51
|
Fagerholm ED, Leech R, Williams S, Zarate CA, Moran RJ, Gilbert JR. Fine-tuning neural excitation/inhibition for tailored ketamine use in treatment-resistant depression. Transl Psychiatry 2021; 11:335. [PMID: 34052834 PMCID: PMC8164631 DOI: 10.1038/s41398-021-01442-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/26/2022] Open
Abstract
The glutamatergic modulator ketamine has been shown to rapidly reduce depressive symptoms in patients with treatment-resistant major depressive disorder (TRD). Although its mechanisms of action are not fully understood, changes in cortical excitation/inhibition (E/I) following ketamine administration are well documented in animal models and could represent a potential biomarker of treatment response. Here, we analyse neuromagnetic virtual electrode time series collected from the primary somatosensory cortex in 18 unmedicated patients with TRD and in an equal number of age-matched healthy controls during a somatosensory 'airpuff' stimulation task. These two groups were scanned as part of a clinical trial of ketamine efficacy under three conditions: (a) baseline; (b) 6-9 h following subanesthetic ketamine infusion; and (c) 6-9 h following placebo-saline infusion. We obtained estimates of E/I interaction strengths by using dynamic causal modelling (DCM) on the time series, thereby allowing us to pinpoint, under each scanning condition, where each subject's dynamics lie within the Poincaré diagram-as defined in dynamical systems theory. We demonstrate that the Poincaré diagram offers classification capability for TRD patients, in that the further the patients' coordinates were shifted (by virtue of ketamine) toward the stable (top-left) quadrant of the Poincaré diagram, the more their depressive symptoms improved. The same relationship was not observed by virtue of a placebo effect-thereby verifying the drug-specific nature of the results. We show that the shift in neural dynamics required for symptom improvement necessitates an increase in both excitatory and inhibitory coupling. We present accompanying MATLAB code made available in a public repository, thereby allowing for future studies to assess individually tailored treatments of TRD.
Collapse
Affiliation(s)
| | - Robert Leech
- Department of Neuroimaging, King's College London, London, UK
| | - Steven Williams
- Department of Neuroimaging, King's College London, London, UK
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, NIMH, NIH, Bethesda, MD, USA
| | - Rosalyn J Moran
- Department of Neuroimaging, King's College London, London, UK
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, NIMH, NIH, Bethesda, MD, USA.
| |
Collapse
|
52
|
Filatova EV, Shadrina MI, Slominsky PA. Major Depression: One Brain, One Disease, One Set of Intertwined Processes. Cells 2021; 10:cells10061283. [PMID: 34064233 PMCID: PMC8224372 DOI: 10.3390/cells10061283] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 01/18/2023] Open
Abstract
Major depressive disorder (MDD) is a heterogeneous disease affecting one out of five individuals and is the leading cause of disability worldwide. Presently, MDD is considered a multifactorial disease with various causes such as genetic susceptibility, stress, and other pathological processes. Multiple studies allowed the formulation of several theories attempting to describe the development of MDD. However, none of these hypotheses are comprehensive because none of them can explain all cases, mechanisms, and symptoms of MDD. Nevertheless, all of these theories share some common pathways, which lead us to believe that these hypotheses depict several pieces of the same big puzzle. Therefore, in this review, we provide a brief description of these theories and their strengths and weaknesses in an attempt to highlight the common mechanisms and relationships of all major theories of depression and combine them together to present the current overall picture. The analysis of all hypotheses suggests that there is interdependence between all the brain structures and various substances involved in the pathogenesis of MDD, which could be not entirely universal, but can affect all of the brain regions, to one degree or another, depending on the triggering factor, which, in turn, could explain the different subtypes of MDD.
Collapse
|
53
|
Ershadi ASB, Amini-Khoei H, Hosseini MJ, Dehpour AR. SAHA Improves Depressive Symptoms, Cognitive Impairment and Oxidative Stress: Rise of a New Antidepressant Class. Neurochem Res 2021; 46:1252-1263. [PMID: 33576938 DOI: 10.1007/s11064-021-03263-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/30/2020] [Accepted: 01/30/2021] [Indexed: 12/28/2022]
Abstract
Depression is a disabling psychiatric disorder affecting millions of people all around the world. Under current therapeutic choices, a portion of patients are not responsive, have relapses, or experience cognitive side effects. Hence, the present study aimed to find other antidepressant compounds lacking the mentioned deficiency. Since epigenetic regulations have attracted more attention in etiology of depression, histone deacetylase (HDAC) inhibitors have gained more importance due to their possible antidepressant activity. We selected a promising member of HDAC inhibitors named suberanilohydroxamic acid (SAHA) to evaluate its antidepressant properties. Early life stress disarrays many neurodevelopmental factors and consequently, leads to the destruction of hippocampus and prefrontal cortex synapses as areas highly related to emotion and memory so that any destruction on them can cause lasting impairments. For that reason, we used maternal separation (MS) paradigm to investigate depression in male mice. To compare the efficacy of SAHA with current treatment options, we also treated a group of MS mice with fluoxetine (FLX) as first-line pharmacological drugs of depression. The results demonstrated that depressive-like behavior, cognitive function and inflammatory response of MS mice were attenuated with SAHA. Our data showed that, besides anti-depressant and cognition-boosting effects similar to FLX, SAHA counteracted inflammatory response caused by depression and reversed the coenzyme Q10 (CoQ10) level in hippocampus. SAHA's effect on alleviating depressive behavior was accompanied with memory enhancement and hippocampus biochemical tests. These findings may propose SAHA as another therapeutic option for depressive symptoms, especially with comorbid cognitive impairment.
Collapse
Affiliation(s)
- Amir Sasan Bayani Ershadi
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, P. O. Box: 45139-56184, Zanjan, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, P. O. Box: 88138-33435, Shahrekord, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, P. O. Box: 45139-56184, Zanjan, Iran.
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, P.O. Box: 13145-784, Tehran, Iran.
| |
Collapse
|
54
|
Misganaw D. Heteromerization of dopaminergic receptors in the brain: Pharmacological implications. Pharmacol Res 2021; 170:105600. [PMID: 33836279 DOI: 10.1016/j.phrs.2021.105600] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/17/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
Dopamine exerts its physiological effects through two subtypes of receptors, i.e. the receptors of the D1 family (D1R and D5R) and the D2 family (D2R, D3R, and D4R), which differ in their pattern of distribution, affinity, and signaling. The D1-like subfamily (D1R and D5R) are coupled to Gαs/olf proteins to activate adenylyl cyclase whereas the D2-like receptors are coupled to Gαi/o subunits and suppress the activity of adenylyl cyclase. Dopamine receptors are capable of forming homodimers, heterodimers, and higher-order oligomeric complexes, resulting in a change in the individual protomers' recognition, signaling, and pharmacology. Heteromerization has the potential to modify the canonical pharmacological features of individual monomeric units such as ligand affinity, activation, signaling, and cellular trafficking through allosteric interactions, reviving the field and introducing a new pharmacological target. Since heteromers are expressed and formed in a tissue-specific manner, they could provide the framework to design selective and effective drug candidates, such as brain-penetrant heterobivalent drugs and interfering peptides, with limited side effects. Therefore, heteromerization could be a promising area of pharmacology research, as it could contribute to the development of novel pharmacological interventions for dopamine dysregulated brain disorders such as addiction, schizophrenia, cognition, Parkinson's disease, and other motor-related disorders. This review is articulated based on the three criteria established by the International Union of Basic and Clinical Pharmacology for GPCR heterodimers (IUPHAR): evidence of co-localization and physical interactions in native or primary tissue, presence of a new physiological and functional property than the individual protomers, and loss of interaction and functional fingerprints upon heterodimer disruption.
Collapse
Affiliation(s)
- Desye Misganaw
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Medicine and Health Science, Wollo University, P.O. Box 1145, Dessie, Ethiopia.
| |
Collapse
|
55
|
Dos Santos RG, Hallak JE, Baker G, Dursun S. Hallucinogenic/psychedelic 5HT2A receptor agonists as rapid antidepressant therapeutics: Evidence and mechanisms of action. J Psychopharmacol 2021; 35:453-458. [PMID: 33740877 DOI: 10.1177/0269881120986422] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Major depressive disorder (MDD) is among the most prevalent mental health disorders worldwide, and it is associated with a reduced quality of life and enormous costs to health care systems. Available drug treatments show low-to-moderate response in most patients, with almost a third of patients being non-responders (treatment-resistant). Furthermore, most currently available medications need several weeks to achieve therapeutic effects, and the long-term use of these drugs is often associated with significant unwanted side effects and resultant reductions in treatment compliance. Therefore, more effective, safer, and faster-acting antidepressants with enduring effects are needed. Together with ketamine, psychedelics (or classic or serotoninergic hallucinogens) such as lysergic acid diethylamide (LSD), psilocybin, and ayahuasca are among the few compounds with recent human evidence of fast-acting antidepressant effects. Several studies in the 1950s to 1970s reported antidepressive and anxiolytic effects of these drugs, which are being confirmed by modern trials (LSD, one trial; psilocybin, five trials; ayahuasca, two trials). The effects of these drugs appear to be produced primarily by their agonism at serotonin (5-hydroxytryptamine, 5-HT) receptors, especially the 5-HT2A receptor. Considering the overall burden of MDD and the necessity of new therapeutic options, the promising (but currently limited) evidence of safety and efficacy of psychedelics has encouraged the scientific community to explore more fully their beneficial effects in MDD.
Collapse
Affiliation(s)
- Rafael Guimarães Dos Santos
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil.,National Institute for Translational Medicine (INCT-TM), CNPq, Brazil
| | - Jaime Ec Hallak
- Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, SP, Brazil.,National Institute for Translational Medicine (INCT-TM), CNPq, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Glen Baker
- National Institute for Translational Medicine (INCT-TM), CNPq, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Serdar Dursun
- National Institute for Translational Medicine (INCT-TM), CNPq, Brazil.,Department of Psychiatry (Neurochemical Research Unit) and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
56
|
Heinrich IA, Freitas AE, Wolin IAV, Nascimento APM, Walz R, Rodrigues ALS, Leal RB. Neuronal activity regulated pentraxin (narp) and GluA4 subunit of AMPA receptor may be targets for fluoxetine modulation. Metab Brain Dis 2021; 36:711-722. [PMID: 33528752 DOI: 10.1007/s11011-021-00675-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/22/2021] [Indexed: 12/28/2022]
Abstract
Fluoxetine is the foremost prescribed antidepressant. Drugs acting on monoaminergic system may also regulate glutamatergic system. Indeed, the investigation of proteins associated with this system, such as Narp (neuronal activity-dependent pentraxin) and GluA4 subunit of AMPA receptor may reveal poorly explored modulations triggered by conventional antidepressants. This study aimed to uncover neurochemical mechanisms underlying the chronic fluoxetine treatment, mainly by evaluating these protein targets in the prefrontal cortex and in the hippocampus. Mice received a daily administration of fluoxetine (0.1, 1 or 10 mg/kg, p.o.) or potable water (vehicle group) for 21 days. These animals were submitted to the forced swim test (FST) to verify antidepressant-like responses and the open-field test (OFT) to assess locomotor activity. Modulation of signaling proteins was analyzed by western blot. Chronic treatment with fluoxetine (1 and 10 mg/kg) was effective, since it reduced the immobility time in the FST, without altering locomotor activity. Fluoxetine 10 mg/kg increased CREB phosphorylation and BDNF expression in the prefrontal cortex and hippocampus. Noteworthy, in the hippocampus fluoxetine also promoted Akt activation and augmented Narp expression. In the prefrontal cortex, a significant decrease in the expression of the GluA4 subunit and Narp were observed following fluoxetine administration (10 mg/kg). The results provide evidence of novel molecular targets potentially involved in the antidepressant effects of fluoxetine, since in mature rodents Narp and GluA4 are mainly expressed in the GABAergic parvalbumin-positive (PV+) interneurons. This may bring new insights into the molecular elements involved in the mechanisms underlying the antidepressant effects of fluoxetine.
Collapse
Affiliation(s)
- Isabella A Heinrich
- Graduate Program in Neuroscience, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Andiara E Freitas
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Ingrid A V Wolin
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Ana Paula M Nascimento
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Roger Walz
- Graduate Program in Neuroscience, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Department of Clinical Medicine, Center of Health Sciences, University Hospital, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Center of Applied Neuroscience (CeNAp), University Hospital, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Graduate Program in Neuroscience, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Rodrigo B Leal
- Graduate Program in Neuroscience, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil.
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil.
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil.
| |
Collapse
|
57
|
Vecchia DD, Kanazawa LKS, Wendler E, Hocayen PDAS, Vital MABF, Takahashi RN, Da Cunha C, Miyoshi E, Andreatini R. Ketamine reversed short-term memory impairment and depressive-like behavior in animal model of Parkinson's disease. Brain Res Bull 2021; 168:63-73. [PMID: 33359641 DOI: 10.1016/j.brainresbull.2020.12.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/01/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022]
Abstract
The most common features of Parkinson's disease (PD) are motor impairments, but many patients also present depression and memory impairment. Ketamine, an N-methyl-d-aspartate (NMDA) receptor antagonist, has been shown to be effective in patients with treatment-resistant major depression. Thus, the present study evaluated the action of ketamine on memory impairment and depressive-like behavior in an animal model of PD. Male Wistar rats received a bilateral infusion of 6 μg/side 6-hydroxydopamine (6-OHDA) into the substantia nigra pars compacta (SNc). Short-term memory was evaluated by the social recognition test, and depressive-like behaviors were evaluated by the sucrose preference and forced swimming tests (FST). Drug treatments included vehicle (i.p., once a week); ketamine (5, 10 and 15 mg/kg, i.p., once a week); and imipramine (20 mg/kg, i.p., daily). The treatments were administered 21 days after the SNc lesion and lasted for 28 days. The SNc lesion impaired short-term social memory, and all ketamine doses reversed the memory impairment and anhedonia (reduction of sucrose preference) induced by 6-OHDA. In the FST, 6-OHDA increased immobility, and all doses of ketamine and imipramine reversed this effect. The anti-immobility effect of ketamine was associated with an increase in swimming but not in climbing, suggesting a serotonergic effect. Ketamine and imipramine did not reverse the 6-OHDA-induced reduction in tyrosine hydroxylase immunohistochemistry in the SNc. In conclusion, ketamine reversed depressive-like behaviors and short-term memory impairment in rats with SNc bilateral lesions, indicating a promising profile for its use in PD patients.
Collapse
Affiliation(s)
- Débora Dalla Vecchia
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil; Uniandrade, Centro Universitário Campos de Andrade, Santa Quiteria, 80310-310, Curitiba, PR, Brazil
| | - Luiz Kae Sales Kanazawa
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil
| | - Etiéli Wendler
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil; Uniandrade, Centro Universitário Campos de Andrade, Santa Quiteria, 80310-310, Curitiba, PR, Brazil
| | - Palloma de Almeida Soares Hocayen
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil
| | - Maria Aparecida Barbato Frazão Vital
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil
| | - Reinaldo Naoto Takahashi
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Trindade, 88049-900, Florianópolis, SC, Brazil
| | - Claudio Da Cunha
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil
| | - Edmar Miyoshi
- Department of Pharmaceutical Sciences, State University of Ponta Grossa, Avenida General Carlos Cavalcanti 4748, 84030-900, Ponta Grossa, PR, Brazil
| | - Roberto Andreatini
- Department of Pharmacology, Setor de Ciências Biológicas, Universidade Federal do Paraná, Centro Politécnico, C.P. 19031, 81531-980, Curitiba, PR, Brazil.
| |
Collapse
|
58
|
Abstract
The therapeutic onset of traditional antidepressants is delayed by several weeks and many depressed patients fail to respond to treatment altogether. In contrast, subanesthetic ketamine can rapidly alleviate symptoms of depression within hours of a single administration, even in patients who are considered treatment-resistant. Ketamine is thought to exert these effects by restoring the integrity of neural circuits that are compromised in depression. This hypothesis stems in part from preclinical observations that ketamine can strengthen synaptic connections by increasing glutamate-mediated neurotransmission and promoting rapid neurotrophic factor release. An improved understanding of how ketamine, and other novel rapid-acting antidepressants, give rise to these processes will help foster future therapeutic innovation. Here, we review the history of antidepressant treatment advances that preceded the ketamine discovery, critically examine mechanistic hypotheses for how ketamine may exert its antidepressant effects, and discuss the impact this knowledge has had on ongoing drug discovery efforts.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA; .,Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA; .,Departments of Pharmacology and Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.,Baltimore Veterans Affairs Medical Center, Veterans Affairs Maryland Health Care System, Baltimore, Maryland 21201, USA
| |
Collapse
|
59
|
Tayab MA, Chowdhury KAA, Jabed M, Mohammed Tareq S, Kamal ATMM, Islam MN, Uddin AMK, Hossain MA, Emran TB, Simal-Gandara J. Antioxidant-Rich Woodfordia fruticosa Leaf Extract Alleviates Depressive-Like Behaviors and Impede Hyperglycemia. PLANTS (BASEL, SWITZERLAND) 2021; 10:287. [PMID: 33546288 PMCID: PMC7913287 DOI: 10.3390/plants10020287] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/12/2021] [Accepted: 01/26/2021] [Indexed: 12/19/2022]
Abstract
Dhaiphul (Woodfordia fruticosa) is a frequently demanded plant in South-East Asian regions for its diverse medicinal values. This study was proposed to examine antioxidant, antidiabetic, and antidepressant potentials of methanol extract of W. fruticosa leaves (MEWF) and its derived n-hexane (NHFMEWF) and ethyl acetate (EAFMEWF) fractions through in vitro, in vivo, and computational models. Among test samples, MEWF and EAFMEWF contained the highest phenolic content and showed maximal antioxidant activity in DPPH radical scavenging and ferric reducing power assays. In comparison, NHFMEWF possessed maximum flavonoid content and a significantly potent α-amylase inhibitory profile comparable with positive control acarbose. In animal models of depression (forced swimming and tail suspension test), EAFMEWF and NHFMEWF demonstrated a dose-dependent antidepressant-like effect; explicitly, the depressive-like behaviors significantly declined in EAFMEWF-treated dosing groups in contrast to the control group. In the computational analysis, previously isolated flavonoid compounds from Dhaiphul leaves manifested potent binding affinity against several key therapeutic target proteins of diabetes and depressive disorders including α-amylase, serotonin transporter, dopamine transporter, and neuronal nitric oxide synthase with varying pharmacokinetics and toxicity profiles. This research's outcomes may provide potential dietary supplements for mitigating hyperglycemia, cellular toxicity, and depressive disorder.
Collapse
Affiliation(s)
- Mohammed Abu Tayab
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (M.A.T.); (K.A.A.C.); (M.J.); (S.M.T.); (A.T.M.M.K.); (A.M.K.U.); (M.A.H.)
| | - Kazi Ashfak Ahmed Chowdhury
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (M.A.T.); (K.A.A.C.); (M.J.); (S.M.T.); (A.T.M.M.K.); (A.M.K.U.); (M.A.H.)
| | - Md. Jabed
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (M.A.T.); (K.A.A.C.); (M.J.); (S.M.T.); (A.T.M.M.K.); (A.M.K.U.); (M.A.H.)
| | - Syed Mohammed Tareq
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (M.A.T.); (K.A.A.C.); (M.J.); (S.M.T.); (A.T.M.M.K.); (A.M.K.U.); (M.A.H.)
| | - A. T. M. Mostafa Kamal
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (M.A.T.); (K.A.A.C.); (M.J.); (S.M.T.); (A.T.M.M.K.); (A.M.K.U.); (M.A.H.)
| | - Mohammad Nazmul Islam
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (M.A.T.); (K.A.A.C.); (M.J.); (S.M.T.); (A.T.M.M.K.); (A.M.K.U.); (M.A.H.)
| | - A. M. Kafil Uddin
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (M.A.T.); (K.A.A.C.); (M.J.); (S.M.T.); (A.T.M.M.K.); (A.M.K.U.); (M.A.H.)
| | - Mohammad Adil Hossain
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; (M.A.T.); (K.A.A.C.); (M.J.); (S.M.T.); (A.T.M.M.K.); (A.M.K.U.); (M.A.H.)
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo—Ourense Campus, E32004 Ourense, Spain
| |
Collapse
|
60
|
Gill H, Gill B, Rodrigues NB, Lipsitz O, Rosenblat JD, El-Halabi S, Nasri F, Mansur RB, Lee Y, McIntyre RS. The Effects of Ketamine on Cognition in Treatment-Resistant Depression: A Systematic Review and Priority Avenues for Future Research. Neurosci Biobehav Rev 2021; 120:78-85. [PMID: 33242561 DOI: 10.1016/j.neubiorev.2020.11.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/12/2020] [Accepted: 11/12/2020] [Indexed: 12/28/2022]
Abstract
Replicated evidence has documented cognitive deficits in populations with treatment-resistant depression (TRD). Approximately 40 % of patients with MDD present with impairment of one or more cognitive domains. As such, there is an unmet need to discover treatments that have pro-cognitive effects in TRD patients. Ketamine has demonstrated efficacy as a rapid-onset intervention for the treatment of depression. The objective of the current review was to assess the effects of ketamine on cognition in TRD patients. We systematically searched PubMed, Google Scholar and PsycINFO between database inception to March 24th, 2020. We identified five studies that evaluated cognition in TRD populations following ketamine treatment. All studies included a 0.5 mg/kg subanesthetic intravenous (IV) administration of ketamine. One study found significant improvements in complex (p = .008) and simple (p = .027) working memory and one study found improvements in visual learning memory following IV ketamine infusions (p = .014). Improvements in speed of processing and verbal learning memory were observed in anxious TRD participants only. Importantly, a subanesthetic dose of IV ketamine does not worsen cognitive function.
Collapse
Affiliation(s)
- Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Barjot Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Nelson B Rodrigues
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Orly Lipsitz
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Joshua Daniel Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Sabine El-Halabi
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Flora Nasri
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Yena Lee
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada.
| |
Collapse
|
61
|
Peng G, Yang L, Wu CY, Zhang LL, Wu CY, Li F, Shi HW, Hou J, Zhang LM, Ma X, Xiong J, Pan H, Zhang GQ. Whole body vibration training improves depression-like behaviors in a rat chronic restraint stress model. Neurochem Int 2021; 142:104926. [PMID: 33276022 DOI: 10.1016/j.neuint.2020.104926] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022]
Abstract
Major depressive disorder (MDD) is a prevalent psychiatric disorder that brings great harm and burden to both patients and society. This study aimed to examine the effects of whole-body vibration (WBV) training on a chronic restraint stress (CRS) induced depression rat model and provide an initial understanding of related molecular mechanisms. Adult Sprague-Dawley male rats were randomly divided into the following three groups: a) control group, b) depressive disorder group, and c) depression with WBV training treatment group. Daily food intake, body weight, sucrose preference test, open field test, elevated plus maze, forced swimming test, and Barnes maze task tests were performed. Immunofluorescence staining and ELISA analysis were used to assess neuronal damage, synaptic proteins, glial cells, and trophic factors. The data of behavioral tests and related biochemical indicators were statistically analyzed and compared between groups. Rats undergoing CRS showed increased anxiety-like behavior and memory impairment, along with synaptic atrophy and neuronal degeneration. WBV could reverse behavioral dysfunction, inhibit the degeneration of neurons, alleviate the damage of neurons and the pathological changes of glial cells, enhance trophic factor expression, and ameliorate the downregulation of dendritic and synaptic proteins after CRS. The effect of WBV in rats may be mediated via the reduction of hippocampal neuronal degeneration and by improving expression of synaptic proteins. WBV training exerts multifactorial benefits on MDD that supports its use as a promising new therapeutic option for improving depression-like behaviors in the depressive and/or potentially depressive.
Collapse
Affiliation(s)
- Guangcong Peng
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Luodan Yang
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Chong Y Wu
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Ling L Zhang
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Chun Y Wu
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Fan Li
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Hai W Shi
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Jun Hou
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Li M Zhang
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Xu Ma
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Jing Xiong
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China
| | - Hongying Pan
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China.
| | - Guang Q Zhang
- Cognitive & Sports Neuroscience Laboratory, National Demonstration Center for Experimental Sports Science Education, College of Physical Education and Sports Science, South China Normal University, China.
| |
Collapse
|
62
|
Sydnor VJ, Lyall AE, Cetin-Karayumak S, Cheung JC, Felicione JM, Akeju O, Shenton ME, Deckersbach T, Ionescu DF, Pasternak O, Cusin C, Kubicki M. Studying pre-treatment and ketamine-induced changes in white matter microstructure in the context of ketamine's antidepressant effects. Transl Psychiatry 2020; 10:432. [PMID: 33319774 PMCID: PMC7738670 DOI: 10.1038/s41398-020-01122-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/23/2020] [Accepted: 11/16/2020] [Indexed: 12/28/2022] Open
Abstract
Ketamine is increasingly being used as a therapeutic for treatment-resistant depression (TRD), yet the effects of ketamine on the human brain remain largely unknown. This pilot study employed diffusion magnetic resonance imaging (dMRI) to examine relationships between ketamine treatment and white matter (WM) microstructure, with the aim of increasing the current understanding of ketamine's neural mechanisms of action in humans. Longitudinal dMRI data were acquired from 13 individuals with TRD two hours prior to (pre-infusion), and four hours following (post-infusion), an intravenous ketamine infusion. Free-water imaging was employed to quantify cerebrospinal fluid-corrected mean fractional anisotropy (FA) in 15 WM bundles pre- and post-infusion. Analyses revealed that higher pre-infusion FA in the left cingulum bundle and the left superior longitudinal fasciculus was associated with greater depression symptom improvement 24 h post-ketamine. Moreover, four hours after intravenous administration of ketamine, FA rapidly increased in numerous WM bundles in the brain; this increase was significantly associated with 24 h symptom improvement in select bundles. Overall, the results of this preliminary study suggest that WM properties, as measured by dMRI, may have a potential impact on clinical improvement following ketamine. Ketamine administration additionally appears to be associated with rapid WM diffusivity changes, suggestive of rapid changes in WM microstructure. This study thus points to pre-treatment WM structure as a potential factor associated with ketamine's clinical efficacy, and to post-treatment microstructural changes as a candidate neuroimaging marker of ketamine's cellular mechanisms.
Collapse
Affiliation(s)
- Valerie J. Sydnor
- Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Amanda E. Lyall
- Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA ,grid.32224.350000 0004 0386 9924Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Suheyla Cetin-Karayumak
- Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Joey C. Cheung
- grid.32224.350000 0004 0386 9924Depression Clinical and Research Program (DCRP), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Julia M. Felicione
- grid.32224.350000 0004 0386 9924Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Boston, MA USA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Martha E. Shenton
- Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA ,Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA ,grid.410370.10000 0004 4657 1992VA Boston Healthcare System, Brockton Division, Brockton, MA USA
| | - Thilo Deckersbach
- grid.32224.350000 0004 0386 9924Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Dawn F. Ionescu
- grid.32224.350000 0004 0386 9924Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.32224.350000 0004 0386 9924Depression Clinical and Research Program (DCRP), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Ofer Pasternak
- Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA ,Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Cristina Cusin
- grid.32224.350000 0004 0386 9924Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.32224.350000 0004 0386 9924Depression Clinical and Research Program (DCRP), Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Marek Kubicki
- Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
63
|
Rana T, Behl T, Sehgal A, Srivastava P, Bungau S. Unfolding the Role of BDNF as a Biomarker for Treatment of Depression. J Mol Neurosci 2020; 71:2008-2021. [PMID: 33230708 DOI: 10.1007/s12031-020-01754-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/08/2020] [Indexed: 12/13/2022]
Abstract
Depression is a well-known disabling mental illness characterized by sadness, loss of interest in activities, and decreased energy. The symptoms of depression are usually recurrent in vulnerable individuals, and persistence of symptoms significantly impairs individuals' quality of life. The exact pathophysiology of depression remains ambiguous, though many hypotheses have been proposed. Brain-derived neurotrophic factor (BDNF) has recently been reported to play a vital role in the pathophysiology of depression. BDNF is an important neurotrophic factor found in the human brain and is involved in neuronal growth and proliferation, synaptic neurotransmission, and neuroplasticity. The neurotrophic theory of depression proposes that depression results from reduced BDNF levels in the brain, which can be treated with antidepressants to alleviate depressive behavior and increase BDNF levels. The aim of this review is to provide broad insight into the role of BDNF in the pathogenesis of depression and in antidepressant therapy. The studies mentioned in this review article greatly support the role of BDNF in the pathogenesis of depression and treatment of this disorder with antidepressants. Since abnormalities in BDNF levels lead to the production of diverse insults that amplify the development or progression of depression, it is important to study and explore BDNF impairment in relation to depression, neuroplasticity, and neurogenesis, and increasing BDNF levels through antidepressant therapy, showing positive response in the management of depression.
Collapse
Affiliation(s)
- Tarapati Rana
- Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | | | - Simona Bungau
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
64
|
Frizzo ME, Ohno Y. Perisynaptic astrocytes as a potential target for novel antidepressant drugs. J Pharmacol Sci 2020; 145:60-68. [PMID: 33357781 DOI: 10.1016/j.jphs.2020.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence suggests that dysfunctions in glutamatergic signaling are associated with the pathophysiology of depression. Several molecules that act on glutamate binding sites, so-called glutamatergic modulators, are rapid-acting antidepressants that stimulate synaptogenesis. Their antidepressant response involves the elevation of both extracellular glutamate and brain-derived neurotrophic factor (BDNF) levels, as well as the postsynaptic activation of the mammalian target of rapamycin complex 1. The mechanisms involved in the antidepressant outcomes of glutamatergic modulators, including ketamine, suggest that astrocytes must be considered a cellular target for developing rapid-acting antidepressants. It is well known that extracellular glutamate levels and glutamate intrasynaptic time-coursing are maintained by perisynaptic astrocytes, where inwardly rectifying potassium channels 4.1 (Kir4.1 channels) regulate both potassium and glutamate uptake. In addition, ketamine reduces membrane expression of Kir4.1 channels, which raises extracellular potassium and glutamate levels, increasing postsynaptic neural activities. Furthermore, inhibition of Kir4.1 channels stimulates BDNF expression in astrocytes, which may enhance synaptic connectivity. In this review, we discuss glutamatergic modulators' actions in regulating extracellular glutamate and BDNF levels, and reinforce the importance of perisynaptic astrocytes for the development of novel antidepressant drugs.
Collapse
Affiliation(s)
- Marcos E Frizzo
- Department of Morphological Sciences, Federal University of Rio Grande Do Sul, Sarmento Leite Street, 500, CEP 90050-170, Porto Alegre, Brazil.
| | - Yukihiro Ohno
- Department of Pharmacology, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka, Japan
| |
Collapse
|
65
|
Neurobiological biomarkers of response to ketamine. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 89:195-235. [PMID: 32616207 DOI: 10.1016/bs.apha.2020.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As a field, psychiatry is undergoing an exciting paradigm shift toward early identification and intervention that will likely minimize both the burden associated with severe mental illnesses as well as their duration. In this context, the rapid-acting antidepressant ketamine has revolutionized our understanding of antidepressant response and greatly expanded the pharmacologic armamentarium for treatment-resistant depression. Efforts to characterize biomarkers of ketamine response support a growing emphasis on early identification, which would allow clinicians to identify biologically enriched subgroups with treatment-resistant depression who are more likely to benefit from ketamine therapy. This chapter presents a broad overview of a range of translational biomarkers, including those drawn from imaging and electrophysiological studies, sleep and circadian rhythms, and HPA axis/endocrine function as well as metabolic, immune, (epi)genetic, and neurotrophic biomarkers related to ketamine response. Ketamine's unique, rapid-acting properties may serve as a model to explore a whole new class of novel rapid-acting treatments with the potential to revolutionize drug development and discovery. However, it should be noted that although several of the biomarkers reviewed here provide promising insights into ketamine's mechanism of action, most studies have focused on acute rather than longer-term antidepressant effects and, at present, none of the biomarkers are ready for clinical use.
Collapse
|
66
|
Aleksandrova LR, Wang YT, Phillips AG. Ketamine and its metabolite, (2R,6R)-HNK, restore hippocampal LTP and long-term spatial memory in the Wistar-Kyoto rat model of depression. Mol Brain 2020; 13:92. [PMID: 32546197 PMCID: PMC7296711 DOI: 10.1186/s13041-020-00627-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/28/2020] [Indexed: 12/28/2022] Open
Abstract
Accumulating evidence implicates dysregulation of hippocampal synaptic plasticity in the pathophysiology of depression. However, the effects of ketamine on synaptic plasticity and their contribution to its mechanism of action as an antidepressant, are still unclear. We investigated ketamine's effects on in vivo dorsal hippocampal (dHPC) synaptic plasticity and their role in mediating aspects of antidepressant activity in the Wistar-Kyoto (WKY) model of depression. dHPC long-term potentiation (LTP) was significantly impaired in WKY rats compared to Wistar controls. Importantly, a single low dose (5 mg/kg, ip) of ketamine or its metabolite, (2R,6R)-HNK, rescued the LTP deficit in WKY rats at 3.5 h but not 30 min following injection, with residual effects at 24 h, indicating a delayed, sustained facilitatory effect on dHPC synaptic plasticity. Consistent with the observed dHPC LTP deficit, WKY rats exhibited impaired hippocampal-dependent long-term spatial memory as measured by the novel object location recognition test (NOLRT), which was effectively restored by pre-treatment with both ketamine or (2R,6R)-HNK. In contrast, in WKYs, which display abnormal stress coping, ketamine, but not (2R,6R)-HNK, had rapid and sustained effects in the forced swim test (FST), a commonly used preclinical screen for antidepressant-like activity. The differential effects of (2R,6R)-HNK observed here reveal a dissociation between drug effects on FST immobility and dHPC synaptic plasticity. Therefore, in the WKY rat model, restoring dHPC LTP was not correlated with ketamine's effects in FST, but importantly, may have contributed to the reversal of hippocampal-dependent cognitive deficits, which are critical features of clinical depression. Our findings support the theory that ketamine may reverse the stress-induced loss of connectivity in key neural circuits by engaging synaptic plasticity processes to "reset the system".
Collapse
Affiliation(s)
- Lily R Aleksandrova
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Yu Tian Wang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Anthony G Phillips
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
67
|
Widman AJ, McMahon LL. Effects of ketamine and other rapidly acting antidepressants on hippocampal excitatory and inhibitory transmission. ADVANCES IN PHARMACOLOGY 2020; 89:3-41. [PMID: 32616211 DOI: 10.1016/bs.apha.2020.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A single sub-anesthetic intravascular dose of the use-dependent NMDAR antagonist, ketamine, improves mood in patients with treatment resistant depression within hours that can last for days, creating an entirely new treatment strategy for the most seriously ill patients. However, the psychomimetic effects and abuse potential of ketamine require that new therapies be developed that maintain the rapid antidepressant effects of ketamine without the unwanted side effects. This necessitates a detailed understanding of what cellular and synaptic mechanisms are immediately activated once ketamine reaches the brain that triggers the needed changes to elicit the improved behavior. Intense research has centered on the effects of ketamine, and the other rapidly acting antidepressants, on excitatory and inhibitory circuits in hippocampus and medial prefrontal cortex to determine common mechanisms, including key modifications in synaptic transmission and the precise location of the NMDARs that mediate the rapid and sustained antidepressant response. We review data comparing the effects of ketamine with other NMDAR receptor modulators and the muscarinic M1 acetylcholine receptor antagonist, scopolamine, together with evidence supporting the disinhibition hypothesis and the direct inhibition hypothesis of ketamine's mechanism of action on synaptic circuits using preclinical models.
Collapse
Affiliation(s)
- Allie J Widman
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lori L McMahon
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
68
|
Kokane SS, Armant RJ, Bolaños-Guzmán CA, Perrotti LI. Overlap in the neural circuitry and molecular mechanisms underlying ketamine abuse and its use as an antidepressant. Behav Brain Res 2020; 384:112548. [PMID: 32061748 PMCID: PMC7469509 DOI: 10.1016/j.bbr.2020.112548] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/18/2020] [Accepted: 02/07/2020] [Indexed: 02/09/2023]
Abstract
Ketamine, a dissociative anesthetic and psychedelic compound, has revolutionized the field of psychopharmacology by showing robust, and rapid-acting antidepressant activity in patients suffering from major depressive disorder (MDD), suicidal tendencies, and treatment-resistant depression (TRD). Ketamine's efficacy, however, is transient, and patients must return to the clinic for repeated treatment as they experience relapse. This is cause for concern because ketamine is known for its abuse liability, and repeated exposure to drugs of abuse often leads to drug abuse/dependence. Though the mechanism(s) underlying its antidepressant activity is an area of current intense research, both clinical and preclinical evidence shows that ketamine's effects are mediated, at least in part, by molecular adaptations resulting in long-lasting synaptic changes in mesolimbic brain regions known to regulate natural and drug reward. This review outlines our limited knowledge of ketamine's neurobiological and biochemical underpinnings mediating its antidepressant effects and correlates them to its abuse potential. Depression and addiction share overlapping neural circuitry and molecular mechanisms, and though speculative, repeated use of ketamine for the treatment of depression could lead to the development of substance use disorder/addiction, and thus should be tempered with caution. There is much that remains to be known about the long-term effects of ketamine, and our lack of understanding of neurobiological mechanisms underlying its antidepressant effects is a clear limiting factor that needs to be addressed systematically before using repeated ketamine in the treatment of depressed patients.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Psychology, The University of Texas at Arlington, United States
| | - Ross J Armant
- Department of Psychology, The University of Texas at Arlington, United States
| | - Carlos A Bolaños-Guzmán
- Department of Psychological and Brain Sciences, Institute for Neuroscience, Texas A&M University, College Station, TX 77840, United States
| | - Linda I Perrotti
- Department of Psychology, The University of Texas at Arlington, United States.
| |
Collapse
|
69
|
Yang T, Nie Z, Shu H, Kuang Y, Chen X, Cheng J, Yu S, Liu H. The Role of BDNF on Neural Plasticity in Depression. Front Cell Neurosci 2020; 14:82. [PMID: 32351365 PMCID: PMC7174655 DOI: 10.3389/fncel.2020.00082] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/20/2020] [Indexed: 01/01/2023] Open
Abstract
Using behavioral, pharmacological, and molecular methods, lots of studies reveal that depression is closely related to the abnormal neural plasticity processes occurring in the prefrontal cortex and limbic system such as the hippocampus and amygdala. Meanwhile, functions of the brain-derived neurotrophic factor (BDNF) and the other neurotrophins in the pathogenesis of depression are well known. The maladaptive neuroplastic in depression may be related to alterations in the levels of neurotrophic factors, which play a central role in plasticity. Enhancement of neurotrophic factors signaling has great potential in therapy for depression. This review highlights the relevance of neurotrophic factors mediated neural plasticity and pathophysiology of depression. These studies reviewed here may suggest new possible targets for antidepressant drugs such as neurotrophins, their receptors, and relevant signaling pathways, and agents facilitating the activation of gene expression and increasing the transcription of neurotrophic factors in the brain.
Collapse
Affiliation(s)
- Tao Yang
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Zheng Nie
- Department of Anatomy and Histology and Embryology, Regeneration Key Lab of Sichuan Province, Chengdu Medical College, Chengdu, China
| | - Haifeng Shu
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Yongqin Kuang
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Xin Chen
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Jingmin Cheng
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Sixun Yu
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China
| | - Huiying Liu
- Department of Respiratory and Critical Care Diseases, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
70
|
Gong W, Zhou Y, Gong W, Qin X. Coniferyl ferulate exerts antidepressant effect via inhibiting the activation of NMDAR-CaMKII-MAPKs and mitochondrial apoptotic pathways. JOURNAL OF ETHNOPHARMACOLOGY 2020; 251:112533. [PMID: 31911178 DOI: 10.1016/j.jep.2019.112533] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/05/2019] [Accepted: 12/27/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoyaosan (XYS), composed of Radix Bupleuri, Radix Angelicae Sinensis, Radix Paeoniae Alba, Rhizoma Atractylodis Macrocephalae, Poria, Herba Menthae, Rhizoma Zingiberis Recens and Radix Glycyrrhizae, is a valuable traditional Chinese medicine (TCM) which is used for the treatment of depression in China. In our previous experiments, we found that coniferyl ferulate (CF) was the main active constituent of Xiaoyaosan based on UPLC-PDA guided isolation technique. However, the antidepressant effect and mechanisms of CF is still unknown. AIM OF THE STUDY In the current study, we aim to explore the possible mechanisms involved in the neuroprotective effect of CF in glutamate-injured PC12 cells, and further to confirm the anti-depressant effect of CF on the model of behavioral despair in vivo. MATERIAL AND METHODS The model of glutamate-injured PC12 cells was employed to investigate the possible mechanisms involved in the neuroprotective effect of CF. The model of behavioral despair was carried out to examine the in vivo anti-depressant effect of CF. RESULTS The results showed that CF significantly attenuated the decrease of cell viability, the release of lactate dehydrogenase (LDH), and the increase of apoptosis rates induced by glutamate. CF could also suppress the influx of Ca2+ and the elevation of p-NR2B, p-CaMK II, p-JNK, and p-p38 level induced by glutamate. Besides, CF could also inhibit the generation of reactive oxygen species (ROS), the decrease of SOD activity, the elevation of malondialdehyde (MDA) level, and suppress the loss of mitochondrial membrane potential (MMPs) and the activation Bcl-2/Bax mediated apoptotic pathways induced by glutamate. Furthermore, CF obviously decreased the immobility time in tail suspension test (TST) and forced swimming test (FST). CONCLUSION In conclusion, CF exert the indeed anti-depressant effect. The inhibition of NMDAR-CaMKII-MAPKs signaling pathway, oxidative stress, and mitochondrial apoptotic pathways were involved in the anti-depressant effect of CF.
Collapse
Affiliation(s)
- Wenxia Gong
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan, 030006, People's Republic of China.
| | - Yuzhi Zhou
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan, 030006, People's Republic of China
| | - Wenlong Gong
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan, 030006, People's Republic of China; College of Chemistry and Chemical Engineering of Shanxi University, No.92, Wucheng Road, Taiyuan, 030006, PR China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan, 030006, People's Republic of China
| |
Collapse
|
71
|
Li M, Woelfer M, Colic L, Safron A, Chang C, Heinze HJ, Speck O, Mayberg HS, Biswal BB, Salvadore G, Fejtova A, Walter M. Default mode network connectivity change corresponds to ketamine's delayed glutamatergic effects. Eur Arch Psychiatry Clin Neurosci 2020; 270:207-216. [PMID: 30353262 DOI: 10.1007/s00406-018-0942-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/10/2018] [Indexed: 12/29/2022]
Abstract
Ketamine exerts rapid antidepressant effects peaking 24 h after a single infusion, which have been suggested to be reflected by both reduced functional connectivity (FC) within default mode network (DMN) and altered glutamatergic levels in the perigenual anterior cingulate cortex (pgACC) at 24 h. Understanding the interrelation and time point specificity of ketamine-induced changes of brain circuitry and metabolism is thus key to future therapeutic developments. We investigated the correlation of late glutamatergic changes with FC changes seeded from the posterior cingulate cortex (PCC) and tested the prediction of the latter by acute fractional amplitude of low-frequency fluctuations (fALFF). In a double-blind, randomized, placebo-controlled study of 61 healthy subjects, we compared effects of subanesthetic ketamine infusion (0.5 mg/kg over 40 min) on resting-state fMRI and MR-Spectroscopy at 7 T 1 h and 24 h post-infusion. FC decrease between PCC and dorsomedial prefrontal cortex (dmPFC) was found at 24 h post-infusion (but not 1 h) and this FC decrease correlated with glutamatergic changes at 24 h in pgACC. Acute increase in fALFF was found in ventral PCC at 1 h which was not observed at 24 h and inversely correlated with the reduced dPCC FC towards the dmPFC at 24 h. The correlation of metabolic and functional markers of delayed ketamine effects and their temporal specificity suggest a potential mechanistic relationship between glutamatergic modulation and reconfiguration of brain regions belonging to the DMN.
Collapse
Affiliation(s)
- Meng Li
- Clinical Affective Neuroimaging Laboratory, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Otto-von-Guericke-University, Magdeburg, Germany
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Otto-von-Guericke-University, Magdeburg, Germany
- New Jersey Institute of Technology, Newark, NJ, USA
| | - Lejla Colic
- Clinical Affective Neuroimaging Laboratory, Leibniz Institute for Neurobiology, Magdeburg, Germany
- Otto-von-Guericke-University, Magdeburg, Germany
| | - Adam Safron
- Department of Psychology, Northwestern University, Evanston, IL, USA
| | - Catie Chang
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA
| | - Hans-Jochen Heinze
- Department of Neurology, Otto-von-Guericke-University, Magdeburg, Germany
- Department Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Oliver Speck
- Department Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Department of Biomedical Magnetic Resonance, Otto-von-Guericke-University, Magdeburg, Germany
| | - Helen S Mayberg
- Department of Neurology and Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Anna Fejtova
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
- RG Presynaptic Plasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Martin Walter
- Clinical Affective Neuroimaging Laboratory, Leibniz Institute for Neurobiology, Magdeburg, Germany.
- Otto-von-Guericke-University, Magdeburg, Germany.
- Department Behavioral Neurology, Leibniz Institute for Neurobiology, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
- Department of Psychiatry and Psychotherapy, University of Tuebingen, Osianderstrasse 24, 72076, Tuebingen, Germany.
| |
Collapse
|
72
|
Tannich F, Barhoumi K, Rejeb A, Aouichri M, Souilem O. Ketamine, at low dose, decrease behavioural alterations in epileptic diseases induced by pilocarpine in mice. Int J Neurosci 2020; 130:1118-1124. [PMID: 32075467 DOI: 10.1080/00207454.2020.1730363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Objective: The objective of the present study was to evaluate the effect of low-dose of ketamine, in short-term, on behavioral impairment and acute neuronal death in the cerebral cortex during the acute phase in a model of epileptic mouse induced by pilocarpine.Methods:Ketamine was administrated (10 mg/kg) intraperitoneally, 30 min before pilocarpine injection (100 mg/kg) in the first group. The second group received the same dose of ketamine 30 min after pilocarpine injection. The effect of ketamine on behavioral disorders and cerebral neuronal integrity in epileptic mice was evaluated.Results:Clinical observations and behavioural tests relate a reduction in behavioural dysfunctions in mice treated with ketamine. Interestingly, treatment of mice with low dose of ketamine decreased the clinical symptoms (movements of the vibrios, nods of the head, and movements of the whiskers), especially when administered before epilepsy induction. Furthermore, the administration of ketamine limits oedema in the hippocampus, neuronal degeneration and gliosis in the different cortical layers. These results could be explained by NMDA receptors inhibition by ketamine.Conclusion:Therefore, it appears that ketamine is endowed with a potential neuroprotective effect and can reduce the severity of neurodegeneration, especially when administrated before Status Epilepticus (SE) installation.
Collapse
Affiliation(s)
- Fatma Tannich
- Laboratory of Physiology and Pharmacology, National School of Veterinary Medicine, Sidi Thabet, University of Manouba, Tunisia.,Neurophysiology Laboratory and Functional Pathology, Department of Biological Sciences, Sciences Faculty of Tunis, University Campus El-Manar, Tunis, Tunisia
| | - Kamel Barhoumi
- Laboratory of Physiology and Pharmacology, National School of Veterinary Medicine, Sidi Thabet, University of Manouba, Tunisia
| | - Ahmed Rejeb
- Laboratory of Anatomic Pathology, National School of Veterinary Medicine, Sidi Thabet, University of Manouba, Tunisia
| | - Mohamed Aouichri
- Higher Institute of Nursing Sciences, Tunis, University Campus El-Manar, Tunis, Tunisia
| | - Ouajdi Souilem
- Laboratory of Physiology and Pharmacology, National School of Veterinary Medicine, Sidi Thabet, University of Manouba, Tunisia
| |
Collapse
|
73
|
Carreno FR, Lodge DJ, Frazer A. Ketamine: Leading us into the future for development of antidepressants. Behav Brain Res 2020; 383:112532. [PMID: 32023492 DOI: 10.1016/j.bbr.2020.112532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 12/28/2022]
Abstract
Numerous randomized double-blind clinical trials have consistently shown that that a single intravenous administration of a subanesthetic dose of ketamine to treatment-resistant depressed patients significantly improved depressive symptomatology rapidly, within two hours, with the effect lasting up to seven days. Despite its very promising effects, ketamine has long been associated with potential for abuse as it can cause psychotropic side effects, such as hallucinations, false beliefs, and severe impairments in judgment and other cognitive processes. Consequently, within the last two decades preclinical research has been carried out aimed at understanding its mechanisms of action and the brain circuits involved in ketamine's antidepressant effects, both of which are discussed in this review. Furthermore, with the hippocampus being a key target for ketamine's beneficial antidepressant effects, we and others have begun to examine behavioral and neurochemical effects of drugs that act selectively on the hippocampus due to the preferential location of their receptor targets. Such drugs are negative allosteric modulators (NAMs) and positive allosteric modulator (PAM) of the α5-GABAA receptor. Such compounds are discussed within the framework of how lessons learned with ketamine point to novel classes of drugs, targeting the GABAergic system, that can recapitulate the antidepressant effects of ketamine without its adverse effects.
Collapse
Affiliation(s)
- Flavia R Carreno
- Department of Pharmacology & Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, United States.
| | - Daniel J Lodge
- Department of Pharmacology & Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, United States; South Texas Veterans Health Care System, Audie L. Murphy Division, United States
| | - Alan Frazer
- Department of Pharmacology & Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, United States; South Texas Veterans Health Care System, Audie L. Murphy Division, United States
| |
Collapse
|
74
|
Zhang G, Xu S, Zhang Z, Zhang Y, Wu Y, An J, Lin J, Yuan Z, Shen L, Si T. Identification of Key Genes and the Pathophysiology Associated With Major Depressive Disorder Patients Based on Integrated Bioinformatics Analysis. Front Psychiatry 2020; 11:192. [PMID: 32317989 PMCID: PMC7146847 DOI: 10.3389/fpsyt.2020.00192] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/27/2020] [Indexed: 12/11/2022] Open
Abstract
Background: At present, laboratory blood tests to support major depressive disorder (MDD) diagnosis are not available. This study aimed to screen potential mRNAs for peripheral blood biomarkers and novel pathophysiology of MDD. Methods: The present study utilized public data from two mRNA microarray datasets to analyze the hub genes changes related to MDD. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of differentially expressed genes (DEGs) were performed. Finally, some potential mRNA quality biomarkers for hub gene expression in blood were identified. Results: A total of 25 significantly co-upregulated DEGs and 98 co-downregulated DEGs were obtained from two datasets. The pathway enrichment analyses showed that co-upregulated genes were significantly enriched in the regulation of cell-matrix adhesion and mitochondrial membrane permeability which were involved in the apoptotic process. Co-downregulated genes were mainly involved in the neutrophil activation which in turn was involved in the immune response, degranulation and cell-mediated immunity, positive regulation of immune response, the Toll-like receptor signaling pathway, and the NOD-like receptor signaling pathway. From the PPI network, 14 hub genes were obtained. Among them, the subnetworks of PLCG1, BCL2A1, TLR8, FADD, and TLR4 screened out from our study have been shown to play a role in immune and inflammation responses. Discussion: The potential molecular mechanisms that have been identified simultaneously include innate immunity, neuroinflammation, and neurotrophic factors for synapse function and development.
Collapse
Affiliation(s)
- Guangyin Zhang
- Department of Psychosomatic Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Key Laboratory of Mental Health, Ministry of Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital and Peking University Institute of Mental Health, Beijing, China
| | - Shixin Xu
- Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | | | - Yu Zhang
- Hebei North University, Hebei, China
| | - Yankun Wu
- Key Laboratory of Mental Health, Ministry of Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital and Peking University Institute of Mental Health, Beijing, China
| | - Jing An
- Key Laboratory of Mental Health, Ministry of Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital and Peking University Institute of Mental Health, Beijing, China
| | - Jinyu Lin
- Key Laboratory of Mental Health, Ministry of Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital and Peking University Institute of Mental Health, Beijing, China
| | - Zhuo Yuan
- Department of Psychosomatic Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li Shen
- Department of Psychosomatic Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tianmei Si
- Key Laboratory of Mental Health, Ministry of Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University Sixth Hospital and Peking University Institute of Mental Health, Beijing, China
| |
Collapse
|
75
|
Functional magnetic resonance spectroscopy in patients with schizophrenia and bipolar affective disorder: Glutamate dynamics in the anterior cingulate cortex during a working memory task. Eur Neuropsychopharmacol 2019; 29:222-234. [PMID: 30558824 DOI: 10.1016/j.euroneuro.2018.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/25/2018] [Accepted: 12/01/2018] [Indexed: 12/16/2022]
Abstract
The glutamate system is implicated in the pathophysiology of schizophrenia and mood disorders. Using functional magnetic resonance spectroscopy (1H-fMRS), it is possible to monitor glutamate dynamically in activated brain areas and may give a closer estimate of glutamatergic neurotransmission than standard magnetic resonance spectroscopy. 14 patients with schizophrenia, 15 patients with bipolar disorder II (BPII) and 14 healthy volunteers underwent a 15 min N-back task in a 48s block design during 1H-fMRS acquisition. Data from the first, second and third 16s group of 8 spectra for each block were analysed to measure levels of glutamate and Glx (glutamate + glutamine), scaled to total creatine (TCr), across averaged 0-back and 2-back conditions. A 6 × 3 repeated-measures analysis of variance (rmANOVA) demonstrated a significant main effect of time for Glx/TCr (P = 0.022). There was a significant increase in Glu/TCr (P = 0.004) and Glx/TCr (P < 0.001) between the final spectra of the 0-back and first spectra of the 2-back condition in the healthy control group only. 2 × 2 rmANOVA revealed a significant time by group interaction for Glx/TCr (P = 0.019) across the 0-back condition, with levels reducing in healthy controls and increasing in the schizophrenia group. While healthy volunteers showed significant increases in glutamatergic measures between task conditions, the lack of such a response in patients with schizophrenia and BPII may reflect deficits in glutamatergic neurotransmission. Abnormal increases during periods of relatively low executive load, without the same dynamic modulation as healthy volunteers with increasing task difficulty, further suggests underlying abnormalities of glutamatergic neurotransmission in schizophrenia.
Collapse
|
76
|
Aleksandrova LR, Wang YT, Phillips AG. Evaluation of the Wistar-Kyoto rat model of depression and the role of synaptic plasticity in depression and antidepressant response. Neurosci Biobehav Rev 2019; 105:1-23. [DOI: 10.1016/j.neubiorev.2019.07.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/10/2019] [Accepted: 07/08/2019] [Indexed: 12/28/2022]
|
77
|
McCoy KT, Costa CB, Pancione K, Hammonds LS. Anticipating Changes for Depression Management in Primary Care. Nurs Clin North Am 2019; 54:457-471. [PMID: 31703773 DOI: 10.1016/j.cnur.2019.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Depression management in primary care settings is the norm, in the United States and globally. As incidence and prevalence of depression continue to mount, there are innovative models of treatment, newer understandings, more open philosophies, and evidence-informed treatments that may address this troubling public health issue. This article attempts to succinctly examine the evidence in identifying and treating this in the United States in an expedient, evidence-informed manner to assist those in need of have care that is patient centered, of high quality, affordable, and readily accessible across the lifespan.
Collapse
Affiliation(s)
- Kathleen T McCoy
- Department of Community Mental Health, University of South Alabama, College of Nursing, HAHN 304/5721 USA Drive North, Mobile, AL 36688-002, USA.
| | - Christine B Costa
- California State University Long Beach, School of Nursing, 1250 Bellflower Boulevard, MS 0301, Long Beach, CA 90804, USA
| | - Kirsten Pancione
- Department of Community Mental Health, University of South Alabama, College of Nursing, HAHN 304/5721 USA Drive North, Mobile, AL 36688-002, USA
| | - Linda Sue Hammonds
- Department of Community Mental Health, University of South Alabama, College of Nursing, HAHN 304/5721 USA Drive North, Mobile, AL 36688-002, USA
| |
Collapse
|
78
|
Fišar Z, Hansíková H, Křížová J, Jirák R, Kitzlerová E, Zvěřová M, Hroudová J, Wenchich L, Zeman J, Raboch J. Activities of mitochondrial respiratory chain complexes in platelets of patients with Alzheimer's disease and depressive disorder. Mitochondrion 2019; 48:67-77. [PMID: 31377247 DOI: 10.1016/j.mito.2019.07.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/03/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022]
Abstract
We analyzed activities of complex I, II, III, and IV, and citrate synthase (CS) in patients with major depressive disorder (MDD) or Alzheimer's disease (AD) presenting with or without depression. Associations of these parameters with disease or disease severity were observed in both AD and MDD; however, mean values of mitochondrial parameters were significantly altered in AD but not in MDD. Potential mitochondrial dysfunction in MDD seems not to be caused by disturbed activity of CS or respiratory complexes. In AD, a decrease in the activity of CS and complex IV may cause mitochondrial dysfunction, whereas an increase in activities of other mitochondrial complexes or their ratios to CS may be an adaptive response. The data indicate that comorbid depression in AD is associated with increased complex II activity. The mitochondrial parameters measured can be included in the panel of biomarkers of AD.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Hana Hansíková
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 120 00 Prague 2, Czech Republic.
| | - Jana Křížová
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 120 00 Prague 2, Czech Republic.
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Eva Kitzlerová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Martina Zvěřová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| | - Jana Hroudová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic; Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Albertov 4, Praha 2 128 00, Prague 2, Czech Republic.
| | - László Wenchich
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 120 00 Prague 2, Czech Republic.
| | - Jiří Zeman
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 2, 120 00 Prague 2, Czech Republic.
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic.
| |
Collapse
|
79
|
Xiao D, Liu L, Li Y, Ruan J, Wang H. Licorisoflavan A Exerts Antidepressant-Like Effect in Mice: Involvement of BDNF-TrkB Pathway and AMPA Receptors. Neurochem Res 2019; 44:2044-2056. [DOI: 10.1007/s11064-019-02840-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/23/2019] [Accepted: 06/30/2019] [Indexed: 12/15/2022]
|
80
|
Suzuki A, Murakami K, Tajima Y, Hara H, Kunugi A, Kimura H. TAK-137, an AMPA receptor potentiator with little agonistic effect, produces antidepressant-like effect without causing psychotomimetic effects in rats. Pharmacol Biochem Behav 2019; 183:80-86. [PMID: 31202810 DOI: 10.1016/j.pbb.2019.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 01/18/2023]
Abstract
Ketamine produces a rapid-onset antidepressant effect in patients with treatment-resistant depression (TRD), although it concurrently causes undesirable psychotomimetic side effects. Accumulating evidence suggests that ketamine produces antidepressant effects via activation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPA-R), with consequent activation of the mammalian target of rapamycin (mTOR) pathway and up-regulation of brain-derived neurotrophic factor (BDNF). We previously reported that TAK-137, an AMPA-R potentiator with little agonistic effect, had potent procognitive effects with lower risks of bell-shaped dose-response and seizure induction. In this study, we characterized the potential of TAK-137 as a novel antidepressant in rats. In rat primary cortical neurons, TAK-137 increased the phosphorylated form of Akt, extracellular signal-regulated kinase, mTOR, and p70S6 kinase, and dose-dependently increased the expression level of BDNF protein. The antidepressant-like effects of ketamine and TAK-137 were assessed on the day after final administration using the novelty-suppressed feeding test in rats. A single intraperitoneal administration of ketamine shortened the latency to feed. Under these conditions, oral administration of TAK-137 for 3 days shortened the feeding latency. Ketamine induced hyperlocomotion and reduced prepulse inhibition, which may be associated with psychotomimetic effects, while TAK-137 did not. TAK-137 may be a safer and rapid-onset therapeutic drug for the treatment of major depressive disorder, including TRD.
Collapse
Affiliation(s)
- Atsushi Suzuki
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Koji Murakami
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yasukazu Tajima
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hiroe Hara
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Akiyoshi Kunugi
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Haruhide Kimura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan.
| |
Collapse
|
81
|
Ionescu DF, Felicione JM, Gosai A, Cusin C, Shin P, Shapero BG, Deckersbach T. Ketamine-Associated Brain Changes: A Review of the Neuroimaging Literature. Harv Rev Psychiatry 2019; 26:320-339. [PMID: 29465479 PMCID: PMC6102096 DOI: 10.1097/hrp.0000000000000179] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Major depressive disorder (MDD) is one of the most prevalent conditions in psychiatry. Patients who do not respond to traditional monoaminergic antidepressant treatments have an especially difficult-to-treat type of MDD termed treatment-resistant depression. Subanesthetic doses of ketamine-a glutamatergic modulator-have shown great promise for rapidly treating patients with the most severe forms of depression. As such, ketamine represents a promising probe for understanding the pathophysiology of depression and treatment response. Through neuroimaging, ketamine's mechanism may be elucidated in humans. Here, we review 47 articles of ketamine's effects as revealed by neuroimaging studies. Some important brain areas emerge, especially the subgenual anterior cingulate cortex. Furthermore, ketamine may decrease the ability to self-monitor, may increase emotional blunting, and may increase activity in reward processing. Further studies are needed, however, to elucidate ketamine's mechanism of antidepressant action.
Collapse
Affiliation(s)
- Dawn F. Ionescu
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | | | - Aishwarya Gosai
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Cristina Cusin
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Philip Shin
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Benjamin G. Shapero
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Thilo Deckersbach
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Boston, MA
| |
Collapse
|
82
|
Tornese P, Sala N, Bonini D, Bonifacino T, La Via L, Milanese M, Treccani G, Seguini M, Ieraci A, Mingardi J, Nyengaard JR, Calza S, Bonanno G, Wegener G, Barbon A, Popoli M, Musazzi L. Chronic mild stress induces anhedonic behavior and changes in glutamate release, BDNF trafficking and dendrite morphology only in stress vulnerable rats. The rapid restorative action of ketamine. Neurobiol Stress 2019; 10:100160. [PMID: 31193464 PMCID: PMC6535630 DOI: 10.1016/j.ynstr.2019.100160] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 12/18/2022] Open
Abstract
Depression is a debilitating mental disease, characterized by persistent low mood and anhedonia. Stress represents a major environmental risk factor for depression; the complex interaction of stress with genetic factors results in different individual vulnerability or resilience to the disorder. Dysfunctions of the glutamate system have a primary role in depression. Clinical neuroimaging studies have consistently reported alterations in volume and connectivity of cortico-limbic areas, where glutamate neurons and synapses predominate. This is confirmed by preclinical studies in rodents, showing that repeated stress induces morphological and functional maladaptive changes in the same brain regions altered in humans. Confirming the key role of glutamatergic transmission in depression, compelling evidence has shown that the non-competitive NMDA receptor antagonist, ketamine, induces, at sub-anesthetic dose, rapid and sustained antidepressant response in both humans and rodents. We show here that the Chronic Mild Stress model of depression induces, only in stress-vulnerable rats, depressed-like anhedonic behavior, together with impairment of glutamate/GABA presynaptic release, BDNF mRNA trafficking in dendrites and dendritic morphology in hippocampus. Moreover, we show that a single administration of ketamine restores, in 24 h, normal behavior and most of the cellular/molecular maladaptive changes in vulnerable rats. Interestingly, ketamine treatment did not restore BDNF mRNA levels reduced by chronic stress but rescued dendritic trafficking of BDNF mRNA. The present results are consistent with a mechanism of ketamine involving rapid restoration of synaptic homeostasis, through re-equilibration of glutamate/GABA release and dendritic BDNF for synaptic translation and reversal of synaptic and circuitry impairment. Chronic mild stress (CMS) induces anhedonic behavior and maladaptive changes in the hippocampus (HPC) of vulnerable rats. CMS reduces basal and evoked release of glutamate in the HPC of vulnerable rats. SCMS reduces evoked release of GABA in the HPC of vulnerable rats. CMS reduces expression of BDNF mRNA and trafficking along dendrites in the HPC of vulnerable rats. CMS reduces length of apical dendrites in CA3 pyramidal neurons of vulnerable rats. Ketamine injection (10 mg/kg) restores in 24h anhedonic behavior and most maladaptive changes, except BDNF expression. The present results suggest that the antidepressant mechanism of ketamine involves restoration of synaptic homeostasis.
Collapse
Affiliation(s)
- Paolo Tornese
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence for Neurodegenerative Diseases, Università degli Studi di Milano, 20133, Milan, Italy
| | - Nathalie Sala
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence for Neurodegenerative Diseases, Università degli Studi di Milano, 20133, Milan, Italy
| | - Daniela Bonini
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148, Genova, Italy
| | - Luca La Via
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148, Genova, Italy
| | - Giulia Treccani
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8240, Risskov, Denmark
| | - Mara Seguini
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence for Neurodegenerative Diseases, Università degli Studi di Milano, 20133, Milan, Italy
| | - Alessandro Ieraci
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence for Neurodegenerative Diseases, Università degli Studi di Milano, 20133, Milan, Italy
| | - Jessica Mingardi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Jens R Nyengaard
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, 8000, Aarhus, Denmark
| | - Stefano Calza
- Unit of Biostatistics and Biomathematics, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148, Genova, Italy
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, 8240, Risskov, Denmark.,Pharmaceutical Research Centre of Excellence, School of Pharmacy, North-West University, 2520, Potchefstroom, South Africa
| | - Alessandro Barbon
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence for Neurodegenerative Diseases, Università degli Studi di Milano, 20133, Milan, Italy
| | - Laura Musazzi
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence for Neurodegenerative Diseases, Università degli Studi di Milano, 20133, Milan, Italy
| |
Collapse
|
83
|
Antidepressant-Like Action of Single Facial Injection of Botulinum Neurotoxin A is Associated with Augmented 5-HT Levels and BDNF/ERK/CREB Pathways in Mouse Brain. Neurosci Bull 2019; 35:661-672. [PMID: 30900142 DOI: 10.1007/s12264-019-00367-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
The present study was designed to examine the therapeutic effects of Botulinum neurotoxin A (BoNT/A) on depression-like behaviors in mice and to explore the potential mechanisms. These results revealed that a single facial injection of BoNT/A induced a rapid and prolonged improvement of depression-like behaviors in naïve and space-restriction-stressed (SRS) mice, reflected by a decreased duration of immobility in behavioral despair tests. BoNT/A significantly increased the 5-hydroxytryptamine (5-HT) levels in several brain regions, including the hippocampus and hypothalamus, in SRS mice. BoNT/A increased the expression of the N-methyl-D-aspartate receptor subunits NR1 and NR2B in the hippocampus, which were significantly decreased in SRS mice. Furthermore, BoNT/A significantly increased the expression of brain-derived neurotrophic factor (BDNF) in the hippocampus, hypothalamus, prefrontal cortex, and amygdala, which were decreased in SRS mice. Finally, BoNT/A transiently increased the levels of phosphorylated extracellular signal-regulated kinase (p-ERK) and cAMP-response element binding protein (p-CREB), which were suppressed in the hippocampus of SRS mice. Collectively, these results demonstrated that BoNT/A treatment has anti-depressant-like activity in mice, and this is associated with increased 5-HT levels and the activation of BDNF/ERK/CREB pathways in the hippocampus, supporting further investigation of BoNT/A therapy in depression.
Collapse
|
84
|
Influence of pharmacological and epigenetic factors to suppress neurotrophic factors and enhance neural plasticity in stress and mood disorders. Cogn Neurodyn 2019; 13:219-237. [PMID: 31168328 DOI: 10.1007/s11571-019-09522-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/17/2018] [Accepted: 01/16/2019] [Indexed: 02/08/2023] Open
Abstract
Stress-induced major depression and mood disorders are characterized by behavioural abnormalities and psychiatric illness, leading to disability and immature mortality worldwide. Neurobiological mechanisms of stress and mood disorders are discussed considering recent findings, and challenges to enhance pharmacological effects of antidepressant, and mood stabilizers. Pharmacological enhancement of ketamine and scopolamine regulates depression at the molecular level, increasing synaptic plasticity in prefrontal regions. Blood-derived neurotrophic factors facilitate mood-deficit symptoms. Epigenetic factors maintain stress-resilience in hippocampal region. Regulation of neurotrophic factors blockades stress, and enhances neuronal survival though it paralyzes limbic regions. Molecular agents and neurotrophic factors also control behavioral and synaptic plasticity in addiction and stress disorders. Future research on neuronal dynamics and cellular actions can be directed to obtain the etiology of synaptic dysregulation in mood disorder and stress. For the first time, the current review contributes to the literature of synaptic plasticity representing the role of epigenetic mechanisms and glucocorticoid receptors to predict depression and anxiety in clinical conditions.
Collapse
|
85
|
Caldieraro MA, Cassano P. Transcranial and systemic photobiomodulation for major depressive disorder: A systematic review of efficacy, tolerability and biological mechanisms. J Affect Disord 2019; 243:262-273. [PMID: 30248638 DOI: 10.1016/j.jad.2018.09.048] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 08/24/2018] [Accepted: 09/15/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Photobiomodulation (PBM) with red and near-infrared light (NIR) -also known as Low-Level Light Therapy-is a low risk, inexpensive treatment-based on non-retinal exposure-under study for several neuropsychiatric conditions. The aim of this paper is to discuss the proposed mechanism of action and to perform a systematic review of pre-clinical and clinical studies on PBM for major depressive disorder (MDD). METHODS A search on MEDLINE and EMBASE databases was performed in July 2017. No time or language restrictions were used. Studies with a primary focus on MDD and presenting original data were included (n = 17). References on the mechanisms of action of PBM also included review articles and studies not focused on MDD. RESULTS Red and NIR light penetrate the skull and modulate brain cortex; an indirect effect of red and NIR light, when delivered non-transcranially, is also postulated. The main proposed mechanism for PBM is the enhancement of mitochondrial metabolism after absorption of NIR energy by the cytochrome C oxidase; however, actions on other pathways relevant to MDD are also reported. Studies on animal models indicate a benefit from PBM that is comparable to antidepressant medications. Clinical studies also indicate a significant antidepressant effect and good tolerability. LIMITATIONS Clinical studies are heterogeneous for population and treatment parameters, and most lack an appropriate control. CONCLUSIONS Preliminary evidence supports the potential of non-retinal PBM as a novel treatment for MDD. Future studies should clarify the ideal stimulation parameters as well as the overall efficacy, effectiveness and safety profile of this treatment.
Collapse
Affiliation(s)
- Marco A Caldieraro
- Serviço de Psiquiatria, Hospital de Clínicas de Porto Alegre. Rua Ramiro Barcelos 2350, Porto Alegre, RS 90035-903, Brazil.
| | - Paolo Cassano
- Depression Clinical and Research Program, Department of Psychiatry, Massachusetts General Hospital. 1 Bowdoin Square, Boston, MA 02114, USA; Center for Anxiety and Traumatic Stress Disorders, Department of Psychiatry, Massachusetts General Hospital, Boston. 1 Bowdoin Square, MA 02114, USA
| |
Collapse
|
86
|
Abstract
For decades, symptoms of depression have been treated primarily with medications that directly target the monoaminergic brain systems, which typically take weeks to exert measurable effects and months to exert remission of symptoms. Low, subanesthetic doses of ( R,S)-ketamine (ketamine) result in the rapid improvement of core depressive symptoms, including mood, anhedonia, and suicidal ideation, occurring within hours following a single administration, with relief from symptoms typically lasting up to a week. The discovery of these actions of ketamine has resulted in a reconceptualization of how depression could be more effectively treated in the future. In this review, we discuss clinical data pertaining to ketamine and other rapid-acting antidepressant drugs, as well as the current state of pharmacological knowledge regarding their mechanism of action. Additionally, we discuss the neurobiological circuits that are engaged by this drug class and that may be targeted by a future generation of medications, for example, hydroxynorketamine; metabotropic glutamate receptor 2/3 antagonists; and N-methyl-d-aspartate, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and γ-aminobutyric acid receptor modulators.
Collapse
Affiliation(s)
- Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
- Departments of Pharmacology and Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA 20892
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| |
Collapse
|
87
|
Novel targets for parkinsonism-depression comorbidity. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 167:1-24. [DOI: 10.1016/bs.pmbts.2019.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
88
|
Ragguett RM, Rong C, Kratiuk K, McIntyre RS. Rapastinel - an investigational NMDA-R modulator for major depressive disorder: evidence to date. Expert Opin Investig Drugs 2018; 28:113-119. [PMID: 30585524 DOI: 10.1080/13543784.2019.1559295] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Major depressive disorder (MDD) is a debilitating disorder with increasing prevalence globally. Despite the development of novel treatments for MDD, many patients present with treatment resistant depression (TRD), identified by treatment non-response following one or more adequate trials of an antidepressant. Rapastinel may prove to be a viable treatment for TRD; it has the potential to produce a rapid antidepressant response without serious adverse events and improve functional symptoms. Areas covered: We review the efficacy of rapastinel via completed and on-going clinical trials. The online databases Pubmed, clinicaltrials.gov and clinicaltrialsregister.eu were searched for rapastinel (GLYX-13) treatment in subjects with MDD. Nine clinical trials were identified. Expert opinion: Rapastinel is a novel and potentially transformative treatment for individuals with TRD. There is a limited number of clinical studies so far, but this compound has the potential to provide rapid, reliable and robust antidepressant effects without psychotomimetic and other unwanted side effects. Alternative formulations such as the oral formulation, provide the opportunity for rapastinel to be administered less frequently, i.e. once weekly. Furthermore, the beneficial effects on measures of cognition and suicidality so far, represent a tremendous advantage.
Collapse
Affiliation(s)
- Renee-Marie Ragguett
- a Mood Disorders Psychopharmacology Unit , University Health Network , Toronto , Canada
| | - Carola Rong
- a Mood Disorders Psychopharmacology Unit , University Health Network , Toronto , Canada.,d Department of Pharmacology , University of Toronto , Toronto , Canada
| | - Kevin Kratiuk
- b Medical Faculty , Poznan University of Medical Sciences , Poznan , Poland
| | - Roger S McIntyre
- a Mood Disorders Psychopharmacology Unit , University Health Network , Toronto , Canada.,c Department of Psychiatry , University of Toronto , Toronto , Canada.,d Department of Pharmacology , University of Toronto , Toronto , Canada
| |
Collapse
|
89
|
Guo W, Machado-Vieira R, Mathew S, Murrough JW, Charney DS, Grunebaum M, Oquendo MA, Kadriu B, Akula N, Henter I, Yuan P, Merikangas K, Drevets W, Furey M, Mann JJ, McMahon FJ, Zarate CA, Shugart YY. Exploratory genome-wide association analysis of response to ketamine and a polygenic analysis of response to scopolamine in depression. Transl Psychiatry 2018; 8:280. [PMID: 30552317 PMCID: PMC6294748 DOI: 10.1038/s41398-018-0311-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/30/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Growing evidence suggests that the glutamatergic modulator ketamine has rapid antidepressant effects in treatment-resistant depressed subjects. The anticholinergic agent scopolamine has also shown promise as a rapid-acting antidepressant. This study applied genome-wide markers to investigate the role of genetic variants in predicting acute antidepressant response to both agents. The ketamine-treated sample included 157 unrelated European subjects with major depressive disorder (MDD) or bipolar disorder (BD). The scopolamine-treated sample comprised 37 unrelated European subjects diagnosed with either MDD or BD who had a current Major Depressive Episode (MDE), and had failed at least two adequate treatment trials for depression. Change in Montgomery-Asberg Depression Rating Scale (MADRS) or the 17-item Hamilton Depression Rating Scale (HAM-D) scale scores at day 1 (24 h post-treatment) was considered the primary outcome. Here, we conduct pilot genome-wide association study (GWAS) analyses to identify potential markers of ketamine response and dissociative side effects. Polygenic risk score analysis of SNPs ranked by the strength of their association with ketamine response was then calculated in order to assess whether common genetic markers from the ketamine study could predict response to scopolamine. Findings require replication in larger samples in light of low power of analyses of these small samples. Neverthless, these data provide a promising illustration of our future potential to identify genetic variants underlying rapid treatment response in mood disorders and may ultimately guide individual patient treatment selection in the future.
Collapse
Affiliation(s)
- Wei Guo
- Statistical Genomics and Data Analysis Core, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sanjay Mathew
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - James W Murrough
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dennis S Charney
- Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Grunebaum
- Columbia University Medical Center/New York State Psychiatric Institute, New York, NY, USA
| | - Maria A Oquendo
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Nirmala Akula
- Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Ioline Henter
- Section on PET Neuroimaging Sciences, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Peixiong Yuan
- Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Kathleen Merikangas
- Genetic Epidemiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Wayne Drevets
- Janssen Pharmaceuticals, Neuroscience Research and Development, La Jolla, CA, USA
| | - Maura Furey
- Janssen Pharmaceuticals, Neuroscience Research and Development, La Jolla, CA, USA
| | - J John Mann
- Departments of Psychiatry and Radiology, College of Physicians and Surgeons, Columbia University, New York State Psychiatric Institute, New York, NY, USA
| | - Francis J McMahon
- Human Genetics Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Yin Yao Shugart
- Statistical Genomics and Data Analysis Core, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
90
|
Thelen C, Flaherty E, Saurine J, Sens J, Mohamed S, Pitychoutis PM. Sex Differences in the Temporal Neuromolecular and Synaptogenic Effects of the Rapid-acting Antidepressant Drug Ketamine in the Mouse Brain. Neuroscience 2018; 398:182-192. [PMID: 30537521 DOI: 10.1016/j.neuroscience.2018.11.053] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 10/31/2018] [Accepted: 11/09/2018] [Indexed: 12/24/2022]
Abstract
Preclinical evidence suggests that ketamine's rapid and sustained antidepressant actions are due to the induction of synaptogenesis in the medial prefrontal cortex (mPFC) and the hippocampus (HIPP), two brain regions implicated in the pathophysiology of major depression. However, research on the neurobiological effects of ketamine has focused almost exclusively on males. Findings from our group and others indicate that female rodents are more reactive to ketamine's antidepressant effects, since they respond to lower doses in antidepressant-predictive behavioral models. The sex-dependent mechanisms that mediate the antidepressant effects of ketamine in the female brain are elusive. Herein, we assessed the neurobiological effects of a single ketamine dose (10 mg/kg; previously shown to induce rapid and sustained antidepressant-like effects in mice of both sexes), on glutamate release in the mPFC, as well as on the expression of synaptic plasticity markers, and spine density in the mPFC and the HIPP of C57BL/6J mice. Our data revealed that ketamine induced a sex-specific "glutamate burst" in the male mPFC. Ketamine activated the mammalian target of rapamycin complex 1 (mTORC1) pathway in prefrontocortical synaptoneurosomes, and enhanced spine formation in the male mPFC and HIPP. In females, ketamine induced a sustained increase in hippocampal spine density. Overall, these data exposed a sharp sex difference in the synaptogenic response to ketamine in stress-naïve mice, and further suggest that the mPFC may play a more important role in mediating the antidepressant effects of the drug in males, while the HIPP may be more important for females.
Collapse
Affiliation(s)
- Connor Thelen
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
| | - Emily Flaherty
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
| | - Joseph Saurine
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
| | - Jonathon Sens
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
| | - Sara Mohamed
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
| | - Pothitos M Pitychoutis
- Department of Biology & Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA.
| |
Collapse
|
91
|
Hesperidin attenuates depression-related symptoms in mice with mild traumatic brain injury. Life Sci 2018; 213:198-205. [DOI: 10.1016/j.lfs.2018.10.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/12/2018] [Accepted: 10/19/2018] [Indexed: 11/20/2022]
|
92
|
Bloomfield MAP, Hindocha C, Green SF, Wall MB, Lees R, Petrilli K, Costello H, Ogunbiyi MO, Bossong MG, Freeman TP. The neuropsychopharmacology of cannabis: A review of human imaging studies. Pharmacol Ther 2018; 195:132-161. [PMID: 30347211 PMCID: PMC6416743 DOI: 10.1016/j.pharmthera.2018.10.006] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The laws governing cannabis are evolving worldwide and associated with changing patterns of use. The main psychoactive drug in cannabis is Δ9-tetrahydrocannabinol (THC), a partial agonist at the endocannabinoid CB1 receptor. Acutely, cannabis and THC produce a range of effects on several neurocognitive and pharmacological systems. These include effects on executive, emotional, reward and memory processing via direct interactions with the endocannabinoid system and indirect effects on the glutamatergic, GABAergic and dopaminergic systems. Cannabidiol, a non-intoxicating cannabinoid found in some forms of cannabis, may offset some of these acute effects. Heavy repeated cannabis use, particularly during adolescence, has been associated with adverse effects on these systems, which increase the risk of mental illnesses including addiction and psychosis. Here, we provide a comprehensive state of the art review on the acute and chronic neuropsychopharmacology of cannabis by synthesizing the available neuroimaging research in humans. We describe the effects of drug exposure during development, implications for understanding psychosis and cannabis use disorder, and methodological considerations. Greater understanding of the precise mechanisms underlying the effects of cannabis may also give rise to new treatment targets.
Collapse
Affiliation(s)
- Michael A P Bloomfield
- Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, United Kingdom; Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, Faculty of Brain Sciences, University College London, United Kingdom; Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, University College Hospital, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, United Kingdom.
| | - Chandni Hindocha
- Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, United Kingdom; Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, Faculty of Brain Sciences, University College London, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, University College Hospital, London, United Kingdom
| | - Sebastian F Green
- Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, United Kingdom
| | - Matthew B Wall
- Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, Faculty of Brain Sciences, University College London, United Kingdom; Centre for Neuropsychopharmacology, Division of Brain Sciences, Faculty of Medicine, Imperial College London, United Kingdom; Invicro UK, Hammersmith Hospital, London, United Kingdom
| | - Rachel Lees
- Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, United Kingdom; Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, Faculty of Brain Sciences, University College London, United Kingdom; Institute of Cognitive Neuroscience, Faculty of Brain Sciences, University College London, United Kingdom
| | - Katherine Petrilli
- Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, United Kingdom; Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, Faculty of Brain Sciences, University College London, United Kingdom; Institute of Cognitive Neuroscience, Faculty of Brain Sciences, University College London, United Kingdom
| | - Harry Costello
- Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, United Kingdom
| | - M Olabisi Ogunbiyi
- Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, United Kingdom
| | - Matthijs G Bossong
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, the Netherlands
| | - Tom P Freeman
- Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, United Kingdom; Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, Faculty of Brain Sciences, University College London, United Kingdom; Department of Psychology, University of Bath, United Kingdom; National Addiction Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom
| |
Collapse
|
93
|
Thomas RK, Baker G, Lind J, Dursun S. Rapid effectiveness of intravenous ketamine for ultraresistant depression in a clinical setting and evidence for baseline anhedonia and bipolarity as clinical predictors of effectiveness. J Psychopharmacol 2018; 32:1110-1117. [PMID: 30182797 DOI: 10.1177/0269881118793104] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Intravenous ketamine has been established as an efficacious and safe treatment, with transient effect, for treatment-resistant depression. However, the effectiveness of intravenous ketamine in non-research settings and with ultraresistant depression patients remains understudied. AIMS This study aims to measure the response and remission rates in ultraresistant depression patients in a clinical setting by means of a retrospective, open label, database study. Secondarily, the study will attempt to support previous findings of clinical predictors of effectiveness with intravenous ketamine treatment. METHODS Fifty patients with ultraresistant depression were treated between May 2015-December 2016, inclusive, in two community hospitals in Edmonton using six ketamine infusions of 0.5 mg/kg over 40 min over 2-3 weeks. Data were collected retrospectively from inpatient and outpatient charts. Statistical analysis to investigate clinical predictors of effectiveness included logistic regression analysis using a dependent variable of a 50% reduction in rating scale score at any point during treatment. RESULTS At baseline, the average treatment resistance was severe, with a Maudsley Staging Method score of 12.1 out of 15, 90.0% were resistant to electroconvulsive therapy, and the average Beck Depression Inventory score was 34.2. The response rate was 44% and remission rate was 16%. As a single predictor, moderate or severe anhedonia at baseline predicted a 55% increased likelihood of response. As a combined predictor, this level of anhedonia at baseline with a diagnosis of bipolar depression predicted a 73% increase in likelihood of response. CONCLUSION In a clinical setting, intravenous ketamine showed effectiveness in a complex, severely treatment-resistant, depressed population on multiple medication profiles concurrently. This study gave support to anhedonia and bipolar depression as clinical predictors of effectiveness.
Collapse
Affiliation(s)
- Rejish K Thomas
- 1 Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Canada
| | - Glen Baker
- 1 Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Canada
- 2 Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - John Lind
- 1 Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Canada
| | - Serdar Dursun
- 1 Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, Canada
- 2 Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
94
|
Caldieraro MA, McKee M, Leistner-Segal S, Vares EA, Kubaski F, Spanemberg L, Brusius-Facchin AC, Fleck MP, Mischoulon D. Val66Met polymorphism association with serum BDNF and inflammatory biomarkers in major depression. World J Biol Psychiatry 2018; 19:402-409. [PMID: 28656803 DOI: 10.1080/15622975.2017.1347713] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Current evidence supports participation of neurotrophic and inflammatory factors in the pathogenesis of major depressive disorder (MDD). Some studies reported an association between the Val66Met polymorphism (rs6265) of brain-derived neurotrophic factor (BDNF) gene with MDD and peripheral BDNF levels. However, no previous studies have examined the association of this polymorphism with inflammation. The present study assessed the association of the Val66Met polymorphism with serum levels of BDNF and inflammatory markers among depressed outpatients. METHODS All participants (n = 73) met DSM-IV criteria for a unipolar depressive episode. The serum levels of BDNF and inflammatory biomarkers (IL-2, IL-4, IL-6, IL-10, TNF-α and IFN-γ) were compared between individuals presenting with at least one Met allele (Met-carriers) and those homozygous for the Val allele. RESULTS In our sample (84.9% female, mean age 52.4 ± 10.3 years), 24.7% (n = 18) were Met-carriers. After Bonferroni correction, the Met allele was significantly associated with higher BDNF and lower TNF-α. These associations persisted after adjusting for potential confounders. CONCLUSIONS The pattern of low BDNF and high inflammation in MDD may be influenced by the Val66Met polymorphism. The association of a polymorphism in the BDNF gene with inflammatory markers in addition to BDNF levels suggests an interaction between these systems.
Collapse
Affiliation(s)
- Marco Antonio Caldieraro
- a Department of Psychiatry , Massachusetts General Hospital , Boston , MA , USA.,b Department of Psychiatry , Hospital de Clínicas de Porto Alegre , Porto Alegre , Brasil
| | - Madison McKee
- c Depression Clinical and Research Program , Massachusetts General Hospital , Boston , MA , USA
| | - Sandra Leistner-Segal
- d Medical Genetics Service, Molecular Genetics Laboratory , Hospital de Clínicas de Porto Alegre , Porto Alegre , Brasil.,e Experimental Research Center , B.R.A.I.N. Laboratory, Hospital de Clínicas de Porto Alegre , Porto Alegre , Brasil
| | - Edgar Arrua Vares
- f Programa de Pós-Graduação Ciências Médicas: Psiquiatria, Department of Psychiatry and Forensic Medicine , Universidade Federal do Rio Grande do Sul (UFRGS) , Porto Alegre , Brasil
| | - Francyne Kubaski
- g Department of Research , Nemours/Alfred I. duPont Hospital for Children , Wilmington , DE , USA.,h Department of Biological Sciences , University of Delaware , Newark , DE , USA.,i Department of Research , Instituto Nacional de Genética Médica Populacional-INAGEMP , Porto Alegre , Brasil
| | - Lucas Spanemberg
- f Programa de Pós-Graduação Ciências Médicas: Psiquiatria, Department of Psychiatry and Forensic Medicine , Universidade Federal do Rio Grande do Sul (UFRGS) , Porto Alegre , Brasil.,j Department of Psychiatry , Núcleo de Formação Específica em Psiquiatria da Escola de Medicina da Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre , Brasil
| | - Ana Carolina Brusius-Facchin
- d Medical Genetics Service, Molecular Genetics Laboratory , Hospital de Clínicas de Porto Alegre , Porto Alegre , Brasil
| | - Marcelo P Fleck
- b Department of Psychiatry , Hospital de Clínicas de Porto Alegre , Porto Alegre , Brasil.,f Programa de Pós-Graduação Ciências Médicas: Psiquiatria, Department of Psychiatry and Forensic Medicine , Universidade Federal do Rio Grande do Sul (UFRGS) , Porto Alegre , Brasil
| | - David Mischoulon
- c Depression Clinical and Research Program , Massachusetts General Hospital , Boston , MA , USA.,k Department of Psychiatry , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
95
|
Rozet I, Rozet M, Borisovskaya A. Anesthesia for Electroconvulsive Therapy: an Update. CURRENT ANESTHESIOLOGY REPORTS 2018. [DOI: 10.1007/s40140-018-0283-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
96
|
Recent insights into antidepressant therapy: Distinct pathways and potential common mechanisms in the treatment of depressive syndromes. Neurosci Biobehav Rev 2018; 88:63-72. [DOI: 10.1016/j.neubiorev.2018.03.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/07/2018] [Accepted: 03/13/2018] [Indexed: 12/13/2022]
|
97
|
Disinhibition of CA1 pyramidal cells by low-dose ketamine and other antagonists with rapid antidepressant efficacy. Proc Natl Acad Sci U S A 2018. [PMID: 29531088 DOI: 10.1073/pnas.1718883115] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Low-dose ketamine, an open-channel N-methyl d-aspartate receptor (NMDAR) antagonist, mediates rapid antidepressant effects in humans that are mimicked in preclinical rodent models. Disinhibition of pyramidal cells via decreased output of fast-spiking GABAergic interneurons has been proposed as a key mechanism that triggers the antidepressant response. Unfortunately, to date, disinhibition has not been directly demonstrated. Furthermore, whether disinhibition is a common mechanism shared among other antagonists with rapid antidepressant properties in humans has not been investigated. Using in vitro electrophysiology in acute slices of dorsal hippocampus from adult male Sprague-Dawley rats, we examined the immediate effects of a clinically relevant concentration of ketamine to directly test the disinhibition hypothesis. As a mechanistic comparison, we also tested the effects of the glycine site NMDAR partial agonist/antagonist GLYX-13 (rapastinel), the GluN2B subunit-selective NMDAR antagonist Ro 25-6981, and the muscarinic acetylcholine receptor (mAChR) antagonist scopolamine. Low-dose ketamine, GLYX-13, and scopolamine reduced inhibitory input onto pyramidal cells and increased synaptically driven pyramidal cell excitability measured at the single-cell and population levels. Conversely, Ro 25-6981 increased the strength of inhibitory transmission and did not change pyramidal cell excitability. These results show a decrease in the inhibition/excitation balance that supports disinhibition as a common mechanism shared among those antagonists with rapid antidepressant properties. These data suggest that pyramidal cell disinhibition downstream of NMDAR antagonism could serve as a possible biomarker for the efficacy of rapid antidepressant therapy.
Collapse
|
98
|
Molina-Jiménez T, Limón-Morales O, Bonilla-Jaime H. Early postnatal treatment with clomipramine induces female sexual behavior and estrous cycle impairment. Pharmacol Biochem Behav 2018; 166:27-34. [PMID: 29407872 DOI: 10.1016/j.pbb.2018.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 01/28/2018] [Accepted: 01/29/2018] [Indexed: 01/10/2023]
Abstract
Administration of clomipramine (CMI), a tricyclic antidepressant, in early stages of development in rats, is considered an animal model for the study of depression. This pharmacological manipulation has induced behavioral and physiological alterations, i.e., less pleasure-seeking behaviors, despair, hyperactivity, cognitive dysfunction, alterations in neurotransmitter systems and in HPA axis. These abnormalities in adult male rats are similar to the symptoms observed in major depressive disorders. One of the main pleasure-seeking behaviors affected in male rats treated with CMI is sexual behavior. However, to date, no effects of early postnatal CMI treatment have been reported on female reproductive cyclicity and sexual behavior. Therefore, we explored CMI administration in early life (8-21 PN) on the estrous cycle and sexual behavior of adult female rats. Compared to the rats in the early postnatal saline treatment (CTRL group), the CMI rats had fewer estrous cycles, fewer days in the estrous stage, and longer cycles during a 20-day period of vaginal cytology analysis. On the behavioral test, the CMI rats displayed fewer proceptive behaviors (hopping, darting) and had lower lordosis quotients. Also, they usually failed to display lordosis and only rarely manifested marginal or normal lordosis. In contrast, the CTRL rats tended to display normal lordosis. These results suggest that early postnatal CMI treatment caused long-term disruptions of the estrous cycle and female sexual behavior, perhaps by alteration in the hypothalamic-pituitary-gonadal (HPG) axes and in neuronal circuits involved in the regulation of the performance and motivational of sexual behavior as the noradrenergic and serotonergic systems.
Collapse
Affiliation(s)
- Tania Molina-Jiménez
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Apartado, Postal 55 535, C.P. 09340 Ciudad de México, Mexico
| | - Ofelia Limón-Morales
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Av Universidad 3000, Cd. Universitaria, Coyoacán, 04510 Ciudad de México, Mexico
| | - Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Apartado Postal 55 535, C.P. 09340 Ciudad de México, Mexico.
| |
Collapse
|
99
|
DIXDC1 contributes to psychiatric susceptibility by regulating dendritic spine and glutamatergic synapse density via GSK3 and Wnt/β-catenin signaling. Mol Psychiatry 2018; 23:467-475. [PMID: 27752079 PMCID: PMC5395363 DOI: 10.1038/mp.2016.184] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/11/2022]
Abstract
Mice lacking DIX domain containing-1 (DIXDC1), an intracellular Wnt/β-catenin signal pathway protein, have abnormal measures of anxiety, depression and social behavior. Pyramidal neurons in these animals' brains have reduced dendritic spines and glutamatergic synapses. Treatment with lithium or a glycogen synthase kinase-3 (GSK3) inhibitor corrects behavioral and neurodevelopmental phenotypes in these animals. Analysis of DIXDC1 in over 9000 cases of autism, bipolar disorder and schizophrenia reveals higher rates of rare inherited sequence-disrupting single-nucleotide variants (SNVs) in these individuals compared with psychiatrically unaffected controls. Many of these SNVs alter Wnt/β-catenin signaling activity of the neurally predominant DIXDC1 isoform; a subset that hyperactivate this pathway cause dominant neurodevelopmental effects. We propose that rare missense SNVs in DIXDC1 contribute to psychiatric pathogenesis by reducing spine and glutamatergic synapse density downstream of GSK3 in the Wnt/β-catenin pathway.
Collapse
|
100
|
Csabai D, Wiborg O, Czéh B. Reduced Synapse and Axon Numbers in the Prefrontal Cortex of Rats Subjected to a Chronic Stress Model for Depression. Front Cell Neurosci 2018; 12:24. [PMID: 29440995 PMCID: PMC5797661 DOI: 10.3389/fncel.2018.00024] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/16/2018] [Indexed: 12/28/2022] Open
Abstract
Stressful experiences can induce structural changes in neurons of the limbic system. These cellular changes contribute to the development of stress-induced psychopathologies like depressive disorders. In the prefrontal cortex of chronically stressed animals, reduced dendritic length and spine loss have been reported. This loss of dendritic material should consequently result in synapse loss as well, because of the reduced dendritic surface. But so far, no one studied synapse numbers in the prefrontal cortex of chronically stressed animals. Here, we examined synaptic contacts in rats subjected to an animal model for depression, where animals are exposed to a chronic stress protocol. Our hypothesis was that long term stress should reduce the number of axo-spinous synapses in the medial prefrontal cortex. Adult male rats were exposed to daily stress for 9 weeks and afterward we did a post mortem quantitative electron microscopic analysis to quantify the number and morphology of synapses in the infralimbic cortex. We analyzed asymmetric (Type I) and symmetric (Type II) synapses in all cortical layers in control and stressed rats. We also quantified axon numbers and measured the volume of the infralimbic cortex. In our systematic unbiased analysis, we examined 21,000 axon terminals in total. We found the following numbers in the infralimbic cortex of control rats: 1.15 × 109 asymmetric synapses, 1.06 × 108 symmetric synapses and 1.00 × 108 myelinated axons. The density of asymmetric synapses was 5.5/μm3 and the density of symmetric synapses was 0.5/μm3. Average synapse membrane length was 207 nm and the average axon terminal membrane length was 489 nm. Stress reduced the number of synapses and myelinated axons in the deeper cortical layers, while synapse membrane lengths were increased. These stress-induced ultrastructural changes indicate that neurons of the infralimbic cortex have reduced cortical network connectivity. Such reduced network connectivity is likely to form the anatomical basis for the impaired functioning of this brain area. Indeed, impaired functioning of the prefrontal cortex, such as cognitive deficits are common in stressed individuals as well as in depressed patients.
Collapse
Affiliation(s)
- Dávid Csabai
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Ove Wiborg
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary.,Institute of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|