1
|
Meng Y, Du J, Liu N, Qiang Y, Xiao L, Lan X, Ma L, Yang J, Yu J, Lu G. Epigenetic modulation: Research progress on histone acetylation levels in major depressive disorders. J Drug Target 2023; 31:142-151. [PMID: 36112185 DOI: 10.1080/1061186x.2022.2125978] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Depression is a serious mental illness and a prevalent condition with multiple aetiologies. The impact of the current therapeutic strategies is limited and the pathogenesis of the illness is not well understood. According to previous studies, depression onset is influenced by a variety of environmental and genetic factors, including chronic stress, aberrant changes in gene expression, and hereditary predisposition. Transcriptional regulation in eukaryotes is closely related to chromosome packing and is controlled by histone post-translational modifications. The development of new antidepressants may proceed along a new path with medications that target epigenetics. Histone deacetylase inhibitors (HDACis) are a class of compounds that interfere with the function of histone deacetylases (HDACs). This review explores the relationship between HDACs and depression and focuses on the current knowledge on their regulatory mechanism in depression and the potential therapeutic use of HDACis with antidepressant efficacy in preclinical research. Future research on inhibitors is also proposed and discussed.
Collapse
Affiliation(s)
- Yuan Meng
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China.,Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Juan Du
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China.,Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Ning Liu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Yuanyuan Qiang
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China
| | - Lifei Xiao
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China
| | - Xiaobing Lan
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Lin Ma
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Jiamei Yang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Jianqiang Yu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, PR China
| | - Guangyuan Lu
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, PR China
| |
Collapse
|
2
|
Mao LM, Mathur N, Wang JQ. Downregulation of surface AMPA receptor expression in the striatum following prolonged social isolation, a role of mGlu5 receptors. IBRO Neurosci Rep 2022; 13:22-30. [PMID: 35711245 PMCID: PMC9193854 DOI: 10.1016/j.ibneur.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/16/2022] [Accepted: 05/28/2022] [Indexed: 11/10/2022] Open
Abstract
Major depressive disorder is a common and serious mood illness. The molecular mechanisms underlying the pathogenesis and symptomatology of depression are poorly understood at present. Multiple neurotransmitter systems are believed to be implicated in depression. Increasing evidence supports glutamatergic transmission as a critical element in depression and antidepressant activity. In this study, we investigated adaptive changes in expression of AMPA receptors in a key limbic reward structure, the striatum, in response to an anhedonic model of depression. Prolonged social isolation in adult rats caused anhedonic/depression- and anxiety-like behavior. In these depressed rats, surface levels of AMPA receptors, mainly GluA1 and GluA3 subunits, were reduced in the nucleus accumbens (NAc). Surface GluA1/A3 expression was also reduced in the caudate putamen (CPu) following chronic social isolation. No change was observed in expression of presynaptic synaptophysin, postsynaptic density-95, and dendritic microtubule-associated protein 2 in the striatum. Noticeably, chronic treatment with the metabotropic glutamate (mGlu) receptor 5 antagonist MTEP reversed the reduction of AMPA receptors in the NAc and CPu. MTEP also prevented depression- and anxiety-like behavior induced by social isolation. These data indicate that adulthood prolonged social isolation induces the adaptive downregulation of GluA1/A3-containing AMPA receptor expression in the limbic striatum. mGlu5 receptor activity is linked to this downregulation, and antagonism of mGlu5 receptors produces an antidepressant effect in this anhedonic model of depression.
Collapse
Key Words
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid;
- ANOVA, analysis of variance
- Antidepressant
- CDH2, Cadherin-2
- CPu, caudate putamen
- Caudate putamen
- GluA1
- MAP-2, microtubule-associated protein 2
- MTEP
- MTEP, 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]-pyridine
- Metabotropic glutamate receptor
- NAc, nucleus accumbens
- NCAD, neural cadherin
- Nucleus accumbens
- PFC, prefrontal cortex
- PSD-95, postsynaptic density-95
- Social isolation
- mGlu, metabotropic glutamate
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Nirav Mathur
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q. Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA,Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA,Correspondence to: Department of Biomedical Sciences, University of Missouri-Kansas City, School of Medicine, 2411 Holmes Street, Kansas City, MO 64108, USA.
| |
Collapse
|
3
|
Luc OT, Pizzagalli DA, Kangas BD. Toward a Quantification of Anhedonia: Unified Matching Law and Signal Detection for Clinical Assessment and Drug Development. Perspect Behav Sci 2021; 44:517-540. [PMID: 35098023 PMCID: PMC8738811 DOI: 10.1007/s40614-021-00288-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2021] [Indexed: 01/22/2023] Open
Abstract
Anhedonia, the loss of pleasure from previously rewarding activities, is a core symptom of several neuropsychiatric conditions, including major depressive disorder (MDD). Despite its transdiagnostic relevance, no effective therapeutics exist to treat anhedonia. This is due, in part, to inconsistent assays across clinical populations and laboratory animals, which hamper treatment development. To bridge this gap, recent work has capitalized on two long-standing research domains dedicated to quantifying responsivity to antecedents and consequences across species: the generalized matching law and signal detection theory. This review traces the integration of these quantitative frameworks, which yielded two empirically derived metrics: response bias (log b) and task discriminability (log d). These metrics serve as primary dependent variables in the Probabilistic Reward Task (PRT). In this computerized task, subjects make visual discriminations and probabilistic contingencies are arranged such that correct responses to one alternative are rewarded more often (rich) than correct responses to the other (lean). Under these conditions, healthy participants consistently develop a response bias in favor of the rich alternative, whereas participants with MDD exhibit blunted biases, which correlate with current and predict future anhedonia. Given the correspondence between anhedonic phenotypes and response bias, the PRT has been reverse-translated for rodents and nonhuman primates. Orderly log b and log d values have been observed across diverse clinical populations and laboratory animals. In addition, pharmacological challenges have produced similar outcomes across species. Taken together, this quantitative framework offers a highly translational approach to assaying reward responsiveness to accelerate treatment development for neuropsychiatric disorders involving anhedonia.
Collapse
Affiliation(s)
- Oanh T. Luc
- Harvard Medical School, McLean Hospital, 115 Mill Street, Belmont, MA 02478 USA
| | - Diego A. Pizzagalli
- Harvard Medical School, McLean Hospital, 115 Mill Street, Belmont, MA 02478 USA
| | - Brian D. Kangas
- Harvard Medical School, McLean Hospital, 115 Mill Street, Belmont, MA 02478 USA
| |
Collapse
|
4
|
Aygun H. Exendin-4 increases absence-like seizures and anxiety-depression-like behaviors in WAG/Rij rats. Epilepsy Behav 2021; 123:108246. [PMID: 34385055 DOI: 10.1016/j.yebeh.2021.108246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022]
Abstract
AIM Epilepsy is a neurological condition affecting millions of people worldwide. Glucagon-like peptide-1 (GLP-1) is a gut hormone, and its neuroprotective effect was investigated in previous studies. In this study, the effects of exendin-4, a GLP-1 receptor agonist, were studied in genetic absence epileptic Wistar Albino Glaxo/Rijswijk rats (WAG/Rij). WAG/Rij rat is a genetic model of the absence epilepsy and depression-like comorbidity. METHOD We examined the effects of exendin-4 (10, 50 and 100 µg/kg) on the absence seizures (Electrocorticography [ECoG] recordings), anxiety level (open-field test [OF]), and depression-like levels (forced swimming test [FST]) in the WAG/Rij rats. Basal ECoG recording was performed for all rats. Then, exendin-4 (10, 50 or 100 µg/kg) was administered intraperitoneally and ECoG recording was made for 180 min. After ECoG recording, forced swimming test and open-field test were applied. RESULTS Administration of 10, 50, or 100 µg/kg exendin-4 increased the duration and number of spike-wave discharges (SWDs) considerably without changing the amplitude. The 100 µg/kg dose of exendin-4 was the most effective in increasing the total duration of SWDs. Additionally, all exendin-4 doses increased anxiety level in OF and depression-like level in FST. CONCLUSION Our results showed that exendin-4 increased SWD incidence and anxiety- and depression-like behaviors in the WAG/Rij rats. Besides, it was also found that high doses caused the most proabsence effect.
Collapse
Affiliation(s)
- Hatice Aygun
- Department of Physiology, Faculty of Medicine, Tokat Gaziosmanpasa University, Tokat, Turkey.
| |
Collapse
|
5
|
Michaels TI, Stone E, Singal S, Novakovic V, Barkin RL, Barkin S. Brain reward circuitry: The overlapping neurobiology of trauma and substance use disorders. World J Psychiatry 2021; 11:222-231. [PMID: 34168969 PMCID: PMC8209534 DOI: 10.5498/wjp.v11.i6.222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/14/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Mental health symptoms secondary to trauma exposure and substance use disorders (SUDs) co-occur frequently in both clinical and community samples. The possibility of a shared aetiology remains an important question in translational neuroscience. Advancements in genetics, basic science, and neuroimaging have led to an improved understanding of the neural basis of these disorders, their frequent comorbidity and high rates of relapse remain a clinical challenge. This project aimed to conduct a review of the field's current understanding regarding the neural circuitry underlying posttraumatic stress disorder and SUD. A comprehensive review was conducted of available published literature regarding the shared neurobiology of these disorders, and is summarized in detail, including evidence from both animal and clinical studies. Upon summarizing the relevant literature, this review puts forth a hypothesis related to their shared neurobiology within the context of fear processing and reward cues. It provides an overview of brain reward circuitry and its relation to the neurobiology, symptomology, and phenomenology of trauma and substance use. This review provides clinical insights and implications of the proposed theory, including the potential development of novel pharmacological and therapeutic treatments to address this shared neurobiology. Limitations and extensions of this theory are discussed to provide future directions and insights for this shared phenomena.
Collapse
Affiliation(s)
- Timothy I Michaels
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, United States
- Department of Psychiatry, The Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, NY 11004, United States
| | - Emily Stone
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, United States
| | - Sonali Singal
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 11004, United States
| | - Vladan Novakovic
- Department of Psychiatry, The Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Glen Oaks, NY 11004, United States
| | - Robert L Barkin
- Department of Anesthesiology, Rush University Medical College, Chicago, IL 60612, United States
| | - Stacy Barkin
- Private Practice, Scottsdale, AZ 85250, United States
| |
Collapse
|
6
|
Mao LM, Wang JQ. Linkage of Non-receptor Tyrosine Kinase Fyn to mGlu5 Receptors in Striatal Neurons in a Depression Model. Neuroscience 2020; 433:11-20. [PMID: 32145272 DOI: 10.1016/j.neuroscience.2020.02.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/22/2022]
Abstract
The Src family kinase (SFK) is a subfamily of non-receptor tyrosine kinases. The SFK member Fyn is enriched at synaptic sites in the limbic reward circuit and plays a pivotal role in the regulation of glutamate receptors. In this study, we investigated changes in phosphorylation and function of the two key SFK members (Fyn and Src) and SFK interactions with a metabotropic glutamate (mGlu) receptor in the limbic striatum of adult rats in response to chronic passive stress, i.e., prolonged social isolation which is a pre-validated animal paradigm modeling depression in adulthood. In rats that showed typical anhedonic/depression-like behavior after chronic social isolation, phosphorylation of SFKs at a conserved and activation-associated autophosphorylation site (Y416) was not altered in the two subdivisions of the striatum, the nucleus accumbens and caudate putamen. The total level of phosphorylation and kinase activity of individual Fyn and Src immunopurified from the striatum also remained stable after social isolation. Noticeably, Fyn and Src were found to interact with a Gαq-coupled mGlu5 receptor in striatal neurons. The interaction of Fyn with mGlu5 receptors was selectively elevated in socially isolated rats. Moreover, social isolation induced an increase in surface expression of striatal mGlu5 receptors, which was reduced by an SFK inhibitor. These results indicate that Fyn interacts with mGlu5 receptors in striatal neurons. Adulthood social isolation in rats enhances the Fyn-mGlu5 interaction, which appears to be critical for the upregulation of surface mGlu5 receptor expression in striatal neurons.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
7
|
Somalwar AR, Choudhary AG, Balasubramanian N, Sakharkar AJ, Subhedar NK, Kokare DM. Cocaine- and amphetamine-regulated transcript peptide promotes reward seeking behavior in socially isolated rats. Brain Res 2019; 1728:146595. [PMID: 31830460 DOI: 10.1016/j.brainres.2019.146595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/06/2019] [Accepted: 12/07/2019] [Indexed: 10/25/2022]
Abstract
Reward deficit, expressed as anhedonia, is one of the major symptoms associated with neuropsychiatric disorders, but the underlying maladaptations have not been understood. Herein, we test the hypothesis that the neuropeptide cocaine- and amphetamine-regulated transcript (CART) may participate in the process. The study is justified since the peptide is a major player in inducing satiety and also processing of reward. The rats were socially isolated to induce reward deficit and conditioned to self-stimulate via an electrode in lateral hypothalamus (LH)-medial forebrain bundle (MFB) region. Compared to group-housed control rats, the socially isolated animals showed decreased lever press activity and elevated ICSS threshold indicating anhedonia-like condition. However, the effects of social isolation were alleviated by CART administered via intracerebroventricular route. The changes in the expression of CART protein and mRNA were screened using immunofluorescence and qRT-PCR methods, respectively. Socially isolated rats showed reduction in the expression of CART in the LH, nucleus accumbens shell (AcbSh) and posterior ventral tegmental area (pVTA) and CART mRNA in the Acb and LH. Double immunostaining with antibodies against CART and synaptophysin revealed significant loss of colabeled elements in LH, AcbSh and pVTA. We suggest that down-regulation of endogenous CARTergic system in the LH-pVTA-AcbSh reward circuitry may be causal to motivational anhedonia like phenotype seen in neuropsychiatric conditions.
Collapse
Affiliation(s)
- Amita R Somalwar
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Amit G Choudhary
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | | | - Amul J Sakharkar
- Department of Biotechnology, Savitribai Phule Pune University, Pune 411 007, India
| | - Nishikant K Subhedar
- Indian Institute of Science Education and Research (IISER), Dr. Homi Bhabha Road, Pune 411 008, India
| | - Dadasaheb M Kokare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India.
| |
Collapse
|
8
|
Antidepressant effects of ketamine on depression-related phenotypes and dopamine dysfunction in rodent models of stress. Behav Brain Res 2019; 379:112367. [PMID: 31739001 DOI: 10.1016/j.bbr.2019.112367] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
Depression, the most prevalent psychiatric disorder, is characterized by increased negative affect (i.e. depressed mood) and reduced positive affect (i.e. anhedonia). Stress is a risk factor for depression in humans, and animal models of chronic stress are typically used to study neurobehavioral alterations relevant to depression. Common behavioral outcomes in rodent models of chronic stress include anhedonia, social dysfunction and behavioral despair. For example, chronically stressed rodents exhibit reduced reward preference, as measured by a loss of preference for sucrose solutions and time spent interacting with a novel conspecific, while also exhibiting less time struggling against inescapable stressors (e.g. forced swim, tail suspension). In both humans and rodents, anhedonia is associated with dysfunction of the dopamine (DA) system. Unlike traditional antidepressants, which are limited by inadequate efficacy and delayed therapeutic response, acute ketamine administration rapidly alleviates depressive symptoms in humans and reverses stress-induced changes in animal models. These effects are partially mediated via actions on the DA system. This review summarizes the clinical effects of ketamine, the neurobiological underpinnings of depression with a focus on DA dysfunction, as well as antidepressant effects of ketamine on depression-related endophenotypes (i.e. anhedonia, despair) and ventral tegmental area (VTA) activity in rodent models of repeated stress. Moreover, we discuss evidence regarding sex differences in ketamine's antidepressant effects, wherein females appear to be more sensitive to lower dose ketamine, as well as novel findings suggesting that ketamine has prophylactic effects with regard to protection against the neurobehavioral impact of future stressors.
Collapse
|
9
|
Conoscenti MA, Fanselow MS. Dissociation in Effective Treatment and Behavioral Phenotype Between Stress-Enhanced Fear Learning and Learned Helplessness. Front Behav Neurosci 2019; 13:104. [PMID: 31156405 PMCID: PMC6529815 DOI: 10.3389/fnbeh.2019.00104] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/29/2019] [Indexed: 01/29/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating disease with relatively high lifetime prevalence. It is marked by a high diversity of symptoms and comorbidity with other psychiatric disease. Furthermore, PTSD has a high level of origin and symptom heterogeneity within the population. These characteristics taken together make it one of the most challenging diseases to effectively model in animals. However, with relatively little headway made in developing effective disease interventions, PTSD remains as a high priority target for animal model study. Learned Helplessness (LH) is a procedure classically used to model depression, but has in recent years transitioned to use as a model of PTSD. Animals in this procedure receive 100 inescapable and unpredictable tailshocks or simple restraint without shock. The following day, the animals are tested in a shuttle box, where inescapably-shocked subjects exhibit exaggerated fear and profound deficit in escape performance. Stress-enhanced fear learning (SEFL) also uses an acute (single session) stressor for modeling PTSD in rodents. The SEFL procedure begins with exposure to 15 footshocks or simple context exposure without shock. Animals that initially received the 15 footshocks exhibit future enhanced fear learning. In this review, we will compare the behavior, physiology, and interventions of these two animal models of PTSD. Despite considerable similarity (a single session containing inescapable and uncontrollable shock) the two procedures produce a very divergent set of behavioral consequences.
Collapse
Affiliation(s)
- Michael A Conoscenti
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Michael S Fanselow
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States.,Staglin Center for Brian and Behavioral Health, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
10
|
Robinson ESJ. Translational new approaches for investigating mood disorders in rodents and what they may reveal about the underlying neurobiology of major depressive disorder. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0036. [PMID: 29352034 PMCID: PMC5790833 DOI: 10.1098/rstb.2017.0036] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2017] [Indexed: 01/02/2023] Open
Abstract
Mood disorders represent one of society's most costly and challenging health burdens. The drug treatments used today were initially discovered serendipitously in the 1950s. Animal models were then developed based on the ability of these drugs to alter specific behaviours. These models have played a major role in the development of the second generation of antidepressants. However, their use has been heavily criticized, particularly in relation to whether they recapitulate similar underlying biology to the psychiatric disorder they are proposed to represent. This article considers our work in the field of affective bias and the development of a translational research programme to try to develop and validate better animal models. We discuss whether the new data that have arisen from these studies support an alternative perspective on the underlying neurobiological processes that lead to major depressive disorder (MDD). Specifically, this article will consider whether a neuropsychological mechanism involving affective biases plays a causal role in the development of MDD and its associated emotional and behavioural symptoms. These animal studies also raise the possibility that neuropsychological mechanisms involving affective biases are a precursor to, rather than a consequence of, the neurotrophic changes linked to MDD. This article is part of a discussion meeting issue ‘Of mice and mental health: facilitating dialogue between basic and clinical neuroscientists’.
Collapse
Affiliation(s)
- Emma S J Robinson
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
11
|
Elman I, Upadhyay J, Langleben DD, Albanese M, Becerra L, Borsook D. Reward and aversion processing in patients with post-traumatic stress disorder: functional neuroimaging with visual and thermal stimuli. Transl Psychiatry 2018; 8:240. [PMID: 30389908 PMCID: PMC6214971 DOI: 10.1038/s41398-018-0292-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/27/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022] Open
Abstract
In patients with post-traumatic stress disorder (PTSD), a decrease in the brain reward function was reported in behavioral- and in neuroimaging studies. While pathophysiological mechanisms underlying this response are unclear, there are several lines of evidence suggesting over-recruitment of the brain reward regions by aversive stimuli rendering them unavailable to respond to reward-related content. The purpose of this study was to juxtapose brain responses to functional neuroimaging probes that reliably produce rewarding and aversive experiences in PTSD subjects and in healthy controls. The stimuli used were pleasant, aversive and neutral images selected from the International Affective Picture System (IAPS) along with pain-inducing heat applied to the dorsum of the left hand; all were administered during 3 T functional magnetic resonance imaging. Analyses of IAPS responses for the pleasant images revealed significantly decreased subjective ratings and brain activations in PTSD subjects that included striatum and medial prefrontal-, parietal- and temporal cortices. For the aversive images, decreased activations were observed in the amygdala and in the thalamus. PTSD and healthy subjects provided similar subjective ratings of thermal sensory thresholds and each of the temperatures. When 46 °C (hot) and 42 °C (neutral) temperatures were contrasted, voxelwise between-group comparison revealed greater activations in the striatum, amygdala, hippocampus and medial prefrontal cortex in the PTSD subjects. These latter findings were for the most part mirrored by the 44 vs. 42 °C contrast. Our data suggest different brain alterations patterns in PTSD, namely relatively diminished corticolimbic response to pleasant and aversive psychosocial stimuli in the face of exaggerated response to heat-related pain. The present findings support the hypothesis that brain sensitization to pain in PTSD may interfere with the processing of psychosocial stimuli whether they are of rewarding or aversive valence.
Collapse
Affiliation(s)
- Igor Elman
- Department of Psychiatry, Cooper Medical School, Rowan University, Glassboro, NJ, USA.
| | - Jaymin Upadhyay
- 000000041936754Xgrid.38142.3cCenter for Pain and the Brain, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Daniel D. Langleben
- 0000 0004 1936 8972grid.25879.31Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Mark Albanese
- 000000041936754Xgrid.38142.3cCambridge Health Alliance, Harvard Medical School, Boston, MA USA
| | - Lino Becerra
- 000000041936754Xgrid.38142.3cCenter for Pain and the Brain, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - David Borsook
- 000000041936754Xgrid.38142.3cCenter for Pain and the Brain, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|
12
|
Scheggi S, De Montis MG, Gambarana C. Making Sense of Rodent Models of Anhedonia. Int J Neuropsychopharmacol 2018; 21:1049-1065. [PMID: 30239762 PMCID: PMC6209858 DOI: 10.1093/ijnp/pyy083] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 09/18/2018] [Indexed: 01/04/2023] Open
Abstract
A markedly reduced interest or pleasure in activities previously considered pleasurable is a main symptom in mood disorder and psychosis and is often present in other psychiatric disorders and neurodegenerative diseases. This condition can be labeled as "anhedonia," although in its most rigorous connotation the term refers to the lost capacity to feel pleasure that is one aspect of the complex phenomenon of processing and responding to reward. The responses to rewarding stimuli are relatively easy to study in rodents, and the experimental conditions that consistently and persistently impair these responses are used to model anhedonia. To this end, long-term exposure to environmental aversive conditions is primarily used, and the resulting deficits in reward responses are often accompanied by other deficits that are mainly reminiscent of clinical depressive symptoms. The different components of impaired reward responses induced by environmental aversive events can be assessed by different tests or protocols that require different degrees of time allocation, technical resources, and equipment. Rodent models of anhedonia are valuable tools in the study of the neurobiological mechanisms underpinning impaired behavioral responses and in the screening and characterization of drugs that may reverse these behavioral deficits. In particular, the antianhedonic or promotivational effects are relevant features in the spectrum of activities of drugs used in mood disorders or psychosis. Thus, more than the model, it is the choice of tests that is crucial since it influences which facets of anhedonia will be detected and should be tuned to the purpose of the study.
Collapse
Affiliation(s)
- Simona Scheggi
- Department of Molecular and Developmental Medicine, University of Siena
| | | | - Carla Gambarana
- Department of Molecular and Developmental Medicine, University of Siena,Correspondence: Carla Gambarana, Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 2 – 53100 Siena, Italy ()
| |
Collapse
|
13
|
Mao LM, Wang JQ. Alterations in mGlu5 receptor expression and function in the striatum in a rat depression model. J Neurochem 2018; 145:287-298. [PMID: 29337350 DOI: 10.1111/jnc.14307] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/27/2017] [Accepted: 01/03/2018] [Indexed: 12/24/2022]
Abstract
Major depressive disorder is a common form of mental illness. Many brain regions are implicated in the pathophysiology and symptomatology of depression. Among key brain areas is the striatum that controls reward and mood and is involved in the development of core depression-like behavior in animal models of depression. While molecular mechanisms in this region underlying depression-related behavior are poorly understood, the glutamatergic input to the striatum is believed to play a role. In this study, we investigated changes in metabotropic glutamate (mGlu) receptor expression and signaling in the striatum of adult rats in response to prolonged (10-12 weeks) social isolation, a pre-validated animal paradigm modeling depression in adulthood. We found that mGlu5 receptor protein levels in the striatum were increased in rats that showed typical depression- and anxiety-like behavior after chronic social isolation. This increase in mGlu5 receptor expression was seen in both subdivisions of the striatum, the nucleus accumbens and caudate putamen. At subcellular and subsynaptic levels, mGlu5 receptor expression was elevated in surface membranes at synaptic sites. In striatal neurons, the mGlu5-associated phosphoinositide signaling pathway was augmented in its efficacy after prolonged social isolation. These data indicate that the mGlu5 receptor is a sensitive substrate of depression. Adulthood social isolation leads to the up-regulation of mGlu5 receptor expression and function in striatal neurons.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - John Q Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA.,Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
14
|
Slaney C, Hinchcliffe JK, Robinson ESJ. Translational Shifts in Preclinical Models of Depression: Implications for Biomarkers for Improved Treatments. Curr Top Behav Neurosci 2018; 40:169-193. [PMID: 29696602 PMCID: PMC7614182 DOI: 10.1007/7854_2018_44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Understanding the neurobiology of major depressive disorder (MDD) remains one of the major challenges in neuroscience. The disease is heterogeneous in nature, and patients present with a varied symptom profile. Studies seeking to identify biomarkers for MDD diagnosis and treatment have not yet found any one candidate which achieves sufficient sensitivity and specificity. In this article, we consider whether neuropsychological impairments, specifically affective biases, could provide a behavioural biomarker. Affective biases are observed when emotional states influence cognitive function. These biases have been shown to influence a number of different cognitive domains with some specific deficits observed in MDD. It has also been possible to use these neuropsychological tests to inform the development of translational tasks for non-human species. The results from studies in rodents suggest that quantification of affective biases is feasible and may provide a reliable method to predict antidepressant efficacy as well as pro-depressant risk. Animal studies suggest that affective state-induced biases in learning and memory operate over a different time course to biases influencing decision-making. The implications for these differences in terms of task validity and future ideas relating to affective biases and MDD are discussed. We also describe our most recent studies which have shown that depression-like phenotypes share a common deficit in reward-related learning and memory which we refer to as a reward-induced positive bias. This deficit is dissociable from more typical measures of hedonic behaviour and motivation for reward and may represent an important and distinct form of reward deficit linked to MDD.
Collapse
Affiliation(s)
- Chloe Slaney
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol, UK
| | - Justyna K Hinchcliffe
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol, UK
| | - Emma S J Robinson
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol, UK.
| |
Collapse
|
15
|
Choi MH, Na JE, Yoon YR, Lee HJ, Yoon S, Rhyu IJ, Baik JH. Role of Dopamine D2 Receptor in Stress-Induced Myelin Loss. Sci Rep 2017; 7:11654. [PMID: 28912499 PMCID: PMC5599541 DOI: 10.1038/s41598-017-10173-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 08/04/2017] [Indexed: 12/24/2022] Open
Abstract
Dopaminergic systems play a major role in reward-related behavior and dysregulation of dopamine (DA) systems can cause several mental disorders, including depression. We previously reported that dopamine D2 receptor knockout (D2R-/-) mice display increased anxiety and depression-like behaviors upon chronic stress. Here, we observed that chronic stress caused myelin loss in wild-type (WT) mice, while the myelin level in D2R-/- mice, which was already lower than that in WT mice, was not affected upon stress. Fewer mature oligodendrocytes (OLs) were observed in the corpus callosum of stressed WT mice, while in D2R-/- mice, both the control and stressed group displayed a decrease in the number of mature OLs. We observed a decrease in the number of active β-catenin (ABC)-expressing and TCF4-expressing cells among OL lineage cells in the corpus callosum of stressed WT mice, while such regulation was not found in D2R-/- mice. Administration of lithium normalized the behavioral impairments and myelin damage induced by chronic stress in WT mice, and restored the number of ABC-positive and TCF4-positive OLs, while such effect was not found in D2R-/- mice. Together, our findings indicate that chronic stress induces myelin loss through the Wnt/β-catenin signaling pathway in association with DA signaling through D2R.
Collapse
Affiliation(s)
- Mi-Hyun Choi
- Molecular Neurobiology Laboratory, Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Ji Eun Na
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Ye Ran Yoon
- Molecular Neurobiology Laboratory, Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Hyo Jin Lee
- Molecular Neurobiology Laboratory, Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Sehyoun Yoon
- Molecular Neurobiology Laboratory, Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Im Joo Rhyu
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Korea
| | - Ja-Hyun Baik
- Molecular Neurobiology Laboratory, Department of Life Sciences, Korea University, Seoul, 02841, Korea.
| |
Collapse
|
16
|
Abstract
Acute trauma can lead to life-long changes in susceptibility to psychiatric disease, such as post-traumatic stress disorder (PTSD). Rats given free access to a concentrated glucose solution for 24 h beginning immediately after trauma failed to show stress-related pathology in the learned helplessness model of PTSD and comorbid major depression. We assessed effective dosing and temporal constraints of the glucose intervention in three experiments. We exposed 120 male Sprague-Dawley rats to 100, 1 mA, 3-15 s, inescapable and unpredictable electric tail shocks (over a 110-min period) or simple restraint in the learned helplessness procedure. Rats in each stress condition had access to a 40% glucose solution or water. We measured fluid consumption under 18-h free access conditions, or limited access (1, 3, 6, 18 h) beginning immediately after trauma, or 3-h access with delayed availability of the glucose solution (0, 1, 3, 6 h). We hypothesized that longer and earlier access following acute stress would improve shuttle-escape performance. Rats exposed to traumatic shock and given 18-h access to glucose failed to show exaggerated fearfulness and showed normal reactivity to foot shock during testing as compared to their water-treated counterparts. At least 3 h of immediate post-stress access to glucose were necessary to see these improvements in test performance. Moreover, delaying access to glucose for more than 3 h post-trauma yielded no beneficial effects. These data clearly identify limits on the post-stress glucose intervention. In conclusion, glucose should be administered almost immediately and at the highest dose after trauma.
Collapse
Affiliation(s)
- M A Conoscenti
- a Department of Psychology , University of California , Los Angeles , Los Angeles , CA , USA
| | - E E Hart
- a Department of Psychology , University of California , Los Angeles , Los Angeles , CA , USA
| | - N J Smith
- a Department of Psychology , University of California , Los Angeles , Los Angeles , CA , USA
| | - T R Minor
- a Department of Psychology , University of California , Los Angeles , Los Angeles , CA , USA
- b UCLA Behavioral Testing Core , Brain Research Institute , Los Angeles , CA , USA
- c Department of Psychiatry and Biobehavioral Sciences , UCLA Integrative Center for Learning and Memory , Los Angeles , CA , USA
- d Stress and Motivated Behavior Institute , New Jersey Medical School , Newark , NJ , USA
| |
Collapse
|
17
|
Abstract
Depression is one of the most common but poorly understood psychiatric conditions. Although drug treatments and psychological therapies are effective in some patients, many do not achieve full remission and some patients receive no apparent benefit. Developing new improved treatments requires a better understanding of the aetiology of symptoms and evaluation of novel therapeutic targets in pre-clinical studies. Recent developments in our understanding of the basic cognitive processes that may contribute to the development of depression and its treatment offer new opportunities for both clinical and pre-clinical research. This chapter discusses the clinical evidence supporting a cognitive neuropsychological model of depression and antidepressant efficacy, and how this information may be usefully translated to pre-clinical investigation. Studies using neuropsychological tests in depressed patients and at risk populations have revealed basic negative emotional biases and disrupted reward and punishment processing, which may also impact on non-affective cognition. These affective biases are sensitive to antidepressant treatments with early onset effects observed, suggesting an important role in recovery. This clinical work into affective biases has also facilitated back-translation to animals and the development of assays to study affective biases in rodents. These animal studies suggest that, similar to humans, rodents in putative negative affective states exhibit negative affective biases on decision-making and memory tasks. Antidepressant treatments also induce positive biases in these rodent tasks, supporting the translational validity of this approach. Although still in the early stages of development and validation, affective biases in depression have the potential to offer new insights into the clinical condition, as well as facilitating the development of more translational approaches for pre-clinical studies.
Collapse
Affiliation(s)
- E S J Robinson
- School of Physiology and Pharmacology, Medical Sciences Building, University Walk, Bristol, BS8 4PX, UK.
| | - J P Roiser
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London, WC1N 3AR, UK
| |
Collapse
|
18
|
How age, sex and genotype shape the stress response. Neurobiol Stress 2016; 6:44-56. [PMID: 28229108 PMCID: PMC5314441 DOI: 10.1016/j.ynstr.2016.11.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/19/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022] Open
Abstract
Exposure to chronic stress is a leading pre-disposing factor for several neuropsychiatric disorders as it often leads to maladaptive responses. The response to stressful events is heterogeneous, underpinning a wide spectrum of distinct changes amongst stress-exposed individuals'. Several factors can underlie a different perception to stressors and the setting of distinct coping strategies that will lead to individual differences on the susceptibility/resistance to stress. Beyond the factors related to the stressor itself, such as intensity, duration or predictability, there are factors intrinsic to the individuals that are relevant to shape the stress response, such as age, sex and genetics. In this review, we examine the contribution of such intrinsic factors to the modulation of the stress response based on experimental rodent models of response to stress and discuss to what extent that knowledge can be potentially translated to humans. Effect of age in the stress response. Effect of sex in the stress response. Effect of genotype in the stress response.
Collapse
|
19
|
Abstract
Depression is one of the most common but poorly understood psychiatric conditions. Although drug treatments and psychological therapies are effective in some patients, many do not achieve full remission and some patients receive no apparent benefit. Developing new improved treatments requires a better understanding of the aetiology of symptoms and evaluation of novel therapeutic targets in pre-clinical studies. Recent developments in our understanding of the basic cognitive processes that may contribute to the development of depression and its treatment offer new opportunities for both clinical and pre-clinical research. This chapter discusses the clinical evidence supporting a cognitive neuropsychological model of depression and antidepressant efficacy, and how this information may be usefully translated to pre-clinical investigation. Studies using neuropsychological tests in depressed patients and at risk populations have revealed basic negative emotional biases and disrupted reward and punishment processing, which may also impact on non-affective cognition. These affective biases are sensitive to antidepressant treatments with early onset effects observed, suggesting an important role in recovery. This clinical work into affective biases has also facilitated back-translation to animals and the development of assays to study affective biases in rodents. These animal studies suggest that, similar to humans, rodents in putative negative affective states exhibit negative affective biases on decision-making and memory tasks. Antidepressant treatments also induce positive biases in these rodent tasks, supporting the translational validity of this approach. Although still in the early stages of development and validation, affective biases in depression have the potential to offer new insights into the clinical condition, as well as facilitating the development of more translational approaches for pre-clinical studies.
Collapse
Affiliation(s)
- E S J Robinson
- School of Physiology and Pharmacology, Medical Sciences Building, University Walk, Bristol, BS8 4PX, UK.
| | - J P Roiser
- Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London, WC1N 3AR, UK
| |
Collapse
|
20
|
Abstract
Drug withdrawal is often conceptualized as an aversive state that motivates drug-seeking and drug-taking behaviors in humans. Stress is more difficult to define, but is also frequently associated with aversive states. Here we describe evidence for the simple theory that drug withdrawal is a stress-like state, on the basis of common effects on behavioral, neurochemical, and molecular endpoints. We also describe data suggesting a more complex relationship between drug withdrawal and stress. As one example, we will highlight evidence that, depending on drug class, components of withdrawal can produce effects that have characteristics consistent with mood elevation. In addition, some stressors can act as positive reinforcers, defined as having the ability to increase the probability of a behavior that produces it. As such, accumulating evidence supports the general principles of opponent process theory, whereby processes that have an affective valence are followed in time by an opponent process that has the opposite valence. Throughout, we identify gaps in knowledge and propose future directions for research. A better understanding of the similarities, differences, and overlaps between drug withdrawal and stress will lead to the development of improved treatments for addiction, as well as for a vast array of neuropsychiatric conditions that are triggered or exacerbated by stress.
Collapse
|
21
|
Abstract
Hormesis is the process by which small stresses build resilience to large stresses. We pre-exposed rats to various parameters of mild-to-moderate stress prior to traumatic stress in the present experiments to assess the potential benefits of hormetic training on resilience to traumatic, uncontrollable stress. Rats underwent varying stress pre-training parameters prior to exposure to uncontrollable traumatic stress in the learned helplessness procedure. The ability to prevent the exaggerated fear responding and escape deficits that normally follow experience with traumatic stress were used as a measure of the benefits of hormetic training. Four experiments examined the effects of number of training sessions, stressor severity and pattern of rest between pre-training stress sessions. Repeated exposure to mild restraint stress or moderate shock stress eliminated both the enhanced fear conditioning and shuttle-escape deficits that result from exposure to traumatic, inescapable shock. The pattern of rest did not contribute to resilience when the pre-exposure stressor was mild, but was vital when the pre-exposure stressor was moderate, with an alternation of stress and rest being the most effective procedure. The data also suggest that the level of resilience may increase with the number of pre-exposure sessions.
Collapse
Affiliation(s)
- Traci N Plumb
- Department of Psychology, University of California , Los Angeles, CA , USA
| | | | | |
Collapse
|
22
|
Hales CA, Stuart SA, Anderson MH, Robinson ESJ. Modelling cognitive affective biases in major depressive disorder using rodents. Br J Pharmacol 2014; 171:4524-38. [PMID: 24467454 PMCID: PMC4199314 DOI: 10.1111/bph.12603] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/03/2014] [Accepted: 01/18/2014] [Indexed: 02/03/2023] Open
Abstract
Major depressive disorder (MDD) affects more than 10% of the population, although our understanding of the underlying aetiology of the disease and how antidepressant drugs act to remediate symptoms is limited. Major obstacles include the lack of availability of good animal models that replicate aspects of the phenotype and tests to assay depression-like behaviour in non-human species. To date, research in rodents has been dominated by two types of assays designed to test for depression-like behaviour: behavioural despair tests, such as the forced swim test, and measures of anhedonia, such as the sucrose preference test. These tests have shown relatively good predictive validity in terms of antidepressant efficacy, but have limited translational validity. Recent developments in clinical research have revealed that cognitive affective biases (CABs) are a key feature of MDD. Through the development of neuropsychological tests to provide objective measures of CAB in humans, we have the opportunity to use ‘reverse translation’ to develop and evaluate whether similar methods are suitable for research into MDD using animals. The first example of this approach was reported in 2004 where rodents in a putative negative affective state were shown to exhibit pessimistic choices in a judgement bias task. Subsequent work in both judgement bias tests and a novel affective bias task suggest that these types of assay may provide translational methods for studying MDD using animals. This review considers recent work in this area and the pharmacological and translational validity of these new animal models of CABs.
Collapse
Affiliation(s)
- Claire A Hales
- School of Physiology and Pharmacology, University of Bristol, University Walk, Bristol, UK
| | | | | | | |
Collapse
|
23
|
Levinstein MR, Samuels BA. Mechanisms underlying the antidepressant response and treatment resistance. Front Behav Neurosci 2014; 8:208. [PMID: 25018708 PMCID: PMC4073308 DOI: 10.3389/fnbeh.2014.00208] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/22/2014] [Indexed: 12/28/2022] Open
Abstract
Depression is a complex and heterogeneous disorder affecting millions of Americans. There are several different medications and other treatments that are available and effective for many patients with depression. However, a substantial percentage of patients fail to achieve remission with these currently available interventions, and relapse rates are high. Therefore, it is necessary to determine both the mechanisms underlying the antidepressant response and the differences between responders and non-responders to treatment. Delineation of these mechanisms largely relies on experiments that utilize animal models. Therefore, this review provides an overview of the various mouse models that are currently used to assess the antidepressant response, such as chronic mild stress, social defeat, and chronic corticosterone. We discuss how these mouse models can be used to advance our understanding of the differences between responders and non-responders to antidepressant treatment. We also provide an overview of experimental treatment modalities that are used for treatment-resistant depression, such as deep brain stimulation and ketamine administration. We will then review the various genetic polymorphisms and transgenic mice that display resistance to antidepressant treatment. Finally, we synthesize the published data to describe a potential neural circuit underlying the antidepressant response and treatment resistance.
Collapse
Affiliation(s)
- Marjorie R Levinstein
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, Research Foundation for Mental Hygiene, Inc. New York, NY, USA
| | - Benjamin A Samuels
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, Research Foundation for Mental Hygiene, Inc. New York, NY, USA
| |
Collapse
|
24
|
Hyper-reactive human ventral tegmental area and aberrant mesocorticolimbic connectivity in overgeneralization of fear in generalized anxiety disorder. J Neurosci 2014; 34:5855-60. [PMID: 24760845 DOI: 10.1523/jneurosci.4868-13.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The ventral tegmental area (VTA) has been primarily implicated in reward-motivated behavior. Recently, aberrant dopaminergic VTA signaling has also been implicated in anxiety-like behaviors in animal models. These findings, however, have yet to be extended to anxiety in humans. Here we hypothesized that clinical anxiety is linked to dysfunction of the mesocorticolimbic circuit during threat processing in humans; specifically, excessive or dysregulated activity of the mesocorticolimbic aversion circuit may be etiologically related to errors in distinguishing cues of threat versus safety, also known as "overgeneralization of fear." To test this, we recruited 32 females with generalized anxiety disorder and 25 age-matched healthy control females. We measured brain activity using fMRI while participants underwent a fear generalization task consisting of pseudo-randomly presented rectangles with systematically varying widths. A mid-sized rectangle served as a conditioned stimulus (CS; 50% electric shock probability) and rectangles with widths of CS ±20%, ±40%, and ±60% served as generalization stimuli (GS; never paired with electric shock). Healthy controls showed VTA reactivity proportional to the cue's perceptual similarity to CS (threat). In contrast, patients with generalized anxiety disorder showed heightened and less discriminating VTA reactivity to GS, a feature that was positively correlated with trait anxiety, as well as increased mesocortical and decreased mesohippocampal coupling. Our results suggest that the human VTA and the mesocorticolimbic system play a crucial role in threat processing, and that abnormalities in this system are implicated in maladaptive threat processing in clinical anxiety.
Collapse
|
25
|
Vargas-Perez H, Bahi A, Bufalino MR, Ting-A-Kee R, Maal-Bared G, Lam J, Fahmy A, Clarke L, Blanchard JK, Larsen BR, Steffensen S, Dreyer JL, van der Kooy D. BDNF signaling in the VTA links the drug-dependent state to drug withdrawal aversions. J Neurosci 2014; 34:7899-909. [PMID: 24899712 PMCID: PMC4099491 DOI: 10.1523/jneurosci.3776-13.2014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 03/08/2014] [Accepted: 03/29/2014] [Indexed: 02/08/2023] Open
Abstract
Drug administration to avoid unpleasant drug withdrawal symptoms has been hypothesized to be a crucial factor that leads to compulsive drug-taking behavior. However, the neural relationship between the aversive motivational state produced by drug withdrawal and the development of the drug-dependent state still remains elusive. It has been observed that chronic exposure to drugs of abuse increases brain-derived neurotrophic factor (BDNF) levels in ventral tegmental area (VTA) neurons. In particular, BDNF expression is dramatically increased during drug withdrawal, which would suggest a direct connection between the aversive state of withdrawal and BDNF-induced neuronal plasticity. Using lentivirus-mediated gene transfer to locally knock down the expression of the BDNF receptor tropomyosin-receptor-kinase type B in rats and mice, we observed that chronic opiate administration activates BDNF-related neuronal plasticity in the VTA that is necessary for both the establishment of an opiate-dependent state and aversive withdrawal motivation. Our findings highlight the importance of a bivalent, plastic mechanism that drives the negative reinforcement underlying addiction.
Collapse
Affiliation(s)
- Hector Vargas-Perez
- Department of Molecular Genetics, Neurobiology Research Group, University of Toronto, Toronto, Ontario, M5S3E1 Canada,
| | - Amine Bahi
- Department of Anatomy, Faculty of Medicine and Health Sciences, United Arab Emirates University, Alabama Ain, 17666 United Arab Emirates
| | - Mary Rose Bufalino
- Department of Medical Biophysics, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S3E1 Canada
| | - Ryan Ting-A-Kee
- Department of Molecular Genetics, Neurobiology Research Group, University of Toronto, Toronto, Ontario, M5S3E1 Canada
| | - Geith Maal-Bared
- Department of Molecular Genetics, Neurobiology Research Group, University of Toronto, Toronto, Ontario, M5S3E1 Canada
| | - Jenny Lam
- Department of Molecular Genetics, Neurobiology Research Group, University of Toronto, Toronto, Ontario, M5S3E1 Canada
| | - Ahmed Fahmy
- Department of Molecular Genetics, Neurobiology Research Group, University of Toronto, Toronto, Ontario, M5S3E1 Canada
| | - Laura Clarke
- Department of Molecular Genetics, Neurobiology Research Group, University of Toronto, Toronto, Ontario, M5S3E1 Canada
| | - Jennifer K Blanchard
- Department of Psychology and Center for Neuroscience, Brigham Young University, Provo, UT 84602, and
| | - Brett R Larsen
- Department of Psychology and Center for Neuroscience, Brigham Young University, Provo, UT 84602, and
| | - Scott Steffensen
- Department of Psychology and Center for Neuroscience, Brigham Young University, Provo, UT 84602, and
| | - Jean-Luc Dreyer
- Department of Medicine, Division of Biochemistry, University of Fribourg, Fribourg 1700, Switzerland
| | - Derek van der Kooy
- Department of Molecular Genetics, Neurobiology Research Group, University of Toronto, Toronto, Ontario, M5S3E1 Canada, Department of Medical Biophysics, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S3E1 Canada
| |
Collapse
|
26
|
Nikulina EM, Johnston CE, Wang J, Hammer RP. Neurotrophins in the ventral tegmental area: Role in social stress, mood disorders and drug abuse. Neuroscience 2014; 282:122-38. [PMID: 24875178 DOI: 10.1016/j.neuroscience.2014.05.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/04/2014] [Accepted: 05/11/2014] [Indexed: 01/19/2023]
Abstract
This review discusses the impact of neurotrophins and other trophic factors, including fibroblast growth factor and glial cell line-derived neurotrophic factor, on mood disorders, weight regulation and drug abuse, with an emphasis on stress- and drug-induced changes in the ventral tegmental area (VTA). Neurotrophins, comprising nerve growth factor, brain-derived neurotrophic factor (BDNF), and neurotrophins 3 and 4/5 play important roles in neuronal plasticity and the development of different psychopathologies. In the VTA, most research has focused on the role of BDNF, because other neurotrophins are not found there in significant quantities. BDNF originating in the VTA provides trophic support to dopamine neurons. The diverse intracellular signaling pathways activated by BDNF may underlie precise physiological functions specific to the VTA. In general, VTA BDNF expression increases after psychostimulant exposures, and enhanced BDNF level in the VTA facilitates psychostimulant effects. The impact of VTA BDNF on the behavioral effects of psychostimulants relies primarily on its action within the mesocorticolimbic circuit. In the case of opiates, VTA BDNF expression and effects seem to be dependent on whether an animal is drug-naïve or has a history of drug use, only the latter of which is related to dopamine mechanisms. Social defeat stress that is continuous in mice or intermittent in rats increases VTA BDNF expression, and is associated with depressive and social avoidance behaviors. Intermittent social defeat stress induces persistent VTA BDNF expression that triggers psychostimulant cross-sensitization. Understanding the cellular and molecular substrates of neurotrophin effects may lead to novel therapeutic approaches for the prevention and treatment of substance use and mood disorders.
Collapse
Affiliation(s)
- E M Nikulina
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA.
| | - C E Johnston
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA; Interdisciplinary Neuroscience Program, Arizona State University, Tempe, AZ, USA
| | - J Wang
- Interdisciplinary Neuroscience Program, Arizona State University, Tempe, AZ, USA
| | - R P Hammer
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA; Interdisciplinary Neuroscience Program, Arizona State University, Tempe, AZ, USA; Department of Pharmacology and Department of Psychiatry, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
27
|
Hamilton DE, Cooke CL, Carter BS, Akil H, Watson SJ, Thompson RC. Basal microRNA expression patterns in reward circuitry of selectively bred high-responder and low-responder rats vary by brain region and genotype. Physiol Genomics 2014; 46:290-301. [PMID: 24569673 PMCID: PMC4035657 DOI: 10.1152/physiolgenomics.00152.2013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/20/2014] [Indexed: 11/22/2022] Open
Abstract
Mental health disorders involving altered reward, emotionality, and anxiety are thought to result from the interaction of individual predisposition (genetic factors) and personal experience (environmental factors), although the mechanisms that contribute to an individual's vulnerability to these disorders remain poorly understood. We used an animal model of individual variation [inbred high-responder/low-responder (bHR-bLR) rodents] known to vary in reward, anxiety, and emotional processing to examine neuroanatomical expression patterns of microRNAs (miRNAs). Laser capture microdissection was used to dissect the prelimbic cortex and the nucleus accumbens core and shell prior to analysis of basal miRNA expression in bHR and bLR male rats. These studies identified 187 miRNAs differentially expressed by genotype in at least one brain region, 10 of which were validated by qPCR. Four of these 10 qPCR-validated miRNAs demonstrated differential expression across multiple brain regions, and all miRNAs with validated differential expression between genotypes had lower expression in bHR animals compared with bLR animals. microRNA (miR)-484 and miR-128a expression differences between the prelimbic cortex of bHR and bLR animals were validated by semiquantitative in situ hybridization. miRNA expression analysis independent of genotype identified 101 miRNAs differentially expressed by brain region, seven of which validated by qPCR. Dnmt3a mRNA, a validated target of miR-29b, varied in a direction opposite that of miR-29b's differential expression between bHR and bLR animals. These data provide evidence that basal central nervous system miRNA expression varies in the bHR-bLR model, implicating microRNAs as potential epigenetic regulators of key neural circuits and individual differences associated with mental health disorders.
Collapse
Affiliation(s)
- David E Hamilton
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | | |
Collapse
|
28
|
Van't Veer A, Carlezon WA. Role of kappa-opioid receptors in stress and anxiety-related behavior. Psychopharmacology (Berl) 2013; 229:435-52. [PMID: 23836029 PMCID: PMC3770816 DOI: 10.1007/s00213-013-3195-5] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/17/2013] [Indexed: 12/15/2022]
Abstract
RATIONALE Accumulating evidence indicates that brain kappa-opioid receptors (KORs) and dynorphin, the endogenous ligand that binds at these receptors, are involved in regulating states of motivation and emotion. These findings have stimulated interest in the development of KOR-targeted ligands as therapeutic agents. As one example, it has been suggested that KOR antagonists might have a wide range of indications, including the treatment of depressive, anxiety, and addictive disorders, as well as conditions characterized by co-morbidity of these disorders (e.g., post-traumatic stress disorder) A general effect of reducing the impact of stress may explain how KOR antagonists can have efficacy in such a variety of animal models that would appear to represent different disease states. OBJECTIVE Here, we review evidence that disruption of KOR function attenuates prominent effects of stress. We will describe behavioral and molecular endpoints including those from studies that characterize the effects of KOR antagonists and KOR ablation on the effects of stress itself, as well as on the effects of exogenously delivered corticotropin-releasing factor, a brain peptide that mediates key effects of stress. CONCLUSION Collectively, available data suggest that KOR disruption produces anti-stress effects and under some conditions can prevent the development of stress-induced adaptations. As such, KOR antagonists may have unique potential as therapeutic agents for the treatment and even prevention of stress-related psychiatric illness, a therapeutic niche that is currently unfilled.
Collapse
MESH Headings
- Animals
- Anti-Anxiety Agents/pharmacology
- Anti-Anxiety Agents/therapeutic use
- Anxiety Disorders/drug therapy
- Anxiety Disorders/metabolism
- Anxiety Disorders/psychology
- Behavior, Animal/drug effects
- Brain/drug effects
- Brain/metabolism
- Brain/physiopathology
- Corticotropin-Releasing Hormone/metabolism
- Dynorphins/genetics
- Dynorphins/metabolism
- Humans
- Ligands
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
- Stress, Psychological/psychology
Collapse
Affiliation(s)
- Ashlee Van't Veer
- Department of Psychiatry, Harvard Medical School, McLean Hospital, MRC 217, 115 Mill Street, Belmont, MA, 02478, USA
| | | |
Collapse
|
29
|
CRF mediates the anxiogenic and anti-rewarding, but not the anorectic effects of PACAP. Neuropsychopharmacology 2013; 38:2160-9. [PMID: 23657440 PMCID: PMC3773665 DOI: 10.1038/npp.2013.113] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 05/03/2013] [Accepted: 05/03/2013] [Indexed: 01/23/2023]
Abstract
Anxiety disorders represent the most common mental disturbances in the world, and they are characterized by an abnormal response to stress. Pituitary adenylate cyclase-activating polypeptide (PACAP) and its receptor PAC1 have been proposed to have a key role in mediating the responses to stress as well as the regulation of food intake and body weight. Corticotropin-releasing factor (CRF), the major stress peptide in the brain, has been hypothesized to be involved in PACAP effects, but the reports are conflicting so far. The present study was aimed at further characterizing the behavioral effects of PACAP in rats and at determining the role of central CRF receptors. We found that intracerebroventricular PACAP treatment induced anxiety-like behavior in the elevated plus maze test and elevated intracranial self-stimulation thresholds; both of these effects were fully blocked by concurrent treatment with the CRF receptor antagonist D-Phe-CRF(12-41). Interestingly, the CRF antagonist had no effect on PACAP-induced increased plasma corticosterone, reduction of food intake, and body weight loss. Finally, we found that PACAP increased CRF levels in the paraventricular nucleus of the hypothalamus and, importantly, in the central nucleus of the amygdala, as measured by solid phase radioimmunoassay and quantitative real-time PCR. Our results strengthen the notion that PACAP is a strong mediator of the behavioral response to stress and prove for the first time that this neuropeptide has anti-rewarding (ie, pro-depressant) effects. In addition, we identified the mechanism by which PACAP exerts its anxiogenic and pro-depressant effects, via the recruitment of the central CRF system and independently from HPA axis activation.
Collapse
|
30
|
Réus GZ, Abelaira HM, dos Santos MAB, Carlessi AS, Tomaz DB, Neotti MV, Liranço JLG, Gubert C, Barth M, Kapczinski F, Quevedo J. Ketamine and imipramine in the nucleus accumbens regulate histone deacetylation induced by maternal deprivation and are critical for associated behaviors. Behav Brain Res 2013; 256:451-6. [PMID: 24004850 DOI: 10.1016/j.bbr.2013.08.041] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/20/2013] [Accepted: 08/24/2013] [Indexed: 10/26/2022]
Abstract
Studies indicate that histone deacetylation is important for long term changes related to stress and antidepressant treatment. The present study aimed to evaluate the effects of the classic antidepressant imipramine, and of an antagonist of the N-methyl-d-asparte (NMDA) receptor, ketamine, on behavior and histone deacetylase (HDAC) activity in the brains of maternally deprived adult rats. To this aim, deprived and non-deprived (control) male Wistar rats were divided into the following groups: non-deprived+saline; non-deprived+imipramine (30 mg/kg); non-deprived+ketamine (15 mg/kg); deprived+saline; deprived+imipramine (30 mg/kg); and deprived+ketamine (15 mg/kg). The drugs were administrated once a day for 14 days during their adult phase. Their behavior were then assessed using the forced swimming and open field tests. In addition, the HDAC activity was evaluated in the prefrontal cortex, hippocampus, amygdala and nucleus accumbens using the kit ELISA-sandwich test. In deprived rats treated with saline, we observed an increase in the immobility time, but treatments with imipramine and ketamine were able to reverse this alteration, decreasing the immobility time. Also, there was a decrease on number of crossings with imipramine treatment in non-deprived rats, and an increase on number of crossings with ketamine treatment in deprived rats. The HDAC activity did not alter in the prefrontal cortex, hippocampus and amygdala by deprivation or via treatment with imipramine or ketamine. However, in the nucleus accumbens we observed an increase of HDAC activity in the deprived rats, and interestingly, imipramine and ketamine treatments were able to decrease HDAC activity in this brain area. These findings provide a novel insight into the epigenetic regulation of histone deacetylase in the nucleus accumbens caused by imipramine and ketamine, and indicate that molecular events are necessary to reverse specific stress-induced behavior.
Collapse
Affiliation(s)
- Gislaine Z Réus
- Laboratório de Neurociências, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), and Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Sim HR, Choi TY, Lee HJ, Kang EY, Yoon S, Han PL, Choi SY, Baik JH. Role of dopamine D2 receptors in plasticity of stress-induced addictive behaviours. Nat Commun 2013; 4:1579. [DOI: 10.1038/ncomms2598] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 02/12/2013] [Indexed: 12/14/2022] Open
|
32
|
Wanat MJ, Bonci A, Phillips PEM. CRF acts in the midbrain to attenuate accumbens dopamine release to rewards but not their predictors. Nat Neurosci 2013; 16:383-5. [PMID: 23416448 PMCID: PMC3609940 DOI: 10.1038/nn.3335] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 01/23/2013] [Indexed: 12/13/2022]
Abstract
Stressors impact dopamine-dependent behaviors such as motivation, though the underlying neurobiological mechanism is not well defined. We report that corticotropin-releasing factor (CRF) acts within the ventral tegmental area (VTA) to reduce the motivation to work for food rewards. CRF in the VTA regulates dopamine output in a stimulus- and pathway-specific manner, collectively offering a mechanism by which acute stress selectively regulates information transmission via the VTA to reprioritize motivated behavior.
Collapse
Affiliation(s)
- Matthew J Wanat
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | | | | |
Collapse
|
33
|
Alcaro A, Panksepp J. The SEEKING mind: Primal neuro-affective substrates for appetitive incentive states and their pathological dynamics in addictions and depression. Neurosci Biobehav Rev 2011; 35:1805-20. [DOI: 10.1016/j.neubiorev.2011.03.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 02/28/2011] [Accepted: 03/01/2011] [Indexed: 01/25/2023]
|
34
|
Canbeyli R. Sensorimotor modulation of mood and depression: An integrative review. Behav Brain Res 2010; 207:249-64. [DOI: 10.1016/j.bbr.2009.11.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 10/29/2009] [Accepted: 11/02/2009] [Indexed: 02/05/2023]
|
35
|
Effect of stress on opioid-seeking behavior: evidence from studies with rats. Ann Behav Med 2009; 18:255-63. [PMID: 18425671 DOI: 10.1007/bf02895287] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Studies concerned with the relation between exposure to stress and the behavioral effects of opioid agonists in animal models of drug use are reviewed. These studies, which primarily utilized male rats, indicate that under certain conditions short-term mild stressors increase self-administration of opioid drugs and reinstate herein-seeking behavior following a drug-free period. On the other hand, there is evidence that long-term chronic inescapable stressors and severe acute stressors reduce the reinforcing effects of morphine as measured by a conditioned place preference procedure and decrease the behavioral effects of other positive reinforcers. The results of the studies reviewed suggest that stressors are important modulators of opioid-taking behavior, especially during drug-free periods. The implications of these findings to the understanding of the neurobiology of relapse to opioid-seeking behavior and for strategies for medication development to prevent relapse to heroin are discussed.
Collapse
|
36
|
Elman I, Lowen S, Frederick BB, Chi W, Becerra L, Pitman RK. Functional neuroimaging of reward circuitry responsivity to monetary gains and losses in posttraumatic stress disorder. Biol Psychiatry 2009; 66:1083-90. [PMID: 19640506 PMCID: PMC9446383 DOI: 10.1016/j.biopsych.2009.06.006] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2008] [Revised: 05/22/2009] [Accepted: 06/05/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND Clinical impressions and preclinical work suggest that posttraumatic stress disorder (PTSD) might be associated with dysfunctional reward processing. To pursue this issue, we administered a validated passive-viewing monetary reward task during functional magnetic resonance imaging (fMRI) to subjects with chronic PTSD and to mentally healthy individuals. METHODS The protocol evaluated fMRI signal changes that anticipated or accompanied monetary gains and losses under varying conditions of controlled expectation. The "expectancy phase" entailed presentation of a promising, unpromising, or intermediate Wheel of Fortune-type spinner, whereas the "outcome phase" was defined by the arrow landing on one of three sectors of that spinner, thereby determining the subjects' gain or loss for that trial. RESULTS Neuroimaging data from 20 PTSD and 26 healthy subjects withstood quality control procedures and were included. In voxelwise and anatomically defined region-of-interest analyses, when gains were contrasted to losses, between-group comparison revealed smaller bilateral striatal activations in the PTSD subjects. In the PTSD group, less striatal activation to gains versus losses was associated with more self-reported motivational and social deficits. CONCLUSIONS The present data support the hypothesis that PTSD is associated with abnormal processing of monetary outcomes and that this alteration might be related to some aspects of emotional numbing.
Collapse
Affiliation(s)
- Igor Elman
- Clinical Psychopathology Laboratory, McLean Hospital and Harvard Medical School, Boston, Massachusetts 02478-9106, USA.
| | - Steven Lowen
- Brain Imaging Center, McLean Hospital and Harvard Medical School
| | | | - Won Chi
- Clinical Psychopathology Laboratory, McLean Hospital and Harvard Medical School
| | - Lino Becerra
- Brain Imaging Center, McLean Hospital and Harvard Medical School
| | - Roger K. Pitman
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
37
|
A Bayesian formulation of behavioral control. Cognition 2009; 113:314-328. [DOI: 10.1016/j.cognition.2009.01.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Revised: 01/25/2009] [Accepted: 01/25/2009] [Indexed: 11/19/2022]
|
38
|
Abstract
Persistent symptoms of depression suggest the involvement of stable molecular adaptations in brain, which may be reflected at the level of chromatin remodeling. We find that chronic social defeat stress in mice causes a transient decrease, followed by a persistent increase, in levels of acetylated histone H3 in the nucleus accumbens, an important limbic brain region. This persistent increase in H3 acetylation is associated with decreased levels of histone deacetylase 2 (HDAC2) in the nucleus accumbens. Similar effects were observed in the nucleus accumbens of depressed humans studied postmortem. These changes in H3 acetylation and HDAC2 expression mediate long-lasting positive neuronal adaptations, since infusion of HDAC inhibitors into the nucleus accumbens, which increases histone acetylation, exerts robust antidepressant-like effects in the social defeat paradigm and other behavioral assays. HDAC inhibitor [N-(2-aminophenyl)-4-[N-(pyridine-3-ylmethoxy-carbonyl)aminomethyl]benzamide (MS-275)] infusion also reverses the effects of chronic defeat stress on global patterns of gene expression in the nucleus accumbens, as determined by microarray analysis, with striking similarities to the effects of the standard antidepressant fluoxetine. Stress-regulated genes whose expression is normalized selectively by MS-275 may provide promising targets for the future development of novel antidepressant treatments. Together, these findings provide new insight into the underlying molecular mechanisms of depression and antidepressant action, and support the antidepressant potential of HDAC inhibitors and perhaps other agents that act at the level of chromatin structure.
Collapse
|
39
|
Morris MJ, Na ES, Grippo AJ, Johnson AK. The effects of deoxycorticosterone-induced sodium appetite on hedonic behaviors in the rat. Behav Neurosci 2009; 120:571-9. [PMID: 16768609 DOI: 10.1037/0735-7044.120.3.571] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The authors tested the hypothesis that chronic treatment with a dose of deoxycorticosterone acetate (DOCA) known to elicit a robust sodium appetite can negatively affect the hedonic state of rats. Daily treatment with DOCA with no opportunity to ingest saline produced a rightward shift in the midpoint (effective current 50) of lateral hypothalamic self-stimulation (LHSS) current-response functions and reduced intakes of a palatable sucrose solution. Providing rats with 0.3 M saline during DOCA treatment prevented the rightward shift in LHSS response functions and the decrease in sucrose intake. The authors concluded that a chronic sodium appetite, with no opportunity to attenuate the appetite, can elicit a reduced responsiveness to reward.
Collapse
Affiliation(s)
- Michael J Morris
- Department of Psychology, Pharmacology, and Exercise Science, and Cardiovascular Center, University of Iowa, Iowa City, IA 52242-1407, USA
| | | | | | | |
Collapse
|
40
|
Imipramine treatment and resiliency exhibit similar chromatin regulation in the mouse nucleus accumbens in depression models. J Neurosci 2009; 29:7820-32. [PMID: 19535594 DOI: 10.1523/jneurosci.0932-09.2009] [Citation(s) in RCA: 196] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although it is a widely studied psychiatric syndrome, major depressive disorder remains a poorly understood illness, especially with regard to the disconnect between treatment initiation and the delayed onset of clinical improvement. We have recently validated chronic social defeat stress in mice as a model in which a depression-like phenotype is reversed by chronic, but not acute, antidepressant administration. Here, we use chromatin immunoprecipitation (ChIP)-chip assays--ChIP followed by genome wide promoter array analyses--to study the effects of chronic defeat stress on chromatin regulation in the mouse nucleus accumbens (NAc), a key brain reward region implicated in depression. Our results demonstrate that chronic defeat stress causes widespread and long-lasting changes in gene regulation, including alterations in repressive histone methylation and in phospho-CREB (cAMP response element-binding protein) binding, in the NAc. We then show similarities and differences in this regulation to that observed in another mouse model of depression, prolonged adult social isolation. In the social defeat model, we observed further that many of the stress-induced changes in gene expression are reversed by chronic imipramine treatment, and that resilient mice-those resistant to the deleterious effects of defeat stress-show patterns of chromatin regulation in the NAc that overlap dramatically with those seen with imipramine treatment. These findings provide new insight into the molecular basis of depression-like symptoms and the mechanisms by which antidepressants exert their delayed clinical efficacy. They also raise the novel idea that certain individuals resistant to stress may naturally mount antidepressant-like adaptations in response to chronic stress.
Collapse
|
41
|
LaPlant Q, Chakravarty S, Vialou V, Mukherjee S, Koo JW, Kalahasti G, Bradbury KR, Taylor SV, Maze I, Kumar A, Graham A, Birnbaum SG, Krishnan V, Truong HT, Neve RL, Nestler EJ, Russo SJ. Role of nuclear factor kappaB in ovarian hormone-mediated stress hypersensitivity in female mice. Biol Psychiatry 2009; 65:874-80. [PMID: 19251249 PMCID: PMC2746634 DOI: 10.1016/j.biopsych.2009.01.024] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2008] [Revised: 01/09/2009] [Accepted: 01/14/2009] [Indexed: 01/29/2023]
Abstract
BACKGROUND The molecular mechanisms of stress-induced depressive behaviors have been characterized extensively in male rodents; however, much less is known about female subjects, despite the fact that human depression is far more prevalent in women. METHODS To gain insight into these mechanisms, we performed microarray analysis in nucleus accumbens (NAc), a key brain reward region implicated in depression, in ovariectomized (OVX) and gonadally intact female mice after chronic unpredictable stress and measured stress-induced depression-like behavior in the forced swim test (FST). Male mice were studied in the FST for comparison. RESULTS We find that stress regulation of genes in NAc of gonadally intact female mice is blunted in OVX mice. This pattern of gene regulation is consistent with behavioral findings on the FST: the pro-depression-like effect of stress in intact female mice is absent in OVX female and gonadally intact male mice. We identified, among many genes regulated by stress, several nuclear factor kappaB (NFkappaB) subunits-a pro-survival transcription factor involved in cellular responses to stress-as being highly upregulated in NAc of OVX mice. Given the role of NFkappaB during stress, we hypothesized that upregulation of NFkappaB by OVX decreases susceptibility to stress. Indeed, we show that inhibition of NFkappaB in NAc of OVX animals increases susceptibility to stress-induced depressive behaviors, whereas activation of NFkappaB in NAc of intact female subjects blocks susceptibility. CONCLUSIONS These results suggest a hormonal mechanism of NFkappaB regulation that contributes to stress-induced depressive behaviors in female subjects and might represent a mechanism for gender differences in prevalence rates of these disorders in humans.
Collapse
Affiliation(s)
- Quincey LaPlant
- Fishberg Department of Neuroscience, Mount Sinai School of Medicine, New York, New York 10029-6574, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zafir A, Ara A, Banu N. Invivo antioxidant status: a putative target of antidepressant action. Prog Neuropsychopharmacol Biol Psychiatry 2009; 33:220-8. [PMID: 19059298 DOI: 10.1016/j.pnpbp.2008.11.010] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Revised: 11/06/2008] [Accepted: 11/07/2008] [Indexed: 12/14/2022]
Abstract
Oxidative stress is a critical route of damage in various psychological stress-induced disorders, such as depression. Antidepressants are widely prescribed to treat these conditions; however, few animal studies have investigated the effect of these drugs on endogenous antioxidant status in the brain. The present study employed a 21-day chronic regimen of random exposure to restraint stress to induce oxidative stress in brain, and behavioural aberrations, in rodents. The forced swimming (FST) and sucrose preference tests were used to identify depression-like phenotypes, and reversal in these indices indicated the effectiveness of treatment with fluoxetine (FLU; 20 mg/kg/day, p.o.; selective serotonin reuptake inhibitor), imipramine (IMI; 10 mg/kg/day, p.o.; tricyclic antidepressant) and venlafaxine (VEN; 10 mg/kg/day, p.o.; dual serotonin/norepinephrine reuptake inhibitor) following restraint stress. The antioxidant status was investigated in the brain of these animals. The results evidenced a significant recovery in the activities of superoxide dismutase (SOD), catalase (CAT), glutathione S-transferase (GST), glutathione reductase (GR) and glutathione (GSH) levels by antidepressant treatments following a restraint stress-induced decline of these parameters. The severely accumulated lipid peroxidation product malondialdehyde (MDA) and protein carbonyl contents in stressed animals were significantly normalized by antidepressant treatments. The altered oxidative status is implicated in various aspects of cellular function affecting the brain. Thus, it is possible that augmentation of in vivo antioxidant defenses could serve as a convergence point for multiple classes of antidepressants as an important mechanism underlying the neuroprotective pharmacological effects of these drugs observed clinically in the treatment of various stress disorders. Consequently, pharmacological modulation of stress-induced oxidative damage as a possible stress-management approach should be an important avenue of further research.
Collapse
Affiliation(s)
- Ayesha Zafir
- Department of Biochemistry, Faculty of Life Sciences, A. M. University, Aligarh 202 002, U.P., India
| | | | | |
Collapse
|
43
|
Smith KL, Jessop DS, Finn DP. Modulation of stress by imidazoline binding sites: implications for psychiatric disorders. Stress 2009; 12:97-114. [PMID: 19006007 DOI: 10.1080/10253890802302908] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
In this review, we present evidence for the involvement of imidazoline binding sites (IBS) in modulating responses to stress, through central control of monoaminergic and hypothalamo-pituitary-adrenal (HPA) axis activity. Pharmacological and physiological evidence is presented for differential effects of different IBS subtypes on serotoninergic and catecholaminergic pathways involved in control of basal and stress-stimulated HPA axis activity. IBS ligands can modulate behavioural and neuroendocrine responses in animal models of stress, depression and anxiety, and a body of evidence exists for alterations in central IBS expression in psychiatric patients, which can be normalised partially or fully by treatment with antidepressants. Dysfunction in monoaminergic systems and the HPA axis under basal and stress-induced activation has been extensively reported in psychiatric illnesses. On the basis of the literature, we suggest a potential therapeutic role for selective IBS ligands in the treatment of depression and anxiety disorders.
Collapse
Affiliation(s)
- Karen L Smith
- Department of Pharmacology and Therapeutics, NCBES Neuroscience Cluster, National University of Ireland, Galway, Ireland
| | | | | |
Collapse
|
44
|
Armario A, Escorihuela RM, Nadal R. Long-term neuroendocrine and behavioural effects of a single exposure to stress in adult animals. Neurosci Biobehav Rev 2008; 32:1121-35. [PMID: 18514314 DOI: 10.1016/j.neubiorev.2008.04.003] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 04/12/2008] [Accepted: 04/14/2008] [Indexed: 11/30/2022]
Abstract
There is now considerable evidence for long-lasting sequels of stress. A single exposure to high intensity predominantly emotional stressors such as immobilisation in wooden-boards (IMO) induces long-term (days to weeks) desensitization of the hypothalamic-pituitary-adrenal (HPA) response to the same (homotypic) stressor, whereas the response to novel (heterotypic) stressors was enhanced. In addition, long-lasting changes in behaviour have been described after a single exposure to brief or more prolonged sessions of shocks, predator, predator odour, underwater stress or a combination of three stressors on 1 day. The most consistent changes are reduced entries into the open arms of the elevated plus-maze and enhanced acoustic startle response, both reflecting enhanced anxiety. However, it is unclear whether there is any relationship between the intensity of the stressors, as evaluated by the main physiological indexes of stress (e.g. HPA axis), the putative traumatic experience they represent and their long-term behavioural consequences. This is particularly critical when trying to model post-traumatic stress disorders (PTSD), which demands a great effort to validate such putative models.
Collapse
Affiliation(s)
- Antonio Armario
- Institut de Neurociències, Universitat Autonòma de Barcelona, 08193 Bellaterra, Barcelona, Spain.
| | | | | |
Collapse
|
45
|
Roy V, Merali Z, Poulter MO, Anisman H. Anxiety responses, plasma corticosterone and central monoamine variations elicited by stressors in reactive and nonreactive mice and their reciprocal F1 hybrids. Behav Brain Res 2007; 185:49-58. [PMID: 17692933 DOI: 10.1016/j.bbr.2007.07.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 07/06/2007] [Accepted: 07/09/2007] [Indexed: 11/25/2022]
Abstract
Stressor-provoked anxiety, plasma corticosterone, and variations of brain monoamine turnover are influenced by genetic factors, but may also be moderated by early life experiences. To evaluate the contribution of maternal influences, behavioral and neurochemical stress responses were assessed in strains of mice that were either stressor-reactive or -resilient (BALB/cByJ and C57BL/6ByJ, respectively) as well as in their reciprocal F(1) hybrids. BALB/cByJ mice demonstrated poorer maternal behaviors than did C57BL/6ByJ dams, irrespective of the pups being raised (inbred or F(1) hybrids). The BALB/cByJ mice appeared more anxious than C57BL/6ByJ mice, exhibiting greater reluctance to step-down from a platform and a greater startle response. Although the F(1) behavior generally resembled that of the C57BL/6ByJ parent strain, in the step-down test the influence of maternal factors were initially evident among the F(1) mice (particularly males) with a BALB/cByJ dam. However, over trials the C57BL/6ByJ-like behavior came to predominate. BALB/cByJ mice also exhibited greater plasma corticosterone elevations, 5-HT utilization in the central amygdala (CeA), and greater NE turnover in the paraventricular nucleus of the hypothalamus (PVN). Interestingly, among the F(1)'s corticosterone and 5-HIAA in the CeA resembled that of the BALB/cByJ parent strain, whereas MHPG accumulation in the PVN was more like that of C57BL/6ByJ mice. It seems that, to some extent, maternal factors influenced anxiety responses in the hybrids, but did not influence the corticosterone or the monoamine variations. The inheritance profiles suggest that anxiety was unrelated to either the corticosterone or monoamine changes.
Collapse
Affiliation(s)
- V Roy
- UPRES PSY.CO EA 1780, Faculté des Sciences, Université de Rouen, Mont Saint Aignan, France
| | | | | | | |
Collapse
|
46
|
Perrine SA, Sheikh IS, Nwaneshiudu CA, Schroeder JA, Unterwald EM. Withdrawal from chronic administration of cocaine decreases delta opioid receptor signaling and increases anxiety- and depression-like behaviors in the rat. Neuropharmacology 2007; 54:355-64. [PMID: 18045627 DOI: 10.1016/j.neuropharm.2007.10.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Revised: 10/10/2007] [Accepted: 10/11/2007] [Indexed: 10/22/2022]
Abstract
Chronic administration of cocaine has been shown to attenuate the functional capacity of delta opioid receptors to inhibit adenylyl cyclase activity. Abuse and withdrawal from cocaine in humans is associated with increases in anxiety and depression. Since recent research supports the role of delta opioid receptors in anxiety- and depression-like behaviors in rodents, we hypothesized that functional desensitization of delta opioid receptors contributes to anxiety- and depression-like behavioral phenotypes following short-term withdrawal from chronic administration of cocaine. To test this hypothesis, delta opioid receptor signaling and behaviors were evaluated 24h after 14days of binge-pattern cocaine administration (15mg/kg three times daily at 1h intervals) in male Sprague-Dawley rats. Results showed that the inhibition of adenylyl cyclase by delta opioid receptor agonists was attenuated in the frontal cortex, nucleus accumbens and caudate putamen 24h after cessation of cocaine administration. One day withdrawal from chronic administration of cocaine resulted in increased anxiety- and depression-like behaviors as measured by the elevated plus maze and the forced swim test respectively, and no change in locomotor activity. The anxiety- and depression-like behaviors were dose-dependently reduced by acute administration of the selective delta opioid receptor agonist, SNC80. These results demonstrate that early withdrawal from cocaine resulted in increased anxiety and depression, which accompanies the desensitization of delta opioid receptor function. Furthermore, cocaine-induced anxiety- and depression-like behaviors were reversible by the delta opioid receptor agonist SNC80.
Collapse
Affiliation(s)
- Shane A Perrine
- Temple University School of Medicine, Department of Pharmacology, Center for Substance Abuse Research, 3420 N. Broad St., Philadelphia, PA 19140, USA.
| | | | | | | | | |
Collapse
|
47
|
Vargas-Perez H, Ting-A-Kee RA, Heinmiller A, Sturgess JE, van der Kooy D. A test of the opponent-process theory of motivation using lesions that selectively block morphine reward. Eur J Neurosci 2007; 25:3713-8. [PMID: 17610590 DOI: 10.1111/j.1460-9568.2007.05599.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The opponent-process theory of motivation postulates that motivational stimuli activate a rewarding process that is followed by an opposed aversive process in a homeostatic control mechanism. Thus, an acute injection of morphine in nondependent animals should evoke an acute rewarding response, followed by a later aversive response. Indeed, the tegmental pedunculopontine nucleus (TPP) mediates the rewarding effects of opiates in previously morphine-naive animals, but not other unconditioned effects of opiates, or learning ability. The aversive opponent process for acute morphine reward was revealed using a place-conditioning paradigm. The conditioned place aversion induced by 16-h spontaneous morphine withdrawal from an acute morphine injection in nondependent rats was abolished by TPP lesions performed prior to drug experience. However, TPP-lesioned rats did show conditioned aversions for an environment paired with the acute administration of the opioid antagonist naloxone, which blocks endogenous opioids. The results show that blocking the rewarding effects of morphine with TPP lesions also blocked the opponent aversive effects of acute morphine withdrawal in nondependent animals. Thus, this spontaneous withdrawal aversion (the opponent process) is induced by the acute rewarding effects of morphine and not by other unconditioned effects of morphine, the pharmacological effects of morphine or endogenous opioids being displaced from opiate receptors.
Collapse
Affiliation(s)
- Hector Vargas-Perez
- Department of Medical Genetics and Microbiology, University of Toronto, Canada.
| | | | | | | | | |
Collapse
|
48
|
Tools to measure and improve animal welfare: reward-related behaviour. Anim Welf 2007. [DOI: 10.1017/s0962728600031742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
AbstractThere is an increasing requirement for tools to assess and improve animal welfare in an objective and scientifically based manner. In our research a concept of welfare is applied which states that welfare is determined by the balance between positive and negative experiences. This concept implies that an interaction exists between stress systems and reward systems in the brain and, as a consequence: (I) negative experiences induce an increased sensitivity (ie need) for positive experiences; and (II) negative experiences can be compensated for by positive experiences. On this basis, two uses of reward-related behaviour can be hypothesised: (I) reward sensitivity may be used as a tool to assess the state of an animal in terms of welfare because it can indicate the current state of the balance that is dependent on previous (stressful) experiences; and (II) regular presentation of rewards may serve as a tool to counteract stress by shifting the balance to the positive side and, thus, to improve welfare. In order to investigate this, we used the rat as a model. Reward sensitivity was determined by the spontaneous behavioural response shown during expectation of a reward (ie anticipatory behaviour). A third (III) use of reward-related behaviour derives from the fact that anticipatory behaviour is influenced by the (rewarding) properties of the forthcoming reward (or other event) and, thus, may serve as a tool to assess the animal's perception of this reward/event. This paper presents a descriptive overview of the evidence obtained thus far for the three proposed uses of reward-related behaviour. The biological background of our concept of welfare can be generalised to all (vertebrate) species, and anticipatory behaviour can be evoked in a wide range of other species. Therefore, this tool for measuring and improving the welfare of captive animals has great potential and will contribute to a good quality of life for captive animals.
Collapse
|
49
|
Corticotropin releasing hormone receptor alterations elicited by acute and chronic unpredictable stressor challenges in stressor-susceptible and resilient strains of mice. Behav Brain Res 2007; 181:180-90. [PMID: 17517441 DOI: 10.1016/j.bbr.2007.04.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Revised: 04/05/2007] [Accepted: 04/07/2007] [Indexed: 11/28/2022]
Abstract
Stressors increase corticotropin releasing hormone (CRH) functioning in hypothalamic and frontal cortical brain regions, and thus may contribute to the provocation of anxiety and depressive disorder. As the effects of stressors on these behavioral changes are more pronounced in some strains of mice (e.g., BALB/cByJ) than in others (e.g., C57BL/6ByJ), the present investigation assessed whether acute and chronic stressors would differentially influence CRH receptor immunoreactivity (ir-CRHr) and CRH receptor mRNA expression (CRH(1) and CRH(2)) in the orbital frontal cortex (OFC) of these strains. An acute noise stressor, and to a greater extent a chronic, variable stressor regimen reduced ir-CRHr in BALB/cByJ mice. In contrast, in the hardier C57BL/6ByJ mice the acute stressor increased ir-CRHr in portions of the OFC, whereas a chronic stressor tended to reduce ir-CRHr. However, whereas the acute stressor did not influence CRH(1) mRNA expression, the chronic stressor increased CRH(1) mRNA expression in both mouse strains. The CRH(2) expression appeared in low abundance in both strains and was unaltered by the stressor. In addition to the OFC variations, quantitative immunohistochemistry indicated that the chronic stressor treatment increased CRH immunoreactivity in the median eminence of C57BL/6ByJ mice, but co-expression of CRH and arginine vasopressin (AVP) immunoreactivity was not provoked by the stressors. The data support the view that stressors provoke marked variations of ir-CRHr in the OFC that might contribute to the differential anxiety/depression-like profiles ordinarily apparent in the stressor-vulnerable and -resilient mouse strains.
Collapse
|
50
|
Millan MJ. Multi-target strategies for the improved treatment of depressive states: Conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacol Ther 2006; 110:135-370. [PMID: 16522330 DOI: 10.1016/j.pharmthera.2005.11.006] [Citation(s) in RCA: 389] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 11/28/2005] [Indexed: 12/20/2022]
Abstract
Major depression is a debilitating and recurrent disorder with a substantial lifetime risk and a high social cost. Depressed patients generally display co-morbid symptoms, and depression frequently accompanies other serious disorders. Currently available drugs display limited efficacy and a pronounced delay to onset of action, and all provoke distressing side effects. Cloning of the human genome has fuelled expectations that symptomatic treatment may soon become more rapid and effective, and that depressive states may ultimately be "prevented" or "cured". In pursuing these objectives, in particular for genome-derived, non-monoaminergic targets, "specificity" of drug actions is often emphasized. That is, priority is afforded to agents that interact exclusively with a single site hypothesized as critically involved in the pathogenesis and/or control of depression. Certain highly selective drugs may prove effective, and they remain indispensable in the experimental (and clinical) evaluation of the significance of novel mechanisms. However, by analogy to other multifactorial disorders, "multi-target" agents may be better adapted to the improved treatment of depressive states. Support for this contention is garnered from a broad palette of observations, ranging from mechanisms of action of adjunctive drug combinations and electroconvulsive therapy to "network theory" analysis of the etiology and management of depressive states. The review also outlines opportunities to be exploited, and challenges to be addressed, in the discovery and characterization of drugs recognizing multiple targets. Finally, a diversity of multi-target strategies is proposed for the more efficacious and rapid control of core and co-morbid symptoms of depression, together with improved tolerance relative to currently available agents.
Collapse
Affiliation(s)
- Mark J Millan
- Institut de Recherches Servier, Centre de Recherches de Croissy, Psychopharmacology Department, 125, Chemin de Ronde, 78290-Croissy/Seine, France.
| |
Collapse
|