.2019.108717] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 02/06/2023] Open
Abstract
The purpose of this study was to explore the possible interaction between ketamine and cannabinoid system in the modulation of depression-related responses using the forced swimming test (FST), tail suspension test (TST) and open-field test (OFT) in mice. Our results revealed that intra-peritoneal (i.p.) injection of ketamine (5 and 10 mg/kg), a non-competitive NMDA antagonist, dose-dependently produced antidepressant-like effect in the FST. Moreover, i.p. administration of both CB1 and CB2 receptor drugs: ACPA (1 mg/kg; CB1 receptor agonist), AM251 (1 mg/kg; CB1 receptor antagonist), GP1a (2 mg/kg; CB2 receptor agonist) and AM630 (0.5 mg/kg; CB2 receptor antagonist) exhibited antidepressant action. Interestingly, the concomitant administration of ineffective doses of ketamine and cannabinoid receptor antagonists provoked the antidepressant-like effects as compared to control group. It should be considered, all above mentioned doses of drugs could not change locomotor activity in the OFT. It seems that possible interaction between ketamine and cannabinoid system may modulate depression-related behavior.
Collapse
Affiliation(s)
- Fatemeh Khakpai
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Sakineh Alijanpour
- Department of Biology, Faculty of Science, Gonbad Kavous University, Gonbad Kavous, Iran.
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran, Iran; Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Dadiomov D, Lee K. The effects of ketamine on suicidality across various formulations and study settings. Ment Health Clin 2019; 9:48-60. [PMID: 30627504 PMCID: PMC6322816 DOI: 10.9740/mhc.2019.01.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Introduction Suicidality and self-injurious behavior afflict patients with a wide variety of psychiatric illnesses. Currently, there are few pharmacologic treatments for suicidality and self-injurious behavior and none that treat these conditions emergently. Recently, ketamine has demonstrated efficacy in treating both depression and acute suicidal ideation. An increasing usage of ketamine, of a variety of formulations, has been studied for these indications. This article reviews the evidence for use of ketamine in self-injurious behavior and suicidality. Methods A review of the MEDLINE database for articles relating to ketamine, self-injurious behavior, suicidality, and self-harm was conducted. Additional articles were assessed via cross-reference. Results A total of 24 articles that included clinical trials, meta-analyses, case series, and case reports were analyzed. The majority of studies of ketamine for suicidal ideation include the intravenous route using a dose of 0.5 mg/kg over 40 minutes. These studies suggest that intravenous ketamine may be effective at reducing suicidal ideation acutely. Data on use of ketamine in the intramuscular, intranasal, and oral forms are limited and of poorer quality. Studies on these formulations contain greater variability of positive and negative results of ketamine for reducing suicidality and self-injurious behavior. The durability of the antisuicidal effects across all formulations is limited. Discussion Ketamine may be an effective option for the treatment of suicidal ideation in patients across inpatient, outpatient, or emergent settings. At this time, more research is needed on the efficacy of ketamine across all formulations being used in clinical practice.
Collapse
|
37
|
Activity-dependent brain-derived neurotrophic factor signaling is required for the antidepressant actions of (2 R,6 R)-hydroxynorketamine. Proc Natl Acad Sci U S A 2018; 116:297-302. [PMID: 30559184 DOI: 10.1073/pnas.1814709116] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ketamine, a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist, produces rapid and long-lasting antidepressant effects in major depressive disorder (MDD) patients. (2R,6R)-Hydroxynorketamine [(2R,6R)-HNK], a metabolite of ketamine, is reported to produce rapid antidepressant effects in rodent models without the side effects of ketamine. Importantly, (2R,6R)-HNK does not block NMDA receptors like ketamine, and the molecular signaling mechanisms for (2R,6R)-HNK remain unknown. Here, we examined the involvement of BDNF/TrkB/mechanistic target of rapamycin complex 1 (mTORC1) signaling in the antidepressant actions of (2R,6R)-HNK. Intramedial prefrontal cortex (intra-mPFC) infusion or systemic (2R,6R)-HNK administration induces rapid and long-lasting antidepressant effects in behavioral tests, identifying the mPFC as a key region for the actions of (2R,6R)-HNK. The antidepressant actions of (2R,6R)-HNK are blocked in mice with a knockin of the BDNF Val66Met allele (which blocks the processing and activity-dependent release of BDNF) or by intra-mPFC microinjection of an anti-BDNF neutralizing antibody. Blockade of L-type voltage-dependent Ca2+ channels (VDCCs), required for activity-dependent BDNF release, also blocks the actions of (2R,6R)-HNK. Intra-mPFC infusion of pharmacological inhibitors of TrkB or mTORC1 signaling, which are downstream of BDNF, also block the actions of (2R,6R)-HNK. Moreover, (2R,6R)-HNK increases synaptic function in the mPFC. These findings indicate that activity-dependent BDNF release and downstream TrkB and mTORC1 signaling, which increase synaptic function in the mPFC, are required for the rapid and long-lasting antidepressant effects of (2R,6R)-HNK, supporting the potential use of this metabolite for the treatment of MDD.
Collapse
|
38
|
Yang Y, Ju W, Zhang H, Sun L. Effect of Ketamine on LTP and NMDAR EPSC in Hippocampus of the Chronic Social Defeat Stress Mice Model of Depression. Front Behav Neurosci 2018; 12:229. [PMID: 30356718 PMCID: PMC6189398 DOI: 10.3389/fnbeh.2018.00229] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
Depression is a common mental disorder that is associated with memory dysfunction. Ketamine has recently been demonstrated to be a rapid antidepressant. The mechanisms underlying how depression induces memory dysfunction and how ketamine relieves depressive symptoms remain poorly understood. This work compared three groups of male C57BL/6J mice: mice exposed to chronic social defeat stress (CSDS) to induce a depression-like phenotype, depression-like mice treated with ketamine, and control mice that were not exposed to CSDS or treated with ketamine. Spatial working memory and long term memory were assessed by spontaneous alternation Y-maze and fear conditioning tests, respectively. We used western blot to analyze the density of N-methyl-D-aspartate receptor (NMDAR) subunits in the hippocampus. We recorded long term potentiation (LTP) and NMDA receptor-mediated excitatory postsynaptic currents (EPSCs) in hippocampal slices. We observed that compared with control mice, depression-like mice had significant reductions in spatial working memory and contextual fear memory. The level of NR2B, LTP and NMDA receptor-mediated EPSCs of depression-like mice were decreased. Ketamine treatment attenuated the memory impairment, and increased the density of NR2B and the amplitude of LTP and NMDA receptor-mediated EPSCs in the hippocampus of depression-like mice. In conclusion, depression-like mice have deficits in working memory and contextual fear memory. The decrease of NR2B, LTP induction and NMDA receptor-mediated EPSCs in the hippocampus may be involved in this process. Ketamine can improve expression of NR2B, LTP induction and NMDA receptor-mediated EPSCs in the hippocampus of depression-like mice, which might be part of the reason why ketamine can alleviate the memory dysfunction induced by depression.
Collapse
Affiliation(s)
- Yu Yang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Changchun, China
| | - Weina Ju
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Changchun, China
| | - Haining Zhang
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
39
|
Abstract
Patients with cancer are more likely to develop depression than the general population, which negatively impacts their quality of life and prognosis. In order to identify effective antidepressants catered toward cancer patients, the biology of depression in the context of cancer must be well-understood. Many theories have emerged postulating the mechanisms underlying the development of depressive disorder. Here, we review the role inflammation, a hyperactive hypothalamic-pituitary-adrenal (HPA) axis, and glutamate excitotoxicity may play in cancer-induced depression. Hopefully, novel therapeutics targeting these dysregulated pathways may be potent in ameliorating depressive symptoms in the cancer population.
Collapse
Affiliation(s)
| | - Gurmit Singh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
40
|
5-HT1A receptor stimulation in the medial prefrontal cortex mediates the antidepressant effects of mGlu2/3 receptor antagonist in mice. Neuropharmacology 2018; 137:96-103. [DOI: 10.1016/j.neuropharm.2018.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 12/11/2022]
|
41
|
Investigation of antidepressant-like and anxiolytic-like actions and cognitive and motor side effects of four N-methyl-D-aspartate receptor antagonists in mice. Behav Pharmacol 2018; 28:37-47. [PMID: 27740963 DOI: 10.1097/fbp.0000000000000266] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Evidence suggests that N-methyl-D-aspartate receptor (NMDAR) antagonists could be efficacious in treating depression and anxiety, but side effects constitute a challenge. This study evaluated the antidepressant-like and anxiolytic-like actions, and cognitive and motor side effects of four NMDAR antagonists. MK-801, ketamine, S-ketamine, RO 25-6981 and the positive control, citalopram, were tested for antidepressant-like and anxiolytic-like effects in mice using the forced-swim test, the elevated zero maze and the novelty-induced hypophagia test. Side effects were assessed using a locomotor activity test, the modified Y-maze and the rotarod test. All compounds increased swim distance in the forced-swim test. In the elevated zero maze, the GluN2B subtype-selective RO 25-6981 affected none of the measured parameters, whereas all other compounds showed anxiolytic-like effects. In the novelty-induced hypophagia test, citalopram and MK-801 showed anxiogenic-like action. All NMDAR antagonists induced hyperactivity. The high doses of ketamine and MK-801 impaired performance in the modified Y-maze test, whereas S-ketamine and RO 25-6891 showed no effects in this test. Only MK-801 impaired rotarod performance. The study supports that NMDARs could be a possible therapeutic target for treating depression and anxiety. However, selective antagonism of GluN2B subunit-containing NMDARs showed no effect on anxiety-like behaviours in this study.
Collapse
|
42
|
Abstract
SummaryKetamine, a synthetic derivative of phencyclidine, is a commonly misused party drug that is restricted in high-income countries because of its addictive potential. Ketamine is also used as an anaesthetic in human and veterinary medicine. In the 1990s, research using ketamine to study the pathophysiology of schizophrenia was terminated owing to ethical concerns. Recently, controversy surrounding the drug has returned, as researchers have demonstrated that intravenous ketamine infusion has a rapid antidepressant effect and have therefore proposed ketamine as a novel antidepressant. This article debates the question of ketamine as an antidepressant, considering the drug's addictive potential, ethical concerns about prescribing a hallucinogen, the evidence base and motives behind ketamine trials.
Collapse
|
43
|
Engaging homeostatic plasticity to treat depression. Mol Psychiatry 2018; 23:26-35. [PMID: 29133952 DOI: 10.1038/mp.2017.225] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/11/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022]
Abstract
Major depressive disorder (MDD) is a complex and heterogeneous mood disorder, making it difficult to develop a generalized, pharmacological therapy that is effective for all who suffer from MDD. Through the fortuitous discovery of N-methyl-D-aspartate receptor (NMDAR) antagonists as effective antidepressants, we have gained key insights into how antidepressant effects can be produced at the circuit and molecular levels. NMDAR antagonists act as rapid-acting antidepressants such that relief from depressive symptoms occurs within hours of a single injection. The mode of action of NMDAR antagonists seemingly relies on their ability to activate protein-synthesis-dependent homeostatic mechanisms that restore top-down excitatory connections. Recent evidence suggests that NMDAR antagonists relieve depressive symptoms by forming new synapses resulting in increased excitatory drive. This event requires the mammalian target of rapamycin complex 1 (mTORC1), a signaling pathway that regulates synaptic protein synthesis. Herein, we review critical studies that shed light on the action of NMDAR antagonists as rapid-acting antidepressants and how they engage a neuron's or neural network's homeostatic mechanisms to self-correct. Recent studies notably demonstrate that a shift in γ-amino-butyric acid receptor B (GABABR) function, from inhibitory to excitatory, is required for mTORC1-dependent translation with NMDAR antagonists. Finally, we discuss how GABABR activation of mTORC1 helps resolve key discrepancies between rapid-acting antidepressants and local homeostatic mechanisms.
Collapse
|
44
|
Distinct retrosplenial cortex cell populations and their spike dynamics during ketamine-induced unconscious state. PLoS One 2017; 12:e0187198. [PMID: 29073221 PMCID: PMC5658186 DOI: 10.1371/journal.pone.0187198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 10/16/2017] [Indexed: 01/11/2023] Open
Abstract
Ketamine is known to induce psychotic-like symptoms, including delirium and visual hallucinations. It also causes neuronal damage and cell death in the retrosplenial cortex (RSC), an area that is thought to be a part of high visual cortical pathways and at least partially responsible for ketamine's psychotomimetic activities. However, the basic physiological properties of RSC cells as well as their response to ketamine in vivo remained largely unexplored. Here, we combine a computational method, the Inter-Spike Interval Classification Analysis (ISICA), and in vivo recordings to uncover and profile excitatory cell subtypes within layers 2&3 and 5&6 of the RSC in mice within both conscious, sleep, and ketamine-induced unconscious states. We demonstrate two distinct excitatory principal cell sub-populations, namely, high-bursting excitatory principal cells and low-bursting excitatory principal cells, within layers 2&3, and show that this classification is robust over the conscious states, namely quiet awake, and natural unconscious sleep periods. Similarly, we provide evidence of high-bursting and low-bursting excitatory principal cell sub-populations within layers 5&6 that remained distinct during quiet awake and sleep states. We further examined how these subtypes are dynamically altered by ketamine. During ketamine-induced unconscious state, these distinct excitatory principal cell subtypes in both layer 2&3 and layer 5&6 exhibited distinct dynamics. We also uncovered different dynamics of local field potential under various brain states in layer 2&3 and layer 5&6. Interestingly, ketamine administration induced high gamma oscillations in layer 2&3 of the RSC, but not layer 5&6. Our results show that excitatory principal cells within RSC layers 2&3 and 5&6 contain multiple physiologically distinct sub-populations, and they are differentially affected by ketamine.
Collapse
|
45
|
Liu Y, Zhao N, Li C, Chang Q, Liu X, Liao Y, Pan R. Longistyline C acts antidepressant in vivo and neuroprotection in vitro against glutamate-induced cytotoxicity by regulating NMDAR/NR2B-ERK pathway in PC12 cells. PLoS One 2017; 12:e0183702. [PMID: 28873095 PMCID: PMC5584824 DOI: 10.1371/journal.pone.0183702] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 08/09/2017] [Indexed: 01/10/2023] Open
Abstract
Depressive disorder is a common psychiatric disease which ranks among the leading cause of disability worldwide. The antidepressants presently used had low cure rate and caused a variety of side-effects. The screening of antidepressant drugs is usually used classic behavioural tests and neuroprotective strategy. Longistyline C, a natural stilbene isolated from the leaves of Cajanuscajan (L.) Millsp, was firstly investigated the antidepressant effect using animal behavioural tests, and studied the neuroprotection and its possible signaling pathways on glutamate-induced injury in PC12 cells. The results of animal test demonstrated that longistyline C had the antidepressant activity, which the effect is similar to the positive control. In current study, we investigated the effect of longistyline C on glutamate-induced injury in PC12 cells and explored its possible signaling pathways. The results demonstrated that pretreatment with longistyline C at the concentrations of 2-8 μmol/L for 24 h had a significant reduction of the cytotoxicity induced by glutamate (15 mmol/L) in PC12 cells using MTT, lactate dehydrogenase (LDH) release assay and Annexin V-PI double staining. Subsequently, we found that pretreatment with longistyline C (8 μmol/L) could drastically down-regulate the over-expression of NMDAR/NR2B and Ca2+/calmodulin-dependent protein kinase II (CaMKII), up-regulate the expressions of p-ERK and p-CREB and alleviate ER stress. In conclusison, longistyline C is most possibly through regulating NMDAR/NR2B-ERK1/2 related pathway and restoring endoplasmic reticulum function to exert neuroprotective effect against glutamate-induced injury in PC12 cells.
Collapse
Affiliation(s)
- Yamin Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| | - Ning Zhao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| | - Chenchen Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| | - Xinmin Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| | - Yonghong Liao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| | - Ruile Pan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science, Peking Union Medical College, Beijing, China
| |
Collapse
|
46
|
Ishikawa C, Shiga T. The postnatal 5-HT 1A receptor regulates adult anxiety and depression differently via multiple molecules. Prog Neuropsychopharmacol Biol Psychiatry 2017; 78:66-74. [PMID: 28483674 DOI: 10.1016/j.pnpbp.2017.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/14/2017] [Accepted: 04/14/2017] [Indexed: 11/16/2022]
Abstract
Serotonin (5-HT) and the 5-HT1A receptor during development are known to modulate anxiety and depression in later life. However, the brain mechanisms linking the postnatal 5-HT system and adult behavior remain unknown. Here, we examined the effects of pharmacological 5-HT1A receptor activation during the postnatal period on anxiety and depression-like behavior in adult BALB/c male mice. To elucidate the underlying mechanisms, we measured mRNA expression of the 5-HT1A receptor, brain-derived neurotrophic factor (BDNF), GABAA receptor subunits, and AMPA receptor subunits in the medial prefrontal cortex (mPFC), amygdala, and hippocampus. Treatment with the selective 5-HT reuptake inhibitor (fluoxetine) and 5-HT1A receptor agonist (8-OH-DPAT) during the postnatal period decreased anxiety-like behavior in adulthood, whereas only 8-OH-DPAT treatment increased depression-like behavior. Concomitantly with the behavioral effects, postnatal treatment with fluoxetine and 8-OH-DPAT decreased the mRNA expression of the GABAA receptor α3 subunit in the mPFC and ventral hippocampus in adulthood, while 8-OH-DPAT, but not fluoxetine, decreased the mRNA expression of the 5-HT1A receptor and BDNF in the mPFC and the GABAA receptor α2 subunit in the mPFC and ventral hippocampus. On the basis of the correlative changes between behavior and mRNA expression, these results suggest that the GABAA receptor α3 subunit in the mPFC and ventral hippocampus may regulate anxiety-like behavior. In contrast, depression-like behavior may be regulated by the 5-HT1A receptor and BDNF in the mPFC and by the GABAA receptor α2 subunit in the mPFC and ventral hippocampus. In summary, activation of the 5-HT1A receptor during the postnatal period may reduce anxiety levels, but increase depression levels during adulthood via different multiple molecules in the mPFC and ventral hippocampus.
Collapse
Affiliation(s)
- Chihiro Ishikawa
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Takashi Shiga
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan; Department of Neurobiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan.
| |
Collapse
|
47
|
Garay RP, Zarate CA, Charpeaud T, Citrome L, Correll CU, Hameg A, Llorca PM. Investigational drugs in recent clinical trials for treatment-resistant depression. Expert Rev Neurother 2017; 17:593-609. [PMID: 28092469 PMCID: PMC5418088 DOI: 10.1080/14737175.2017.1283217] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The authors describe the medications for treatment-resistant depression (TRD) in phase II/III of clinical development in the EU and USA and provide an opinion on how current treatment can be improved in the near future. Areas covered: Sixty-two trials were identified in US and EU clinical trial registries that included six investigational compounds in recent phase III development and 12 others in recent phase II clinical trials. Glutamatergic agents have been the focus of many studies. A single intravenous dose of the glutamatergic modulator ketamine produces a robust and rapid antidepressant effect in persons with TRD; this effect continues to remain significant for 1 week. This observation was a turning point that opened the way for other, more selective glutamatergic modulators (intranasal esketamine, AVP-786, AVP-923, AV-101, and rapastinel). Of the remaining compounds, monoclonal antibodies open highly innovative therapeutic options, based on new pathophysiological approaches to depression. Expert commentary: Promising new agents are emerging for TRD treatment. Glutamatergic modulators likely represent a very promising alternative to monoaminergic antidepressant monotherapy. We could see the arrival of the first robust and rapid acting antidepressant drug in the near future, which would strongly facilitate the ultimate goal of recovery in persons with TRD.
Collapse
Affiliation(s)
- Ricardo P. Garay
- Pharmacology and Therapeutics, Craven, Villemoisson-sur-Orge, France
| | - Carlos A. Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Charpeaud
- Centre Médico-Psychologique B, CHU, Université d’Auvergne, Clermont-Ferrand, France
| | - Leslie Citrome
- Department of Psychiatry and Behavioral Sciences, New York Medical College, Valhalla, NY, USA
| | - Christoph U. Correll
- Psychiatry Research, Northwell Health, The Zucker Hillside Hospital, Glen Oaks, New York, USA
- Hofstra Northwell Health School of Medicine, Hempstead, New York, USA
| | - Ahcène Hameg
- Pharmacology and Therapeutics, Craven, Villemoisson-sur-Orge, France
| | - Pierre-Michel Llorca
- Centre Médico-Psychologique B, CHU, Université d’Auvergne, Clermont-Ferrand, France
| |
Collapse
|
48
|
Dinis-Oliveira RJ. Metabolism and metabolomics of ketamine: a toxicological approach. Forensic Sci Res 2017; 2:2-10. [PMID: 30483613 PMCID: PMC6197107 DOI: 10.1080/20961790.2017.1285219] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/18/2017] [Indexed: 01/03/2023] Open
Abstract
Ketamine is a phencyclidine derivative and a non-competitive antagonist of N-methyl-D-aspartate (NMDA) receptor for which glutamate is the full agonist. It produces a functional dissociation between the thalamocortical and limbic systems, a state that has been termed as dissociative anaesthesia. Considerable variability in the pharmacokinetics and pharmacodynamics between individuals that can affect dose-response and toxicological profile has been reported. This review aims to discuss pharmacokinetics of ketamine, namely focusing on all major and minor, active and inactive metabolites. Both ketamine optical isomers undergo hepatic biotransformation through the cytochrome P450, specially involving the isoenzymes 3A4 and 2B6. It is first N-demethylated to active metabolite norketamine. Different minor pathways have been described, namely hydroxylation of the cyclohexanone ring of ketamine and norketamine, and further conjugation with glucuronic acid to increase renal excretion. More recently, metabolomics data evidenced the alteration of several biological pathways after ketamine administration such as glycolysis, tricarboxylic acid cycle, amino acids metabolism and mitochondrial β-oxidation of fatty acids. It is expected that knowing the metabolism and metabolomics of ketamine may provide further insights aiming to better characterize ketamine from a clinical and forensic perspective.
Collapse
Affiliation(s)
- Ricardo Jorge Dinis-Oliveira
- Department of Sciences, IINFACTS - Institute of Research and Advanced Training in Health Sciences and Technologies, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal.,Department of Biological Sciences, UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Porto, Portugal.,Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
49
|
Nashed MG, Ungard RG, Young K, Zacal NJ, Seidlitz EP, Fazzari J, Frey BN, Singh G. Behavioural Effects of Using Sulfasalazine to Inhibit Glutamate Released by Cancer Cells: A Novel target for Cancer-Induced Depression. Sci Rep 2017; 7:41382. [PMID: 28120908 PMCID: PMC5264609 DOI: 10.1038/srep41382] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022] Open
Abstract
Despite the lack of robust evidence of effectiveness, current treatment options for cancer-induced depression (CID) are limited to those developed for non-cancer related depression. Here, anhedonia-like and coping behaviours were assessed in female BALB/c mice inoculated with 4T1 mammary carcinoma cells. The behavioural effects of orally administered sulfasalazine (SSZ), a system xc− inhibitor, were compared with fluoxetine (FLX). FLX and SSZ prevented the development of anhedonia-like behaviour on the sucrose preference test (SPT) and passive coping behaviour on the forced swim test (FST). The SSZ metabolites 5-aminosalicylic acid (5-ASA) and sulfapyridine (SP) exerted an effect on the SPT but not on the FST. Although 5-ASA is a known anti-inflammatory agent, neither treatment with SSZ nor 5-ASA/SP prevented tumour-induced increases in serum levels of interleukin-1β (IL-1β) and IL-6, which are indicated in depressive disorders. Thus, the observed antidepressant-like effect of SSZ may primarily be attributable to the intact form of the drug, which inhibits system xc−. This study represents the first attempt at targeting cancer cells as a therapeutic strategy for CID, rather than targeting downstream effects of tumour burden on the central nervous system. In doing so, we have also begun to characterize the molecular pathways of CID.
Collapse
Affiliation(s)
- Mina G Nashed
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Robert G Ungard
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Kimberly Young
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Natalie J Zacal
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Eric P Seidlitz
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Jennifer Fazzari
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, L8N 3K7, Canada.,Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, L8P 3K7, Canada
| | - Gurmit Singh
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
50
|
Ketamine modulates hippocampal neurogenesis and pro-inflammatory cytokines but not stressor induced neurochemical changes. Neuropharmacology 2017; 112:210-220. [DOI: 10.1016/j.neuropharm.2016.04.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 04/13/2016] [Accepted: 04/17/2016] [Indexed: 12/28/2022]
|