1
|
Acero-Castillo MC, Correia MBM, Caixeta FV, Motta V, Barros M, Maior RS. Is the antidepressant effect of ketamine separate from its psychotomimetic effect? A review of rodent models. Neuropharmacology 2024; 258:110088. [PMID: 39032814 DOI: 10.1016/j.neuropharm.2024.110088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Ketamine is an NMDA (N-methyl-d-aspartate) glutamate receptor antagonist, which has a myriad of dose-dependent pharmacological and behavioral effects, including anesthetic, sedative, amnestic, analgesic, and anti-inflammatory properties. Intriguingly, ketamine at subanesthetic doses displays a relevant profile both in mimicking symptoms of schizophrenia and also as the first fast-acting treatment for depression. Here, we present an overview of the state-of-the-art knowledge about ketamine as an antidepressant as well as a pharmacological model of schizophrenia in animal models and human participants. Ketamine's dual effect appears to arise from its mechanism of action involving NMDA receptors, with both immediate and downstream consequences being triggered as a result. Finally, we discuss the feasibility of a unified approach linking the glutamatergic hypothesis of schizophrenia to the promising preclinical and clinical success of ketamine in the treatment of refractory depression.
Collapse
Affiliation(s)
- M C Acero-Castillo
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M B M Correia
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil; Department of Anthropology, Emory University, Atlanta GA, ZIP 30322, USA
| | - F V Caixeta
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - V Motta
- Department of Basic Psychological Processes, Institute of Psychology, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - M Barros
- Department of Pharmacy, School of Health Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil
| | - R S Maior
- Laboratory of Neuroscience, Metabolism, and Behavior, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia, ZIP 70910-900, Brasilia-DF, Brazil.
| |
Collapse
|
2
|
Arrabal-Gómez C, Beltran-Casanueva R, Hernández-García A, Bayolo-Guanche JV, Barbancho-Fernández MA, Serrano-Castro PJ, Narváez M. Enhancing Cognitive Functions and Neuronal Growth through NPY1R Agonist and Ketamine Co-Administration: Evidence for NPY1R-TrkB Heteroreceptor Complexes in Rats. Cells 2024; 13:669. [PMID: 38667284 PMCID: PMC11049095 DOI: 10.3390/cells13080669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
This study investigates the combined effects of the neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31-Pro34]NPY at a dose of 132 µg and Ketamine at 10 mg/Kg on cognitive functions and neuronal proliferation, against a backdrop where neurodegenerative diseases present an escalating challenge to global health systems. Utilizing male Sprague-Dawley rats in a physiological model, this research employed a single-dose administration of these compounds and assessed their impact 24 h after treatment on object-in-place memory tasks, alongside cellular proliferation within the dorsal hippocampus dentate gyrus. Methods such as the in situ proximity ligation assay and immunohistochemistry for proliferating a cell nuclear antigen (PCNA) and doublecortin (DCX) were utilized. The results demonstrated that co-administration significantly enhanced memory consolidation and increased neuronal proliferation, specifically neuroblasts, without affecting quiescent neural progenitors and astrocytes. These effects were mediated by the potential formation of NPY1R-TrkB heteroreceptor complexes, as suggested by receptor co-localization studies, although further investigation is required to conclusively prove this interaction. The findings also highlighted the pivotal role of brain-derived neurotrophic factor (BDNF) in mediating these effects. In conclusion, this study presents a promising avenue for enhancing cognitive functions and neuronal proliferation through the synergistic action of the NPY1R agonist and Ketamine, potentially via NPY1R-TrkB heteroreceptor complex formation, offering new insights into therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Carlos Arrabal-Gómez
- NeuronLab, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain; (C.A.-G.); (M.A.B.-F.)
- Facultad de Psicología, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain
- Unit of Neurology, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, 29010 Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, 29016 Málaga, Spain
| | - Rasiel Beltran-Casanueva
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; (R.B.-C.); (A.H.-G.); (J.V.B.-G.)
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain
| | - Aracelis Hernández-García
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; (R.B.-C.); (A.H.-G.); (J.V.B.-G.)
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain
| | - Juan Vicente Bayolo-Guanche
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden; (R.B.-C.); (A.H.-G.); (J.V.B.-G.)
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain
| | - Miguel Angel Barbancho-Fernández
- NeuronLab, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain; (C.A.-G.); (M.A.B.-F.)
| | - Pedro Jesús Serrano-Castro
- NeuronLab, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain; (C.A.-G.); (M.A.B.-F.)
- Unit of Neurology, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, 29010 Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, 29016 Málaga, Spain
| | - Manuel Narváez
- NeuronLab, Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, 29071 Málaga, Spain; (C.A.-G.); (M.A.B.-F.)
- Unit of Neurology, Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga, 29010 Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, 29016 Málaga, Spain
| |
Collapse
|
3
|
Abstract
Major depressive disorder (MDD) is a leading cause of suicide in the world. Monoamine-based antidepressant drugs are a primary line of treatment for this mental disorder, although the delayed response and incomplete efficacy in some patients highlight the need for improved therapeutic approaches. Over the past two decades, ketamine has shown rapid onset with sustained (up to several days) antidepressant effects in patients whose MDD has not responded to conventional antidepressant drugs. Recent preclinical studies have started to elucidate the underlying mechanisms of ketamine's antidepressant properties. Herein, we describe and compare recent clinical and preclinical findings to provide a broad perspective of the relevant mechanisms for the antidepressant action of ketamine.
Collapse
Affiliation(s)
- Ji-Woon Kim
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Institute of Regulatory Innovation through Science, Kyung Hee University, Seoul, Republic of Korea
| | - Kanzo Suzuki
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Tokyo, Japan
| | - Ege T Kavalali
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
| | - Lisa M Monteggia
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, Tennessee, USA;
| |
Collapse
|
4
|
Kim JW, Suzuki K, Kavalali ET, Monteggia LM. Bridging rapid and sustained antidepressant effects of ketamine. Trends Mol Med 2023; 29:364-375. [PMID: 36907686 PMCID: PMC10101916 DOI: 10.1016/j.molmed.2023.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/05/2023] [Accepted: 02/15/2023] [Indexed: 03/12/2023]
Abstract
Acute administration of (R,S)-ketamine (ketamine) produces rapid antidepressant effects that in some patients can be sustained for several days to more than a week. Ketamine blocks N-methyl-d-asparate (NMDA) receptors (NMDARs) to elicit specific downstream signaling that induces a novel form of synaptic plasticity in the hippocampus that has been linked to the rapid antidepressant action. These signaling events lead to subsequent downstream transcriptional changes that are involved in the sustained antidepressant effects. Here we review how ketamine triggers this intracellular signaling pathway to mediate synaptic plasticity which underlies the rapid antidepressant effects and links it to downstream signaling and the sustained antidepressant effects.
Collapse
Affiliation(s)
- Ji-Woon Kim
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA; College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea; Department of Regulatory Science, Gradaute School, Kyung Hee University, Seoul, Republic of Korea; Institute of Regulatory Innovation through Science, Kyung Hee University, Seoul, Republic of Korea
| | - Kanzo Suzuki
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA; Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika-ku, Japan
| | - Ege T Kavalali
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA
| | - Lisa M Monteggia
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA.
| |
Collapse
|
5
|
Colwell MJ, Tagomori H, Chapman S, Gillespie AL, Cowen PJ, Harmer CJ, Murphy SE. Pharmacological targeting of cognitive impairment in depression: recent developments and challenges in human clinical research. Transl Psychiatry 2022; 12:484. [PMID: 36396622 PMCID: PMC9671959 DOI: 10.1038/s41398-022-02249-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
Impaired cognition is often overlooked in the clinical management of depression, despite its association with poor psychosocial functioning and reduced clinical engagement. There is an outstanding need for new treatments to address this unmet clinical need, highlighted by our consultations with individuals with lived experience of depression. Here we consider the evidence to support different pharmacological approaches for the treatment of impaired cognition in individuals with depression, including treatments that influence primary neurotransmission directly as well as novel targets such as neurosteroid modulation. We also consider potential methodological challenges in establishing a strong evidence base in this area, including the need to disentangle direct effects of treatment on cognition from more generalised symptomatic improvement and the identification of sensitive, reliable and objective measures of cognition.
Collapse
Affiliation(s)
- Michael J Colwell
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Hosana Tagomori
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Sarah Chapman
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Amy L Gillespie
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Philip J Cowen
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Catherine J Harmer
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Susannah E Murphy
- University Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK.
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK.
| |
Collapse
|
6
|
Onaolapo AY, Onaolapo OJ. Glutamate and depression: Reflecting a deepening knowledge of the gut and brain effects of a ubiquitous molecule. World J Psychiatry 2021; 11:297-315. [PMID: 34327123 PMCID: PMC8311508 DOI: 10.5498/wjp.v11.i7.297] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
The versatility of glutamate as the brain’s foremost excitatory neurotransmitter and modulator of neurotransmission and function is considered common knowledge. Years of research have continued to uncover glutamate’s effects and roles in several neurological and neuropsychiatric disorders, including depression. It had been considered that a deeper understanding of the roles of glutamate in depression might open a new door to understanding the pathological basis of the disorder, improve the approach to patient management, and lead to the development of newer drugs that may benefit more patients. This review examines our current understanding of the roles of endogenous and exogenous sources of glutamate and the glutamatergic system in the aetiology, progression and management of depression. It also examines the relationships that link the gut-brain axis, glutamate and depression; as it emphasizes how the gut-brain axis could impact depression pathogenesis and management via changes in glutamate homeostasis. Finally, we consider what the likely future of glutamate-based therapies and glutamate-based therapeutic manipulations in depression are, and if with them, we are now on the final chapter of understanding the neurochemical milieu of depressive disorders.
Collapse
Affiliation(s)
- Adejoke Yetunde Onaolapo
- Behavioural Neuroscience Unit, Neurobiology Subdivision, Department of Anatomy, Ladoke Akintola University of Technology, Oyo State 234, Nigeria
| | - Olakunle James Onaolapo
- Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Department of Pharmacology, Ladoke Akintola University of Technology, Oyo State 234, Nigeria
| |
Collapse
|
7
|
Castrén E, Monteggia LM. Brain-Derived Neurotrophic Factor Signaling in Depression and Antidepressant Action. Biol Psychiatry 2021; 90:128-136. [PMID: 34053675 DOI: 10.1016/j.biopsych.2021.05.008] [Citation(s) in RCA: 261] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 12/19/2022]
Abstract
Neurotrophic factors, particularly BDNF (brain-derived neurotrophic factor), have been associated with depression and antidepressant drug action. A variety of preclinical and clinical studies have implicated impaired BDNF signaling through its receptor TrkB (neurotrophic receptor tyrosine kinase 2) in the pathophysiology of mood disorders, but many of the initial findings have not been fully supported by more recent meta-analyses, and more both basic and clinical research is needed. In contrast, increased expression and signaling of BDNF has been repeatedly implicated in the mechanisms of both typical and rapid-acting antidepressant drugs, and recent findings have started to elucidate the mechanisms through which antidepressants regulate BDNF signaling. BDNF is a critical regulator of various types of neuronal plasticities in the brain, and plasticity has increasingly been connected with antidepressant action. Although some equivocal data exist, the hypothesis of a connection between neurotrophic factors and neuronal plasticity with mood disorders and antidepressant action has recently been further strengthened by converging evidence from a variety of more recent data reviewed here.
Collapse
Affiliation(s)
- Eero Castrén
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - Lisa M Monteggia
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
8
|
Małgorzata P, Paweł K, Iwona ML, Brzostek T, Andrzej P. Glutamatergic dysregulation in mood disorders: opportunities for the discovery of novel drug targets. Expert Opin Ther Targets 2020; 24:1187-1209. [PMID: 33138678 DOI: 10.1080/14728222.2020.1836160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Recently, a considerable attention has been paid to glutamatergic conception of mood disorders. The development of new treatment strategies targeted at glutamate provides new opportunities for the treatment of mood disorders. It is expected that these novel therapeutic options will provide a fast and sustained antidepressant effect and will be better tolerated by patients than the currently available antidepressants. AREAS COVERED This paper discusses glutamatergic abnormalities in mood disorders and reviews novel glutamate-based drugs developed for the treatment of these disorders. We have searched the PubMed and EMBASE databases, presented the results of relevant clinical studies and also describe novel glutamate-based agents that are under investigation. EXPERT OPINION The glutamatergic system plays many important roles in energy metabolism of the brain and neurotransmission; therefore, any attempt to identify novel therapeutic targets within this system seems justified. The effective development of new glutamate-based drugs requires, among others, a more in-depth exploration and understanding of the anatomy, function, and localization of different glutamatergic receptors in the brain. In our opinion, novel glutamate-based antidepressants will find application in the treatment of mood disorders and present an option will be widely used in clinical practice in the future.
Collapse
Affiliation(s)
- Panek Małgorzata
- Department of Biotechnology and General Technology of Food, Faculty of Food Technology, University of Agriculture , Kraków, Poland
| | - Kawalec Paweł
- Department of Nutrition and Drug Research, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University , Kraków, Poland
| | - Malinowska Lipień Iwona
- Department of Internal Medicine and Community Nursing, Faculty of Health Sciences, Jagiellonian University Medical College , Kraków, Poland
| | - Tomasz Brzostek
- Department of Internal Medicine and Community Nursing, Faculty of Health Sciences, Jagiellonian University Medical College , Kraków, Poland
| | - Pilc Andrzej
- Department of Nutrition and Drug Research, Institute of Public Health, Faculty of Health Sciences, Jagiellonian University , Kraków, Poland.,Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences , Kraków, Poland
| |
Collapse
|
9
|
Shin C, Kim YK. Ketamine in Major Depressive Disorder: Mechanisms and Future Perspectives. Psychiatry Investig 2020; 17:181-192. [PMID: 32209965 PMCID: PMC7113176 DOI: 10.30773/pi.2019.0236] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/18/2019] [Indexed: 02/08/2023] Open
Abstract
Major depressive disorder (MDD) is a serious psychiatric illness that causes functional impairment in many people. While monoaminergic antidepressants have been used to effectively treat MDD, these antidepressants have limitations in that they have delayed onset of action and many patients remain treatment-resistant. Therefore, there is a need to develop antidepressants with a novel target, and researchers have directed their attention to the glutamatergic system. Ketamine, although developed as an anesthetic, has been found to produce an antidepressant effect at sub-anesthetic doses via N-Methyl-D-aspartic acid (NMDA) receptor blockade as well as NMDA receptor- independent pathways. A single infusion of ketamine produced rapid improvement in clinical symptoms to a considerable level and led to the resolution of serious depressive symptoms, including imminent suicidal ideation, in patients with MDD. A series of recent randomized controlled trials have provided a high level of evidence for the therapeutic efficacy of ketamine treatment in MDD and presented new insights on the dose, usage, and route of administration of ketamine as an antidepressant. With this knowledge, it is expected that ketamine treatment protocols for MDD will be established as a treatment option available in clinical practice. However, long-term safety must be taken into consideration as ketamine has abuse potential and it is associated with psychological side effects such as dissociative or psychotomimetic effects.
Collapse
Affiliation(s)
- Cheolmin Shin
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| |
Collapse
|
10
|
Yang H, Hong W, Chen L, Tao Y, Peng Z, Zhou H. Analysis of risk factors for depression in Alzheimer's disease patients. Int J Neurosci 2020; 130:1136-1141. [PMID: 32053409 DOI: 10.1080/00207454.2020.1730369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose: Depression, which affects about 52% of Alzheimer's disease (AD) patients, can worsen cognitive impairment and increase mortality and suicide rates. We hope to provide clinical evidence for the prevention and treatment of depression in AD patients by investigating related risk factors of depression in AD patients.Methods: 158 AD inpatients of the Department of Neurology, Daping Hospital from September 2017 to March 2019 were enrolled. General information, laboratory tests, cognitive and emotional function assessments of the inpatients were collected. Logistic regression was used to analyze the risk factors of depression in AD patients, and the relationship between 17 Hamilton depression scale scores and HbA1c levels in AD patients was further analyzed.Results: The prevalence of age, gender, hypertension, hyperlipidemia, Type 2 diabetes mellitus (T2DM), and white matter lesions (WML) in the AD with depression group was significantly different from without depression group. Hypertension, T2DM, and WML are independent risk factors for depression in AD patients. The depression scores of AD patients with HbA1c>6.5% were significantly higher than AD patients with HbA1c ≤ 6.5%, and there were significant difference in depression scale scores between using anti-diabetes drugs group and not using anti-diabetes drugs group whose HbA1c level is >6.5%, while no difference in depression scores between using anti-diabetes drugs group and not using anti-diabetes drugs group whose HbA1c level is ≤6.5%.Conclusion: T2DM is an independent risk factor for AD patients with depression. Increased HbA1c levels aggravate depression in AD patients, and controlling HbA1c levels and anti-diabetes drugs can reduce the severity of depression in AD patients.
Collapse
Affiliation(s)
- Hai Yang
- Department of Neurology, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Wenjuan Hong
- Graduate School of Bengbu Medical College, Bengbu City, China
| | - Le Chen
- Graduate School of Bengbu Medical College, Bengbu City, China
| | - Yong Tao
- Department of Neurology, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Zeyan Peng
- Department of Neurology, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Huadong Zhou
- Department of Neurology, Daping Hospital, Army Military Medical University, Chongqing, China
| |
Collapse
|
11
|
Lavretsky H, Laird KT, Krause-Sorio B, Heimberg BF, Yeargin J, Grzenda A, Wu P, Thana-Udom K, Ercoli LM, Siddarth P. A Randomized Double-Blind Placebo-Controlled Trial of Combined Escitalopram and Memantine for Older Adults With Major Depression and Subjective Memory Complaints. Am J Geriatr Psychiatry 2020; 28:178-190. [PMID: 31519517 PMCID: PMC6997044 DOI: 10.1016/j.jagp.2019.08.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/07/2019] [Accepted: 08/13/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Geriatric depression is difficult to treat and frequently accompanied by cognitive complaints that increase risk for dementia. New treatment strategies targeting both depression and cognition are urgently needed. METHODS We conducted a 6-month double-blind placebo-controlled trial to assess the efficacy and tolerability of escitalopram + memantine (ESC/MEM) compared to escitalopram + placebo (ESC/PBO) for improving mood and cognitive functioning in depressed older adults with subjective memory complaints (NCT01902004). Primary outcome was change in depression as assessed by the HAM-D post-treatment (at 6 months). Remission was defined as HAM-D ≤6; naturalistic follow-up continued until 12 months. RESULTS Of the 95 randomized participants, 62 completed the 6-month assessment. Dropout and tolerability did not differ between groups. Mean daily escitalopram dose was 11.1 mg (SD = 3.7; range: 5-20 mg). Mean daily memantine dose was 19.3 mg (SD = 2.6; range 10-20 mg). Remission rate within ESC/MEM was 45.8% and 47.9%, compared to 38.3% and 31.9% in ESC/PBO, at 3 and 6 months, respectively (χ2(1) = 2.0, p = 0.15). Both groups improved significantly on the HAM-D at 3, 6, and 12 months, with no observed between-group differences. ESC/MEM demonstrated greater improvement in delayed recall (F(2,82) = 4.3, p = 0.02) and executive functioning (F(2,82) = 5.1, p = 0.01) at 12 months compared to ESC/PBO. CONCLUSIONS The combination of memantine with escitalopram was well tolerated and as effective as escitalopram and placebo in improving depression using HAM-D. Combination memantine and escitalopram was significantly more effective than escitalopram and placebo in improving cognitive outcomes at 12 months. Future reports will address the role of biomarkers of aging in treatment response.
Collapse
Affiliation(s)
- Helen Lavretsky
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA.
| | - Kelsey T Laird
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA
| | - Beatrix Krause-Sorio
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA
| | - Brandon F Heimberg
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA
| | - Jillian Yeargin
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA
| | - Adrienne Grzenda
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA
| | - Pauline Wu
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA
| | - Kitikan Thana-Udom
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA; Department of Psychiatry, Faculty of Medicine Siriraj Hospital, Mahidol University (KT-U), Bangkok, Thailand
| | - Linda M Ercoli
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA
| | - Prabha Siddarth
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, UCLA (HL, KTL, BK-S, BFH, JY, AG, PW, KT-U, LME, PS), Los Angeles, CA
| |
Collapse
|
12
|
Suzuki K, Monteggia LM. The role of eEF2 kinase in the rapid antidepressant actions of ketamine. RAPID ACTING ANTIDEPRESSANTS 2020; 89:79-99. [DOI: 10.1016/bs.apha.2020.04.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
13
|
Abstract
This article covers current research on the relationship between depression and cognitive impairment in older adults. First, it approaches the clinical assessment of late-life depression and comorbid cognitive impairment. Cognitive risk factors for suicide are discussed. Research is then provided on neuropsychological changes associated with depression, discussing subjective cognitive impairment, mild cognitive impairment, and dementia profiles. In addition, literature regarding neuroimaging and biomarker findings in depressed older adults is presented. Finally, therapeutic models for treatment of late-life depression are discussed, including psychotherapy models, holistic treatments, pharmacologic approaches, and brain stimulation therapies.
Collapse
Affiliation(s)
- Ryan D Greene
- Department of Psychology in Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush VA Medical Center, Indianapolis, IN, USA; University of Indianapolis, Indianapolis, IN, USA.
| | - Alex Cook
- University of Indianapolis, Indianapolis, IN, USA
| | - Dustin Nowaskie
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sophia Wang
- Roudebush VA Medical Center, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA; Center of Health Innovation and Implementation Science, Center for Translational Science and Innovation, Indianapolis, IN, USA; Sandra Eskenazi Center for Brain Care Innovation, Eskenazi Hospital, Indianapolis, IN, USA
| |
Collapse
|
14
|
Pham TH, Gardier AM. Fast-acting antidepressant activity of ketamine: highlights on brain serotonin, glutamate, and GABA neurotransmission in preclinical studies. Pharmacol Ther 2019; 199:58-90. [DOI: 10.1016/j.pharmthera.2019.02.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/25/2019] [Indexed: 12/13/2022]
|
15
|
Simning A, Kittel J, Conwell Y. Late-Life Depressive and Anxiety Symptoms Following Rehabilitation Services in Medicare Beneficiaries. Am J Geriatr Psychiatry 2019; 27:381-390. [PMID: 30655031 PMCID: PMC6431271 DOI: 10.1016/j.jagp.2018.12.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim of this study was to determine whether patients who received rehabilitation services had an increased risk of having late-life depressive or anxiety symptoms within the year following termination of services. METHODS The National Health and Aging Trends Study (NHATS) is a population-based, longitudinal cohort survey of a nationally representative sample of Medicare beneficiaries aged 65years and older. This study involved 5,979 participants from the 2016 NHATS survey. The Patient Health Questionnaire-2 and Generalized Anxiety Disorder 2-item assessed for clinically significant depressive and anxiety symptoms. RESULTS The prevalence of depressive and anxiety symptoms was higher in older adults who had received rehabilitation services in the year prior and varied by site: no rehabilitation (depressive and anxiety symptoms): 10.4% and 8.8%; nursing home or inpatient rehabilitation: 38.8% and 23.8%; outpatient rehabilitation: 8.6% and 5.5%; in-home rehabilitation: 35.3% and 20.5%; multiple rehabilitation sites: 20.3% and 14.4%; and any rehabilitation site: 18.4% and 11.8%. In multiple logistic regression analyses, nursing home and inpatient and in-home rehabilitation services, respectively, were associated with an increased risk of having subsequent depressive symptoms (odds ratio: 3.51; 95% confidence interval [CI]: 1.85-6.63; OR: 2.15; 95% CI: 1.08-4.30) but not anxiety symptoms. CONCLUSION Older adults who receive rehabilitation services are at risk of having depressive and anxiety symptoms after these services have terminated. As mental illness is associated with considerable morbidity and may affect rehabilitation outcomes, additional efforts to identify and treat depression and anxiety in these older adults may be warranted.
Collapse
Affiliation(s)
- Adam Simning
- Department of Psychiatry (AS, YC), University of Rochester School of Medicine and Dentistry, Rochester, NY.
| | | | - Yeates Conwell
- University of Rochester School of Medicine and Dentistry (URSMD), Department of Psychiatry,University of Rochester Medical Center, Office for Aging Research and Health Services
| |
Collapse
|
16
|
Depression as a Neuroendocrine Disorder: Emerging Neuropsychopharmacological Approaches beyond Monoamines. Adv Pharmacol Sci 2019; 2019:7943481. [PMID: 30719038 PMCID: PMC6335777 DOI: 10.1155/2019/7943481] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 01/26/2023] Open
Abstract
Depression is currently recognized as a crucial problem in everyday clinical practice, in light of ever-increasing rates of prevalence, as well as disability, morbidity, and mortality related to this disorder. Currently available antidepressant drugs are notoriously problematic, with suboptimal remission rates and troubling side-effect profiles. Their mechanisms of action focus on the monoamine hypothesis for depression, which centers on the disruption of serotonergic, noradrenergic, and dopaminergic neurotransmission in the brain. Nevertheless, views on the pathophysiology of depression have evolved notably, and the comprehension of depression as a complex neuroendocrine disorder with important systemic implications has sparked interest in a myriad of novel neuropsychopharmacological approaches. Innovative pharmacological targets beyond monoamines include glutamatergic and GABAergic neurotransmission, brain-derived neurotrophic factor, various endocrine axes, as well as several neurosteroids, neuropeptides, opioids, endocannabinoids and endovanilloids. This review summarizes current knowledge on these pharmacological targets and their potential utility in the clinical management of depression.
Collapse
|
17
|
Abstract
UNLABELLED This paper reviews recent research on late-life depression (LLD) pharmacotherapy, focusing on updated information for monotherapy and augmentation treatments. We then review new research on moderators of clinical response and how to use the information for improved efficacy. RECENT FINDINGS A recent review shows that sertraline, paroxetine, and duloxetine were superior to placebo for the treatment of LLD. There is concern that paroxetine could have adverse outcomes in the geriatric population due to anticholinergic properties; however, studies show no increases in mortality, dementia risk, or cognitive measures. Among newer antidepressants, vortioxetine has demonstrated efficacy in LLD, quetiapine has demonstrated efficacy especially for patients with sleep disturbances, and aripiprazole augmentation for treatment resistance in LLD was found to be safe and effective. Researchers have also been identifying moderators of LLD that can guide treatment. Researchers are learning how to associate moderators, neuroanatomical models, and antidepressant response. SSRI/SNRIs remain first-line treatment for LLD. Aripiprazole is an effective and safe augmentation for treatment resistance. Studies are identifying actionable moderators that can increase treatment response.
Collapse
|
18
|
Abstract
Clinical studies have demonstrated that a single sub-anesthetic dose of the dissociative anesthetic ketamine induces rapid and sustained antidepressant actions. Although this finding has been met with enthusiasm, ketamine's widespread use is limited by its abuse potential and dissociative properties. Recent preclinical research has focused on unraveling the molecular mechanisms underlying the antidepressant actions of ketamine in an effort to develop novel pharmacotherapies, which will mimic ketamine's antidepressant actions but lack its undesirable effects. Here we review hypotheses for the mechanism of action of ketamine as an antidepressant, including synaptic or GluN2B-selective extra-synaptic N-methyl-D-aspartate receptor (NMDAR) inhibition, inhibition of NMDARs localized on GABAergic interneurons, inhibition of NMDAR-dependent burst firing of lateral habenula neurons, and the role of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor activation. We also discuss links between ketamine's antidepressant actions and downstream mechanisms regulating synaptic plasticity, including brain-derived neurotrophic factor (BDNF), eukaryotic elongation factor 2 (eEF2), mechanistic target of rapamycin (mTOR) and glycogen synthase kinase-3 (GSK-3). Mechanisms that do not involve direct inhibition of the NMDAR, including a role for ketamine's (R)-ketamine enantiomer and hydroxynorketamine (HNK) metabolites, specifically (2R,6R)-HNK, are also discussed. Proposed mechanisms of ketamine's action are not mutually exclusive and may act in a complementary manner to exert acute changes in synaptic plasticity, leading to sustained strengthening of excitatory synapses, which are necessary for antidepressant behavioral actions. Understanding the molecular mechanisms underpinning ketamine's antidepressant actions will be invaluable for the identification of targets, which will drive the development of novel, effective, next-generation pharmacotherapies for the treatment of depression.
Collapse
|
19
|
Abstract
Traditional pharmacological treatments for depression have a delayed therapeutic onset, ranging from several weeks to months, and there is a high percentage of individuals who never respond to treatment. In contrast, ketamine produces rapid-onset antidepressant, anti-suicidal, and anti-anhedonic actions following a single administration to patients with depression. Proposed mechanisms of the antidepressant action of ketamine include N-methyl-D-aspartate receptor (NMDAR) modulation, gamma aminobutyric acid (GABA)-ergic interneuron disinhibition, and direct actions of its hydroxynorketamine (HNK) metabolites. Downstream actions include activation of the mechanistic target of rapamycin (mTOR), deactivation of glycogen synthase kinase-3 and eukaryotic elongation factor 2 (eEF2), enhanced brain-derived neurotrophic factor (BDNF) signaling, and activation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPARs). These putative mechanisms of ketamine action are not mutually exclusive and may complement each other to induce potentiation of excitatory synapses in affective-regulating brain circuits, which results in amelioration of depression symptoms. We review these proposed mechanisms of ketamine action in the context of how such mechanisms are informing the development of novel putative rapid-acting antidepressant drugs. Such drugs that have undergone pre-clinical, and in some cases clinical, testing include the muscarinic acetylcholine receptor antagonist scopolamine, GluN2B-NMDAR antagonists (i.e., CP-101,606, MK-0657), (2R,6R)-HNK, NMDAR glycine site modulators (i.e., 4-chlorokynurenine, pro-drug of the glycineB NMDAR antagonist 7-chlorokynurenic acid), NMDAR agonists [i.e., GLYX-13 (rapastinel)], metabotropic glutamate receptor 2/3 (mGluR2/3) antagonists, GABAA receptor modulators, and drugs acting on various serotonin receptor subtypes. These ongoing studies suggest that the future acute treatment of depression will typically occur within hours, rather than months, of treatment initiation.
Collapse
Affiliation(s)
- Panos Zanos
- Department of Psychiatry, University of Maryland School of Medicine, Rm. 934F MSTF, 685 W. Baltimore St., Baltimore, MD, 21201, USA.
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, St. BRB 5-007, 655 W. Baltimore St., Baltimore, MD, 21201, USA, Baltimore, MD, 21201, USA
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Rm. 936 MSTF, 685 W. Baltimore St., Baltimore, MD, 21201, USA
| |
Collapse
|
20
|
Abstract
This article covers current research on the relationship between depression and cognitive impairment in older adults. First, it approaches the clinical assessment of late-life depression and comorbid cognitive impairment. Cognitive risk factors for suicide are discussed. Research is then provided on neuropsychological changes associated with depression, discussing subjective cognitive impairment, mild cognitive impairment, and dementia profiles. Additionally, literature regarding neuroimaging and biomarker findings in depressed older adults is presented. Finally, therapeutic models for treatment of late-life depression are also discussed, including psychotherapy models, holistic treatments, pharmacologic approaches, and brain-stimulation therapies.
Collapse
|
21
|
Ghasemi M, Phillips C, Fahimi A, McNerney MW, Salehi A. Mechanisms of action and clinical efficacy of NMDA receptor modulators in mood disorders. Neurosci Biobehav Rev 2017; 80:555-572. [DOI: 10.1016/j.neubiorev.2017.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 06/23/2017] [Accepted: 07/08/2017] [Indexed: 12/22/2022]
|
22
|
Nicotine and networks: Potential for enhancement of mood and cognition in late-life depression. Neurosci Biobehav Rev 2017; 84:289-298. [PMID: 28859996 DOI: 10.1016/j.neubiorev.2017.08.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/11/2017] [Accepted: 08/25/2017] [Indexed: 11/23/2022]
Abstract
Late-life depression is characterized by both lower mood and poor cognitive performance, symptoms that often do not fully respond to current antidepressant medications. Nicotinic acetylcholine receptor (nAChR) agonists such as nicotine may serve as a novel therapeutic approach for this population. Both preclinical and preliminary clinical studies suggest that nAChR agonists can improve depressive behavior in animal models and improve mood in depressed individuals. Substantial literature also supports that nAChR agonists benefit cognitive performance, particularly in older populations. These potential benefits may be mediated by the effects of nAChR stimulation on neural network function and connectivity. Functional neuroimaging studies detail effects of nAChR agonists on the default mode network, central-executive network, and salience network that may oppose or reverse network changes seen in depression. We propose that, given the existent literature and the clinical presentation of late-life depression, nicotine or other nAChR agonists may have unique therapeutic benefits in this population and that clinical trials examining nicotine effects on mood, cognition, and network dynamics in late-life depression are justified.
Collapse
|
23
|
Harraz MM, Snyder SH. Antidepressant Actions of Ketamine Mediated by the Mechanistic Target of Rapamycin, Nitric Oxide, and Rheb. Neurotherapeutics 2017; 14:728-733. [PMID: 28612328 PMCID: PMC5509634 DOI: 10.1007/s13311-017-0540-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The weeks/months it takes for traditional antidepressants to act pose an obstacle in the management of depression. Ketamine's prompt and sustained antidepressant effects constitute a major advance. Multiple studies implicate glutamatergic signaling to protein synthesis machinery and synapse formation in ketamine's antidepressant effects. Here we review evidence linking ketamine to glutamate receptor subtypes and protein homeostasis. We describe a signaling cascade wherein nitric oxide drives the formation of a ternary protein complex comprised of glyceraldehyde 3-phosphate dehydrogenase, seven in absentia homolog 1, and Ras homolog enriched in brain downstream of the glutamate N-methyl-D-aspartate receptor. Seven in absentia homolog 1 ubiquitylates and degrades Ras homolog enriched in brain leading to inhibition of mechanistic target of rapamycin. Ketamine inhibits this molecular cascade leading to activation of mechanistic target of rapamycin and, in turn, to antidepressant actions.
Collapse
Affiliation(s)
- Maged M Harraz
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
24
|
Simning A, Simons KV. Treatment of depression in nursing home residents without significant cognitive impairment: a systematic review. Int Psychogeriatr 2017; 29:209-226. [PMID: 27758728 PMCID: PMC5469363 DOI: 10.1017/s1041610216001733] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Depression in nursing facilities is widespread and has been historically under-recognized and inadequately treated. Many interventions have targeted depression among residents with dementia in these settings. Less is known about depression treatment in residents without dementia who may be more likely to return to community living. Our study aimed to systematically evaluate randomized control trials (RCTs) in nursing facilities that targeted depression within samples largely comprised of residents without dementia. METHODS The following databases were evaluated with searches covering January 1991 to December 2015 (PubMed, PsycINFO) and March 2016 (CINAHL). We also examined national and international clinical trial registries including ClinicalTrials.gov. RCTs were included if they were published in English, evaluated depression or depressive symptoms as primary or secondary outcomes, and included a sample with a mean age of 65 years and over for which most had no or only mild cognitive impairment. RESULTS A total of 32 RCTs met our criteria including those testing psychotherapeutic interventions (n=13), psychosocial and recreation interventions (n=9), and pharmacologic or other biologic interventions (n=10). Seven psychotherapeutic, six psychosocial and recreation, and four pharmacologic or other biologic interventions demonstrated a treatment benefit. CONCLUSIONS Many studies had small samples, were of poor methodological quality, and did not select for depressed residents. There is limited evidence suggesting that cognitive behavioral therapies, reminiscence, interventions to reduce social isolation, and exercise-based interventions have some promise for decreasing depression in cognitively intact nursing home residents; little can be concluded from the pharmacologic or other biologic RCTs.
Collapse
Affiliation(s)
- Adam Simning
- Department of Psychiatry,University of Rochester School of Medicine and Dentistry,Rochester,New York,USA
| | - Kelsey V Simons
- Department of Psychiatry,University of Rochester School of Medicine and Dentistry,Rochester,New York,USA
| |
Collapse
|
25
|
Potential involvement of serotonergic signaling in ketamine's antidepressant actions: A critical review. Prog Neuropsychopharmacol Biol Psychiatry 2016; 71:27-38. [PMID: 27262695 DOI: 10.1016/j.pnpbp.2016.05.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/24/2016] [Accepted: 05/31/2016] [Indexed: 02/07/2023]
Abstract
A single i.v. infusion of ketamine, classified as an N-methyl-d-aspartate (NMDA) receptor antagonist, may alleviate depressive symptoms within hours of administration in treatment resistant depressed patients, and the antidepressant effect may last for several weeks. These unique therapeutic properties have prompted researchers to explore the mechanisms mediating the antidepressant effects of ketamine, but despite many efforts, no consensus on its antidepressant mechanism of action has been reached. Recent preclinical reports have associated the neurotransmitter serotonin (5-hydroxytryptamine; 5-HT) with the antidepressant-like action of ketamine. Here, we review the current evidence for a serotonergic role in ketamine's antidepressant effects. The pharmacological profile of ketamine may include equipotent activity on several non-NMDA targets, and the current hypotheses for the mechanisms responsible for ketamine's antidepressant activity do not appear to preclude the possibility that non-glutamate neurotransmitters are involved in the antidepressant effects. At multiple levels, the serotonergic and glutamatergic systems interact, and such crosstalk could support the notion that changes in serotonergic neurotransmission may impact ketamine's antidepressant potential. In line with these prospects, ketamine may increase 5-HT levels in the prefrontal cortex of rats, plausibly via hippocampal NMDA receptor inhibition and activation of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors. In addition, a number of preclinical studies suggest that the antidepressant-like effects of ketamine may depend on endogenous activation of 5-HT receptors. Recent imaging and behavioral data predominantly support a role for 5-HT1A or 5-HT1B receptors, but the full range of 5-HT receptors has currently not been systematically investigated in this context. Furthermore, the nature of any 5-HT dependent mechanism in ketamine's antidepressant effect is currently not understood, and therefore, more studies are warranted to confirm this hypothesis and explore the specific pathways that might implicate 5-HT.
Collapse
|
26
|
Zhang K, Yamaki VN, Wei Z, Zheng Y, Cai X. Differential regulation of GluA1 expression by ketamine and memantine. Behav Brain Res 2016; 316:152-159. [PMID: 27599619 DOI: 10.1016/j.bbr.2016.09.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 12/20/2022]
Abstract
Evidence from preclinical and clinical studies shows that ketamine, a noncompetitive NMDA receptor antagonist, exerts rapid and sustained antidepressant responses. However, ketamine's psychotomimetic side effects and abuse liability limit the clinical use of the compound. Interestingly, memantine, another NMDA receptor channel blocker, processes no defined antidepressant property but is much safer and clinical tolerated. Understanding why ketamine but not memantine exhibits rapid antidepressant responses is important to elucidate the cellular signaling underlying the fast antidepressant actions of ketamine and to design a new safer generation of fast-acting antidepressants. Here we show that ketamine but memantine caused a rapid and sustained antidepressant-like responses in forced swim test (FST). Both drugs enhanced GluA1 S845 phosphorylation and potentiated Schaffer collateral-CA1 synaptic transmission. However, ketamine but not memantine elevated the expression of GluA1. Incubating acutely prepared hippocampal slices with ketamine but not memantine enhanced mTOR phosphorylation in a time course parallel to the time course of GluA1 elevation. Our results suggest that distinct properties in regulation of mTOR phosphorylation and synaptic protein expression may underlie the differential effectiveness of ketamine and memantine in their antidepressant responses.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Vitor Nagai Yamaki
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Zhisheng Wei
- The Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong, 51030, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Yu Zheng
- The Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong, 51030, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Xiang Cai
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA; Center for Integrated Research in Cognitive & Neural Sciences, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA.
| |
Collapse
|
27
|
Demontis F, Serra G. Failure of memantine to “reverse” quinpirole-induced hypomotility. World J Psychiatry 2016; 6:215-220. [PMID: 27354963 PMCID: PMC4919260 DOI: 10.5498/wjp.v6.i2.215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/28/2016] [Accepted: 03/16/2016] [Indexed: 02/05/2023] Open
Abstract
AIM: To evaluate antidepressant-like effect of memantine in a rat model.
METHODS: Male Wistar rats were treated intraperitoneally with either vehicle, memantine (10 mg/kg) or imipramine (20 mg/kg), for 3 wk. Twenty-four hour after the last treatment animals were challenged with quinpirole (0.3 mg/kg s.c.) and tested for motor activity. After 1 h habituation to the motility cages, the motor response was recorded for the following 45-min and the data were collected in 5-min time bins.
RESULTS: As expected, chronic treatment with imipramine potentiated the locomotor stimulant effect of quinpirole. On the contrary, chronic memantine administration failed to induce the behavioral supersensitivity to the dopamine agonist.
CONCLUSION: The results show that memantine, at variance with antidepressant treatments, fails to induce dopaminergic behavioral supersensitivity. This observation is consistent with the results of preclinical and clinical studies suggesting that memantine does not have an acute antidepressant action but does have an antimanic and mood-stabilizing effect.
Collapse
|
28
|
Papp M, Gruca P, Lason-Tyburkiewicz M, Willner P. Antidepressant, anxiolytic and procognitive effects of rivastigmine and donepezil in the chronic mild stress model in rats. Psychopharmacology (Berl) 2016; 233:1235-43. [PMID: 26769042 PMCID: PMC4801996 DOI: 10.1007/s00213-016-4206-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/04/2016] [Indexed: 01/28/2023]
Abstract
BACKGROUND The treatment of depression in old age is complicated by frequent co-morbidity with cognitive impairment. Anti-dementia drugs have some efficacy to improve cognitive performance and there is an inconsistent literature regarding the effect of such drugs on depressive symptoms. Here, we have investigated whether anti-dementia drugs would have antidepressant-like and pro-cognitive effects in a well-validated animal model of depression and cognitive impairment, chronic mild stress (CMS). METHODS Rats were subjected to CMS for a total of 8 weeks. After 2 weeks, subgroups of stressed and non-stressed animals were treated daily, for 5 weeks followed by 1 week of drug withdrawal, with vehicle, imipramine (10 mg/kg), rivastigmine (2 mg/kg), donepezil (0.3 mg/kg) or memantine (5 mg/kg). Sucrose intake was tested weekly, and animals were also tested in the elevated plus maze (at week 7) and in an object recognition task (at weeks 7 and 8). RESULTS CMS decreased sucrose intake, had an anxiogenic effect in the elevated plus maze, and impaired performance in the object recognition test. Imipramine, rivastigmine and donepezil normalized performance in all three tests. Memantine had anxiolytic and pro-cognitive effects, but did not reverse CMS-induced anhedonia. DISCUSSION The fact that all three anti-dementia drugs reversed CMS-induced cognitive impairment and that cholinesterase inhibitors, but not memantine, have antidepressant-like effects in this model suggest that different mechanisms may underlie CMS-induced anhedonia and cognitive impairment. We discuss the clinical implications of these findings.
Collapse
Affiliation(s)
- Mariusz Papp
- Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343, Krakow, Poland.
| | - Piotr Gruca
- />Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | | | - Paul Willner
- />Department of Psychology, Swansea University, Swansea, UK
| |
Collapse
|
29
|
Björkholm C, Monteggia LM. BDNF - a key transducer of antidepressant effects. Neuropharmacology 2015; 102:72-9. [PMID: 26519901 DOI: 10.1016/j.neuropharm.2015.10.034] [Citation(s) in RCA: 675] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/13/2015] [Accepted: 10/25/2015] [Indexed: 12/25/2022]
Abstract
How do antidepressants elicit an antidepressant response? Here, we review accumulating evidence that the neurotrophin brain-derived neurotrophic factor (BDNF) serves as a transducer, acting as the link between the antidepressant drug and the neuroplastic changes that result in the improvement of the depressive symptoms. Over the last decade several studies have consistently highlighted BDNF as a key player in antidepressant action. An increase in hippocampal and cortical expression of BDNF mRNA parallels the antidepressant-like response of conventional antidepressants such as SSRIs. Subsequent studies showed that a single bilateral infusion of BDNF into the ventricles or directly into the hippocampus is sufficient to induce a relatively rapid and sustained antidepressant-like effect. Importantly, the antidepressant-like response to conventional antidepressants is attenuated in mice where the BDNF signaling has been disrupted by genetic manipulations. Low dose ketamine, which has been found to induce a rapid antidepressant effect in patients with treatment-resistant depression, is also dependent on increased BDNF signaling. Ketamine transiently increases BDNF translation in hippocampus, leading to enhanced synaptic plasticity and synaptic strength. Ketamine has been shown to increase BDNF translation by blocking NMDA receptor activity at rest, thereby inhibiting calcium influx and subsequently halting eukaryotic elongation factor 2 (eEF2) kinase leading to a desuppression of protein translation, including BDNF translation. The antidepressant-like response of ketamine is abolished in BDNF and TrkB conditional knockout mice, eEF2 kinase knockout mice, in mice carrying the BDNF met/met allele, and by intra-cortical infusions of BDNF-neutralizing antibodies. In summary, current data suggests that conventional antidepressants and ketamine mediate their antidepressant-like effects by increasing BDNF in forebrain regions, in particular the hippocampus, making BDNF an essential determinant of antidepressant efficacy.
Collapse
Affiliation(s)
- Carl Björkholm
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lisa M Monteggia
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
30
|
Cummings J, Friedman JH, Garibaldi G, Jones M, Macfadden W, Marsh L, Robert PH. Apathy in Neurodegenerative Diseases: Recommendations on the Design of Clinical Trials. J Geriatr Psychiatry Neurol 2015; 28:159-73. [PMID: 25809634 DOI: 10.1177/0891988715573534] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 10/08/2014] [Indexed: 12/18/2022]
Abstract
Apathy is a common feature of neurodegenerative disorders but is difficult to study in a clinical trial setting due to practical and conceptual barriers. Principal challenges include a paucity of data regarding apathy in these disorders, an absence of established diagnostic criteria, the presence of confounding factors (eg, coexisting depression), use of concomitant medications, and an absence of a gold-standard apathy assessment scale. Based on a literature search and ongoing collaboration among the authors, we present recommendations for the design of future clinical trials of apathy, suggesting Alzheimer disease and Parkinson disease as models with relevance across a wider array of neuropsychiatric disorders. Recommendations address clarification of the targeted study population (apathy diagnosis and severity at baseline), confounding factors (mood/cognition, behavior, and treatment), outcome measures, study duration, use of comparators and considerations around environment, and the role of the caregiver and patient assent. This review contributes to the search for an optimal approach to study treatment of apathy in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Joseph H Friedman
- Department of Neurology, Movement Disorders Program, Butler Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - George Garibaldi
- Clinical Development, Neurosciences, F. Hoffman-La Roche AG, Basel, Switzerland
| | - Martin Jones
- Bridge Medical Consulting Ltd, London, United Kingdom
| | - Wayne Macfadden
- Clinical Development, Neurosciences, F. Hoffman-La Roche AG, Basel, Switzerland
| | - Laura Marsh
- Mental Health Care Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA Menninger Department of Psychiatry, Baylor College of Medicine, Houston, TX, USA Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philippe H Robert
- CoBTeK, Research Memory Center CMRR CHU, University of Sophia Antipolis, Nice, France
| |
Collapse
|
31
|
Weiser M, Garibaldi G. Quantifying motivational deficits and apathy: a review of the literature. Eur Neuropsychopharmacol 2015; 25:1060-81. [PMID: 25863434 DOI: 10.1016/j.euroneuro.2014.08.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/23/2014] [Indexed: 11/28/2022]
Abstract
Varying definitions of apathy in the published literature and a lack of a consensus regarding diagnostic criteria make the identification and quantification of apathy difficult in both clinical trials and clinical practice. The Apathy Evaluation Scale was developed specifically to assess apathy, but variations in the threshold values defined for clinically significant apathy diminish its use as a screening tool in clinical trials, although it has demonstrated sensitivity to changes in treatment in a number of studies. The Neuropsychiatric Inventory contains an Apathy subscale, which has been used to identify clinical trial populations (with a consistent threshold value) and measure changes following treatment. Few of the other assessment tools currently used in patients with neuropsychiatric disorders are specific for apathy or explore it in any depth, most have not been validated in the general population, do not have cut-off points representing clinically significant apathy, and its changes over time and in response to treatment. Further research is required to address these issues in order to facilitate the quantification of apathy and its natural history. Such research should be conducted with the aim of developing new, specific tools for use across neuropsychiatric disorders.
Collapse
Affiliation(s)
- Mark Weiser
- Departments of Psychiatry, Tel Aviv University and Sheba Medical Center, Israel.
| | | |
Collapse
|
32
|
Affiliation(s)
- Sophia Wang
- Department of Psychiatry and Behavior Sciences, Duke University Medical Center, Durham, North Carolina 27710; ,
- Durham Veterans Affairs Medical Center, Durham, North Carolina 27705
| | - Dan G. Blazer
- Department of Psychiatry and Behavior Sciences, Duke University Medical Center, Durham, North Carolina 27710; ,
- Center for the Study of Aging, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
33
|
Demontis F, Falconi M, Canu D, Serra G. Memantine prevents "bipolar-like" behavior induced by chronic treatment with imipramine in rats. Eur J Pharmacol 2015; 752:49-54. [PMID: 25661848 DOI: 10.1016/j.ejphar.2015.01.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 01/11/2015] [Accepted: 01/22/2015] [Indexed: 12/15/2022]
Abstract
A great deal of evidence suggests that virtually all antidepressant treatments induce a dopaminergic behavioral supersensitivity. We have suggested that this effect may play a key role not only in the antidepressant effect of these treatments, but also in their ability to induce a switch from depression to mania. In 2003-4 we found that the sensitization of dopamine receptors induced by imipramine is followed, after imipramine withdrawal, by a desensitization of these receptors associated with a depressive-like behavior assessed in the forced swimming test. The dopamine receptor sensitization can be prevented by MK-801, an NMDA receptor antagonist, but not by currently used mood stabilizers (lithium, carbamazepine, valproate). These observations led us to suggest - and later confirm - with preliminary clinical observations that memantine may have an acute antimanic and a long-lasting mood-stabilizing effect in treatment-resistant bipolar disorder patients. Here we present data showing that memantine prevents not only the dopamine receptor sensitization induced by imipramine, as observed with MK-801, but also the ensuing desensitization and the associated depressive-like behaviorq observed after antidepressant withdrawal.
Collapse
Affiliation(s)
| | - Marcella Falconi
- Dipartimento di Scienze Biomediche, Università di Sassari, Italy
| | - Desirèe Canu
- Dipartimento di Scienze Biomediche, Università di Sassari, Italy
| | - Gino Serra
- Dipartimento di Scienze Biomediche, Università di Sassari, Italy.
| |
Collapse
|
34
|
Hillhouse TM, Porter JH. A brief history of the development of antidepressant drugs: from monoamines to glutamate. Exp Clin Psychopharmacol 2015; 23:1-21. [PMID: 25643025 PMCID: PMC4428540 DOI: 10.1037/a0038550] [Citation(s) in RCA: 319] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Major depressive disorder (MDD) is a chronic, recurring, and debilitating mental illness that is the most common mood disorder in the United States. It has been almost 50 years since the monoamine hypothesis of depression was articulated, and just over 50 years since the first pharmacological treatment for MDD was discovered. Several monoamine-based pharmacological drug classes have been developed and approved for the treatment of MDD; however, remission rates are low (often less than 60%) and there is a delayed onset before remission of depressive symptoms is achieved. As a result of a "proof-of-concept" study in 2000 with the noncompetitive NMDA antagonist ketamine, a number of studies have examined the glutamatergic systems as viable targets for the treatment of MDD. This review will provide a brief history on the development of clinically available antidepressant drugs, and then review the possible role of glutamatergic systems in the pathophysiology of MDD. Specifically, the glutamatergic review will focus on the N-methyl-D-aspartate (NMDA) receptor and the efficacy of drugs that target the NMDA receptor for the treatment of MDD. The noncompetitive NMDA receptor antagonist ketamine, which has consistently produced rapid and sustained antidepressant effects in MDD patients in a number of clinical studies, has shown the most promise as a novel glutamatergic-based treatment for MDD. However, compounds that target other glutamatergic mechanisms, such as GLYX-13 (a glycine-site partial agonist at NMDA receptors) appear promising in early clinical trials. Thus, the clinical findings to date are encouraging and support the continued search for and the development of novel compounds that target glutamatergic mechanisms.
Collapse
Affiliation(s)
- Todd M. Hillhouse
- the Department of Psychology at Virginia Commonwealth University at the time this review was written and is now at the University of Michigan in the Department of Pharmacology
| | - Joseph H. Porter
- the Department of Psychology at Virginia Commonwealth University
| |
Collapse
|
35
|
Antidepressant actions of ketamine: from molecular mechanisms to clinical practice. Curr Opin Neurobiol 2015; 30:139-43. [PMID: 25562451 DOI: 10.1016/j.conb.2014.12.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 12/01/2014] [Accepted: 12/03/2014] [Indexed: 12/11/2022]
Abstract
In the past decade the emergence of glutamate N-methyl-d-aspartate (NMDA) receptor blockers such as ketamine as fast-acting antidepressants fostered a major conceptual advance by demonstrating the possibility of a rapid antidepressant response. This discovery brings unique mechanistic insight into antidepressant action, as there is a substantial amount of basic knowledge on glutamatergic neurotransmission and how blockade of NMDA receptors may elicit plasticity. The combination of this basic knowledge base and the growing clinical findings will facilitate the development of novel fast acting antidepressants.
Collapse
|
36
|
Drewniany E, Han J, Hancock C, Jones RL, Lim J, Nemat Gorgani N, Sperry JK, Yu HJ, Raffa RB. Rapid-onset antidepressant action of ketamine: potential revolution in understanding and future pharmacologic treatment of depression. J Clin Pharm Ther 2014; 40:125-30. [PMID: 25545040 DOI: 10.1111/jcpt.12238] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/09/2014] [Indexed: 12/16/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE The current pharmacotherapeutic treatment of major depressive disorder (MDD) generally takes weeks to be effective. As the molecular action of these drugs is immediate, the mechanistic basis for this lag is unclear. A drug that has a more rapid onset of action would be a major therapeutic advance and also be a useful comparator to provide valuable mechanistic insight into the disorder and its treatment. COMMENT Recent evidence suggests that ketamine produces rapid-onset antidepressant action. Important questions are as follows: is it specific or coincidental to other effects; is there a dose-response relationship; and is the mechanism related to that of current antidepressants. NMDA receptor antagonism is unlikely the explanation for ketamine's antidepressant action. WHAT IS NEW AND CONCLUSION It is not an exaggeration to state that the new findings, if validated, might produce a revolution in understanding and treating depressive disorders.
Collapse
Affiliation(s)
- E Drewniany
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
How does ketamine elicit a rapid antidepressant response? Curr Opin Pharmacol 2014; 20:35-9. [PMID: 25462290 DOI: 10.1016/j.coph.2014.11.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 11/22/2022]
Abstract
A single sub-psychotomimetic dose of ketamine, an ionotropic glutamatergic n-methyl-D-aspartate (NMDA) receptor antagonist, produces a fast-acting antidepressant response in patients suffering from major depressive disorder. Depressed patients report alleviation of core symptoms within 2 h of a single low-dose intravenous infusion of ketamine with effects lasting up to 2 weeks. The rapidity of ketamine action implies that major symptoms of depression can be alleviated without substantial structural plasticity or circuit rewiring. Therefore, the ability of ketamine to exert a rapid effect provides a unique opportunity to elucidate the types of acute synaptic plasticity changes that can be recruited to counter depression symptoms.
Collapse
|
38
|
Niciu MJ, Henter ID, Luckenbaugh DA, Zarate CA, Charney DS. Glutamate receptor antagonists as fast-acting therapeutic alternatives for the treatment of depression: ketamine and other compounds. Annu Rev Pharmacol Toxicol 2014; 54:119-39. [PMID: 24392693 DOI: 10.1146/annurev-pharmtox-011613-135950] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The N-methyl-D-aspartate (NMDA) receptor antagonist ketamine has rapid and potent antidepressant effects in treatment-resistant major depressive disorder and bipolar depression. These effects are in direct contrast to the more modest effects seen after weeks of treatment with classic monoaminergic antidepressants. Numerous open-label and case studies similarly validate ketamine's antidepressant properties. These clinical findings have been reverse-translated into preclinical models in an effort to elucidate ketamine's antidepressant mechanism of action, and three important targets have been identified: mammalian target of rapamycin (mTOR), eukaryotic elongation factor 2 (eEF2), and glycogen synthase kinase-3 (GSK-3). Current clinical and preclinical research is focused on (a) prolonging/maintaining ketamine's antidepressant effects, (b) developing more selective NMDA receptor antagonists free of ketamine's adverse effects, and (c) identifying predictor, mediator/moderator, and treatment response biomarkers of ketamine's antidepressant effects.
Collapse
Affiliation(s)
- Mark J Niciu
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institutes of Health/National Institute of Mental Health, Bethesda, Maryland 20814-9692;
| | | | | | | | | |
Collapse
|
39
|
Omranifard V, Shirzadi E, Samandari S, Afshar H, Maracy MR. Memantine add on to citalopram in elderly patients with depression: A double-blind placebo-controlled study. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2014; 19:525-30. [PMID: 25197294 PMCID: PMC4155707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 11/03/2013] [Accepted: 01/16/2014] [Indexed: 11/13/2022]
Abstract
BACKGROUND Proper management of depression in elderly population would improve the outcome of the disease and reduce its related disability and mortality. Use of memantine with minimal side effects and drug interaction seems reasonable in the elderly but its antidepressant activity is controversial. The aim of the current research is to investigate the effects of add-on memantine during citalopram therapy in elderly patients with depression, in Isfahan. MATERIALS AND METHODS In this double-blind, placebo controlled trial study; elderly patients aged more than 60 years who were recently diagnosed with depression, were enrolled. The selected patients were randomlysplit into two groups, viz. intervention and placebo groups. The intervention was memantine (20 mg daily) or identical placebo plus citalopram for 8 weeks. The severity of depression and quality of life was evaluated using Geriatric Depression Scale (GDS-15), Hamilton Rating Scale for depression (HRSD) and World Health Organization Quality of Life WHOQOL-BREF respectively. The mentioned scores were evaluated at baseline, 4 weeks and 8 weeks, after initiating the trial in two studied groups and compared with each other. RESULTS 28 and 29 patients were studied in the intervention and placebo groups, respectively. Score of GDS-15, HRSD and WHO-QOL-BREF scales at baseline, 4 weeks and 8 weeks, after initiating trial did not change significantly after use of memantine (P > 0.05). There was no significant difference in mean +/- SD of GDS-15, HRSD and WHO-QOL-BREF scales among intervention and placebo groups (P > 0.05). CONCLUSION The outcome of this clinical trial did not support the antidepressant effect of add-on memantine in elderly patients with depression receiving citalopram. It is recommended to design further studies considering the limitations of the current study mentioned herein and the effect of memantine with other anti-depressant agents.
Collapse
Affiliation(s)
- Victoria Omranifard
- Department of Psychiatry, Behavioral Science Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Shirzadi
- Department of Psychiatry, Behavioral Science Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeid Samandari
- Department of Psychiatry, Behavioral Science Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Afshar
- Department of Psychiatry, Psychosomatic Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran,Address for correspondence: Dr. Hamid Afshar, Psychosomatic Research Center, Department of Psychiatry, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran. E-mail:
| | - Mohammad Reza Maracy
- Environment Research Center, Faculty of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
40
|
Mechanisms underlying differential effectiveness of memantine and ketamine in rapid antidepressant responses. Proc Natl Acad Sci U S A 2014; 111:8649-54. [PMID: 24912158 DOI: 10.1073/pnas.1323920111] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Ketamine is an NMDA receptor (NMDAR) antagonist that elicits rapid antidepressant responses in patients with treatment-resistant depression. However, ketamine can also produce psychotomimetic effects that limit its utility as an antidepressant, raising the question of whether the clinically tolerated NMDAR antagonist memantine possesses antidepressant properties. Despite its similar potency to ketamine as an NMDAR antagonist, clinical data suggest that memantine does not exert rapid antidepressant actions for reasons that are poorly understood. In this study, we recapitulate the ketamine and memantine clinical findings in mice, showing that ketamine, but not memantine, has antidepressant-like effects in behavioral models. Using electrophysiology in cultured hippocampal neurons, we show that ketamine and memantine effectively block NMDAR-mediated miniature excitatory postsynaptic currents in the absence of Mg(2+). However, in physiological levels of extracellular Mg(2+), we identified key functional differences between ketamine and memantine in their ability to block NMDAR function at rest. This differential effect of ketamine and memantine extends to intracellular signaling coupled to NMDAR at rest, in that memantine does not inhibit the phosphorylation of eukaryotic elongation factor 2 or augment subsequent expression of BDNF, which are critical determinants of ketamine-mediated antidepressant efficacy. These results demonstrate significant differences between the efficacies of ketamine and memantine on NMDAR-mediated neurotransmission that have impacts on downstream intracellular signaling, which we hypothesize is the trigger for rapid antidepressant responses. These data provide a novel framework on the necessary functional requirements of NMDAR-mediated neurotransmission as a critical determinant necessary to elicit rapid antidepressant responses.
Collapse
|
41
|
Niciu MJ, Ionescu DF, Richards EM, Zarate CA. Glutamate and its receptors in the pathophysiology and treatment of major depressive disorder. J Neural Transm (Vienna) 2013; 121:907-24. [PMID: 24318540 DOI: 10.1007/s00702-013-1130-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/25/2013] [Indexed: 12/13/2022]
Abstract
Monoaminergic neurotransmitter (serotonin, norepinephrine and dopamine) mechanisms of disease dominated the research landscape in the pathophysiology and treatment of major depressive disorder (MDD) for more than 50 years and still dominate available treatment options. However, the sum of all brain neurons that use monoamines as their primary neurotransmitter is <20%. In addition, most patients treated with monoaminergic antidepressants are left with significant residual symptoms and psychosocial disability not to mention side effects, e.g., sexual dysfunction. In the past several decades, there has been greater focus on the major excitatory neurotransmitter in the human brain, glutamate, in the pathophysiology and treatment of MDD. Although several preclinical and human magnetic resonance spectroscopy studies had already implicated glutamatergic abnormalities in the human brain, it was rocketed by the discovery that the N-methyl-D-aspartate receptor antagonist ketamine has rapid and potent antidepressant effects in even the most treatment-resistant MDD patients, including those who failed to respond to electroconvulsive therapy and who have active suicidal ideation. In this review, we will first provide a brief introduction to glutamate and its receptors in the mammalian brain. We will then review the clinical evidence for glutamatergic dysfunction in MDD, the discovery and progress-to-date with ketamine as a rapidly acting antidepressant, and other glutamate receptor modulators (including proprietary medications) for treatment-resistant depression. We will finally conclude by offering potential future directions necessary to realize the enormous therapeutic promise of glutamatergic antidepressants.
Collapse
Affiliation(s)
- Mark J Niciu
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, Department of Health and Human Services, National Institute of Mental Health, National Institutes of Health, 10 Center Drive, Building 10/CRC, Room 7-5545, Bethesda, MD, 20892, USA,
| | | | | | | |
Collapse
|
42
|
Abstract
Late life depression (LLD) frequently presents with cognitive impairment, and growing evidence suggests that these disease processes are "linked" in multiple ways. For some individuals, LLD may be a recurrence of a long-standing depressive illness, while for others it may be the leading symptom of a developing neuropathological disorder. Overall, studies investigating the relationship between treatment of LLD and improvement in cognitive functioning have yielded mixed results. Research suggests that a subset of individuals with LLD and cognitive dysfunction will experience an improvement in cognitive function after antidepressant treatment, though a significant proportion will continue to exhibit cognitive impairment following resolution of their depressive symptoms. From a treatment standpoint, it is critical to ensure that an individual's depressive symptoms have been treated to remission, measured by a standardized rating scale such as the Geriatric Depression Scale (GDS). SSRI or SNRI monotherapy is often effective, and may be enhanced by employing an evidence-based psychotherapy such as Problem Solving Therapy (PST) or Interpersonal Therapy (IPT), modified to accommodate cognitive impairments that may be present. With respect to specific treatment of cognitive dysfunction, cognitive augmentation or training strategies can be helpful for some patients, and may be explored in combination with treatment of the primary depressive episode. While the introduction of a cholinesterase inhibitor (e.g. donepezil) may be considered, the potential benefit (modest improvement in cognition and functioning) must be weighed against an increased risk for worsening or recurrent depression. Finally, lifestyle factors-such as aerobic exercise, follow-up with a primary care physician for management of co-morbid medical illnesses, and regular participation in stimulating activities (such as through a senior center)-are important and should be included as part of the overall treatment plan.
Collapse
Affiliation(s)
- Aaron M Koenig
- Department of Psychiatry, University of Pittsburgh School of Medicine (Pittsburgh, PA, USA)
| | - Meryl A Butters
- Department of Psychiatry, University of Pittsburgh School of Medicine (Pittsburgh, PA, USA)
| |
Collapse
|
43
|
Pałucha-Poniewiera A, Wierońska JM, Brański P, Burnat G, Chruścicka B, Pilc A. Is the mGlu5 receptor a possible target for new antidepressant drugs? Pharmacol Rep 2013; 65:1506-11. [DOI: 10.1016/s1734-1140(13)71511-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 09/23/2013] [Indexed: 01/23/2023]
|
44
|
Zurkovsky L, Taylor WD, Newhouse PA. Cognition as a therapeutic target in late-life depression: potential for nicotinic therapeutics. Biochem Pharmacol 2013; 86:1133-44. [PMID: 23933385 PMCID: PMC3856552 DOI: 10.1016/j.bcp.2013.07.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 12/19/2022]
Abstract
Depression is associated with impairments to cognition and brain function at any age, but such impairments in the elderly are particularly problematic because of the additional burden of normal cognitive aging and in some cases, structural brain pathology. Individuals with late-life depression exhibit impairments in cognition and brain structural integrity, alongside mood dysfunction. Antidepressant treatment improves symptoms in some but not all patients, and those who benefit may not return to the cognitive and functional level of nondepressed elderly. Thus, for comprehensive treatment of late-life depression, it may be necessary to address both the affective and cognitive deficits. In this review, we propose a model for the treatment of late-life depression in which nicotinic stimulation is used to improve cognitive performance and improve the efficacy of an antidepressant treatment of the syndrome of late-life depression. The cholinergic system is well-established as important to cognition. Although muscarinic stimulation may exacerbate depressive symptoms, nicotinic stimulation may improve cognition and neural functioning without a detriment to mood. While some studies of nicotinic subtype specific receptor agonists have shown promise in improving cognitive performance, less is known regarding how nicotinic receptor stimulation affects cognition in depressed elderly patients. Late-life depression thus represents a new therapeutic target for the development of nicotinic agonist drugs. Parallel treatment of cognitive dysfunction along with medical and psychological approaches to treating mood dysfunction may be necessary to ensure full resolution of depressive illness in aging.
Collapse
Affiliation(s)
- Lilia Zurkovsky
- Center for Cognitive Medicine, Department of Psychiatry, Vanderbilt University School of Medicine, 1601 23rd Avenue South, Nashville, TN 37212, United States
| | | | | |
Collapse
|
45
|
Smith EG, Deligiannidis KM, Ulbricht CM, Landolin CS, Patel JK, Rothschild AJ. Antidepressant augmentation using the N-methyl-D-aspartate antagonist memantine: a randomized, double-blind, placebo-controlled trial. J Clin Psychiatry 2013; 74:966-73. [PMID: 24229746 PMCID: PMC4000742 DOI: 10.4088/jcp.12m08252] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 03/25/2013] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Intravenous N-methyl-d-aspartate (NMDA) antagonists have shown promising results in rapidly ameliorating depression symptoms, but placebo-controlled trials of oral NMDA antagonists as monotherapy have not observed efficacy. We conducted a randomized, double-blind, placebo-controlled trial of the NMDA antagonist memantine as an augmentation treatment for patients with DSM-IV major depressive disorder. METHOD Adult outpatients with major depressive disorder and partial response or nonresponse to their current antidepressant (as indicated by a 17-item Hamilton Depression Rating Scale score of ≥ 16 at baseline) were randomized (from July 2006-December 2011) to add memantine (flexible dose 5-20 mg/d, with all memantine group participants reaching the dose of 20 mg/d) (n = 15) or placebo (n = 16) to their existing treatment for 8 weeks. The primary outcome, change in Montgomery-Asberg Depression Rating Score (MADRS), was evaluated with repeated-measures mixed effects models using last-observation-carried-forward methods. Secondary outcomes included other depression and anxiety rating scales, suicidal and delusional ideation, and other adverse effects. RESULTS 84% of participants completed the trial, including 93% of participants receiving memantine. Participants receiving memantine did not show a statistically or clinically significant change in MADRS scores compared to placebo, either over the entire study (β = 0.133, favoring placebo, P = .74) or at study completion (week 8 mean [SD] MADRS score change = -7.13 [6.61] [memantine]; -7.25 [11.14] [placebo]; P = .97). A minimal to small effect size (comparing change to baseline variability) favoring placebo was observed (Cohen d = 0.19). Similarly, no substantial effect sizes favoring memantine nor statistically significant between-group differences were observed on secondary efficacy outcomes. CONCLUSIONS This trial did not detect significant statistical or effect size differences between memantine and placebo augmentation among nonresponders or poor responders to conventional antidepressants. While the small number of participants is a limitation, this study suggests memantine lacks substantial efficacy as an augmentation treatment for major depressive disorder. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT00344682.
Collapse
|
46
|
Affiliation(s)
- Daniel C. Mathews
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, and Department of Health and Human Services, Bethesda, Maryland
| | - Carlos A. Zarate
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, and Department of Health and Human Services, Bethesda, Maryland
| |
Collapse
|
47
|
Mathews DC, Henter ID, Zarate CA. Targeting the glutamatergic system to treat major depressive disorder: rationale and progress to date. Drugs 2012; 72:1313-33. [PMID: 22731961 DOI: 10.2165/11633130-000000000-00000] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a severe, debilitating medical illness that affects millions of individuals worldwide. The young age of onset and chronicity of the disorder has a significant impact on the long-term disability that affected individuals face. Most existing treatments have focused on the 'monoamine hypothesis' for rational design of compounds. However, patients continue to experience low remission rates, residual subsyndromal symptoms, relapses and overall functional impairment. In this context, growing evidence suggests that the glutamatergic system is uniquely central to the neurobiology and treatment of MDD. Here, we review data supporting the involvement of the glutamatergic system in the pathophysiology of MDD, and discuss the efficacy of glutamatergic agents as novel therapeutics. Preliminary clinical evidence has been promising, particularly with regard to the N-methyl-D-aspartate (NMDA) antagonist ketamine as a 'proof-of-concept' agent. The review also highlights potential molecular and inflammatory mechanisms that may contribute to the rapid antidepressant response seen with ketamine. Because existing pharmacological treatments for MDD are often insufficient for many patients, the next generation of treatments needs to be more effective, rapid acting and better tolerated than currently available medications. There is extant evidence that the glutamatergic system holds considerable promise for developing the next generation of novel and mechanistically distinct agents for the treatment of MDD.
Collapse
Affiliation(s)
- Daniel C Mathews
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
48
|
Mathews DC, Henter ID, Zarate CA. Targeting the glutamatergic system to treat major depressive disorder: rationale and progress to date. Drugs 2012. [PMID: 22731961 DOI: 10.2165/11633130‐000000000‐00000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Major depressive disorder (MDD) is a severe, debilitating medical illness that affects millions of individuals worldwide. The young age of onset and chronicity of the disorder has a significant impact on the long-term disability that affected individuals face. Most existing treatments have focused on the 'monoamine hypothesis' for rational design of compounds. However, patients continue to experience low remission rates, residual subsyndromal symptoms, relapses and overall functional impairment. In this context, growing evidence suggests that the glutamatergic system is uniquely central to the neurobiology and treatment of MDD. Here, we review data supporting the involvement of the glutamatergic system in the pathophysiology of MDD, and discuss the efficacy of glutamatergic agents as novel therapeutics. Preliminary clinical evidence has been promising, particularly with regard to the N-methyl-D-aspartate (NMDA) antagonist ketamine as a 'proof-of-concept' agent. The review also highlights potential molecular and inflammatory mechanisms that may contribute to the rapid antidepressant response seen with ketamine. Because existing pharmacological treatments for MDD are often insufficient for many patients, the next generation of treatments needs to be more effective, rapid acting and better tolerated than currently available medications. There is extant evidence that the glutamatergic system holds considerable promise for developing the next generation of novel and mechanistically distinct agents for the treatment of MDD.
Collapse
Affiliation(s)
- Daniel C Mathews
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|