1
|
Page CE, Epperson CN, Novick AM, Duffy KA, Thompson SM. Beyond the serotonin deficit hypothesis: communicating a neuroplasticity framework of major depressive disorder. Mol Psychiatry 2024:10.1038/s41380-024-02625-2. [PMID: 38816586 DOI: 10.1038/s41380-024-02625-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
The serotonin deficit hypothesis explanation for major depressive disorder (MDD) has persisted among clinicians and the general public alike despite insufficient supporting evidence. To combat rising mental health crises and eroding public trust in science and medicine, researchers and clinicians must be able to communicate to patients and the public an updated framework of MDD: one that is (1) accessible to a general audience, (2) accurately integrates current evidence about the efficacy of conventional serotonergic antidepressants with broader and deeper understandings of pathophysiology and treatment, and (3) capable of accommodating new evidence. In this article, we summarize a framework for the pathophysiology and treatment of MDD that is informed by clinical and preclinical research in psychiatry and neuroscience. First, we discuss how MDD can be understood as inflexibility in cognitive and emotional brain circuits that involves a persistent negativity bias. Second, we discuss how effective treatments for MDD enhance mechanisms of neuroplasticity-including via serotonergic interventions-to restore synaptic, network, and behavioral function in ways that facilitate adaptive cognitive and emotional processing. These treatments include typical monoaminergic antidepressants, novel antidepressants like ketamine and psychedelics, and psychotherapy and neuromodulation techniques. At the end of the article, we discuss this framework from the perspective of effective science communication and provide useful language and metaphors for researchers, clinicians, and other professionals discussing MDD with a general or patient audience.
Collapse
Affiliation(s)
- Chloe E Page
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - C Neill Epperson
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Family Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Helen and Arthur E. Johnson Depression Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew M Novick
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Korrina A Duffy
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Scott M Thompson
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
2
|
Bajaj S, Mahesh R. Converged avenues: depression and Alzheimer's disease- shared pathophysiology and novel therapeutics. Mol Biol Rep 2024; 51:225. [PMID: 38281208 DOI: 10.1007/s11033-023-09170-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024]
Abstract
Depression, a highly prevalent disorder affecting over 280 million people worldwide, is comorbid with many neurological disorders, particularly Alzheimer's disease (AD). Depression and AD share overlapping pathophysiology, and the search for accountable biological substrates made it an essential and intriguing field of research. The paper outlines the neurobiological pathways coinciding with depression and AD, including neurotrophin signalling, the hypothalamic-pituitary-adrenal axis (HPA), cellular apoptosis, neuroinflammation, and other aetiological factors. Understanding overlapping pathways is crucial in identifying common pathophysiological substrates that can be targeted for effective management of disease state. Antidepressants, particularly monoaminergic drugs (first-line therapy), are shown to have modest or no clinical benefits. Regardless of the ineffectiveness of conventional antidepressants, these drugs remain the mainstay for treating depressive symptoms in AD. To overcome the ineffectiveness of traditional pharmacological agents in treating comorbid conditions, a novel therapeutic class has been discussed in the paper. This includes neurotransmitter modulators, glutamatergic system modulators, mitochondrial modulators, antioxidant agents, HPA axis targeted therapy, inflammatory system targeted therapy, neurogenesis targeted therapy, repurposed anti-diabetic agents, and others. The primary clinical challenge is the development of therapeutic agents and the effective diagnosis of the comorbid condition for which no specific diagnosable scale is present. Hence, introducing Artificial Intelligence (AI) into the healthcare system is revolutionary. AI implemented with interdisciplinary strategies (neuroimaging, EEG, molecular biomarkers) bound to have accurate clinical interpretation of symptoms. Moreover, AI has the potential to forecast neurodegenerative and psychiatric illness much in advance before visible/observable clinical symptoms get precipitated.
Collapse
Affiliation(s)
- Shivanshu Bajaj
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani, 333031, Rajasthan, India
| | - Radhakrishnan Mahesh
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani, 333031, Rajasthan, India.
| |
Collapse
|
3
|
Terreros G, Pérez MÁ, Muñoz-LLancao P, D’Espessailles A, Martínez EA, Dagnino-Subiabre A. The Neuroprotective Role of Quinoa ( Chenopodium quinoa, Wild) Supplementation in Hippocampal Morphology and Memory of Adolescent Stressed Rats. Nutrients 2024; 16:381. [PMID: 38337665 PMCID: PMC10857380 DOI: 10.3390/nu16030381] [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: 11/10/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 02/12/2024] Open
Abstract
Brain physiology and morphology are vulnerable to chronic stress, impacting cognitive performance and behavior. However, functional compounds found in food may alleviate these alterations. White quinoa (Chenopodium quinoa, Wild) seeds contain a high content of n-3 fatty acids, including alpha-linolenic acid. This study aimed to evaluate the potential neuroprotective role of a quinoa-based functional food (QFF) in rats. Prepubertal male Sprague-Dawley rats were fed with rat chow or QFF (50% rat chow + 50% dehydrated quinoa seeds) and exposed or not to restraint stress protocol (2 h/day; 15 days). Four experimental groups were used: Non-stressed (rat chow), Non-stressed + QFF, Stressed (rat chow) and Stressed + QFF. Weight gain, locomotor activity (open field), anxiety (elevated plus maze, light-dark box), spatial memory (Y-maze), and dendritic length in the hippocampus were measured in all animals. QFF intake did not influence anxiety-like behaviors, while the memory of stressed rats fed with QFF improved compared to those fed with rat chow. Additionally, QFF intake mitigated the stress-induced dendritic atrophy in pyramidal neurons located in the CA3 area of the hippocampus. The results suggest that a quinoa-supplemented diet could play a protective role in the memory of chronically stressed rats.
Collapse
Affiliation(s)
- Gonzalo Terreros
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 8370993, Chile; (G.T.); (A.D.)
- Auditory and Cognition Center (AUCO), Santiago 8320000, Chile
| | - Miguel Ángel Pérez
- Health Sciences School, Universidad Viña del Mar, Viña del Mar 2580022, Chile;
| | - Pablo Muñoz-LLancao
- Department of Cell Biology, School of Medicine, Yale University, New Haven, CT 06510, USA;
| | - Amanda D’Espessailles
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 8370993, Chile; (G.T.); (A.D.)
| | | | - Alexies Dagnino-Subiabre
- Auditory and Cognition Center (AUCO), Santiago 8320000, Chile
- Laboratory of Stress Neurobiology, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso 2360102, Chile
| |
Collapse
|
4
|
Radley JJ, Herman JP. Preclinical Models of Chronic Stress: Adaptation or Pathology? Biol Psychiatry 2023; 94:194-202. [PMID: 36631383 PMCID: PMC10166771 DOI: 10.1016/j.biopsych.2022.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/15/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022]
Abstract
The experience of prolonged stress changes how individuals interact with their environment and process interoceptive cues, with the end goal of optimizing survival and well-being in the face of a now-hostile world. The chronic stress response includes numerous changes consistent with limiting further damage to the organism, including development of passive or active behavioral strategies and metabolic adjustments to alter energy mobilization. These changes are consistent with symptoms of pathology in humans, and as a result, chronic stress has been used as a translational model for diseases such as depression. While it is of heuristic value to understand symptoms of pathology, we argue that the chronic stress response represents a defense mechanism that is, at its core, adaptive in nature. Transition to pathology occurs only after the adaptive capacity of an organism is exhausted. We offer this perspective as a means of framing interpretations of chronic stress studies in animal models and how these data relate to adaptation as opposed to pathology.
Collapse
Affiliation(s)
- Jason J Radley
- Department of Psychological and Brain Sciences, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa
| | - James P Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio; Cincinnati Veterans Administration Medical Center, Cincinnati, Ohio.
| |
Collapse
|
5
|
Morais FA, Lemos IS, Matiola RT, Freitas MLS, Alano CG, Cabral J, Wessler LB, Generoso JS, Scaini G, Réus GZ, Streck EL. Coadministration of tianeptine alters behavioral parameters and levels of neurotrophins in a chronic model of Maple Syrup Urine disease. Metab Brain Dis 2022; 37:1585-1596. [PMID: 35394251 DOI: 10.1007/s11011-022-00969-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/21/2022] [Indexed: 10/18/2022]
Abstract
Maple Syrup Urine Disease (MSUD) is caused by the deficiency in the activity of the branched-chain α-ketoacid dehydrogenase complex (BCKDC), resulting in the accumulation of the branched-chain amino acids (BCAA) leucine, isoleucine, and valine, and their respective branched-chain α-keto acids. Patients with MSUD are at high risk of developing chronic neuropsychiatric disorders; however, the pathophysiology of brain damage in these patients remains unclear. We hypothesize that MSUD can cause depressive symptoms in patients. To test our hypothesis, Wistar rats were submitted to the BCAA and tianeptine (antidepressant) administration for 21 days, starting seven days postnatal. Depression-like symptoms were assessed by testing for anhedonia and forced swimming after treatments. After the last test, the brain structures were dissected for the evaluation of neutrophins. We demonstrate that chronic BCAA administration induced depressive-like behavior, increased BDNF levels, and decreased NGF levels, suggesting a relationship between BCAA toxicity and brain damage, as observed in patients with MSUD. However, the administration of tianeptine was effective in preventing behavioral changes and restoring neurotrophins levels.
Collapse
Affiliation(s)
- Fábio A Morais
- Laboratório de Doenças Neurometabólicas, Laboratório de Neurologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Isabela S Lemos
- Laboratório de Doenças Neurometabólicas, Laboratório de Neurologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Rafaela T Matiola
- Laboratório de Doenças Neurometabólicas, Laboratório de Neurologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Maria Luísa S Freitas
- Laboratório de Doenças Neurometabólicas, Laboratório de Neurologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Carolina G Alano
- Laboratório de Doenças Neurometabólicas, Laboratório de Neurologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Julia Cabral
- Laboratório de Doenças Neurometabólicas, Laboratório de Neurologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Leticia B Wessler
- Laboratório de Doenças Neurometabólicas, Laboratório de Neurologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Jaqueline S Generoso
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Giselli Scaini
- Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Gislaine Z Réus
- Laboratório de Psiquiatria Translacional, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil
| | - Emilio L Streck
- Laboratório de Doenças Neurometabólicas, Laboratório de Neurologia Experimental, Programa de Pós-Graduação Em Ciências da Saúde, Universidade Do Extremo Sul Catarinense, Criciúma, SC, Brasil.
| |
Collapse
|
6
|
Impact of stress on inhibitory neuronal circuits, our tribute to Bruce McEwen. Neurobiol Stress 2022; 19:100460. [PMID: 35734023 PMCID: PMC9207718 DOI: 10.1016/j.ynstr.2022.100460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/22/2022] [Accepted: 05/10/2022] [Indexed: 12/03/2022] Open
Abstract
This manuscript is dedicated to the memory of Bruce S. McEwen, to commemorate the impact he had on how we understand stress and neuronal plasticity, and the profound influence he exerted on our scientific careers. The focus of this review is the impact of stressors on inhibitory circuits, particularly those of the limbic system, but we also consider other regions affected by these adverse experiences. We revise the effects of acute and chronic stress during different stages of development and lifespan, taking into account the influence of the sex of the animals. We review first the influence of stress on the physiology of inhibitory neurons and on the expression of molecules related directly to GABAergic neurotransmission, and then focus on specific interneuron subpopulations, particularly on parvalbumin and somatostatin expressing cells. Then we analyze the effects of stress on molecules and structures related to the plasticity of inhibitory neurons: the polysialylated form of the neural cell adhesion molecule and perineuronal nets. Finally, we review the potential of antidepressants or environmental manipulations to revert the effects of stress on inhibitory circuits.
Collapse
|
7
|
Erythropoietin attenuates locomotor and cognitive impairments in male rats subjected to physical and psychological stress. IBRO Neurosci Rep 2022; 12:303-308. [PMID: 35519433 PMCID: PMC9062441 DOI: 10.1016/j.ibneur.2022.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/17/2022] [Accepted: 04/17/2022] [Indexed: 11/01/2022] Open
Abstract
Physical and cognitive problems associated with stress are believed to result from stress-related damage to neurons involved in motor and cognitive control. In general, there are two types of stress, physical and psychological which both negatively impact neuronal function. Erythropoietin (EPO) has been shown to exert a neuroprotective effect in various models of physical brain injury; however, its actions on stress-related changes in behavior are unknown. The aim of the current study was to determine whether EPO ameliorated stress-induced locomotor and cognitive impairments, and to compare the effects of EPO on behavioral changes induced by the two different types of stressors. In this study, male Wistar rats were randomly divided into five groups and placed under physical or psychological stress for 10 consecutive days while erythropoietin was injected intraperitoneally (i.p.) every other day (500 U/kg/i.p.) 30 min before stress induction. Exploratory, anxiety-related behaviors, learning and memory were assessed by using open field, plus maze and Morris Water Maze (MWM) tests respectively. Our data showed physical and psychological stress induced dysfunction in locomotion, reduced explorative skills, heightened anxiety-like behavior and reduced memory, which could be partly reversed by EPO. We conclude that EPO reduces adverse effects of both psychological and physical stress, putatively through protection of locomotor and cognitive-controlling neurons vulnerable to the damaging effects of stress. However, future studies need to elucidate the neural mechanisms of the protective effects of EPO. Anxiety like behavior and spatial memory impaired in stress-exposed rats. Physical and Psychological stress had the same impact on behavioral function EPO could improve memory retrieval and lessen anxiety-like behaviors
Collapse
|
8
|
Pekarskaya EA, Holt ES, Gingrich JA, Ansorge MS, Javitch JA, Canetta SE. Tianeptine, but not fluoxetine, decreases avoidant behavior in a mouse model of early developmental exposure to fluoxetine. Sci Rep 2021; 11:22852. [PMID: 34819526 PMCID: PMC8613176 DOI: 10.1038/s41598-021-02074-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/14/2021] [Indexed: 01/12/2023] Open
Abstract
Depression and anxiety, two of the most common mental health disorders, share common symptoms and treatments. Most pharmacological agents available to treat these disorders target monoamine systems. Currently, finding the most effective treatment for an individual is a process of trial and error. To better understand how disease etiology may predict treatment response, we studied mice exposed developmentally to the selective serotonin reuptake inhibitor (SSRI) fluoxetine (FLX). These mice show the murine equivalent of anxiety- and depression-like symptoms in adulthood and here we report that these mice are also behaviorally resistant to the antidepressant-like effects of adult SSRI administration. We investigated whether tianeptine (TIA), which exerts its therapeutic effects through agonism of the mu-opioid receptor instead of targeting monoaminergic systems, would be more effective in this model. We found that C57BL/6J pups exposed to FLX from postnatal day 2 to 11 (PNFLX, the mouse equivalent in terms of brain development to the human third trimester) showed increased avoidant behaviors as adults that failed to improve, or were even exacerbated, by chronic SSRI treatment. By contrast, avoidant behaviors in these same mice were drastically improved following chronic treatment with TIA. Overall, this demonstrates that TIA may be a promising alternative treatment for patients that fail to respond to typical antidepressants, especially in patients whose serotonergic system has been altered by in utero exposure to SSRIs.
Collapse
Affiliation(s)
- Elizabeth A Pekarskaya
- Department of Neuroscience, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA
| | - Emma S Holt
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA
| | - Jay A Gingrich
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA
- Sackler Institute for Developmental Psychobiology, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA
| | - Mark S Ansorge
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA
- Sackler Institute for Developmental Psychobiology, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA
| | - Jonathan A Javitch
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA.
- Department of Molecular Pharmacology and Therapeutics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Sarah E Canetta
- Division of Molecular Therapeutics, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA.
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA.
- Sackler Institute for Developmental Psychobiology, Columbia University Vagelos College of Physicians and Surgeons and the New York State Psychiatric Institute, New York, NY, USA.
| |
Collapse
|
9
|
Rădulescu I, Drăgoi AM, Trifu SC, Cristea MB. Neuroplasticity and depression: Rewiring the brain's networks through pharmacological therapy (Review). Exp Ther Med 2021; 22:1131. [PMID: 34504581 PMCID: PMC8383338 DOI: 10.3892/etm.2021.10565] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
In modern society, depression is one of the most common mental illness; however, its pathophysiology is not yet fully understood. A great body of evidence suggests that depression causes changes in neuroplasticity in specific regions of the brain which are correlated to symptom severity, negative emotional rumination as well as fear learning. Depression is correlated with atrophy of neurons in the cortical and limbic brain regions that control mood and emotion. Antidepressant therapy can exhibit effects on neuroplasticity and reverse the neuroanatomical changes found in depressed patients. The investigation of fast-acting agents that reverse behavioral and neuronal deficiencies of chronic depression, especially the glutamate receptor antagonist NMDA ketamine, and the cellular mechanisms underlying the rapid antidepressant actions of ketamine and related agents are of real interest in current research. Actual medication such as serotonin (5-HT) selective reuptake inhibitor (SSRI) antidepressants, require weeks to months of administration before a clear therapeutic response. The current review aimed to underline the negative effects of depression on neuroplasticity and present the current findings on the effects of antidepressant medication.
Collapse
Affiliation(s)
- Ioana Rădulescu
- Faculty of Psychology and Educational Sciences, University of Bucharest, 050663 Bucharest, Romania
| | - Ana Miruna Drăgoi
- Department of Psychiatry, 'Prof. Dr. Alex. Obregia' Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| | - Simona Corina Trifu
- Department of Clinical Neurosciences, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihai Bogdan Cristea
- Department of Morphological Sciences, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
10
|
Macro- and Microscale Stress-Associated Alterations in Brain Structure: Translational Link With Depression. Biol Psychiatry 2021; 90:118-127. [PMID: 34001371 DOI: 10.1016/j.biopsych.2021.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023]
Abstract
Major depressive disorder (MDD) is a stress-related disorder associated with many cytoarchitectural and neurochemical changes. However, the majority of these changes cannot be reliably detected in the living brain. The examination of animal stress models and postmortem human brain tissue has significantly contributed to our understanding of the pathophysiology of MDD. Ronald Duman's work in humans and in rodent models was critical to the investigation of the contribution of synaptic deficits to MDD and chronic stress pathology, their role in the development and expression of depressive-like behavior, and reversal by novel drugs. Here, we review evidence from magnetic resonance imaging in humans and animals that suggests that corticolimbic alterations are associated with depression symptomatology. We also discuss evidence of cytoarchitectural alterations affecting neurons, astroglia, and synapses in MDD and highlight how similar changes are described in rodent chronic stress models and are linked to the emotion-related behavioral deficits. Finally, we report on the latest approaches developed to measure the synaptic and astroglial alterations in vivo, using positron emission tomography, and how it can inform on the contribution of MDD-associated cytoarchitectural alterations to the symptomatology and the treatment of stress-related disorders.
Collapse
|
11
|
Czéh B, Simon M. Benefits of animal models to understand the pathophysiology of depressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110049. [PMID: 32735913 DOI: 10.1016/j.pnpbp.2020.110049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022]
Abstract
Major depressive disorder (MDD) is a potentially life-threatening mental disorder imposing severe social and economic burden worldwide. Despite the existence of effective antidepressant treatment strategies the exact pathophysiology of the disease is still unknown. Large number of animal models of MDD have been developed over the years, but all of them suffer from significant shortcomings. Despite their limitations these models have been extensively used in academic research and drug development. The aim of this review is to highlight the benefits of animal models of MDD. We focus here on recent experimental data where animal models were used to examine current theories of this complex disease. We argue, that despite their evident imperfections, these models provide invaluable help to understand cellular and molecular mechanisms contributing to the development of MDD. Furthermore, animal models are utilized in research to find clinically useful biomarkers. We discuss recent neuroimaging and microRNA studies since these investigations yielded promising candidates for biomarkers. Finally, we briefly summarize recent progresses in drug development, i.e. the FDA approval of two novel antidepressant drugs: S-ketamine and brexanolone (allopregnanolone). Deeper understanding of the exact molecular and cellular mechanisms of action responsible for the antidepressant efficacy of these rapid acting drugs could aid us to design further compounds with similar effectiveness, but less side effects. Animal studies are likely to provide valuable help in this endeavor.
Collapse
Affiliation(s)
- Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary; Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary.
| | - Maria Simon
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary; Department of Psychiatry and Psychotherapy, Medical School, University of Pécs, Hungary
| |
Collapse
|
12
|
Huzian O, Baka J, Csakvari E, Dobos N, Leranth C, Siklos L, Duman RS, Farkas T, Hajszan T. Stress Resilience is Associated with Hippocampal Synaptoprotection in the Female Rat Learned Helplessness Paradigm. Neuroscience 2021; 459:85-103. [PMID: 33524494 DOI: 10.1016/j.neuroscience.2021.01.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 01/11/2021] [Accepted: 01/21/2021] [Indexed: 12/28/2022]
Abstract
The synaptogenic hypothesis of major depressive disorder implies that preventing the onset of depressive-like behavior also prevents the loss of hippocampal spine synapses. By applying the psychoactive drugs, diazepam and fluoxetine, we investigated whether blocking the development of helpless behavior by promoting stress resilience in the rat learned helplessness paradigm is associated with a synaptoprotective action in the hippocampus. Adult ovariectomized and intact female Sprague-Dawley rats (n = 297) were treated with either diazepam, fluoxetine, or vehicle, exposed to inescapable footshocks or sham stress, and tested in an active escape task to assess helpless behavior. Escape-evoked corticosterone secretion, as well as remodeling of hippocampal spine synapses at a timepoint representing the onset of escape testing were also analyzed. In ovariectomized females, treatment with diazepam prior to stress exposure prevented helpless behavior, blocked the loss of hippocampal spine synapses, and muted the corticosterone surge evoked by escape testing. Although fluoxetine stimulated escape performance and hippocampal synaptogenesis under non-stressed conditions, almost all responses to fluoxetine were abolished following exposure to inescapable stress. Only a much higher dose of fluoxetine was capable of partly reproducing the strong protective actions of diazepam. Importantly, these protective actions were retained in the presence of ovarian hormones. Our findings indicate that stress resilience is associated with the preservation of spine synapses in the hippocampus, raising the possibility that, besides synaptogenesis, hippocampal synaptoprotection is also implicated in antidepressant therapy.
Collapse
Affiliation(s)
- Orsolya Huzian
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Judith Baka
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Eszter Csakvari
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Nikoletta Dobos
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Csaba Leranth
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - Laszlo Siklos
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, United States; Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, United States
| | - Tamas Farkas
- Department of Physiology, Anatomy, and Neuroscience, University of Szeged Faculty of Science and Informatics, Kozep Fasor 52, 6726 Szeged, Hungary
| | - Tibor Hajszan
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States.
| |
Collapse
|
13
|
Tai HH, Cha J, Vedaei F, Dunlop BW, Craighead WE, Mayberg HS, Choi KS. Treatment-Specific Hippocampal Subfield Volume Changes With Antidepressant Medication or Cognitive-Behavior Therapy in Treatment-Naive Depression. Front Psychiatry 2021; 12:718539. [PMID: 35002790 PMCID: PMC8739262 DOI: 10.3389/fpsyt.2021.718539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/08/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Hippocampal atrophy has been consistently reported in major depressive disorder with more recent focus on subfields. However, literature on hippocampal volume changes after antidepressant treatment has been limited. The first-line treatments for depression include antidepressant medication (ADM) or cognitive-behavior therapy (CBT). To understand the differential effects of CBT and ADM on the hippocampus, we investigated the volume alterations of hippocampal subfields with treatment, outcome, and chronicity in treatment-naïve depression patients. Methods: Treatment-naïve depressed patients from the PReDICT study were included in this analysis. A total of 172 patients who completed 12 weeks of randomized treatment with CBT (n = 45) or ADM (n = 127) were included for hippocampal subfield volume analysis. Forty healthy controls were also included for the baseline comparison. Freesurfer 6.0 was used to segment 26 hippocampal substructures and bilateral whole hippocampus from baseline and week 12 structural MRI scans. A generalized linear model with covariates of age and gender was used for group statistical tests. A linear mixed model for the repeated measures with covariates of age and gender was used to examine volumetric changes over time and the contributing effects of treatment type, outcome, and illness chronicity. Results: Of the 172 patients, 85 achieved remission (63/127 ADM, 22/45 CBT). MDD patients showed smaller baseline volumes than healthy controls in CA1, CA3, CA4, parasubiculum, GC-ML-DG, Hippocampal Amygdala Transition Area (HATA), and fimbria. Over 12 weeks of treatment, further declines in the volumes of CA1, fimbria, subiculum, and HATA were observed regardless of treatment type or outcome. CBT remitters, but not ADM remitters, showed volume reduction in the right hippocampal tail. Unlike ADM remitters, ADM non-responders had a decline in volume in the bilateral hippocampal tails. Baseline volume of left presubiculum (regardless of treatment type) and right fimbria and HATA in CBT patients were correlated with a continuous measure of clinical improvement. Chronicity of depression had no effect on any measures of hippocampal subfield volumes. Conclusion: Two first-line antidepressant treatments, CBT and ADM, have different effects on hippocampal tail after 12 weeks. This finding suggests that remission achieved via ADM may protect against progressive hippocampal atrophy by altering neuronal plasticity or supporting neurogenesis. Studies with multimodal neuroimaging, including functional and structural analysis, are needed to assess further the impact of two different antidepressant treatments on hippocampal subfields.
Collapse
Affiliation(s)
- Hua-Hsin Tai
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jungho Cha
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Faezeh Vedaei
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - W Edward Craighead
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Helen S Mayberg
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ki Sueng Choi
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
14
|
Understanding stress: Insights from rodent models. CURRENT RESEARCH IN NEUROBIOLOGY 2021; 2:100013. [PMID: 36246514 PMCID: PMC9559100 DOI: 10.1016/j.crneur.2021.100013] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 02/01/2023] Open
Abstract
Through incorporating both physical and psychological forms of stressors, a variety of rodent models have provided important insights into the understanding of stress physiology. Rodent models also have provided significant information with regards to the mechanistic basis of the pathophysiology of stress-related disorders such as anxiety disorders, depressive illnesses, cognitive impairment and post-traumatic stress disorder. Additionally, rodent models of stress have served as valuable tools in the area of drug screening and drug development for treatment of stress-induced conditions. Although rodent models do not accurately reproduce the biochemical or physiological parameters of stress response and cannot fully mimic the natural progression of human disorders, yet, animal research has provided answers to many important scientific questions. In this review article, important studies utilizing a variety of stress models are described in terms of their design and apparatus, with specific focus on their capabilities to generate reliable behavioral and biochemical read-out. The review focusses on the utility of rodent models by discussing examples in the literature that offer important mechanistic insights into physiologically relevant questions. The review highlights the utility of rodent models of stress as important tools for advancing the mission of scientific research and inquiry. Stressful life events may lead to the onset of severe psychopathologies in humans. Rodents may model many features of stress exposure in human populations. Induction of stress via pharmacological and psychological manipulations alter rodent behavior. Mechanistic rodent studies reveal key molecular targets critical for new therapeutic targets.
Collapse
|
15
|
Tianeptine Enhances Energy-related Processes in the Hippocampal Non-synaptic Mitochondria in a Rat Model of Depression. Neuroscience 2020; 451:111-125. [DOI: 10.1016/j.neuroscience.2020.09.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022]
|
16
|
Misztal T, Kowalczyk P, Młotkowska P, Marciniak E. The Effect of Allopregnanolone on Enzymatic Activity of the DNA Base Excision Repair Pathway in the Sheep Hippocampus and Amygdala under Natural and Stressful Conditions. Int J Mol Sci 2020; 21:E7762. [PMID: 33092287 PMCID: PMC7589085 DOI: 10.3390/ijms21207762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 01/24/2023] Open
Abstract
The neurosteroid allopregnanolone (AL) has many beneficial functions in the brain. This study tested the hypothesis that AL administered for three days into the third brain ventricle would affect the enzymatic activity of the DNA base excision repair (BER) pathway in the hippocampal CA1 and CA3 fields and the central amygdala in luteal-phase sheep under both natural and stressful conditions. Acute stressful stimuli, including isolation and partial movement restriction, were used on the last day of infusion. The results showed that stressful stimuli increased N-methylpurine DNA glycosylase (MPG), thymine DNA glycosylase (TDG), 8-oxoguanine glycosylase (OGG1), and AP-endonuclease 1 (APE1) mRNA expression, as well as repair activities for 1,N6-ethenoadenine (εA), 3,N4-ethenocytosine (εC), and 8-oxoguanine (8-oxoG) compared to controls. The stimulated events were lower in stressed and AL-treated sheep compared to sheep that were only stressed (except MPG mRNA expression in the CA1 and amygdala, as well as TDG mRNA expression in the CA1). AL alone reduced mRNA expression of all DNA repair enzymes (except TDG in the amygdala) relative to controls and other groups. DNA repair activities varied depending on the tissue-AL alone stimulated the excision of εA in the amygdala, εC in the CA3 and amygdala, and 8-oxoG in all tissues studied compared to controls. However, the excision efficiency of lesioned bases in the AL group was lower than in the stressed and stressed and AL-treated groups, with the exception of εA in the amygdala. In conclusion, the presented modulating effect of AL on the synthesis of BER pathway enzymes and their repair capacity, both under natural and stressful conditions, indicates another functional role of this neurosteroid in brain structures.
Collapse
Affiliation(s)
- Tomasz Misztal
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland; (P.K.); (P.M.); (E.M.)
| | | | | | | |
Collapse
|
17
|
Yoshino K, Oda Y, Kimura M, Kimura H, Nangaku M, Shirayama Y, Iyo M. The alterations of glutamate transporter 1 and glutamine synthetase in the rat brain of a learned helplessness model of depression. Psychopharmacology (Berl) 2020; 237:2547-2553. [PMID: 32445055 DOI: 10.1007/s00213-020-05555-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 05/11/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Although glutamate transmission via astrocytes has been proposed to contribute to the pathophysiology of depression, the precise mechanisms are unknown. Herein, we investigated the levels of glutamate transporter-1 (GLT-1) and glutamine synthetase (GS) of astrocytes in learned helplessness (LH) rats (an animal model of depression) and non-LH rats (an animal model of resilience). METHODS We administered inescapable mild electric shock to rats and then discriminated the LH and non-LH rats by a post-shock test. Almost 55% of the rats acquired LH. We then measured the expressions of GLT-1 and GS in several brain regions of LH and non-LH rats by Western blot analysis. RESULTS The levels of GLT-1 and GS in the CA-1, CA-3, dentate gyrus (DG), medial prefrontal cortex (mPF), and nucleus accumbens (NAc) of the LH group were significantly higher than those of the control group. The GS levels in the amygdala of the LH rats were significantly decreased compared to the controls. There were significant differences in GLT-1 and GS levels between the non-LH and LH rats in the CA-1 and CA-3. CONCLUSIONS These results suggest that the LH rats experienced up-regulations of GLT-1 and GS in the CA-1, CA-3, DG, mPF, and NAc and a down-regulation of GS in the amygdala. It is possible that the effects of the GLT-1 and GS levels on astrocytes in the CA-1 and CA-3 are critical for the differentiation of resilience from vulnerability.
Collapse
Affiliation(s)
- Kouhei Yoshino
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana Chuou-ku, Chiba, Chiba, 260-8670, Japan
| | - Yasunori Oda
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana Chuou-ku, Chiba, Chiba, 260-8670, Japan.
| | - Makoto Kimura
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana Chuou-ku, Chiba, Chiba, 260-8670, Japan
| | - Hiroshi Kimura
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana Chuou-ku, Chiba, Chiba, 260-8670, Japan
| | - Masahito Nangaku
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana Chuou-ku, Chiba, Chiba, 260-8670, Japan
| | - Yukihiko Shirayama
- Department of Psychiatry, Teikyo University Chiba Medical Center, 3426-3 Anesaki, Ichihara, Chiba, 290-0111, Japan
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana Chuou-ku, Chiba, Chiba, 260-8670, Japan
| |
Collapse
|
18
|
Cambiaghi M, Crupi R, Bautista EL, Elsamadisi A, Malik W, Pozdniakova H, Han Z, Buffelli M, Battaglia F. The Effects of 1-Hz rTMS on Emotional Behavior and Dendritic Complexity of Mature and Newly Generated Dentate Gyrus Neurons in Male Mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17114074. [PMID: 32521613 PMCID: PMC7312937 DOI: 10.3390/ijerph17114074] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Low-frequency repetitive transcranial magnetic stimulation (1-Hz rTMS) is a promising noninvasive tool for the treatment of depression. Hippocampal neuronal plasticity is thought to play a pivotal role in the pathophysiology of depressive disorders and the mechanism of action of antidepressant treatments. We investigated the effect of 1-Hz rTMS treatment on hippocampal dentate gyrus structural plasticity and related emotional behaviors modifications. Experimentally, adult male mice received either five days of 1-Hz rTMS or Sham stimulation. After stimulation, the mice underwent a battery of tests for anxiety-like and depression-like behaviors. We also tested the effect of treatment on mature and newly generated granule cell dendritic complexity. Our data showed that 1-Hz rTMS induced structural plasticity in mature granule cells, as evidenced by increased dendritic length and number of intersections. However, the stimulation did not increase the proliferation of the dentate gyrus progenitor cells. On the contrary, the stimulated mice showed increased dendritic complexity of newly generated neurons. Moreover, 1-Hz rTMS resulted in antidepressant-like effects in the tail suspension test, but it did not affect anxiety-like behaviors. Therefore, our results indicate that 1-Hz rTMS modulates dentate gyrus morphological plasticity in mature and newly generated neurons. Furthermore, our data provide some evidence of an association between the antidepressant-like activity of 1-Hz rTMS and structural plasticity in the hippocampus.
Collapse
Affiliation(s)
- Marco Cambiaghi
- Department of Neurosciences, Biomedicine and Movement Sciences-University of Verona, 37134 Verona, Italy; (M.C.); (M.B.)
| | - Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98125 Messina, Italy;
| | - Erick Larios Bautista
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Amir Elsamadisi
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Wasib Malik
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Helen Pozdniakova
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Zhiyong Han
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
| | - Mario Buffelli
- Department of Neurosciences, Biomedicine and Movement Sciences-University of Verona, 37134 Verona, Italy; (M.C.); (M.B.)
| | - Fortunato Battaglia
- Department of Medical Sciences, Neurology and Psychiatry, Hackensack Meridian School of Medicine, Seton Hall University, Nutley, NJ 07110, USA; (E.L.B.); (A.E.); (W.M.); (H.P.); (Z.H.)
- Correspondence: ; Tel.: +97-3761-9605
| |
Collapse
|
19
|
Tyrtyshnaia A, Manzhulo I, Konovalova S, Zagliadkina A. Neuropathic Pain Causes a Decrease in the Dendritic Tree Complexity of Hippocampal CA3 Pyramidal Neurons. Cells Tissues Organs 2020; 208:89-100. [DOI: 10.1159/000506812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 02/26/2020] [Indexed: 11/19/2022] Open
Abstract
The International Pain Association defines neuropathic pain as “an unpleasant sensory and emotional experience associated with actual or potential tissue damage.” Recent studies show that chronic neuropathic pain causes both morphological and functional changes within brain structures. Due to the impact of supraspinal centers on pain signal processing, patients with chronic pain often suffer from depression, anxiety, memory impairment, and learning disabilities. Changes in hippocampal neuronal and glial plasticity can play a substantial role in the development of these symptoms. Given the special role of the CA3 hippocampal area in chronic stress reactions, we suggested that this region may undergo significant morphological changes as a result of persistent pain. Since the CA3 area is involved in the implementation of hippocampus-dependent memory, changes in the neuronal morphology can cause cognitive impairment observed in chronic neuropathic pain. This study aimed to elucidate the structural and plastic changes within the hippocampus associated with dendritic tree atrophy of CA3 pyramidal neurons in mice with chronic sciatic nerve constriction. Behavioral testing revealed impaired working and long-term memory in mice with a chronic constriction injury. Using the Golgi-Cox method, we revealed a decrease in the number of branches and dendritic length of CA3 pyramidal neurons. The dendritic spine number was decreased, predominantly due to a reduction in mushroom spines. An immunohistochemical study showed changes in astro- and microglial activity, which could affect the morphology of neurons both directly and indirectly via the regulation of neurotrophic factor synthesis. Using ELISA, we found a decrease in brain-derived neurotrophic factor production and an increase in neurotrophin-3 production. Morphological and biochemical changes in the CA3 area are accompanied by impaired working and long-term memory of animals. Thus, we can conclude that morphological and biochemical changes within the CA3 hippocampal area may underlie the cognitive impairment in neuropathic pain.
Collapse
|
20
|
Dafsari FS, Jessen F. Depression-an underrecognized target for prevention of dementia in Alzheimer's disease. Transl Psychiatry 2020; 10:160. [PMID: 32433512 PMCID: PMC7239844 DOI: 10.1038/s41398-020-0839-1] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022] Open
Abstract
It is broadly acknowledged that the onset of dementia in Alzheimer's disease (AD) may be modifiable by the management of risk factors. While several recent guidelines and multidomain intervention trials on prevention of cognitive decline address lifestyle factors and risk diseases, such as hypertension and diabetes, a special reference to the established risk factor of depression or depressive symptoms is systematically lacking. In this article we review epidemiological studies and biological mechanisms linking depression with AD and cognitive decline. We also emphasize the effects of antidepressive treatment on AD pathology including the molecular effects of antidepressants on neurogenesis, amyloid burden, tau pathology, and inflammation. We advocate moving depression and depressive symptoms into the focus of prevention of cognitive decline and dementia. We constitute that early treatment of depressive symptoms may impact on the disease course of AD and affect the risk of developing dementia and we propose the need for clinical trials.
Collapse
Affiliation(s)
- Forugh S Dafsari
- Department of Psychiatry and Psychotherapy, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany.
- Max-Planck-Institute for Metabolism Research, Gleueler Str. 50, 50931, Cologne, Germany.
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany
- German Center for Neurodegenerative Disease (DZNE), Sigmund-Freud-Str. 27, 53127, Bonn, Germany
| |
Collapse
|
21
|
Abstract
SummaryThe first effective antidepressants (monoamine oxidase inhibitors and tricyclic antidepressants) relied on their ability to augment serotonin and noradrenaline levels at the synapse. Forty years later, the same biological model led to the supremacy of the serotonergic hypothesis to explain not only the pathophysiology of depressive illness, but also the neuropharmacological basis for obsessive compulsive disorder, phobias, posttraumatic stress disorder, and even generalized anxiety disorder. It could be argued that the blinkered view of depression as a solely serotonergic phenomenon has not only restrained and limited research into other potential systems, but has also slowed down the discovery of putative antidepressant drugs. While some might argue that the hypothalamic-pituitary-adrenal (HPA) axis explains an individual’s sensitivity to depression, there are others who equally claim that the most likely explanations are to be found in the neuropsychopharmacology of the immune system or even through reductions in hippocampal volume. There is a richness of possibilities regarding the mechanisms for antidepressant activity embracing theoretical, pharmacological and clinical data. However, the methods by which putative antidepressants are assessed and their clinical efficacy demonstrated are not always robust. That current clinical comparisons of antidepressants rarely show major differences in efficacy between existing molecules could be taken as an indication that “all drugs are the same” or perhaps, more insightfully, as an indication that the ubiquitous Hamilton depression (HAM-D) rating scales are not sensitive to inter-drug differences, even though pronounced pharmacodynamic differences between molecules are easily demonstrated. Any advances in the development of new antidepressants will have to find not only original compounds but also unique psychometric tests by which the drugs can be assessed in a sensitive, reliable, and valid manner.
Collapse
Affiliation(s)
- I Hindmarch
- HPRU Medical Research Centre, University of Surrey, Egerton Road, Guildford, Surrey, UK
| |
Collapse
|
22
|
Guirado R, Perez-Rando M, Ferragud A, Gutierrez-Castellanos N, Umemori J, Carceller H, Nacher J, Castillo-Gómez E. A Critical Period for Prefrontal Network Configurations Underlying Psychiatric Disorders and Addiction. Front Behav Neurosci 2020; 14:51. [PMID: 32317945 PMCID: PMC7155216 DOI: 10.3389/fnbeh.2020.00051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/19/2020] [Indexed: 12/24/2022] Open
Abstract
The medial prefrontal cortex (mPFC) has been classically defined as the brain region responsible for higher cognitive functions, including the decision-making process. Ample information has been gathered during the last 40 years in an attempt to understand how it works. We now know extensively about the connectivity of this region and its relationship with neuromodulatory ascending projection areas, such as the dorsal raphe nucleus (DRN) or the ventral tegmental area (VTA). Both areas are well-known regulators of the reward-based decision-making process and hence likely to be involved in processes like evidence integration, impulsivity or addiction biology, but also in helping us to predict the valence of our future actions: i.e., what is “good” and what is “bad.” Here we propose a hypothesis of a critical period, during which the inputs of the mPFC compete for target innervation, establishing specific prefrontal network configurations in the adult brain. We discuss how these different prefrontal configurations are linked to brain diseases such as addiction or neuropsychiatric disorders, and especially how drug abuse and other events during early life stages might lead to the formation of more vulnerable prefrontal network configurations. Finally, we show different promising pharmacological approaches that, when combined with the appropriate stimuli, will be able to re-establish these functional prefrontocortical configurations during adulthood.
Collapse
Affiliation(s)
- Ramon Guirado
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Valencia, Spain.,Neuroscience Center, University of Helsinki, Helsinki, Finland.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Dirección General de Universidades, Gobierno de Aragón, Zaragoza, Spain
| | - Marta Perez-Rando
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Antonio Ferragud
- Department of Psychology, Cambridge University, Cambridge, United Kingdom
| | | | - Juzoh Umemori
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Hector Carceller
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Valencia, Spain
| | - Juan Nacher
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de Valencia, Valencia, Spain.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| |
Collapse
|
23
|
Feng M, Crowley NA, Patel A, Guo Y, Bugni SE, Luscher B. Reversal of a Treatment-Resistant, Depression-Related Brain State with the Kv7 Channel Opener Retigabine. Neuroscience 2019; 406:109-125. [PMID: 30858110 DOI: 10.1016/j.neuroscience.2019.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/27/2022]
Abstract
Neuroinflammation is associated with increased vulnerability to diverse psychiatric conditions, including treatment-resistant major depressive disorder (MDD). Here we assessed whether high fat diet (HFD) induced neuroinflammation may be suitable to model a treatment-resistant depressive-like brain state in mice. Male and female mice were fed a HFD for 18 weeks, followed by quantitation of glucose tolerance, inflammatory markers of brain tissue (TNFα, IL-6, IL-1β, Iba-1), neural excitability in the prelimbic cortex (PLC), as well as assessment of emotional reactivity and hedonic behavior in a battery of behavioral tests. In addition, we assessed the behavioral responsiveness of mice to fluoxetine, desipramine, ketamine, and the Kv7 channel opener and anticonvulsant retigabine. HFD exposure led to glucose intolerance and neuroinflammation in male mice, with similar but non-significant trends in females. Neuroinflammation of males was associated with anxious-depressive-like behavior and defects in working memory, along with neural hyperexcitability and increased Ih currents of pyramidal cells in the PLC. The behavioral changes were largely resistant to chronic treatment with fluoxetine and desipramine, as well as ketamine. By contrast, retigabine (also known as ezogabine) normalized neural excitability and Ih currents recorded from slices of HFD-treated animals and significantly ameliorated most of the behavioral impairments, without effects in control diet exposed animals. Thus, treatment resistant depressive-like brain states that are associated with chronic neuroinflammation may involve hyperexcitability of pyramidal neurons and may be effectively treated by retigabine.
Collapse
Affiliation(s)
- Mengyang Feng
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Nicole A Crowley
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Akshilkumar Patel
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Yao Guo
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Sierra E Bugni
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Bernhard Luscher
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802.
| |
Collapse
|
24
|
Indersmitten T, Schachter MJ, Young S, Welty N, Otte S, Nassi JJ, Lovenberg T, Bonaventure P, Wyatt RM. In vivo Calcium Imaging Reveals That Cortisol Treatment Reduces the Number of Place Cells in Thy1-GCaMP6f Transgenic Mice. Front Neurosci 2019; 13:176. [PMID: 30881283 PMCID: PMC6405689 DOI: 10.3389/fnins.2019.00176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/14/2019] [Indexed: 01/16/2023] Open
Abstract
The hippocampus, a structure essential for spatial navigation and memory undergoes anatomical and functional changes during chronic stress. Here, we investigate the effects of chronic stress on the ability of place cells to encode the neural representation of a linear track. To model physiological conditions of chronic stress on hippocampal function, transgenic mice expressing the genetically encoded calcium indicator GCaMP6f in CA1 pyramidal neurons were chronically administered with 40 μg/ml of cortisol for 8 weeks. Cortisol-treated mice exhibited symptoms typically observed during chronic stress, including diminished reward seeking behavior and reduced adrenal gland and spleen weights. In vivo imaging of hippocampal cellular activity during linear track running behavior revealed a reduced number of cells that could be recruited to encode spatial position, despite an unchanged overall number of active cells, in cortisol-treated mice. The properties of the remaining place cells that could be recruited to encode spatial information, however, was unperturbed. Bayesian decoders trained to estimate the mouse’s position on the track using single neuron activity data demonstrated reduced performance in a cue richness-dependent fashion in cortisol-treated animals. The performance of decoders utilizing data from the entire neuronal ensemble was unaffected by cortisol treatment. Finally, to test the hypothesis that an antidepressant drug could prevent the effects of cortisol, we orally administered a group of mice with 10 mg/kg citalopram during cortisol administration. Citalopram prevented the cortisol-induced decrease in single-neuron decoder performance but failed to significantly prevent anhedonia and the cortisol-induced reduction in the proportion place cells. The dysfunction observed at the single-neuron level indicates that chronic stress may impair the ability of the hippocampus to encode individual neural representations of the mouse’s spatial position, a function pivotal to forming an accurate navigational map of the mouse’s external environment; however, the hippocampal ensemble as a whole is resilient to any cortisol-induced insults to single neuronal place cell function on the linear track.
Collapse
Affiliation(s)
- Tim Indersmitten
- Janssen Research & Development, LLC., San Diego, CA, United States
| | | | - Stephanie Young
- Janssen Research & Development, LLC., San Diego, CA, United States
| | - Natalie Welty
- Janssen Research & Development, LLC., San Diego, CA, United States
| | - Stephani Otte
- Janssen Research & Development, LLC., San Diego, CA, United States
| | - Jonathan J Nassi
- Janssen Research & Development, LLC., San Diego, CA, United States
| | | | | | - Ryan M Wyatt
- Janssen Research & Development, LLC., San Diego, CA, United States
| |
Collapse
|
25
|
Trujillo V, Durando PE, Suárez MM. Maternal separation induces long-term changes in mineralocorticoid receptor in rats subjected to chronic stress and treated with tianeptine. Int J Neurosci 2018; 129:540-550. [PMID: 30485752 DOI: 10.1080/00207454.2018.1550398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE The aim of this study was to analyze whether early maternal separation would result in long-term, persistent alterations in stress response in adulthood, altering mineralocorticoid receptor immunoreactivity (MR-ir) in the dorsal hippocampal areas [CA1, CA2, CA3 and dentate gyrus (DG)], paraventricular nucleus of the hypothalamus and medial and central nucleus of the amygdala, key structures involved in stress response regulation. We also analyzed whether chronic treatment with the antidepressant tianeptine reverses these possible changes. MATERIAL AND METHODS Male Wistar rats were subjected to daily maternal separation for 4.5 h during 3 weeks or left undisturbed. As adults, they were exposed to chronic stress during 24 days or left undisturbed, and they were also daily treated with tianeptine (10 mg/kg i.p.) or isotonic solution. RESULTS In the CA2 and DG areas of the dorsal hippocampus, there was an increase in MR-ir in non-maternally separated and chronic stressed groups. Tianeptine raised MR-ir in the CA3. In the DG, control and maternally separated + chronic stress groups treated with tianeptine showed more MR-ir than their respective vehicle groups. In the paraventricular nucleus, tianeptine decreased MR-ir in non-separated groups, but not in maternally separated rats. CONCLUSIONS Our results support findings that early-life events induce long-term changes in stress response regulation, persistent into adulthood, which are manifested during challenges in later life, and that treatment with tianeptine, which tends to attenuate the hypothalamus-pituitary-adrenal axis dysregulation, depends on the individual experience of each rat.
Collapse
Affiliation(s)
- Verónica Trujillo
- a Facultad de Ciencias Exactas, Físicas y Naturales , Universidad Nacional de Córdoba, Laboratorio de Fisiología Animal , Córdoba , Argentina
| | - Patricia Evelina Durando
- a Facultad de Ciencias Exactas, Físicas y Naturales , Universidad Nacional de Córdoba, Laboratorio de Fisiología Animal , Córdoba , Argentina
| | - Marta Magdalena Suárez
- a Facultad de Ciencias Exactas, Físicas y Naturales , Universidad Nacional de Córdoba, Laboratorio de Fisiología Animal , Córdoba , Argentina.,b Facultad de Ciencias Médicas , Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Universidad Nacional de Córdoba Ciudad Universitaria , Córdoba , Argentina
| |
Collapse
|
26
|
Hu X, Zhang L, Hu X, Lu L, Tang S, Li H, Bu X, Gong Q, Huang X. Abnormal Hippocampal Subfields May Be Potential Predictors of Worse Early Response to Antidepressant Treatment in Drug‐Naïve Patients With Major Depressive Disorder. J Magn Reson Imaging 2018; 49:1760-1768. [PMID: 30295348 DOI: 10.1002/jmri.26520] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 09/07/2018] [Accepted: 09/07/2018] [Indexed: 02/05/2023] Open
Affiliation(s)
- Xiaoxiao Hu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Lianqing Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Xinyu Hu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Lu Lu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Shi Tang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Hailong Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Xuan Bu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Xiaoqi Huang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan Province, P.R. China
| |
Collapse
|
27
|
Chen CC, Lu J, Yang R, Ding JB, Zuo Y. Selective activation of parvalbumin interneurons prevents stress-induced synapse loss and perceptual defects. Mol Psychiatry 2018; 23:1614-1625. [PMID: 28761082 PMCID: PMC5794672 DOI: 10.1038/mp.2017.159] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/16/2017] [Accepted: 06/20/2017] [Indexed: 01/10/2023]
Abstract
Stress, a prevalent experience in modern society, is a major risk factor for many psychiatric disorders. Although sensorimotor abnormalities are often present in these disorders, little is known about how stress affects the sensory cortex. Combining behavioral analyses with in vivo synaptic imaging, we show that stressful experiences lead to progressive, clustered loss of dendritic spines along the apical dendrites of layer (L) 5 pyramidal neurons (PNs) in the mouse barrel cortex, and such spine loss closely associates with deteriorated performance in a whisker-dependent texture discrimination task. Furthermore, the activity of parvalbumin-expressing inhibitory interneurons (PV+ INs) decreases in the stressed mouse due to reduced excitability of these neurons. Importantly, both behavioral defects and structural changes of L5 PNs are prevented by selective pharmacogenetic activation of PV+INs in the barrel cortex during stress. Finally, stressed mice raised under environmental enrichment (EE) maintain normal activation of PV+ INs, normal texture discrimination, and L5 PN spine dynamics similar to unstressed EE mice. Our findings suggest that the PV+ inhibitory circuit is crucial for normal synaptic dynamics in the mouse barrel cortex and sensory function. Pharmacological, pharmacogenetic and environmental approaches to prevent stress-induced maladaptive behaviors and synaptic malfunctions converge on the regulation of PV+ IN activity, pointing to a potential therapeutic target for stress-related disorders.
Collapse
Affiliation(s)
- Chia-Chien Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Ju Lu
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Renzhi Yang
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Jun B. Ding
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA 94304, USA,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA,Correspondence: Dr. Yi Zuo, Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA, , Phone: +1-831-459-3812, Fax: +1-831-459-3139
| |
Collapse
|
28
|
Rusconi F, Battaglioli E. Acute Stress-Induced Epigenetic Modulations and Their Potential Protective Role Toward Depression. Front Mol Neurosci 2018; 11:184. [PMID: 29904343 PMCID: PMC5990609 DOI: 10.3389/fnmol.2018.00184] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/14/2018] [Indexed: 11/16/2022] Open
Abstract
Psychiatric disorders entail maladaptive processes impairing individuals’ ability to appropriately interface with environment. Among them, depression is characterized by diverse debilitating symptoms including hopelessness and anhedonia, dramatically impacting the propensity to live a social and active life and seriously affecting working capability. Relevantly, besides genetic predisposition, foremost risk factors are stress-related, such as experiencing chronic psychosocial stress—including bullying, mobbing and abuse—, and undergoing economic crisis or chronic illnesses. In the last few years the field of epigenetics promised to understand core mechanisms of gene-environment crosstalk, contributing to get into pathogenic processes of many disorders highly influenced by stressful life conditions. However, still very little is known about mechanisms that tune gene expression to adapt to the external milieu. In this Perspective article, we discuss a set of protective, functionally convergent epigenetic processes induced by acute stress in the rodent hippocampus and devoted to the negative modulation of stress-induced immediate early genes (IEGs) transcription, hindering stress-driven morphostructural modifications of corticolimbic circuitry. We also suggest that chronic stress damaging protective epigenetic mechanisms, could bias the functional trajectory of stress-induced neuronal morphostructural modification from adaptive to maladaptive, contributing to the onset of depression in vulnerable individuals. A better understanding of the epigenetic response to stress will be pivotal to new avenues of therapeutic intervention to treat depression, especially in light of limited efficacy of available antidepressant drugs.
Collapse
Affiliation(s)
- Francesco Rusconi
- Department of Medical Biotechnologies and Translational Medicine, University of Milan Via Fratelli Cervi, Segrate, Italy
| | - Elena Battaglioli
- Department of Medical Biotechnologies and Translational Medicine, University of Milan Via Fratelli Cervi, Segrate, Italy.,CNR Institute of Neuroscience Via Vanvitelli, Milan, Italy
| |
Collapse
|
29
|
Lee H, Kim HK, Kwon JT, Kim YO, Seo J, Lee S, Cho IH, Kim HJ. Effects of Tianeptine on Adult Rats Following Prenatal Stress. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2018; 16:197-208. [PMID: 29739134 PMCID: PMC5953020 DOI: 10.9758/cpn.2018.16.2.197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/01/2017] [Accepted: 05/25/2017] [Indexed: 01/24/2023]
Abstract
Objective Exposing a pregnant female to stress during the critical period of embryonic fetal brain development increases the risk of psychiatric disorders in the offspring. The objective of this study was to investigate the effect of antidepressant tianeptine on prenatally stressed (PNS) rats. Methods In this study, a repeated variable stress paradigm was applied to pregnant rats during the last week of gestation. To investigate the effects of antidepressant tianeptine on PNS rats, behavioral and protein expression analyses were performed. Forced swim test, open field test, and social interaction test were performed to determine changes in PNS rats compared to non-stressed offspring. Haloperidol was used as a positive control as an antipsychotic drug based on previous studies. Results Behavioral changes were restored after treatment with tianeptine or haloperidol. Western blot and immunohistochemical analyses of the prefrontal cortex revealed downregulation of several neurodevelopmental proteins in PNS rats. After treatment with tianeptine or haloperidol, their expression levels were increased. Conclusion Downregulation of several proteins in PNS rats might have caused subsequent behavioral changes in PNS rats. After tianeptine or haloperidol treatment, behavioral changes in PNS rats were restored. Therefore, tianeptine might decrease incidence of prenatal stress related-psychiatric disorders such as depression and schizophrenia.
Collapse
Affiliation(s)
- Hwayoung Lee
- Department of Clinical Pharmacology and Soonchunhyang Medical Research Institute, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Hyung-Ki Kim
- Department of Clinical Pharmacology and Soonchunhyang Medical Research Institute, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Jun-Tack Kwon
- Department of Clinical Pharmacology and Soonchunhyang Medical Research Institute, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Young Ock Kim
- Department of Development of Ginseng and Medical Plants Research Institute, Rural Administration, Eumseong, Korea
| | - Jonghoon Seo
- Department of Clinical Pharmacology and Soonchunhyang Medical Research Institute, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sanghyun Lee
- Department of Integrative Plant Science, Chung-Ang University, Anseong, Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, Brain Korea 21 Plus Program, and Institute of Korean Medicine, College of Oriental Medicine, Kyung Hee University, Seoul, Korea
| | - Hak-Jae Kim
- Department of Clinical Pharmacology and Soonchunhyang Medical Research Institute, Soonchunhyang University College of Medicine, Cheonan, Korea
| |
Collapse
|
30
|
Maynard KR, Hobbs JW, Rajpurohit SK, Martinowich K. Electroconvulsive seizures influence dendritic spine morphology and BDNF expression in a neuroendocrine model of depression. Brain Stimul 2018; 11:856-859. [PMID: 29674117 DOI: 10.1016/j.brs.2018.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/30/2018] [Accepted: 04/03/2018] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Electroconvulsive therapy (ECT) is a rapid and effective treatment for major depressive disorder. Chronic stress-induced depression causes dendrite atrophy and deficiencies in brain-derived neurotrophic factor (BDNF), which are reversed by anti-depressant drugs. Electroconvulsive seizures (ECS), an animal model of ECT, robustly increase BDNF expression and stimulate dendritic outgrowth. OBJECTIVE The present study aims to understand cellular and molecular plasticity mechanisms contributing to the efficacy of ECS following chronic stress-induced depression. METHODS We quantify Bdnf transcript levels and dendritic spine density and morphology on cortical pyramidal neurons in mice exposed to vehicle or corticosterone and receiving either Sham or ECS treatment. RESULTS ECS rescues corticosterone-induced defects in spine morphology and elevates Bdnf exon 1 and exon 4-containing transcripts in cortex. CONCLUSIONS Dendritic spine remodeling and induction of activity-induced BDNF in the cortex represent important cellular and molecular plasticity mechanisms underlying the efficacy of ECS for treatment of chronic stress-induced depression.
Collapse
Affiliation(s)
- Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, United States
| | - John W Hobbs
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, United States
| | - Sumita K Rajpurohit
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, United States
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, United States; Departments of Neuroscience, Psychiatry and Behavioral Sciences, Johns Hopkins Medical School, Baltimore, MD, 21205, United States.
| |
Collapse
|
31
|
Ortiz JB, Conrad CD. The impact from the aftermath of chronic stress on hippocampal structure and function: Is there a recovery? Front Neuroendocrinol 2018; 49:114-123. [PMID: 29428548 DOI: 10.1016/j.yfrne.2018.02.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 12/18/2022]
Abstract
Chronic stress results in functional and structural changes to the brain and especially the hippocampus. Decades of research have provided insights into the mechanisms by which chronic stress impairs hippocampal-mediated cognition and the corresponding reduction of hippocampal CA3 apical dendritic complexity. Yet, when chronic stress ends and time passes, which we refer to as a "post-stress rest period," hippocampal-mediated spatial memory deficits begin to improve and CA3 apical dendritic arbors increase in complexity. The processes by which the hippocampus improves from a chronically stressed state are not simply the reversal of the mechanisms that produced spatial memory deficits and CA3 apical dendritic retraction. This review will discuss our current understanding of how a chronically stressed hippocampus improves after a post-stress rest period. Untangling the mechanisms that allow for this post-stress plasticity is a critical next step in understanding how to promote resilience in the face of stressors.
Collapse
Affiliation(s)
- J Bryce Ortiz
- Department of Psychology, Arizona State University, Box 1104, Tempe, AZ 85287-1104, United States.
| | - Cheryl D Conrad
- Department of Psychology, Arizona State University, Box 1104, Tempe, AZ 85287-1104, United States
| |
Collapse
|
32
|
Cameron HA, Schoenfeld TJ. Behavioral and structural adaptations to stress. Front Neuroendocrinol 2018; 49:106-113. [PMID: 29421158 PMCID: PMC5963997 DOI: 10.1016/j.yfrne.2018.02.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/20/2018] [Accepted: 02/03/2018] [Indexed: 10/18/2022]
Abstract
Unpredictable aversive experiences, or stressors, lead to changes in depression- and anxiety-related behavior and to changes in hippocampal structure including decreases in adult neurogenesis, granule cell and pyramidal cell dendritic morphology, and volume. Here we review the relationship between these behavioral and structural changes and discuss the possibility that these changes may be largely adaptive. Specifically, we suggest that new neurons in the dentate gyrus enhance behavioral adaptability to changes in the environment, biasing behavior in novel situations based on previous experience with stress. Conversely, atrophy-like changes in the hippocampus and decreased adult neurogenesis following chronic stress may serve to limit stress responses and stabilize behavior during chronic stress.
Collapse
Affiliation(s)
- Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
33
|
Nava N, Treccani G, Alabsi A, Kaastrup Mueller H, Elfving B, Popoli M, Wegener G, Nyengaard JR. Temporal Dynamics of Acute Stress-Induced Dendritic Remodeling in Medial Prefrontal Cortex and the Protective Effect of Desipramine. Cereb Cortex 2018; 27:694-705. [PMID: 26523035 DOI: 10.1093/cercor/bhv254] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Stressful events are associated with increased risk of mood disorders. Volumetric reductions have been reported in brain areas critical for the stress response, such as medial prefrontal cortex (mPFC), and dendritic remodeling has been proposed as an underlying factor. Here, we investigated the time-dependent effects of acute stress on dendritic remodeling within the prelimbic (PL) region of the PFC, and whether treatment with the antidepressant desipramine (DMI) may interfere. Rodents were subjected to foot-shock stress: dendritic length and spine density were analyzed 1 day, 7 days, and 14 days after stress. Acute stress produced increased spine density and decreased cofilin phosphorylation at 1 day, paralleled with dendritic retraction. An overall shift in spine population was observed at 1 day, resulting in a stress-induced increase in small spines. Significant atrophy of apical dendrites was observed at 1 day, which was prevented by chronic DMI, and at 14 days after stress exposure. Chronic DMI resulted in dendritic elaboration at 7 days but did not prevent the effects of FS-stress. Collectively, these data demonstrate that 1) acute stressors may induce rapid and sustained changes of PL neurons; and 2) chronic DMI may protect neurons from rapid stress-induced synaptic changes.
Collapse
Affiliation(s)
- Nicoletta Nava
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, Aarhus C 8000, Denmark.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark
| | - Giulia Treccani
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark.,Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Milan 20133, Italy
| | - Abdelrahman Alabsi
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging,Aarhus University Hospital, Aarhus C 8000, Denmark
| | - Heidi Kaastrup Mueller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Milan 20133, Italy
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov 8240, Denmark.,Pharmaceutical Research Center of Excellence, School of Pharmacy, North-West University, Potchefstroom, South Africa
| | - Jens Randel Nyengaard
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging,Aarhus University Hospital, Aarhus C 8000, Denmark
| |
Collapse
|
34
|
Rubio-Casillas A, Fernández-Guasti A. The dose makes the poison: from glutamate-mediated neurogenesis to neuronal atrophy and depression. Rev Neurosci 2018; 27:599-622. [PMID: 27096778 DOI: 10.1515/revneuro-2015-0066] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/04/2016] [Indexed: 12/21/2022]
Abstract
Experimental evidence has demonstrated that glutamate is an essential factor for neurogenesis, whereas another line of research postulates that excessive glutamatergic neurotransmission is associated with the pathogenesis of depression. The present review shows that such paradox can be explained within the framework of hormesis, defined as biphasic dose responses. Low glutamate levels activate adaptive stress responses that include proteins that protect neurons against more severe stress. Conversely, abnormally high levels of glutamate, resulting from increased release and/or decreased removal, cause neuronal atrophy and depression. The dysregulation of the glutamatergic transmission in depression could be underlined by several factors including a decreased inhibition (γ-aminobutyric acid or serotonin) or an increased excitation (primarily within the glutamatergic system). Experimental evidence shows that the activation of N-methyl-D-aspartate receptor (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPAR) can exert two opposite effects on neurogenesis and neuron survival depending on the synaptic or extrasynaptic concentration. Chronic stress, which usually underlies experimental and clinical depression, enhances glutamate release. This overactivates NMDA receptors (NMDAR) and consequently impairs AMPAR activity. Various studies show that treatment with antidepressants decreases plasma glutamate levels in depressed individuals and regulates glutamate receptors by reducing NMDAR function by decreasing the expression of its subunits and by potentiating AMPAR-mediated transmission. Additionally, it has been shown that chronic treatment with antidepressants having divergent mechanisms of action (including tricyclics, selective serotonin reuptake inhibitors, and ketamine) markedly reduced depolarization-evoked glutamate release in the hippocampus. These data, taken together, suggest that the glutamatergic system could be a final common pathway for antidepressant treatments.
Collapse
|
35
|
The recent progress in animal models of depression. Prog Neuropsychopharmacol Biol Psychiatry 2017; 77:99-109. [PMID: 28396255 PMCID: PMC5605906 DOI: 10.1016/j.pnpbp.2017.04.008] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/28/2017] [Accepted: 04/07/2017] [Indexed: 12/11/2022]
Abstract
Major depression disorder (MDD) is a debilitating mental illness with significant morbidity and mortality. Despite the growing number of studies that have emerged, the precise underlying mechanisms of MDD remain unknown. When studying MDD, tissue samples like peripheral blood or post-mortem brain samples are used to elucidate underlying mechanisms. Unfortunately, there are many uncontrollable factors with such samples such as medication history, age, time after death before post-mortem tissue was collected, age, sex, race, and living conditions. Although these factors are critical, they introduce confounding variables that can influence the outcome profoundly. In this regard, animal models provide a crucial approach to examine neural circuitry and molecular and cellular pathways in a controlled environment. Further, manipulations with pharmacological agents and gene editing are accepted methods of studying depression in animal models, which is impossible to employ in human patient studies. Here, we have reviewed the most widely used animal models of depression and delineated the salient features of each model in terms of behavioral and neurobiological outcomes. We have also illustrated the current challenges in using these models and have suggested strategies to delineate the underlying mechanism associated with vulnerability or resilience to developing depression.
Collapse
|
36
|
Hare BD, Ghosal S, Duman RS. Rapid Acting Antidepressants in Chronic Stress Models: Molecular and Cellular Mechanisms. CHRONIC STRESS 2017. [PMID: 28649673 PMCID: PMC5482287 DOI: 10.1177/2470547017697317] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Stress-associated disorders, including depression and anxiety, impact nearly 20% of individuals in the United States. The social, health, and economic burden imposed by stress-associated disorders requires in depth research efforts to identify suitable treatment strategies. Traditional medications (e.g., selective serotonin reuptake inhibitors, monoamine oxidase inhibitors) have significant limitations, notably a time lag for therapeutic response that is compounded by low rates of efficacy. Excitement over ketamine, a rapid acting antidepressant effective in treatment resistant patients, is tempered by transient dissociative and psychotomimetic effects, as well as abuse potential. Rodent stress models are commonly used to produce behavioral abnormalities that resemble those observed in stress-associated disorders. Stress models also produce molecular and cellular morphological changes in stress sensitive brain regions, including the prefrontal cortex and hippocampus that resemble alterations observed in depression. Rapid acting antidepressants such as ketamine can rescue stress-associated morphological and behavioral changes in rodent models. Here, we review the literature supporting a role for rapid acting antidepressants in opposing the effects of stress, and summarize research efforts seeking to elucidate the molecular, cellular, and circuit level targets of these agents.
Collapse
Affiliation(s)
- Brendan D Hare
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Sriparna Ghosal
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ronald S Duman
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
37
|
Cussotto S, Cryan JF, O'Leary OF. The hippocampus and dorsal raphe nucleus are key brain areas associated with the antidepressant effects of lithium augmentation of desipramine. Neurosci Lett 2017; 648:14-20. [PMID: 28351776 DOI: 10.1016/j.neulet.2017.03.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/09/2017] [Accepted: 03/23/2017] [Indexed: 11/25/2022]
Abstract
Approximately 50% of depressed individuals fail to achieve remission with first-line antidepressant drugs and a third remain treatment-resistant. When first-line antidepressant treatment is unsuccessful, second-line strategies include dose optimisation, switching to another antidepressant, combination with another antidepressant, or augmentation with a non-antidepressant medication. Much of the evidence for the efficacy of augmentation strategies comes from studies using lithium to augment the effects of tricyclic antidepressants. The neural circuitry underlying the therapeutic effects of lithium augmentation is not yet fully understood. Recently, we reported that chronic treatment with a combination of lithium and the antidepressant desipramine, exerted antidepressant-like behavioural effects in a mouse strain (BALB/cOLaHsd) that did not exhibit an antidepressant-like behavioural response to either drug alone. In the present study, we used this model in combination with ΔFosB/FosB immunohistochemistry to identify brain regions chronically affected by lithium augmentation of desipramine when compared to either treatment alone. The data suggest that the dorsal raphe nucleus and the CA3 regions of the dorsal hippocampus are key nodes in the neural circuitry underlying antidepressant action of lithium augmentation of desipramine. These data give new insight into the neurobiology underlying the mechanism of lithium augmentation in the context of treatment-resistant depression.
Collapse
Affiliation(s)
- Sofia Cussotto
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
38
|
Gao Y, Ma J, Tang J, Liang X, Huang CX, Wang SR, Chen LM, Wang FF, Tan CX, Chao FL, Zhang L, Qiu X, Luo YM, Xiao Q, Du L, Xiao Q, Tang Y. White matter atrophy and myelinated fiber disruption in a rat model of depression. J Comp Neurol 2017; 525:1922-1933. [PMID: 28118485 DOI: 10.1002/cne.24178] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 01/13/2017] [Accepted: 01/16/2017] [Indexed: 01/17/2023]
Abstract
Brain imaging and postmortem studies have indicated that white matter abnormalities may contribute to the pathology and pathogenesis of depression. However, until now, no study has quantitatively investigated white matter changes in depression in rats. The current study used the chronic unpredictable stress (CUS) model of depression. Body weight and sucrose preference test (SPT) scores were assessed weekly. Upon successfully establishing the CUS animal model, all animals were tested using the SPT and the open field test (OFT). Then, transmission electron microscopy and unbiased stereological methods were used to investigate white matter changes in the rats. Compared with the control group, the body weight and sucrose preference of the CUS rats were significantly decreased (p < .001, p < .001, respectively). In the OFT, the total time spent and the total distance traveled in the inner area by the CUS rats were significantly lower than those of the control group (p = .002, p = .001, respectively). The stereological results revealed that white matter volume, the total volume, and the total length and mean diameter of myelinated fibers in the white matter of the CUS rats were significantly decreased compared to the control rats (p = .042, p = .038, p = .035, p = .019, respectively). The results of this study suggested that white matter atrophy and disruption of myelinated fibers in the white matter may contribute to the pathophysiology underlying depression, which might provide new targets for the development of novel therapeutic interventions for depression.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Geriatrics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P. R. China.,Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Jing Ma
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Jing Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Xin Liang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Chun-Xia Huang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China.,Department of Physiology, Chongqing Medical University, Chongqing, P. R. China
| | - San-Rong Wang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Lin-Mu Chen
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Fei-Fei Wang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Chuan-Xue Tan
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Feng-Lei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Xuan Qiu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yan-Min Luo
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Qian Xiao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Lian Du
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China.,Department of Psychiatry, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Qian Xiao
- Department of Geriatrics, The First Affiliated Hospital, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P. R. China.,Laboratory of Stem Cells and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
39
|
The effects of desipramine, fluoxetine, or tianeptine on changes in bulbar BDNF levels induced by chronic social instability stress and inflammation. Pharmacol Rep 2017; 69:520-525. [PMID: 31994095 DOI: 10.1016/j.pharep.2017.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/09/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUND Stress is a major predisposing factor in the development of psychiatric disorders and potential source of augmented inflammatory processes in the brain. Increasing body of evidence shows an important role of alterations in the olfactory bulbs (OBs) function in stress-related disorders. The aim of the present study was to investigate the impact of antidepressants on the alterations of brain-derived neurotrophic factor (BDNF) induced by lipopolysaccharide (LPS) in female rats subjected to chronic social instability stress (CSIS). METHODS 9 weeks old female rats were subjected to CSIS and injected ip once daily with desipramine (10 mg/kg), fluoxetine (5 mg/kg), or tianeptine (10 mg/kg) for 4 weeks. On the last day of the experiment, rats being at the estrus phase of cycle were injected ip with LPS (1 mg/kg) or saline. RESULTS The BDNF mRNA and protein levels were evaluated in the olfactory bulbs. and the BDNF protein levels were measured in plasma. A single LPS administration in the stressed rats resulted in significant decrease in the bulbar BDNF mRNA, but not in the protein level. Chronic administration of desipramine, fluoxetine, or tianeptine increased the BDNF mRNA expression and protein levels in the LPS-injected stressed rats. There was no effect of the studied antidepressants on the reduction of the plasma BDNF protein level induced by CSIS and LPS. CONCLUSIONS These results suggest that studied antidepressants were effective in inhibiting the impact of LPS on BDNF expression in the stressed rats what may be significant for beneficial action of this drugs.
Collapse
|
40
|
Effect of amitriptyline treatment on neurofilament-H protein in an experimental model of depression. Brain Res Bull 2017; 128:1-6. [DOI: 10.1016/j.brainresbull.2016.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/19/2016] [Accepted: 11/01/2016] [Indexed: 02/04/2023]
|
41
|
Shirayama Y, Takahashi M, Osone F, Hara A, Okubo T. Myo-inositol, Glutamate, and Glutamine in the Prefrontal Cortex, Hippocampus, and Amygdala in Major Depression. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2016; 2:196-204. [PMID: 29560915 DOI: 10.1016/j.bpsc.2016.11.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 11/13/2016] [Accepted: 11/28/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND The brains of patients with depression exhibit many changes in various regions. Recently, proton magnetic resonance spectroscopy has been used to measure brain metabolites, using saturation bands to shape the volume of interest. Our a priori hypothesis was that myo-inositol and glutamate were downregulated in the hippocampus and amygdala in depression. METHODS We measured brain metabolites from the medial prefrontal cortex, hippocampus, and amygdala of 22 drug-naïve, first-episode patients with major depressive disorder and 27 healthy control subjects using 3T proton magnetic resonance spectroscopy. RESULTS Compared with healthy control subjects, patients showed statistically significant reductions in myo-inositol levels in all three regions and reductions in glutamate levels in the medial prefrontal cortex. Furthermore, we found significant decreases in the ratios of glutamate to creatine plus phosphocreatine in the medial prefrontal cortex and amygdala. Additionally, the ratios of glutamine to creatine plus phosphocreatine were also decreased in all three regions examined, although not all the participants presented reliable data. Finally, glutamate levels in the medial prefrontal cortex and amygdala have significant correlations with executive function and those in the hippocampus with memory function. Hippocampal myo-inositol was significantly related to blood cortisol. CONCLUSIONS Our findings indicated abnormal myo-inositol, glutamate, and glutamine levels in the brains of major depressive disorder patients.
Collapse
Affiliation(s)
- Yukihiko Shirayama
- Department of Psychiatry, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan.
| | - Michio Takahashi
- Department of Psychiatry, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan
| | - Fumio Osone
- Department of Radiology, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan
| | - Akira Hara
- Department of Radiology, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan
| | - Toshiyuki Okubo
- Department of Radiology, Teikyo University Chiba Medical Center, Ichihara, Chiba, Japan
| |
Collapse
|
42
|
Pollano A, Zalosnik MI, Durando PE, Suárez MM. Differential effects of tianeptine on the dorsal hippocampal volume of rats submitted to maternal separation followed by chronic unpredictable stress in adulthood. Stress 2016; 19:599-608. [PMID: 27604299 DOI: 10.1080/10253890.2016.1224842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Early maternal separation (MS) may produce lasting effects in the dorsal hippocampus (DH) that can change its response to chronic stress in adulthood. Chronic stress affects DH morphology and function, but tianeptine (an anti-depressant) can reverse the stress-induced morphological impairments. Morphologic alterations of hippocampus can affect contextual memory. Therefore, we evaluated the effect of tianeptine in MS and chronically stressed rats on: 1) volume of the DH and its areas using stereology and 2) hippocampal-dependent memory using a fear conditioning test. Male Wistar rats were subjected to daily MS for 4.5 h between postnatal days (PND) 1-21, or to animal facility rearing (AFR). Between (PND) days 50 and 74, rats were exposed to chronic unpredictable stress and were treated daily with tianeptine (10 mg/kg) or vehicle, providing eight groups: AFR-unstressed/vehicle (n = 5 for stereology, n = 18 for fear conditioning test); AFR unstressed/tianeptine (n = 6 and n = 10); AFR-chronic stress/vehicle (n = 6 and n = 14); AFR-chronic stress/tianeptine (n = 6 and n = 10), MS-unstressed/vehicle (n = 5 and n = 19), MS-unstressed/tianeptine (n = 6 and n = 10), MS-chronic stress/vehicle (n = 6 and n = 18), and MS-chronic stress/tianeptine (n = 6 and n = 10). MS-chronic stress/tianeptine rats showed a diminished CA1 area than the corresponding MS-unstressed/tianeptine rats. The combination of stressors produced a freezing response similar to those of the control group during postconditioning. During retrieval, MS led to a diminished freezing response compared to the AFR-unstressed groups. Tianeptine had no effect on freezing behavior. Our results show that tianeptine can affect the CA1 area volume differently depending on the nature and quantity of stressors but cannot alter freezing to context.
Collapse
Affiliation(s)
- Antonella Pollano
- a Laboratorio de Fisiología Animal, Facultad de Ciencias Exactas, Físicas y Naturales , Universidad Nacional de Córdoba , Córdoba , Argentina
| | - María I Zalosnik
- a Laboratorio de Fisiología Animal, Facultad de Ciencias Exactas, Físicas y Naturales , Universidad Nacional de Córdoba , Córdoba , Argentina
| | - Patricia E Durando
- a Laboratorio de Fisiología Animal, Facultad de Ciencias Exactas, Físicas y Naturales , Universidad Nacional de Córdoba , Córdoba , Argentina
| | - Marta M Suárez
- a Laboratorio de Fisiología Animal, Facultad de Ciencias Exactas, Físicas y Naturales , Universidad Nacional de Córdoba , Córdoba , Argentina
| |
Collapse
|
43
|
Zhang D, Wang X, Lu XY. Adiponectin Exerts Neurotrophic Effects on Dendritic Arborization, Spinogenesis, and Neurogenesis of the Dentate Gyrus of Male Mice. Endocrinology 2016; 157:2853-69. [PMID: 27187175 PMCID: PMC4929553 DOI: 10.1210/en.2015-2078] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The hippocampus, a brain region critical for learning, memory and emotional processing, maintains its capacity to undergo structural plasticity throughout life. Hippocampal structural plasticity can be modulated by a number of intrinsic and extrinsic factors. This study investigated the effects of adiponectin, an adipocyte-derived hormone, on dendritic growth, arborization, and spinogenesis in mature granule neurons of the hippocampal dentate gyrus generated during embryonic (early-born) or early postnatal (late-born) stages. We found that adiponectin deficiency reduced dendritic length, branching and spine density of granule neurons. The reduction was more evident in early-born granule neurons than in late-born granule neurons. Intracerebroventricular infusion of adiponectin for 1 week increased of dendritic spines and arbor complexity in late-born granule neurons. Moreover, adiponectin deficiency decreased the production of adult-born new granule neurons through suppressing neural progenitor cell proliferation and differentiation, whereas intracerebroventricular adiponectin infusion increased the proliferation of neural progenitor cells in adult dentate gyrus. These results suggest that adiponectin plays an important role in dendritic spine remodeling and neurogenesis in the dentate gyrus.
Collapse
Affiliation(s)
- Di Zhang
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Xuezhen Wang
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| | - Xin-Yun Lu
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, Texas 78229
| |
Collapse
|
44
|
Seo MK, McIntyre RS, Cho HY, Lee CH, Park SW, Mansur RB, Kim GM, Baek JH, Woo YS, Lee JG, Kim YH. Tianeptine induces mTORC1 activation in rat hippocampal neurons under toxic conditions. Psychopharmacology (Berl) 2016; 233:2617-27. [PMID: 27129862 DOI: 10.1007/s00213-016-4309-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/22/2016] [Indexed: 12/23/2022]
Abstract
RATIONALE Recent studies have demonstrated that mTORC1 activation may be related to antidepressant action. However, the relationship between mTORC1 signaling activation and currently prescribed antidepressants remains unclear. OBJECTIVE The aim of the present study was to determine whether alterations in mTORC1 signaling are observable following treatment with tianeptine under toxic conditions induced by B27 deprivation. Additionally, we investigated whether this drug affects synaptic proteins, neurite outgrowth, and spine density via mTORC1 signaling. METHODS Using Western blotting, we measured the phosphorylation levels of mTORC1, 4E-BP-1, p70S6K, Akt, and ERK in rat primary hippocampal neurons. Changes in BDNF, dendritic outgrowth, spine density, and synaptic proteins (PSD-95, synaptophysin, and GluR1) were measured. RESULTS Tianeptine significantly increased the phosphorylation of mTORC1, 4E-BP-1, p70S6K, Akt, and ERK. The increase in mTOR phosphorylation was blocked by the PI3K, MEK, and mTORC1 inhibitors. Tianeptine increased BDNF, dendritic outgrowth, spine density, and synaptic proteins; all of these effects were blocked by the mTORC1 inhibitor. CONCLUSIONS In this study, we demonstrated that tianeptine activates the mTORC1 signaling pathway and increases dendritic outgrowth, spine density, and synaptic proteins through mTORC1 signaling under toxic conditions in rat primary hippocampal neurons.
Collapse
Affiliation(s)
- Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Hye Yeon Cho
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Chan Hong Lee
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
- Department of Health Science and Technology, Graduate School, Inje University, Busan, Republic of Korea
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Gyung-Mee Kim
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea
| | - Jun Hyung Baek
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea
| | - Young Sup Woo
- Department of Psychiatry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea.
- Mood Disorders Psychopharmacology Unit, University Health Network, University of Toronto, Toronto, ON, Canada.
- Department of Health Science and Technology, Graduate School, Inje University, Busan, Republic of Korea.
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea.
- Department of Psychiatry, Inje University Haeundae Paik Hospital, 1435, Jwa-dong, Haeundae-gu, Busan, 612-030, Republic of Korea.
| | - Young Hoon Kim
- Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea.
- Department of Health Science and Technology, Graduate School, Inje University, Busan, Republic of Korea.
- Department of Psychiatry, School of Medicine, Haeundae Paik Hospital, Inje University, Busan, Republic of Korea.
- Department of Psychiatry, Inje University Haeundae Paik Hospital, 1435, Jwa-dong, Haeundae-gu, Busan, 612-030, Republic of Korea.
| |
Collapse
|
45
|
Pro-apoptotic Action of Corticosterone in Hippocampal Organotypic Cultures. Neurotox Res 2016; 30:225-38. [PMID: 27189478 PMCID: PMC4947107 DOI: 10.1007/s12640-016-9630-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 01/31/2023]
Abstract
Elevated levels of glucocorticoids exert neurotoxic effects, and the hippocampus is particularly sensitive to the effects of glucocorticoids. Because some data have indicated that an increased action of glucocorticoids in the perinatal period enhances the susceptibility of brain tissue to adverse substances later in life, the main purpose of the present study was to compare necrotic/apoptotic corticosterone action in hippocampal organotypic cultures obtained from control animals with the effect of this steroid in tissue from prenatally stressed rats. Because the adverse effects of glucocorticoid action on nerve cell viability appear to result mainly from an increase in the intensity of the effects of glutamate and changes in growth factor and pro-inflammatory cytokine synthesis, the involvement of these factors in corticosterone action were also determined. In stress-like concentration (1 μM), corticosterone, when added to hippocampal cultures for 1 and 3 days, alone or jointly with glutamate, did not induce necrosis. In contrast, in 3-day cultures, corticosterone (1 μM) increased caspase-3 activity and the mRNA expression of the pro-apoptotic Bax. Moreover, corticosterone’s effect on caspase-3 activity was stronger in hippocampal cultures from prenatally stressed compared to control rats. Additionally, 24 h of exposure to corticosterone and glutamate, when applied separately and together, increased Bdnf, Ngf, and Tnf-α expression. In contrast, after 72 h, a strong decrease in the expression of both growth factors was observed, while the expression of TNF-α remained high. The present study showed that in stress-like concentrations, corticosterone exerted pro-apoptotic but not necrotic effects in hippocampal organotypic cultures. Prenatal stress increased the pro-apoptotic effects of corticosterone. Increased synthesis of the pro-inflammatory cytokine TNF-α may be connected with the adverse effects of corticosterone on brain cell viability.
Collapse
|
46
|
Chen F, du Jardin KG, Waller JA, Sanchez C, Nyengaard JR, Wegener G. Vortioxetine promotes early changes in dendritic morphology compared to fluoxetine in rat hippocampus. Eur Neuropsychopharmacol 2016; 26:234-245. [PMID: 26711685 DOI: 10.1016/j.euroneuro.2015.12.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/20/2015] [Accepted: 12/01/2015] [Indexed: 12/28/2022]
Abstract
Preclinical studies reveal that the multimodal antidepressant vortioxetine enhances long-term potentiation and dendritic branching compared to a selective serotonin reuptake inhibitor (SSRI). In the present study, we investigated vortioxetine׳s effects on spines and dendritic morphology in rat hippocampus at two time points compared to the SSRI, fluoxetine. Rats were dosed for 1 and 4 weeks with vortioxetine and fluoxetine at doses relevant for antidepressant activity. Dendritic morphology of pyramidal neurons (i.e., dendritic length, dendritic branch, spine number and density, and Sholl analysis) was examined in Golgi-stained sections from hippocampal CA1. After 1 week of treatment, vortioxetine significantly increased spine number (apical and basal dendrites), spine density (only basal), dendritic length (only apical), and dendritic branch number (apical and basal), whereas fluoxetine had no effect. After 4 weeks of treatment, vortioxetine significantly increased all measures of dendritic spine morphology as did fluoxetine except for spine density of basal dendrites. The number of intersections in the apical and basal dendrites was also significantly increased for both treatments after 4 weeks compared to control. In addition, 4 weeks of vortioxetine treatment, but not fluoxetine, promoted a decrease in spine neck length. In conclusion, 1-week vortioxetine treatment induced changes in spine number and density and dendritic morphology, whereas an equivalent dose of fluoxetine had no effects. Decreased spine neck length following 4-week vortioxetine treatment suggests a transition to mature spine morphology. This implies that vortioxetine׳s effects on spine and dendritic morphology are mediated by mechanisms that go beyond serotonin reuptake inhibition.
Collapse
Affiliation(s)
- Fenghua Chen
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, DK-8000 Aarhus C, Denmark; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, DK-8240 Risskov, Denmark.
| | - Kristian Gaarn du Jardin
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, DK-8240 Risskov, Denmark
| | - Jessica A Waller
- Sourcing and Scientific Excellence at Lundbeck Research USA, Inc., Paramus, NJ 07652-1431, USA
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, DK-8240 Risskov, Denmark; Sourcing and Scientific Excellence at Lundbeck Research USA, Inc., Paramus, NJ 07652-1431, USA
| | - Jens R Nyengaard
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, DK-8240 Risskov, Denmark; Centre for Pharmaceutical Excellence, School of Pharmacy, North-West University, Potchefstroom 2520, South Africa
| |
Collapse
|
47
|
Malykhin N, Coupland N. Hippocampal neuroplasticity in major depressive disorder. Neuroscience 2015; 309:200-13. [DOI: 10.1016/j.neuroscience.2015.04.047] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/17/2015] [Accepted: 04/21/2015] [Indexed: 01/31/2023]
|
48
|
Miskowiak KW, Vinberg M, Macoveanu J, Ehrenreich H, Køster N, Inkster B, Paulson OB, Kessing LV, Skimminge A, Siebner HR. Effects of Erythropoietin on Hippocampal Volume and Memory in Mood Disorders. Biol Psychiatry 2015; 78:270-7. [PMID: 25641635 DOI: 10.1016/j.biopsych.2014.12.013] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Persistent cognitive dysfunction in depression and bipolar disorder (BD) impedes patients' functional recovery. Erythropoietin (EPO) increases neuroplasticity and reduces cognitive difficulties in treatment-resistant depression (TRD) and remitted BD. This magnetic resonance imaging study assessed the neuroanatomical basis for these effects. METHODS Patients with TRD who were moderately depressed or BD in partial remission were randomized to 8 weekly EPO (40,000 IU) or saline infusions in a double-blind, parallel-group design. Patients underwent magnetic resonance imaging, memory assessment with the Rey Auditory Verbal Learning Test, and mood ratings with the Beck Depression Inventory, Hamilton Depression Rating Scale, and Young Mania Rating Scale at baseline and week 14. Hippocampus segmentation and analysis of hippocampal volume, shape, and gray matter density were conducted with FMRIB Software Library tools. Memory change was analyzed with repeated-measures analysis of covariance adjusted for depression symptoms, diagnosis, age, and gender. RESULTS Eighty-four patients were randomized; 1 patient withdrew and data collection was incomplete for 14 patients; data were thus analyzed for 69 patients (EPO: n = 35, saline: n = 34). Compared with saline, EPO was associated with mood-independent memory improvement and reversal of brain matter loss in the left hippocampal cornu ammonis 1 to cornu ammonis 3 and subiculum. Using the entire sample, memory improvement was associated with subfield hippocampal volume increase independent of mood change. CONCLUSIONS EPO-associated memory improvement in TRD and BD may be mediated by reversal of brain matter loss in a subfield of the left hippocampus. EPO may provide a therapeutic option for patients with mood disorders who have impaired neuroplasticity and cognition.
Collapse
Affiliation(s)
- Kamilla W Miskowiak
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Maj Vinberg
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Julian Macoveanu
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Nicolai Køster
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Becky Inkster
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Olaf B Paulson
- Neurobiological Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Lars V Kessing
- Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Arnold Skimminge
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre
| | - Hartwig R Siebner
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital Hvidovre, Hvidovre; Department of Neurology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| |
Collapse
|
49
|
Carrier N, Saland SK, Duclot F, He H, Mercer R, Kabbaj M. The Anxiolytic and Antidepressant-like Effects of Testosterone and Estrogen in Gonadectomized Male Rats. Biol Psychiatry 2015; 78:259-69. [PMID: 25683735 PMCID: PMC4501899 DOI: 10.1016/j.biopsych.2014.12.024] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/03/2014] [Accepted: 12/19/2014] [Indexed: 01/01/2023]
Abstract
BACKGROUND While the influence of testosterone levels on vulnerability to affective disorders is not straightforward, research suggests this hormone may confer some degree of resiliency in men. We recently demonstrated a role for the dentate gyrus in mediating testosterone's protective effects on depressive-like behavior in gonadectomized male rats. Here, testosterone may exert its effects through androgen receptor-mediated mechanisms or via local aromatization to estradiol. METHODS Gonadectomized male rats were implanted with a placebo, testosterone, or estradiol pellet, and subsequent protective anxiolytic- and antidepressant-like effects of testosterone and its aromatized metabolite, estradiol, were then investigated in the open field and sucrose preference tests, respectively. Moreover, their influence on gene expression in the hippocampus was analyzed by genome-wide complementary DNA microarray analysis. Finally, the contribution of testosterone's aromatization within the dentate gyrus was assessed by local infusion of the aromatase inhibitor fadrozole, whose efficacy was confirmed by liquid chromatography-tandem mass spectrometry. RESULTS Both hormones had antidepressant-like effects associated with a substantial overlap in transcriptional regulation, particularly in synaptic plasticity- and mitogen-activated protein kinase pathway-related genes. Further, chronic aromatase inhibition within the dentate gyrus blocked the protective effects of testosterone. CONCLUSIONS Both testosterone and estradiol exhibit anxiolytic- and antidepressant-like effects in gonadectomized male rats, while similarly regulating critical mediators of these behaviors, suggesting common underlying mechanisms. Accordingly, we demonstrated that testosterone's protective effects are mediated, in part, by its aromatization in the dentate gyrus. These findings thus provide further insight into a role for estradiol in mediating the protective anxiolytic- and antidepressant-like effects of testosterone.
Collapse
Affiliation(s)
| | | | | | | | | | - Mohamed Kabbaj
- Department of Biomedical Sciences, Program in Neurosciences.
| |
Collapse
|
50
|
Potential roles for Homer1 and Spinophilin in the preventive effect of electroconvulsive seizures on stress-induced CA3c dendritic retraction in the hippocampus. Eur Neuropsychopharmacol 2015; 25:1324-31. [PMID: 25935093 DOI: 10.1016/j.euroneuro.2015.04.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 03/31/2015] [Accepted: 04/10/2015] [Indexed: 11/21/2022]
Abstract
Electroconvulsive therapy (ECT) remains the treatment of choice for patients with severe or drug-resistant depressive disorders, yet the mechanism behind its efficacy remains poorly characterized. In the present study, we used electroconvulsive seizures (ECS), an animal model of ECT, to identify proteins possibly involved in the preventive effect of ECS on stress-induced neuronal atrophy in the hippocampus. Rats were stressed daily using the 21-day 6h daily restraint stress paradigm and subjected to sham seizures, a single ECS on the last day of the restraint period or daily repeated seizures for 10 consecutive days during the end of the restraint period. Consistent with previous findings, dendritic atrophy was observed in the CA3c hippocampal region of chronically stressed rats. In addition, we confirmed our recent findings of increased spine density in the CA1 region following chronic restraint stress. The morphological alterations in the CA3c area were prevented by treatment with ECS. On the molecular level, we showed that the synaptic proteins Homer1 and Spinophilin are targeted by ECS. Repeated ECS blocked stress-induced up-regulation of Spinophilin protein levels and further increased the stress-induced up-regulation of Homer1. Given the roles of Spinophilin in the regulation of AMPA receptors and Homer1 in the regulation of metabotropic glutamate receptors (mGluRs), our data imply the existence of a mechanism where ECS regulate cell excitability by modulating AMPA receptor function and mGluR related calcium homeostasis. These molecular changes could potentially contribute to the mechanism induced by ECS which prevents the stress-induced morphological changes in the CA3c region.
Collapse
|