1
|
Potokiri A, Omeiza NA, Ajayi AM, Adeleke PA, Alagbonsi AI, Iwalewa EO. Yeast supplementation potentiates fluoxetine's anti-depressant effect in mice via modulation of oxido-inflammatory, CREB, and MAPK signaling pathways. Curr Res Physiol 2024; 7:100132. [PMID: 39483857 PMCID: PMC11526068 DOI: 10.1016/j.crphys.2024.100132] [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: 08/11/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction The therapeutic potential of yeast in the management of depression is unknown. Thus, we evaluated the modulatory effect of nutritional yeast supplementation on antidepressant activity of fluoxetine in mice models of depressive-like behaviors (DLB). Methods A total of 112 mice were divided into 16 groups (n = 7 each) for a 3-stage study. Stage I (non-DLB study) had groups Ia (10 mL/kg vehicle), Ib (20 mg/kg fluoxetine), Ic - If (2% yeast diet for all, but Id - If additionally received 5 mg/kg, 10 mg/kg, and 20 mg/kg fluoxetine respectively). Stage II (lipopolysaccharide [LPS] model of DLB) had groups IIa - IIb (10 mL/kg vehicle), IIc (20 mg/kg fluoxetine), IId (yeast) and IIe (yeast + 20 mg/kg fluoxetine). After these treatments for 24 days, animals in IIb - IIe received 0.83 mg/kg of LPS on the 25th day. Except for group IIIa (10 mL/kg vehicle), animals in other groups of stage III (unpredictable chronic mild stress [UCMS] model) were exposed to UCMS for 24 days along with 10 mL/kg vehicle (IIIb), 20 mg/kg fluoxetine (IIIc), yeast (IIId), or yeast + fluoxetine (IIIe). Results Yeast and fluoxetine attenuated LPS- and UCMS-induced immobility, derangement of oxido-inflammatory (TNF-α, IL-6, NO, MDA, SOD, GSH, CAT, and AChE) and CREB/MAPK pathways. While fluoxetine had more potent effect than yeast when used separately, pre-treatment of mice with their combination had more pronounced effect than either of them. Conclusion Yeast supplementation improves the antidepressant activity of fluoxetine in mice by modulating oxido-inflammatory, CREB, and MAPK pathways.
Collapse
Affiliation(s)
- Augustina Potokiri
- Department of Pharmacology and Therapeutics, Neuropharmacology and Toxicology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Noah A. Omeiza
- Department of Pharmacology and Therapeutics, Neuropharmacology and Toxicology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, Academia Sinica, Taipei, Taiwan
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Abayomi M. Ajayi
- Department of Pharmacology and Therapeutics, Neuropharmacology and Toxicology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Paul A. Adeleke
- Department of Pharmacology and Therapeutics, Neuropharmacology and Toxicology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Abdullateef I. Alagbonsi
- Department of Physiology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye, Southern Province, Rwanda
| | - Ezekiel O. Iwalewa
- Department of Pharmacology and Therapeutics, Neuropharmacology and Toxicology Unit, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
2
|
Kamenish K, Robinson ESJ. Neuropsychological Effects of Antidepressants: Translational Studies. Curr Top Behav Neurosci 2024; 66:101-130. [PMID: 37955824 DOI: 10.1007/7854_2023_446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Pharmacological treatments that improve mood were first identified serendipitously, but more than half a century later, how these drugs induce their antidepressant effects remains largely unknown. With the help of animal models, a detailed understanding of their pharmacological targets and acute and chronic effects on brain chemistry and neuronal function has been achieved, but it remains to be elucidated how these effects translate to clinical efficacy. Whilst the field has been dominated by the monoamine and neurotrophic hypotheses, the idea that the maladaptive cognitive process plays a critical role in the development and perpetuation of mood disorders has been discussed since the 1950s. Recently, studies using objective methods to quantify changes in emotional processing found acute effects with conventional antidepressants in both healthy volunteers and patients. These positive effects on emotional processing and cognition occur without a change in the subjective ratings of mood. Building from these studies, behavioural methods for animals that quantify similar cognitive affective processes have been developed. Integrating these behavioural approaches with pharmacology and targeted brain manipulations, a picture is beginning to emerge of the underlying mechanisms that may link the pharmacology of antidepressants, these neuropsychological constructs and clinical efficacy. In this chapter, we discuss findings from animal studies, experimental medicine and patients investigating the neuropsychological effects of antidepressant drugs. We discuss the possible neural circuits that contribute to these effects and discuss whether a neuropsychological model of antidepressant effects could explain the temporal differences in clinical benefits observed with conventional delayed-onset antidepressants versus rapid-acting antidepressants.
Collapse
Affiliation(s)
- Katie Kamenish
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK
| | - Emma S J Robinson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, UK.
| |
Collapse
|
3
|
Bian LH, Wang SQ, Li WJ, Li J, Yin Y, Ye FF, Guo JY. Cryptotanshinone regulates gut microbiota and PI3K-AKT pathway in rats to alleviate CUMS induced depressive symptoms. Biomed Pharmacother 2023; 169:115921. [PMID: 38011787 DOI: 10.1016/j.biopha.2023.115921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/09/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
Cryptotanshinone (CPT), a bioactive compound derived from the traditional Chinese herb Salvia miltiorrhiza, exhibits promising antidepressant properties. Employing a rat model subjected to Chronic Unpredictable Mild Stress (CUMS), behavioral analyses (open field experiment, elevated cross maze experiment, sugar water preference experiment, forced swimming experiment) and inflammatory factor assessments were conducted to assess the efficacy of CPT in alleviating depressive symptoms and inflammatory responses induced by CUMS. Moreover, 16 S rDNA analysis revealed alterations in the gut microbiota of rats exposed to both CUMS and CPT administration. Notably, CPT administration was found to mitigate harmful bacterial shifts associated with depression. Preliminary exploration of the molecular mechanism underlying CPT's antidepressant effects via transcriptomics analysis and molecular docking indicated that CPT might exert its influence by regulating the PI3K-AKT pathway. This study sheds light on the potential therapeutic role of CPT in managing depressive disorders, offering a comprehensive understanding of its impact on behavior, inflammation, gut microbiota, and molecular pathways.
Collapse
Affiliation(s)
- Li-Hua Bian
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Si-Qi Wang
- University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 4A DatunRoad, Chaoyang District, Beijing 100101, China
| | - Wen-Jing Li
- University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 4A DatunRoad, Chaoyang District, Beijing 100101, China
| | - Jie Li
- University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 4A DatunRoad, Chaoyang District, Beijing 100101, China
| | - Yi Yin
- University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 4A DatunRoad, Chaoyang District, Beijing 100101, China
| | - Fang-Fu Ye
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian-You Guo
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 4A DatunRoad, Chaoyang District, Beijing 100101, China.
| |
Collapse
|
4
|
Marques KL, Moreira ML, Thiele MC, Cunha-Rodrigues MC, Barradas PC. Depressive-like behavior and impaired synaptic plasticity in the prefrontal cortex as later consequences of prenatal hypoxic-ischemic insult in rats. Behav Brain Res 2023; 452:114571. [PMID: 37421988 DOI: 10.1016/j.bbr.2023.114571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Perinatal hypoxia-ischemia (HI) is a leading cause of morbidity and mortality among newborns. Infants with HI encephalopathy may experience lasting consequences, such as depression, in adulthood. In this study, we examined depressive-like behavior, neuronal population, and markers of monoaminergic and synaptic plasticity in the prefrontal cortex (PFC) of adolescent rats subjected to a prenatal HI model. Pregnant rats underwent a surgery in which uterine and ovarian blood flow was blocked for 45 min at E18 (HI procedure). Sham-operated subjects were also generated (SH procedure). Behavioral tests were conducted on male and female pups from P41 to P43, and animals were histologically processed or dissected for western blotting at P45. We found that the HI groups consumed less sucrose in the sucrose preference test and remained immobile for longer periods in the forced swim test. Additionally, we observed a significant reduction in neuronal density and PSD95 levels in the HI group, as well as a smaller number of synaptophysin-positive cells. Our results underscore the importance of this model in investigating the effects of HI-induced injuries, as it reproduces an increase in depressive-like behavior and suggests that the HI insult affects circuits involved in mood modulation.
Collapse
Affiliation(s)
- Kethely L Marques
- Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Milena L Moreira
- Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Maria C Thiele
- Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marta C Cunha-Rodrigues
- Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Penha C Barradas
- Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
5
|
Batsukh S, Oh S, Rheu K, Lee BJ, Choi CH, Son KH, Byun K. Rice Germ Attenuates Chronic Unpredictable Mild Stress-Induced Muscle Atrophy. Nutrients 2023; 15:2719. [PMID: 37375622 DOI: 10.3390/nu15122719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic stress leads to hypothalamic-pituitary-adrenal axis dysfunction, increasing cortisol levels. Glucocorticoids (GCs) promote muscle degradation and inhibit muscle synthesis, eventually causing muscle atrophy. In this study, we aimed to evaluate whether rice germ supplemented with 30% γ-aminobutyric acid (RG) attenuates muscle atrophy in an animal model of chronic unpredictable mild stress (CUMS). We observed that CUMS raised the adrenal gland weight and serum adrenocorticotropic hormone (ACTH) and cortisol levels, and these effects were reversed by RG. CUMS also enhanced the expression of the GC receptor (GR) and GC-GR binding in the gastrocnemius muscle, which were attenuated by RG. The expression levels of muscle degradation-related signaling pathways, such as the Klf15, Redd-1, FoxO3a, Atrogin-1, and MuRF1 pathways, were enhanced by CUMS and attenuated by RG. Muscle synthesis-related signaling pathways, such as the IGF-1/AKT/mTOR/s6k/4E-BP1 pathway, were reduced by CUMS and enhanced by RG. Moreover, CUMS raised oxidative stress by enhancing the levels of iNOS and acetylated p53, which are involved in cell cycle arrest, whereas RG attenuated both iNOS and acetylated p53 levels. Cell proliferation in the gastrocnemius muscle was reduced by CUMS and enhanced by RG. The muscle weight, muscle fiber cross-sectional area, and grip strength were reduced by CUMS and enhanced by RG. Therefore, RG attenuated ACTH levels and cortisol-related muscle atrophy in CUMS animals.
Collapse
Affiliation(s)
- Sosorburam Batsukh
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kyoungmin Rheu
- Marine Bioprocess Co., Ltd., Smart Marine BioCenter, Busan 46048, Republic of Korea
| | - Bae-Jin Lee
- Marine Bioprocess Co., Ltd., Smart Marine BioCenter, Busan 46048, Republic of Korea
| | - Chang Hu Choi
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
6
|
Rice Germ Ameliorated Chronic Unpredictable Mild Stress-Induced Depressive-like Behavior by Reducing Neuroinflammation. Nutrients 2022; 14:nu14245382. [PMID: 36558541 PMCID: PMC9780988 DOI: 10.3390/nu14245382] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Stress-induced neuroinflammation is widely regarded as one of the primary causes of depression. Gamma-aminobutyric acid (GABA)-enriched foods relieve stress and reduce inflammatory reactions. This study aimed to evaluate whether rice germ with 30% GABA (RG) reduced neuroinflammation in mice exposed to chronic unpredictable mild stress (CUMS). CUMS mice were administered 40, 90, and 140 mg/kg of RG. CUMS increased serum and hypothalamic pro-inflammatory cytokine (TNF-α and IL-6) levels, which were decreased by RG. In the hypothalamus, CUMS elevated M1-type microglia markers of CD86 and NF-κB, whereas RG lowered these levels. The expression levels of NLRP3 inflammasome complex (NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1), IL-1β, and IL-18 were increased in the hypothalamus of CUMS mice and decreased by RG. RG attenuated depressive-like behaviors in CUMS mice, as measured by the forced swim test and tail suspension test. In conclusion, RG decreased hypothalamic inflammation-related signals, such as TNF-α, IL-6, M1 polarization, NF-κB, NLRP3 inflammasome complex, caspase-1, IL-1β, and IL-18, to diminish depressive-like behavior.
Collapse
|
7
|
Sucrose Preference Test as a Measure of Anhedonic Behavior in a Chronic Unpredictable Mild Stress Model of Depression: Outstanding Issues. Brain Sci 2022; 12:brainsci12101287. [PMID: 36291221 PMCID: PMC9599556 DOI: 10.3390/brainsci12101287] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 12/20/2022] Open
Abstract
Despite numerous studies on the neurobiology of depression, the etiological and pathophysiological mechanisms of this disorder remain poorly understood. A large number of animal models and tests to evaluate depressive-like behavior have been developed. Chronic unpredictable mild stress (CUMS) is the most common and frequently used model of depression, and the sucrose preference test (SPT) is one of the most common tests for assessing anhedonia. However, not all laboratories can reproduce the main effects of CUMS, especially when this refers to a decrease in sucrose preference. It is also unknown how the state of anhedonia, assessed by the SPT, relates to the state of anhedonia in patients with depression. We analyzed the literature available in the PubMed database using keywords relevant to the topic of this narrative review. We hypothesize that the poor reproducibility of the CUMS model may be due to differences in sucrose consumption, which may be influenced by such factors as differences in sucrose preference concentration threshold, water and food deprivation, and differences in animals’ susceptibility to stress. We also believe that comparisons between animal and human states of anhedonia should be made with caution because there are many inconsistencies between the two, including in assessment methods. We also tried to offer some recommendations that should improve the reproducibility of the CUMS model and provide a framework for future research.
Collapse
|
8
|
Lu X, Qi C, Zheng J, Sun M, Jin L, Sun J. The Antidepressant Effect of Deoiled Sunflower Seeds on Chronic Unpredictable Mild Stress in Mice Through Regulation of Microbiota–Gut–Brain Axis. Front Nutr 2022; 9:908297. [PMID: 35859751 PMCID: PMC9289741 DOI: 10.3389/fnut.2022.908297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/10/2022] [Indexed: 11/30/2022] Open
Abstract
Objectives Sunflower seeds provide tryptophan-rich proteins with the potential to protect against depression. Tryptophan is a precursor of serotonin and a substrate for the production of indole derivatives by gut microbiota. This study aimed to investigate the association between the depression-alleviating effects of deoiled and dechlorogenic sunflower seeds (DSFS) and regulation of gut microbiota. Materials and Methods Male C57BL/6J mice were fed a diet comprising a source of soy protein (normal and model control), DSFS or whey protein concentrate (positive control) for 7 weeks, and chronic stress-induced depression was induced. Results Feeding the DSFS diet prevented depression-like behaviors, intestinal barrier damage, elevated plasma corticosterone, and reduced hippocampal serotonin levels in mice. Meanwhile, Feeding the DSFS diet significantly altered the gut microbiota structure, characterized by elevated relative abundances of Ileibacterium valens, Ruminococcus flavefaciens, Clostridium scindens, and Olsenella massiliensis, which were inversely associated with depressive behaviors and markers of mucosal barrier damage. DSFS also altered the gut metabolite profile, prevented depression-induced gut L-tryptophan depletion, and upregulated its metabolite indoleacetaldehyde. Conclusion Feeding the DSFS diet prevented depression in mice by remodeling the gut microbiota and bacterial tryptophan metabolism.
Collapse
Affiliation(s)
- Xiaomeng Lu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
- National R&D Center for Nuts Processing Technology, Qiaqia Food Co., Ltd., Hefei, China
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Jie Zheng
- National R&D Center for Nuts Processing Technology, Qiaqia Food Co., Ltd., Hefei, China
| | - Mei Sun
- National R&D Center for Nuts Processing Technology, Qiaqia Food Co., Ltd., Hefei, China
| | - Long Jin
- National R&D Center for Nuts Processing Technology, Qiaqia Food Co., Ltd., Hefei, China
- *Correspondence: Long Jin,
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
- Jin Sun,
| |
Collapse
|
9
|
Pizzagalli DA, Roberts AC. Prefrontal cortex and depression. Neuropsychopharmacology 2022; 47:225-246. [PMID: 34341498 PMCID: PMC8617037 DOI: 10.1038/s41386-021-01101-7] [Citation(s) in RCA: 236] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 01/03/2023]
Abstract
The prefrontal cortex (PFC) has emerged as one of the regions most consistently impaired in major depressive disorder (MDD). Although functional and structural PFC abnormalities have been reported in both individuals with current MDD as well as those at increased vulnerability to MDD, this information has not translated into better treatment and prevention strategies. Here, we argue that dissecting depressive phenotypes into biologically more tractable dimensions - negative processing biases, anhedonia, despair-like behavior (learned helplessness) - affords unique opportunities for integrating clinical findings with mechanistic evidence emerging from preclinical models relevant to depression, and thereby promises to improve our understanding of MDD. To this end, we review and integrate clinical and preclinical literature pertinent to these core phenotypes, while emphasizing a systems-level approach, treatment effects, and whether specific PFC abnormalities are causes or consequences of MDD. In addition, we discuss several key issues linked to cross-species translation, including functional brain homology across species, the importance of dissecting neural pathways underlying specific functional domains that can be fruitfully probed across species, and the experimental approaches that best ensure translatability. Future directions and clinical implications of this burgeoning literature are discussed.
Collapse
Affiliation(s)
- Diego A Pizzagalli
- Department of Psychiatry, Harvard Medical School & McLean Hospital, Belmont, MA, USA.
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
10
|
Borbély É, Simon M, Fuchs E, Wiborg O, Czéh B, Helyes Z. Novel drug developmental strategies for treatment-resistant depression. Br J Pharmacol 2021; 179:1146-1186. [PMID: 34822719 PMCID: PMC9303797 DOI: 10.1111/bph.15753] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/17/2021] [Accepted: 11/14/2021] [Indexed: 11/30/2022] Open
Abstract
Major depressive disorder is a leading cause of disability worldwide. Because conventional therapies are ineffective in many patients, novel strategies are needed to overcome treatment‐resistant depression (TRD). Limiting factors of successful drug development in the last decades were the lack of (1) knowledge of pathophysiology, (2) translational animal models and (3) objective diagnostic biomarkers. Here, we review novel drug targets and drug candidates currently investigated in Phase I–III clinical trials. The most promising approaches are inhibition of glutamatergic neurotransmission by NMDA and mGlu5 receptor antagonists, modulation of the opioidergic system by κ receptor antagonists, and hallucinogenic tryptamine derivates. The only registered drug for TRD is the NMDA receptor antagonist, S‐ketamine, but add‐on therapies with second‐generation antipsychotics, certain nutritive, anti‐inflammatory and neuroprotective agents seem to be effective. Currently, there is an intense research focus on large‐scale, high‐throughput omics and neuroimaging studies. These results might provide new insights into molecular mechanisms and potential novel therapeutic strategies.
Collapse
Affiliation(s)
- Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Hungary.,Molecular Pharmacology Research Group, Szentágothai János Research Centre, University of Pécs, Pécs, Hungary
| | - Mária Simon
- Department of Psychiatry and Psychotherapy, Clinical Centre, Medical School, University of Pécs, Hungary
| | - Eberhard Fuchs
- German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai János Research Centre, University of Pécs, Pécs, Hungary.,Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Hungary.,Molecular Pharmacology Research Group, Szentágothai János Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
11
|
Kopra E, Mondelli V, Pariante C, Nikkheslat N. Ketamine's effect on inflammation and kynurenine pathway in depression: A systematic review. J Psychopharmacol 2021; 35:934-945. [PMID: 34180293 PMCID: PMC8358579 DOI: 10.1177/02698811211026426] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ketamine is a novel rapid-acting antidepressant with high efficacy in treatment-resistant patients. Its exact therapeutic mechanisms of action are unclear; however, in recent years its anti-inflammatory properties and subsequent downstream effects on tryptophan (TRP) metabolism have sparked research interest. AIM This systematic review examined the effect of ketamine on inflammatory markers and TRP-kynurenine (KYN) pathway metabolites in patients with unipolar and bipolar depression and in animal models of depression. METHODS MEDLINE, Embase, and PsycINFO databases were searched on October 2020 (1806 to 2020). RESULTS Out of 807 initial results, nine human studies and 22 animal studies on rodents met the inclusion criteria. Rodent studies provided strong support for ketamine-induced decreases in pro-inflammatory cytokines, namely in interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α and indicated anti-inflammatory effects on TRP metabolism, including decreases in the enzyme indoleamine 2,3-dioxygenase (IDO). Clinical evidence was less robust with high heterogeneity between sample characteristics, but most experiments demonstrated decreases in peripheral inflammation including in IL-1β, IL-6, and TNF-α. Preliminary support was also found for reduced activation of the neurotoxic arm of the KYN pathway. CONCLUSION Ketamine appears to induce anti-inflammatory effects in at least a proportion of depressed patients. Suggestions for future research include investigation of markers in the central nervous system and examination of clinical relevance of inflammatory changes.
Collapse
Affiliation(s)
- Emma Kopra
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Carmine Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Naghmeh Nikkheslat
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| |
Collapse
|
12
|
Abreu L, Koebach A, Díaz O, Carleial S, Hoeffler A, Stojetz W, Freudenreich H, Justino P, Brück T. Life With Corona: Increased Gender Differences in Aggression and Depression Symptoms Due to the COVID-19 Pandemic Burden in Germany. Front Psychol 2021; 12:689396. [PMID: 34385959 PMCID: PMC8353131 DOI: 10.3389/fpsyg.2021.689396] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/11/2021] [Indexed: 12/30/2022] Open
Abstract
Gender differences (GD) in mental health have come under renewed scrutiny during the COVID-19 pandemic. While rapidly emerging evidence indicates a deterioration of mental health in general, it remains unknown whether the pandemic will have an impact on GD in mental health. To this end, we investigate the association of the pandemic and its countermeasures affecting everyday life, labor, and households with changes in GD in aggression, anxiety, depression, and the somatic symptom burden. We analyze cross-sectional data from 10,979 individuals who live in Germany and who responded to the online survey "Life with Corona" between October 1, 2020 and February 28, 2021. We estimate interaction effects from generalized linear models. The analyses reveal no pre-existing GD in aggression but exposure to COVID-19 and COVID-19 countermeasures is associated with sharper increases in aggression in men than in women. GD in anxiety decreased among participants with children in the household (with men becoming more anxious). We also observe pre-existing and increasing GD with regards to the severity of depression, with women presenting a larger increase in symptoms during the hard lockdown or with increasing stringency. In contrast to anxiety, GD in depression increased among participants who lived without children (women > men), but decreased for individuals who lived with children; here, men converged to the levels of depression presented by women. Finally, GD in somatic symptoms decreased during the hard lockdown (but not with higher stringency), with men showing a sharper increase in symptoms, especially when they lived with children or alone. Taken together, the findings indicate an increase in GD in mental health as the pandemic unfolded in Germany, with rising female vulnerability to depression and increasing male aggression. The combination of these two trends further suggests a worrying mental health situation for singles and families. Our results have important policy implications for the German health system and public health policy. This public health challenge requires addressing the rising burden of pandemic-related mental health challenges and the distribution of this burden between women and men, within families and for individuals who live alone.
Collapse
Affiliation(s)
- Liliana Abreu
- Development Research Group, Department of Politics and Public Administration, University of Konstanz, Konstanz, Germany
| | - Anke Koebach
- Development Research Group, Department of Politics and Public Administration, University of Konstanz, Konstanz, Germany
- Clinical Neuropsychology, Department of Psychology, University of Konstanz, Konstanz, Germany
| | - Oscar Díaz
- ISDC – International Security and Development Center, Berlin, Germany
| | - Samuel Carleial
- Clinical Neuropsychology, Department of Psychology, University of Konstanz, Konstanz, Germany
| | - Anke Hoeffler
- Development Research Group, Department of Politics and Public Administration, University of Konstanz, Konstanz, Germany
| | - Wolfgang Stojetz
- ISDC – International Security and Development Center, Berlin, Germany
| | - Hanna Freudenreich
- Leibniz Institute of Vegetable and Ornamental Crops, Großbeeren, Germany
| | - Patricia Justino
- World Institute for Development Economic Research, United Nations University, Helsinki, Finland
| | - Tilman Brück
- ISDC – International Security and Development Center, Berlin, Germany
- Leibniz Institute of Vegetable and Ornamental Crops, Großbeeren, Germany
- Natural Resources Institute, University of Greenwich, Chatham Maritime, United Kingdom
| |
Collapse
|
13
|
Du Preez A, Onorato D, Eiben I, Musaelyan K, Egeland M, Zunszain PA, Fernandes C, Thuret S, Pariante CM. Chronic stress followed by social isolation promotes depressive-like behaviour, alters microglial and astrocyte biology and reduces hippocampal neurogenesis in male mice. Brain Behav Immun 2021; 91:24-47. [PMID: 32755644 DOI: 10.1016/j.bbi.2020.07.015] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/29/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022] Open
Abstract
Unpredictable chronic mild stress (UCMS) is one of the most commonly used, robust and translatable models for studying the neurobiological basis of major depression. Although the model currently has multiple advantages, it does not entirely follow the trajectory of the disorder, whereby depressive symptomology can often present months after exposure to stress. Furthermore, patients with depression are more likely to withdraw in response to their stressful experience, or as a symptom of their depression, and, in turn, this withdrawal/isolation can further exacerbate the stressful experience and the depressive symptomology. Therefore, we investigated the effect(s) of 6 weeks of UCMS followed by another 6 weeks of social isolation (referred to as UCMSI), on behaviour, corticosterone stress responsivity, immune system functioning, and hippocampal neurogenesis, in young adult male mice. We found that UCMSI induced several behavioural changes resembling depression but did not induce peripheral inflammation. However, UCMSI animals showed increased microglial activation in the ventral dentate gyrus (DG) of the hippocampus and astrocyte activation in both the dorsal and ventral DG, with increased GFAP-positive cell immunoreactivity, GFAP-positive cell hypertrophy and process extension, and increased s100β-positive cell density. Moreover, UCMSI animals had significantly reduced neurogenesis in the DG and reduced levels of peripheral vascular endothelial growth factor (VEGF) - a trophic factor produced by astrocytes and that stimulates neurogenesis. Finally, UCMSI mice also had normal baseline corticosterone levels but a smaller increase in corticosterone following acute stress, that is, the Porsolt Swim Test. Our work gives clinically relevant insights into the role that microglial and astrocyte functioning, and hippocampal neurogenesis may play in the context of stress, social isolation and depression, offering a potentially new avenue for therapeutic target.
Collapse
Affiliation(s)
- Andrea Du Preez
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK.
| | - Diletta Onorato
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Inez Eiben
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Ksenia Musaelyan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Martin Egeland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Patricia A Zunszain
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Sandrine Thuret
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| |
Collapse
|
14
|
Lu X, Ce Q, Jin L, Zheng J, Sun M, Tang X, Li D, Sun J. Deoiled sunflower seeds ameliorate depression by promoting the production of monoamine neurotransmitters and inhibiting oxidative stress. Food Funct 2020; 12:573-586. [PMID: 33367360 DOI: 10.1039/d0fo01978j] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We aimed to evaluate the antidepressant activity of deoiled sunflower seeds (SFS), which are rich in tryptophan, in our mouse model and explored a possible mechanism of action. Male C57BL/6J mice were subjected to chronic unpredictable mild stress (CUMS) and were administered a diet containing SFS as the main protein source. SFS alleviated CUMS-induced depression-like behaviors, compared to the effects of a whey protein-based diet. This effect was related to increases in the levels of serotonin, dopamine, norepinephrine, acetylcholine, and brain-derived neurotrophic factor in SFS-fed mice. These changes accompanied the amelioration of inflammatory abnormalities and oxidative stress. SFS increased the aromatic amino acid levels, and the ratio of tryptophan to neutral amino acids. Furthermore, the antidepressant-like effects of SFS were involved in lipid, nucleotide, and amino acid metabolism. In summary, SFS was found to attenuate depression-like symptoms in mice. These antidepressant effects may be related to the increase in the levels of aromatic amino acids and neurotransmitters, amelioration of oxidative stress and inflammation, and the regulation of the levels of abnormal metabolites to the normal levels.
Collapse
Affiliation(s)
- Xiaomeng Lu
- Institute of Nutrition and Health, School of Public Health, Qingdao University, Qingdao 266071, Shandong, China.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
de Abreu MS, Giacomini ACVV, Genario R, Rech N, Carboni J, Lakstygal AM, Amstislavskaya TG, Demin KA, Leonard BE, Vlok M, Harvey BH, Piato A, Barcellos LJG, Kalueff AV. Non-pharmacological and pharmacological approaches for psychiatric disorders: Re-appraisal and insights from zebrafish models. Pharmacol Biochem Behav 2020; 193:172928. [PMID: 32289330 DOI: 10.1016/j.pbb.2020.172928] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022]
Abstract
Acute and chronic stressors are common triggers of human mental illnesses. Experimental animal models and their cross-species translation to humans are critical for understanding of the pathogenesis of stress-related psychiatric disorders. Mounting evidence suggests that both pharmacological and non-pharmacological approaches can be efficient in treating these disorders. Here, we analyze human, rodent and zebrafish (Danio rerio) data to compare the impact of non-pharmacological and pharmacological therapies of stress-related psychopathologies. Emphasizing the likely synergism and interplay between pharmacological and environmental factors in mitigating daily stress both clinically and in experimental models, we argue that environmental enrichment emerges as a promising complementary therapy for stress-induced disorders across taxa. We also call for a broader use of novel model organisms, such as zebrafish, to study such treatments and their potential interplay.
Collapse
Affiliation(s)
- Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA.
| | - Ana C V V Giacomini
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil; Postgraduate Program in Environmental Sciences, University of Passo Fundo (UPF), Passo Fundo, Brazil
| | - Rafael Genario
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Nathália Rech
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Júlia Carboni
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Anton M Lakstygal
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, St. Petersburg, Russia; Granov Russian Scientific Center of Radiology and Surgical Technologies, St. Petersburg, Russia
| | - Tamara G Amstislavskaya
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia; Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, St. Petersburg, Russia
| | - Brian E Leonard
- University College Galway, Pharmacology Department, Galway, Ireland
| | - Marli Vlok
- Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Potchefstroom, South Africa
| | - Brian H Harvey
- Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Potchefstroom, South Africa
| | - Angelo Piato
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA; Postgraduate Program in Neurosciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Leonardo J G Barcellos
- Postgraduate Program in Environmental Sciences, University of Passo Fundo (UPF), Passo Fundo, Brazil; Postgraduate Program in Bio-Experimentation, University of Passo Fundo (UPF), Passo Fundo, Brazil; Postgraduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
16
|
Sartim AG, Sartim MA, Cummings RD, Dias-Baruffi M, Joca SR. Impaired emotional response to stress in mice lacking galectin-1 or galectin-3. Physiol Behav 2020; 220:112862. [PMID: 32156558 DOI: 10.1016/j.physbeh.2020.112862] [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: 12/27/2019] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 12/15/2022]
Abstract
Galectin-1 (Gal-1) and galectin-3 (Gal-3) are multifunctional glycan-binding proteins, expressed in the brain and in its limbic structures that are involved in behavioral control. Gal-1 induces the expression of the brain-derived neurotrophic factor (BDNF) and promotes adult neural stem cells proliferation, biological events impaired in stress-related psychiatric disorders, such as depression and anxiety. Despite that, there is no evidence regarding galectin involvement in emotional control during stressful situations. Thus, we analyzed the behavioral phenotype of Gal-1 or Gal-3 knock-out mice (Gal-1 KO or Gal-3 KO) in different experimental models predictive of depressive and compulsive-like behaviors. METHODS C57BL-6 Gal-1 KO, Gal-3 KO, and wild-type mice (WT) were analyzed under the open field test (OFT) and, 6 h later, under the forced swim test (FST). Additionally, independent groups of male mice, lacking galectins or not, were exposed to the tail suspension test (TST) or to the marble burying test (MBT). The hippocampus and prefrontal cortex (PFC) of the mice submitted to MBT were dissected to access BDNF levels. RESULTS Both Gal-1 and Gal-3 KO mice showed increased time of immobility in the FST and in the TST compared to WT animals, thus reflecting an impaired stress-coping behavior. Additionally, Gal-1 and Gal-3 KO female mice presented increased compulsive-like behavior in the MBT, without significant changes in the locomotor activity. BDNF levels were found to be decreased in the PFC of Gal-1 KO mice. DISCUSSION Our results demonstrate that the absence of either endogenous Gal-1 and Gal-3 impairs stress-coping and increases compulsive-like behavior, suggesting that Gal-1 and Gal-3 are involved in the neurobiology of depression and obsessive-compulsive-like disorder.
Collapse
Affiliation(s)
- A G Sartim
- Department of Biomolecular Sciences, School of Pharmaceutical Science of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - M A Sartim
- Basic and Applied Immunology Graduate Program, Institute of Biological Sciences, Federal University of Amazonas, Manaus, AM, Brazil
| | - R D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, Room 11087, Boston, MA, 02115, United States
| | - M Dias-Baruffi
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo. Ribeirão Preto, SP, Brazil.
| | - S R Joca
- Department of Biomolecular Sciences, School of Pharmaceutical Science of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus Denmark.
| |
Collapse
|
17
|
Riemann D, Krone LB, Wulff K, Nissen C. Sleep, insomnia, and depression. Neuropsychopharmacology 2020; 45:74-89. [PMID: 31071719 PMCID: PMC6879516 DOI: 10.1038/s41386-019-0411-y] [Citation(s) in RCA: 375] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 12/23/2022]
Abstract
Since ancient times it is known that melancholia and sleep disturbances co-occur. The introduction of polysomnography into psychiatric research confirmed a disturbance of sleep continuity in patients with depression, revealing not only a decrease in Slow Wave Sleep, but also a disinhibition of REM (rapid eye movement) sleep, demonstrated as a shortening of REM latency, an increase of REM density, as well as total REM sleep time. Initial hopes that these abnormalities of REM sleep may serve as differential-diagnostic markers for subtypes of depression were not fulfilled. Almost all antidepressant agents suppress REM sleep and a time-and-dose-response relationship between total REM sleep suppression and therapeutic response to treatment seemed apparent. The so-called Cholinergic REM Induction Test revealed that REM sleep abnormalities can be mimicked by administration of cholinomimetic agents. Another important research avenue is the study of chrono-medical timing of sleep deprivation and light exposure for their positive effects on mood in depression. Present day research takes the view on insomnia, i.e., prolonged sleep latency, problems to maintain sleep, and early morning awakening, as a transdiagnostic symptom for many mental disorders, being most closely related to depression. Studying insomnia from different angles as a transdiagnostic phenotype has opened many new perspectives for research into mechanisms but also for clinical practice. Thus, the question is: can the early and adequate treatment of insomnia prevent depression? This article will link current understanding about sleep regulatory mechanisms with knowledge about changes in physiology due to depression. The review aims to draw the attention to current and future strategies in research and clinical practice to the benefits of sleep and depression therapeutics.
Collapse
Affiliation(s)
- Dieter Riemann
- Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Lukas B Krone
- Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK
| | - Katharina Wulff
- Departments of Radiation Sciences & Molecular Biology, Umea University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umea University, Umeå, Sweden
| | - Christoph Nissen
- University Hospital of Psychiatry and Psychotherapy, Bern, Switzerland
| |
Collapse
|
18
|
Loss of control over mild aversive events produces significant helplessness in mice. Behav Brain Res 2019; 376:112173. [PMID: 31445976 DOI: 10.1016/j.bbr.2019.112173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/05/2019] [Accepted: 08/21/2019] [Indexed: 02/08/2023]
Abstract
Most of the pathophysiology of depression are still unknown because of its numerous disease states of distinct etiology and pathogenesis. Stressful rodent models have been used to test a number of hypotheses regarding the etiology of depression. The learned helplessness rodent model demonstrates that having no control at all over aversive events produces helplessness and depression, but the role of loss of control over aversive events in helplessness is still not reliably modelled or deeply investigated. A rodent model of helplessness produced by loss of control is closer to human conditions and is therefore more useful for novel mechanistic and pre-clinic studies. The present work proposed a triadic experimental design in which a Loss Of Control (LOC) group of mice was firstly exposed to escapable mild footshocks to acquire control, and then to inescapable shocks to lose control, with a yoked (L-Yoked) group receiving identical but always uncontrollable shocks. Although both the LOC and the L-Yoked groups developed helplessness, as compared with the naive control group, the helplessness exhibited in the LOC group was significantly more serious than that in the L-Yoked group. The difference in severity between the LOC and the L-Yoked groups demonstrates the effects of loss of control over aversive events, in addition to the effects of the aversive events per se. The LOC paradigm can be used to reproduce pathology of depression induced by loss of control over aversive life events, with a good constructive validity.
Collapse
|
19
|
Kasten CR, Holmgren EB, Wills TA. Metabotropic Glutamate Receptor Subtype 5 in Alcohol-Induced Negative Affect. Brain Sci 2019; 9:E183. [PMID: 31366097 PMCID: PMC6721373 DOI: 10.3390/brainsci9080183] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 02/07/2023] Open
Abstract
Allosteric modulators of metabotropic glutamate 5 receptors (mGlu5 receptors) have been identified as a promising treatment to independently alleviate both negative affective states and ethanol-seeking and intake. However, these conditions are often comorbid and might precipitate one another. Acute and protracted ethanol withdrawal can lead to negative affective states. In turn, these states are primary drivers of alcohol relapse, particularly among women. The current review synthesizes preclinical studies that have observed the role of mGlu5 receptor modulation in negative affective states following ethanol exposure. The primary behavioral assays discussed are ethanol-seeking and intake, development and extinction of ethanol-associated cues and contexts, behavioral despair, and anxiety-like activity. The work done to-date supports mGlu5 receptor modulation as a promising target for mediating negative affective states to reduce ethanol intake or prevent relapse. Limitations in interpreting these data include the lack of models that use alcohol-dependent animals, limited use of adolescent and female subjects, and a lack of comprehensive evaluations of negative affective-like behavior.
Collapse
Affiliation(s)
- Chelsea R Kasten
- LSU Health Sciences Center-New Orleans, Department of Cell Biology and Anatomy, Medical Education Building, 1901 Perdido Street, Room 6103, New Orleans, LA 70112, USA
| | - Eleanor B Holmgren
- LSU Health Sciences Center-New Orleans, Department of Cell Biology and Anatomy, Medical Education Building, 1901 Perdido Street, Room 6103, New Orleans, LA 70112, USA
| | - Tiffany A Wills
- LSU Health Sciences Center-New Orleans, Department of Cell Biology and Anatomy, Medical Education Building, 1901 Perdido Street, Room 6103, New Orleans, LA 70112, USA.
| |
Collapse
|
20
|
Lakstygal AM, de Abreu MS, Lifanov DA, Wappler-Guzzetta EA, Serikuly N, Alpsyshov ET, Wang D, Wang M, Tang Z, Yan D, Demin KA, Volgin AD, Amstislavskaya TG, Wang J, Song C, Alekseeva P, Kalueff AV. Zebrafish models of diabetes-related CNS pathogenesis. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:48-58. [PMID: 30476525 DOI: 10.1016/j.pnpbp.2018.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/18/2018] [Accepted: 11/22/2018] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus (DM) is a common metabolic disorder that affects multiple organ systems. DM also affects brain processes, contributing to various CNS disorders, including depression, anxiety and Alzheimer's disease. Despite active research in humans, rodent models and in-vitro systems, the pathogenetic link between DM and brain disorders remains poorly understood. Novel translational models and new model organisms are therefore essential to more fully study the impact of DM on CNS. The zebrafish (Danio rerio) is a powerful novel model species to study metabolic and CNS disorders. Here, we discuss how DM alters brain functions and behavior in zebrafish, and summarize their translational relevance to studying DM-related CNS pathogenesis in humans. We recognize the growing utility of zebrafish models in translational DM research, as they continue to improve our understanding of different brain pathologies associated with DM, and may foster the discovery of drugs that prevent or treat these diseases.
Collapse
Affiliation(s)
- Anton M Lakstygal
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Dmitry A Lifanov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; School of Pharmacy, Southwest University, Chongqing, China
| | | | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | | | - DongMei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - MengYao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - ZhiChong Tang
- School of Pharmacy, Southwest University, Chongqing, China
| | - DongNi Yan
- School of Pharmacy, Southwest University, Chongqing, China
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | | | - JiaJia Wang
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Polina Alekseeva
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia; Ural Federal University, Ekaterinburg, Russia; Russian Scientific Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; ZENEREI Research Center, Slidell, LA, USA.
| |
Collapse
|
21
|
Bowman MA, Daws LC. Targeting Serotonin Transporters in the Treatment of Juvenile and Adolescent Depression. Front Neurosci 2019; 13:156. [PMID: 30872996 PMCID: PMC6401641 DOI: 10.3389/fnins.2019.00156] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 02/11/2019] [Indexed: 01/01/2023] Open
Abstract
Depression is a serious public health concern. Many patients are not effectively treated, but in children and adolescents this problem is compounded by limited pharmaceutical options. Currently, the Food and Drug Administration approves only two antidepressants for use in these young populations. Both are selective serotonin reuptake inhibitors (SSRIs). Compounding matters further, they are therapeutically less efficacious in children and adolescents than in adults. Here, we review clinical and preclinical literature describing the antidepressant efficacy of SSRIs in juveniles and adolescents. Since the high-affinity serotonin transporter (SERT) is the primary target of SSRIs, we then synthesize these reports with studies of SERT expression/function during juvenile and adolescent periods. Preclinical literature reveals some striking parallels with clinical studies, primary among them is that, like humans, juvenile and adolescent rodents show reduced antidepressant-like responses to SSRIs. These findings underscore the utility of preclinical assays designed to screen drugs for antidepressant efficacy across ages. There is general agreement that SERT expression/function is lower in juveniles and adolescents than in adults. It is well established that chronic SSRI treatment decreases SERT expression/function in adults, but strikingly, SERT expression/function in adolescents is increased following chronic treatment with SSRIs. Finally, we discuss a putative role for organic cation transporters and/or plasma membrane monoamine transporter in serotonergic homeostasis in juveniles and adolescents. Taken together, fundamental differences in SERT, and putatively in other transporters capable of serotonin clearance, may provide a mechanistic basis for the relative inefficiency of SSRIs to treat pediatric depression, relative to adults.
Collapse
Affiliation(s)
- Melodi A Bowman
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Lynette C Daws
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
22
|
Demin KA, Sysoev M, Chernysh MV, Savva AK, Koshiba M, Wappler-Guzzetta EA, Song C, De Abreu MS, Leonard B, Parker MO, Harvey BH, Tian L, Vasar E, Strekalova T, Amstislavskaya TG, Volgin AD, Alpyshov ET, Wang D, Kalueff AV. Animal models of major depressive disorder and the implications for drug discovery and development. Expert Opin Drug Discov 2019; 14:365-378. [PMID: 30793996 DOI: 10.1080/17460441.2019.1575360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Depression is a highly debilitating psychiatric disorder that affects the global population and causes severe disabilities and suicide. Depression pathogenesis remains poorly understood, and the disorder is often treatment-resistant and recurrent, necessitating the development of novel therapies, models and concepts in this field. Areas covered: Animal models are indispensable for translational biological psychiatry, and markedly advance the study of depression. Novel approaches continuously emerge that may help untangle the disorder heterogeneity and unclear categories of disease classification systems. Some of these approaches include widening the spectrum of model species used for translational research, using a broader range of test paradigms, exploring new pathogenic pathways and biomarkers, and focusing more closely on processes beyond neural cells (e.g. glial, inflammatory and metabolic deficits). Expert opinion: Dividing the core symptoms into easily translatable, evolutionarily conserved phenotypes is an effective way to reevaluate current depression modeling. Conceptually novel approaches based on the endophenotype paradigm, cross-species trait genetics and 'domain interplay concept', as well as using a wider spectrum of model organisms and target systems will enhance experimental modeling of depression and antidepressant drug discovery.
Collapse
Affiliation(s)
- Konstantin A Demin
- a Institute of Experimental Medicine , Almazov National Medical Research Centre , St. Petersburg , Russia.,b Institute of Translational Biomedicine , St. Petersburg State University , St. Petersburg , Russia
| | - Maxim Sysoev
- c Laboratory of Preclinical Bioscreening , Russian Research Center for Radiology and Surgical Technologies , St. Petersburg , Russia.,d Institute of Experimental Medicine , St. Petersburg , Russia
| | - Maria V Chernysh
- b Institute of Translational Biomedicine , St. Petersburg State University , St. Petersburg , Russia
| | - Anna K Savva
- e Faculty of Biology , St. Petersburg State University , St. Petersburg , Russia
| | | | | | - Cai Song
- h Research Institute of Marine Drugs and Nutrition , Guangdong Ocean University , Zhanjiang , China.,i Marine Medicine Development Center, Shenzhen Institute , Guangdong Ocean University , Shenzhen , China
| | - Murilo S De Abreu
- j Bioscience Institute , University of Passo Fundo (UPF) , Passo Fundo , Brazil
| | | | - Matthew O Parker
- l Brain and Behaviour Lab , School of Pharmacy and Biomedical Science, University of Portsmouth , Portsmouth , UK
| | - Brian H Harvey
- m Center of Excellence for Pharmaceutical Sciences , Division of Pharmacology, School of Pharmacy, North-West University , Potchefstroom , South Africa
| | - Li Tian
- n Institute of Biomedicine and Translational Medicine , University of Tartu , Tartu , Estonia
| | - Eero Vasar
- n Institute of Biomedicine and Translational Medicine , University of Tartu , Tartu , Estonia
| | - Tatyana Strekalova
- o Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, and Department of Normal Physiology , Sechenov First Moscow State Medical University , Moscow , Russia.,p Laboratory of Cognitive Dysfunctions , Institute of General Pathology and Pathophysiology , Moscow , Russia.,q Department of Neuroscience , Maastricht University , Maastricht , The Netherlands
| | | | - Andrey D Volgin
- g The International Zebrafish Neuroscience Research Consortium (ZNRC) , Slidell , LA , USA.,r Scientific Research Institute of Physiology and Basic Medicine , Novosibirsk , Russia
| | - Erik T Alpyshov
- s School of Pharmacy , Southwest University , Chongqing , China
| | - Dongmei Wang
- s School of Pharmacy , Southwest University , Chongqing , China
| | - Allan V Kalueff
- s School of Pharmacy , Southwest University , Chongqing , China.,t Almazov National Medical Research Centre , St. Petersburg , Russia.,u Ural Federal University , Ekaterinburg , Russia.,v Granov Russian Research Center of Radiology and Surgical Technologies , St. Petersburg , Russia.,w Laboratory of Biological Psychiatry, Institute of Translational Biomedicine , St. Petersburg State University , St. Petersburg , Russia.,x Laboratory of Translational Biopsychiatry , Scientific Research Institute of Physiology and Basic Medicine , Novosibirsk , Russia.,y ZENEREI Institute , Slidell , LA , USA.,z The International Stress and Behavior Society (ISBS), US HQ , New Orleans , LA , USA
| |
Collapse
|
23
|
Harro J. Animal models of depression: pros and cons. Cell Tissue Res 2018; 377:5-20. [PMID: 30560458 DOI: 10.1007/s00441-018-2973-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022]
Abstract
Animal models of depression are certainly needed but the question in the title has been raised owing to the controversies in the interpretation of the readout in a number of tests, to the perceived lack of progress in the development of novel treatments and to the expressed doubts in whether animal models can offer anything to make a true breakthrough in understanding the neurobiology of depression and producing novel drugs against depression. Herewith, it is argued that if anything is wrong with animal models, including those for depression, it is not about the principle of modelling complex human disorder in animals but in the way the tests are selected, conducted and interpreted. Further progress in the study of depression and in developing new treatments, will be supported by animal models of depression if these were more critically targeted to drug screening vs. studies of underlying neurobiology, clearly stratified to vulnerability and pathogenetic models, focused on well-defined endophenotypes and validated for each setting while bearing the existing limits to validation in mind. Animal models of depression need not to rely merely on behavioural readouts but increasingly incorporate neurobiological measures as the understanding of depression as human brain disorder advances. Further developments would be fostered by cross-fertilizinga translational approach that is bidirectional, research on humans making more use of neurobiological findings in animals.
Collapse
Affiliation(s)
- Jaanus Harro
- Division of Neuropsychopharmacology, Department of Psychology, Estonian Centre of Behavioural and Health Sciences, University of Tartu, Ravila 14A Chemicum, 50411, Tartu, Estonia.
| |
Collapse
|
24
|
Godoy LD, Umeoka EHL, Ribeiro DE, Santos VR, Antunes-Rodrigues J, Joca SRL, Garcia-Cairasco N. Multimodal early-life stress induces biological changes associated to psychopathologies. Horm Behav 2018; 100:69-80. [PMID: 29548783 DOI: 10.1016/j.yhbeh.2018.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/07/2018] [Accepted: 03/11/2018] [Indexed: 01/21/2023]
Abstract
Evidences suggest the contributive role of early-life stress (ELS) to affective and anxiety disorders. Chronic exposure to the same stressor may generate habituation, while the exposure to different and repeated stressors gradually promotes maladaptive plasticity. Therefore, to further understand the effects of heterotypic stressors during early life period, male Wistar rat pups (P1-P21) were exposed to Multimodal ELS paradigm. Results indicate pups did not habituate to multimodal ELS and neonates respond to both physical and psychogenic stressors. Adult rats that underwent ELS protocol showed significant lower sucrose intake, decreased latency to immobility in the forced swim test and increased latency to light compartment in the light-dark test when compared to control group. Although it has been shown that ELS-induced changes in hippocampus can be used as biomarkers, multimodal ELS did not significantly alter BDNF, Tyrosine Kinase B (TrkB) receptor expression or neurogenesis in the hippocampus. Taken together, these findings indicate that multimodal ELS protocol can be an interesting experimental model for understanding long-term psychiatric disorders associated with stress. Indeed, our data with neurogenesis, BDNF and TrkB, and conflicting data from the literature, suggest that additional studies on synaptic plasticity/intracellular cascades would help to detect the underlying mechanisms.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Eduardo H L Umeoka
- Neurosciences and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Deidiane Elisa Ribeiro
- Pharmacology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | | | - José Antunes-Rodrigues
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Samia Regiane Lourenço Joca
- Physics and Chemistry Department, Ribeirão Preto School of Pharmacy, University of São Paulo, Brazil; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Norberto Garcia-Cairasco
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; Neurosciences and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil.
| |
Collapse
|
25
|
Dadomo H, Gioiosa L, Cigalotti J, Ceresini G, Parmigiani S, Palanza P. What is stressful for females? Differential effects of unpredictable environmental or social stress in CD1 female mice. Horm Behav 2018; 98:22-32. [PMID: 29187314 DOI: 10.1016/j.yhbeh.2017.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/14/2017] [Accepted: 11/21/2017] [Indexed: 01/01/2023]
Abstract
Stressful life events are a major factor in the etiology of several diseases, such as cardiovascular, inflammatory and psychiatric disorders (i.e., depression and anxiety), with the two sexes greatly differing in vulnerability. In humans and other animals, physiological and behavioral responses to stress are strongly dependent on gender, and conditions that are stressful for males are not necessarily stressful for females. Hence the need of an animal model of social chronic stress specifically designed for females. In the present study we aimed to compare the effects of two different chronic stress procedures in female mice, by investigating the impact of 4weeks of nonsocial unpredictable, physical stress by the Chronic Mild Stress paradigm (CMS; Exp.1) or of Social Instability Stress (SIS; Exp.2) on physiological, endocrine and behavioral parameters in adult female mice. CMS had a pronounced effect on females' response to novelty (i.e., either novel environment or novel social stimulus), body weight growth and hormonal profile. Conversely, 4weeks of social instability did not alter females' response to novelty nor hormonal levels but induced anhedonia. Our findings thus showed that female mice were more sensitive to nonsocial stress due to unpredictable physical environment than to social instability stressors. Neither of these stress paradigms, however, induced a consistent behavioral and physiological stress response in female mice comparable to that induced by chronic stress procedures in male mice, thus confirming the difficulties of developing a robust and validated model of chronic psychosocial stress in female mice.
Collapse
Affiliation(s)
- Harold Dadomo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Laura Gioiosa
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Jenny Cigalotti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Graziano Ceresini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Parmigiani
- Department of Chemistry, Life Sciences and Environmental Sustainaibility, University of Parma, Parma, Italy
| | - Paola Palanza
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| |
Collapse
|
26
|
Benatti C, Alboni S, Blom JMC, Mendlewicz J, Tascedda F, Brunello N. Molecular changes associated with escitalopram response in a stress-based model of depression. Psychoneuroendocrinology 2018; 87:74-82. [PMID: 29049934 DOI: 10.1016/j.psyneuen.2017.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/15/2017] [Accepted: 10/12/2017] [Indexed: 11/30/2022]
Abstract
Converging evidence points at hypothalamus-pituitary-adrenal (HPA) axis hyperactivity and neuroinflammation as important factors involved in the etiopathogenesis of major depressive disorder (MDD) and in therapeutic efficacy of antidepressants. In this study, we examined the molecular effects associated with a response to a week-long treatment with escitalopram in the chronic escape deficit (CED) model, a validated model of depression based on the induction of an escape deficit after exposure of rats to an unavoidable stress. We confirmed our previous result that a treatment with escitalopram (10mg/kg) was effective after 7days in reverting the stress-induced escape deficit in approximately 50% of the animals, separating responders from non-responders. Expression of markers of HPA axis functionality as well as several inflammatory mediators were evaluated in the hypothalamus, a key structure integrating signals from the neuro, immune, endocrine systems. In the hypothalamus of responder animals we observed a decrease in the expression of CRH and its receptors and an increase in GR protein in total and nuclear extracts; this effect was accompanied by a significant decrease in circulating corticosterone in the same cohort. Hypothalamic IL-1β and TNFα expression were increased in stressed animals, while CXCL2, IL-6, and ADAM17 mRNA levels were decreased in escitalopram treated rats regardless of the treatment response. These data suggest that efficacy of a one week treatment with escitalopram may be partially mediated by a decrease HPA axis activity, while in the hypothalamus the drug-induced effects on the expression of immune modulators did not correlate with the behavioural outcome.
Collapse
Affiliation(s)
- Cristina Benatti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology University of Modena and Reggio Emilia, Modena, Italy.
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy
| | - Joan M C Blom
- Center for Neuroscience and Neurotechnology University of Modena and Reggio Emilia, Modena, Italy; Department of Education and Human Sciences, University of Modena and Reggio Emilia, Viale Antonio Allegri 9, 42121, Reggio Emilia, Italy
| | - Julien Mendlewicz
- Department of Psychiatry, University Clinics of Brussels, Erasme Hospital, Free University of Brussels, 808 Route de Lennik, Brussels, Belgium
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology University of Modena and Reggio Emilia, Modena, Italy
| | - Nicoletta Brunello
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125, Modena, Italy; Center for Neuroscience and Neurotechnology University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
27
|
Hales CA, Houghton CJ, Robinson ESJ. Behavioural and computational methods reveal differential effects for how delayed and rapid onset antidepressants effect decision making in rats. Eur Neuropsychopharmacol 2017; 27:1268-1280. [PMID: 29100819 PMCID: PMC5720479 DOI: 10.1016/j.euroneuro.2017.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/18/2017] [Accepted: 09/25/2017] [Indexed: 12/16/2022]
Abstract
Major depressive disorder (MDD) is one of the most prevalent psychiatric disorders. Until the recent discovery of the rapid onset antidepressant action of ketamine, pharmacological treatments for MDD were limited to conventional antidepressant drugs with delayed clinical efficacy. Using a judgement bias task, this study has investigated whether the temporal differences observed in patients would be reflected in affective biases and decision making behaviour in rodents. The diffusion model was also used to investigate the underlying decision making processes. Positive biases were induced in this task over timeframes that mirror the rapid versus delayed antidepressant efficacy of the drugs in clinical populations. Diffusion modelling revealed that the antidepressants tested also have different effects on decision making processes, suggesting they may act through different neurobiological substrates. This combination of behaviour and computational modelling may provide a useful approach to further investigate the mechanisms underlying rapid antidepressant effect and assess potential new treatments.
Collapse
Affiliation(s)
- Claire A Hales
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Conor J Houghton
- Department of Computer Science, Faculty of Engineering, University of Bristol, Bristol BS8 1UB, UK
| | - Emma S J Robinson
- School of Physiology, Pharmacology & Neuroscience, Faculty of Biomedical Sciences, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
28
|
Unpredictable chronic mild stress differentially impairs social and contextual discrimination learning in two inbred mouse strains. PLoS One 2017; 12:e0188537. [PMID: 29166674 PMCID: PMC5699833 DOI: 10.1371/journal.pone.0188537] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/08/2017] [Indexed: 12/28/2022] Open
Abstract
Alterations in the social and cognitive domain are considered important indicators for increased disability in many stress-related disorders. Similar impairments have been observed in rodents chronically exposed to stress, mimicking potential endophenotypes of stress-related psychopathologies such as major depression disorder (MDD), anxiety, conduct disorder, and posttraumatic stress disorder (PTSD). Data from numerous studies suggest that deficient plasticity mechanisms in hippocampus (HC) and prefrontal cortex (PFC) might underlie these social and cognitive deficits. Specifically, stress-induced deficiencies in neural plasticity have been associated with a hypodopaminergic state and reduced neural plasticity persistence. Here we assessed the effects of unpredictable chronic mild stress (UCMS) on exploratory, social and cognitive behavior of females of two inbred mouse strains (C57BL/6J and DBA/2J) that differ in their dopaminergic profile. Exposure to chronic stress resulted in impaired circadian rhythmicity, sociability and social cognition in both inbred strains, but differentially affected activity patterns and contextual discrimination performance. These stress-induced behavioral impairments were accompanied by reduced expression levels of brain derived neurotrophic factor (BDNF) in the prefrontal cortex. The strain-specific cognitive impairment was coexistent with enhanced plasma corticosterone levels and reduced expression of genes related to dopamine signaling in hippocampus. These results underline the importance of assessing different strains with multiple test batteries to elucidate the neural and genetic basis of social and cognitive impairments related to chronic stress.
Collapse
|
29
|
Phillips C. Brain-Derived Neurotrophic Factor, Depression, and Physical Activity: Making the Neuroplastic Connection. Neural Plast 2017; 2017:7260130. [PMID: 28928987 PMCID: PMC5591905 DOI: 10.1155/2017/7260130] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/18/2017] [Indexed: 12/14/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin that is vital to the survival, growth, and maintenance of neurons in key brain circuits involved in emotional and cognitive function. Convergent evidence indicates that neuroplastic mechanisms involving BDNF are deleteriously altered in major depressive disorder (MDD) and animal models of stress. Herein, clinical and preclinical evidence provided that stress-induced depressive pathology contributes to altered BDNF level and function in persons with MDD and, thereby, disruptions in neuroplasticity at the regional and circuit level. Conversely, effective therapeutics that mitigate depressive-related symptoms (e.g., antidepressants and physical activity) optimize BDNF in key brain regions, promote neuronal health and recovery of function in MDD-related circuits, and enhance pharmacotherapeutic response. A greater knowledge of the interrelationship between BDNF, depression, therapeutic mechanisms of action, and neuroplasticity is important as it necessarily precedes the derivation and deployment of more efficacious treatments.
Collapse
|
30
|
Ordway GA, Szebeni A, Hernandez LJ, Crawford JD, Szebeni K, Chandley MJ, Burgess KC, Miller C, Bakkalbasi E, Brown RW. Antidepressant-Like Actions of Inhibitors of Poly(ADP-Ribose) Polymerase in Rodent Models. Int J Neuropsychopharmacol 2017; 20:994-1004. [PMID: 29016792 PMCID: PMC5716178 DOI: 10.1093/ijnp/pyx068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Many patients suffering from depressive disorders are refractory to treatment with currently available antidepressant medications, while many more exhibit only a partial response. These factors drive research to discover new pharmacological approaches to treat depression. Numerous studies demonstrate evidence of inflammation and elevated oxidative stress in major depression. Recently, major depression has been shown to be associated with elevated levels of DNA oxidation in brain cells, accompanied by increased gene expression of the nuclear base excision repair enzyme, poly(ADP-ribose) polymerase-1. Given these findings and evidence that drugs that inhibit poly(ADP-ribose) polymerase-1 activity have antiinflammatory and neuroprotective properties, the present study was undertaken to examine the potential antidepressant properties of poly(ADP-ribose) polymerase inhibitors. METHODS Two rodent models, the Porsolt swim test and repeated exposure to psychological stressors, were used to test the poly(ADP-ribose) polymerase inhibitor, 3-aminobenzamide, for potential antidepressant activity. Another poly(ADP-ribose) polymerase inhibitor, 5-aminoisoquinolinone, was also tested. RESULTS Poly(ADP-ribose) polymerase inhibitors produced antidepressant-like effects in the Porsolt swim test, decreasing immobility time, and increasing latency to immobility, similar to the effects of fluoxetine. In addition, 3-aminobenzamide treatment increased sucrose preference and social interaction times relative to vehicle-treated control rats following repeated exposure to combined social defeat and unpredictable stress, mediating effects similar to fluoxetine treatment. CONCLUSIONS The poly(ADP-ribose) polymerase inhibitors 3-aminobenzamide and 5-aminoisoquinolinone exhibit antidepressant-like activity in 2 rodent stress models and uncover poly(ADP-ribose) polymerase as a unique molecular target for the potential development of a novel class of antidepressants.
Collapse
Affiliation(s)
- Gregory A Ordway
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi),Correspondence: Gregory A. Ordway, PhD, East Tennessee State University, PO Box 70577, Johnson City, 37614 ()
| | - Attila Szebeni
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Liza J Hernandez
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Jessica D Crawford
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Katalin Szebeni
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Michelle J Chandley
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Katherine C Burgess
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Corwin Miller
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Erol Bakkalbasi
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| | - Russell W Brown
- Department of Biomedical Sciences (Drs Ordway and Szebeni, Ms Hernandez, Drs Crawford and Szebeni, Ms Burgess, and Dr Brown) and Department of Psychiatry and Behavioral Sciences (Dr Ordway), James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee; Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson City, Tennessee (Dr Chandley); DS Therapeutics, Houston, Texas (Dr Miller and Dr Bakkalbasi)
| |
Collapse
|
31
|
Slattery DA, Cryan JF. Modelling depression in animals: at the interface of reward and stress pathways. Psychopharmacology (Berl) 2017; 234:1451-1465. [PMID: 28224183 DOI: 10.1007/s00213-017-4552-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/27/2017] [Indexed: 12/13/2022]
Abstract
RATIONALE Despite substantial research efforts the aetiology of major depressive disorder (MDD) remains poorly understood, which is due in part to the heterogeneity of the disorder and the complexity of designing appropriate animal models. However, in the last few decades, a focus on the development of novel stress-based paradigms and a focus on using hedonic/anhedonic behaviour have led to renewed optimism in the use of animal models to assess aspects of MDD. OBJECTIVES Therefore, in this review article, dedicated to Athina Markou, we summarise the use of stress-based animal models for studying MDD in rodents and how reward-related readouts can be used to validate/assess the model and/or treatment. RESULTS We reveal the use and limitations of chronic stress paradigms, which we split into non-social (i.e. chronic mild stress), social (i.e. chronic social defeat) and drug-withdrawal paradigms for studying MDD and detail numerous reward-related readouts that are employed in preclinical research. Finally, we finish with a section regarding important factors to consider when using animal models. CONCLUSIONS One of the most consistent findings following chronic stress exposure in rodents is a disruption of the brain reward system, which can be easily assessed using sucrose, social interaction, food, drug of abuse or intracranial self-stimulation as a readout. Probing the underlying causes of such alterations is providing a greater understanding of the potential systems and processes that are disrupted in MDD.
Collapse
Affiliation(s)
- D A Slattery
- Laboratory of Translational Psychiatry, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Heinrich-Hoffmann-Str. 10, 60528, Frankfurt, Germany.
| | - J F Cryan
- APC Microbiome Institute, Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
32
|
Cussotto S, Cryan JF, O'Leary OF. The hippocampus and dorsal raphe nucleus are key brain areas associated with the antidepressant effects of lithium augmentation of desipramine. Neurosci Lett 2017; 648:14-20. [PMID: 28351776 DOI: 10.1016/j.neulet.2017.03.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/09/2017] [Accepted: 03/23/2017] [Indexed: 11/25/2022]
Abstract
Approximately 50% of depressed individuals fail to achieve remission with first-line antidepressant drugs and a third remain treatment-resistant. When first-line antidepressant treatment is unsuccessful, second-line strategies include dose optimisation, switching to another antidepressant, combination with another antidepressant, or augmentation with a non-antidepressant medication. Much of the evidence for the efficacy of augmentation strategies comes from studies using lithium to augment the effects of tricyclic antidepressants. The neural circuitry underlying the therapeutic effects of lithium augmentation is not yet fully understood. Recently, we reported that chronic treatment with a combination of lithium and the antidepressant desipramine, exerted antidepressant-like behavioural effects in a mouse strain (BALB/cOLaHsd) that did not exhibit an antidepressant-like behavioural response to either drug alone. In the present study, we used this model in combination with ΔFosB/FosB immunohistochemistry to identify brain regions chronically affected by lithium augmentation of desipramine when compared to either treatment alone. The data suggest that the dorsal raphe nucleus and the CA3 regions of the dorsal hippocampus are key nodes in the neural circuitry underlying antidepressant action of lithium augmentation of desipramine. These data give new insight into the neurobiology underlying the mechanism of lithium augmentation in the context of treatment-resistant depression.
Collapse
Affiliation(s)
- Sofia Cussotto
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
33
|
Moussaoui N, Jacobs JP, Larauche M, Biraud M, Million M, Mayer E, Taché Y. Chronic Early-life Stress in Rat Pups Alters Basal Corticosterone, Intestinal Permeability, and Fecal Microbiota at Weaning: Influence of Sex. J Neurogastroenterol Motil 2017; 23:135-143. [PMID: 27829577 PMCID: PMC5216644 DOI: 10.5056/jnm16105] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 12/16/2022] Open
Abstract
Background/Aims Wistar rat dams exposed to limited nesting stress (LNS) from post-natal days (PND) 2 to 10 display erratic maternal behavior, and their pups show delayed maturation of the hypothalamic-pituitary-adrenal axis and impaired epithelial barrier at PND10 and a visceral hypersensitivity at adulthood. Little is known about the impact of early life stress on the offspring before adulthood and the influence of sex. We investigated whether male and female rats previously exposed to LNS displays at weaning altered corticosterone, intestinal permeability, and microbiota. Methods Wistar rat dams and litters were maintained from PND2 to 10 with limited nesting/bedding materials and thereafter reverted to normal housing up to weaning (PND21). Control litters had normal housing. At weaning, we monitored body weight, corticosterone plasma levels (enzyme immunoassay), in vivo intestinal to colon permeability (fluorescein isothiocyanate-dextran 4 kDa) and fecal microbiota (DNA extraction and amplification of the V4 region of the 16S ribosomal RNA gene). Results At weaning, LNS pups had hypercorticosteronemia and enhanced intestinal permeability with females > males while body weights were similar. LNS decreased fecal microbial diversity and induced a distinct composition characterized by increased abundance of Gram positive cocci and reduction of fiber-degrading, butyrate-producing, and mucus-resident microbes. Conclusions These data indicate that chronic exposure to LNS during the first week post-natally has sustained effects monitored at weaning including hypercorticosteronemia, a leaky gut, and dysbiosis. These alterations may impact on the susceptibility to develop visceral hypersensitivity in adult rats and have relevance to the development of irritable bowel syndrome in childhood.
Collapse
Affiliation(s)
- Nabila Moussaoui
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Jonathan P Jacobs
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Muriel Larauche
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Mandy Biraud
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Mulugeta Million
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Emeran Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Yvette Taché
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
34
|
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
|
35
|
Ng E, Browne CJ, Samsom JN, Wong AHC. Depression and substance use comorbidity: What we have learned from animal studies. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 43:456-474. [PMID: 27315335 DOI: 10.1080/00952990.2016.1183020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Depression and substance use disorders are often comorbid, but the reasons for this are unclear. In human studies, it is difficult to determine how one disorder may affect predisposition to the other and what the underlying mechanisms might be. Instead, animal studies allow experimental induction of behaviors relevant to depression and drug-taking, and permit direct interrogation of changes to neural circuits and molecular pathways. While this field is still new, here we review animal studies that investigate whether depression-like states increase vulnerability to drug-taking behaviors. Since chronic psychosocial stress can precipitate or predispose to depression in humans, we review studies that use psychosocial stressors to produce depression-like phenotypes in animals. Specifically, we describe how postweaning isolation stress, repeated social defeat stress, and chronic mild (or unpredictable) stress affect behaviors relevant to substance abuse, especially operant self-administration. Potential brain changes mediating these effects are also discussed where available, with an emphasis on mesocorticolimbic dopamine circuits. Postweaning isolation stress and repeated social defeat generally increase acquisition or maintenance of drug self-administration, and alter dopamine sensitivity in various brain regions. However, the effects of chronic mild stress on drug-taking have been much less studied. Future studies should consider standardizing stress-induction protocols, including female subjects, and using multi-hit models (e.g. genetic vulnerabilities and environmental stress).
Collapse
Affiliation(s)
- Enoch Ng
- a Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Canada.,b Institute of Medical Science, University of Toronto , Toronto , Canada
| | - Caleb J Browne
- c Department of Psychology , University of Toronto , Toronto , Canada.,d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada
| | - James N Samsom
- d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada.,e Department of Pharmacology , University of Toronto , Toronto , Canada
| | - Albert H C Wong
- b Institute of Medical Science, University of Toronto , Toronto , Canada.,d Campbell Family Health Institute , Centre for Addiction and Mental Health , Toronto , Canada.,e Department of Pharmacology , University of Toronto , Toronto , Canada.,f Department of Psychiatry , University of Toronto , Toronto , Canada
| |
Collapse
|
36
|
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
|
37
|
Tucker LB, Burke JF, Fu AH, McCabe JT. Neuropsychiatric Symptom Modeling in Male and Female C57BL/6J Mice after Experimental Traumatic Brain Injury. J Neurotrauma 2016; 34:890-905. [PMID: 27149139 PMCID: PMC5314988 DOI: 10.1089/neu.2016.4508] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Psychiatric symptoms such as anxiety and depression are frequent and persistent complaints following traumatic brain injury (TBI). Modeling these symptoms in animal models of TBI affords the opportunity to determine mechanisms underlying behavioral pathologies and to test potential therapeutic agents. However, testing these symptoms in animal models of TBI has yielded inconsistent results. The goal of the current study was to employ a battery of tests to measure multiple anxiety- and depressive-like symptoms following TBI in C57BL/6J mice, and to determine if male and female mice are differentially affected by the injury. Following controlled cortical impact (CCI) at a parietal location, neither male nor female mice showed depressive-like symptoms as measured by the Porsolt forced-swim test and sucrose preference test. Conclusions regarding anxiety-like behaviors were dependent upon the assay employed; CCI-induced thigmotaxis in the open field suggested an anxiogenic effect of the injury; however, results from the elevated zero maze, light-dark box, and marble-burying tests indicated that CCI reduced anxiety-like behaviors. Fewer anxiety-like behaviors were also associated with the female sex. Increased levels of activity were also measured in female mice and injured mice in these tests, and conclusions regarding anxiety should be taken with caution when experimental manipulations induce changes in baseline activity. These results underscore the irreconcilability of results from studies attempting to model TBI-induced neuropsychiatric symptoms. Changes in injury models or better attempts to replicate the clinical syndrome may improve the translational applicability of rodent models of TBI-induced anxiety and depression.
Collapse
Affiliation(s)
- Laura B Tucker
- 1 Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Service University of the Health Sciences , Bethesda, Maryland.,2 Department of Anatomy, Physiology, and Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - John F Burke
- 1 Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Service University of the Health Sciences , Bethesda, Maryland.,2 Department of Anatomy, Physiology, and Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Amanda H Fu
- 1 Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Service University of the Health Sciences , Bethesda, Maryland.,2 Department of Anatomy, Physiology, and Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Joseph T McCabe
- 1 Pre-Clinical Studies Core, Center for Neuroscience and Regenerative Medicine, Uniformed Service University of the Health Sciences , Bethesda, Maryland.,2 Department of Anatomy, Physiology, and Genetics, F.E. Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| |
Collapse
|
38
|
McCammon JM, Sive H. Challenges in understanding psychiatric disorders and developing therapeutics: a role for zebrafish. Dis Model Mech 2016; 8:647-56. [PMID: 26092527 PMCID: PMC4486859 DOI: 10.1242/dmm.019620] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The treatment of psychiatric disorders presents three major challenges to the research and clinical community: defining a genotype associated with a disorder, characterizing the molecular pathology of each disorder and developing new therapies. This Review addresses how cellular and animal systems can help to meet these challenges, with an emphasis on the role of the zebrafish. Genetic changes account for a large proportion of psychiatric disorders and, as gene variants that predispose to psychiatric disease are beginning to be identified in patients, these are tractable for study in cellular and animal systems. Defining cellular and molecular criteria associated with each disorder will help to uncover causal physiological changes in patients and will lead to more objective diagnostic criteria. These criteria should also define co-morbid pathologies within the nervous system or in other organ systems. The definition of genotypes and of any associated pathophysiology is integral to the development of new therapies. Cell culture-based approaches can address these challenges by identifying cellular pathology and by high-throughput screening of gene variants and potential therapeutics. Whole-animal systems can define the broadest function of disorder-associated gene variants and the organismal impact of candidate medications. Given its evolutionary conservation with humans and its experimental tractability, the zebrafish offers several advantages to psychiatric disorder research. These include assays ranging from molecular to behavioural, and capability for chemical screening. There is optimism that the multiple approaches discussed here will link together effectively to provide new diagnostics and treatments for psychiatric patients. Summary: In this review, we discuss strengths and limitations of prevalent laboratory models that are used for understanding psychiatric disorders and developing therapeutics, with emphasis on the zebrafish.
Collapse
Affiliation(s)
- Jasmine M McCammon
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, MA 02142, USA Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
39
|
Frisbee JC, Brooks SD, Stanley SC, d'Audiffret AC. An Unpredictable Chronic Mild Stress Protocol for Instigating Depressive Symptoms, Behavioral Changes and Negative Health Outcomes in Rodents. J Vis Exp 2015. [PMID: 26650668 DOI: 10.3791/53109] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Chronic, unresolved stress is a major risk factor for the development of clinical depression. While many preclinical models of stress-induced depression have been reported, the unpredictable chronic mild stress (UCMS) protocol is an established translationally-relevant model for inducing behavioral symptoms commonly associated with clinical depression, such as anhedonia, altered grooming behavior, and learned helplessness in rodents. The UCMS protocol also induces physiological (e.g., hypercortisolemia, hypertension) and neurological (e.g., anhedonia, learned helplessness) changes that are clinically associated with depression. Importantly, UCMS-induced depressive symptoms can be ameliorated through chronic, but not acute, treatment with common SSRIs. As such, the UCMS protocol offers many advantages over acute stress protocols or protocols that utilize more extreme stressors. Our protocol involves randomized, daily exposures to 7 distinct stressors: damp bedding, removal of bedding, cage tilt, alteration of light/dark cycles, social stresses, shallow water bath, and predator sounds/smells. By subjecting rodents 3-4 hr daily to these mild stressors for 8 weeks, we demonstrate both significant behavioral changes and poor health outcomes to the cardiovascular system. This approach allows for in-depth interrogation of the neurological, behavioral, and physiological alterations associated with chronic stress-induced depression, as well as for testing of new potential therapeutic agents or intervention strategies.
Collapse
Affiliation(s)
- Jefferson C Frisbee
- Department of Physiology and Pharmacology, West Virginia University Health Sciences Center;
| | - Steven D Brooks
- Department of Physiology and Pharmacology, West Virginia University Health Sciences Center
| | - Shyla C Stanley
- Department of Physiology and Pharmacology, West Virginia University Health Sciences Center
| | - Alexandre C d'Audiffret
- Division of Vascular and Endovascular Surgery, Center for Cardiovascular and Respiratory Sciences, West Virginia University Health Sciences Center
| |
Collapse
|
40
|
O'Brien FE, Moloney GM, Scott KA, O'Connor RM, Clarke G, Dinan TG, Griffin BT, Cryan JF. Chronic P-glycoprotein inhibition increases the brain concentration of escitalopram: potential implications for treating depression. Pharmacol Res Perspect 2015; 3:e00190. [PMID: 27022464 PMCID: PMC4777256 DOI: 10.1002/prp2.190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/23/2015] [Accepted: 09/02/2015] [Indexed: 12/26/2022] Open
Abstract
Recent preclinical studies have revealed a functionally important role for the drug efflux pump P‐glycoprotein (P‐gp) at the blood–brain barrier in limiting brain levels and thus antidepressant‐like activity of certain antidepressant drugs. Specifically, acute administration of P‐gp inhibitors, such as verapamil and cyclosporin A (CsA), has been shown to augment brain concentrations and functional activity of the antidepressant escitalopram in rodents. However, depression is a chronic disorder and current treatments require prolonged administration to elicit their full therapeutic effect. Thus, it is important to investigate whether acute findings in relation to P‐gp inhibition translate to chronic paradigms. To this end, the present study investigates whether chronic treatment with the P‐gp inhibitor verapamil and the antidepressant escitalopram results in enhanced brain distribution and antidepressant‐like effects of escitalopram. Verapamil (10 mg·kg−1 i.p.) and escitalopram (0.1 mg·kg−1 i.p.) were administered once daily for 22 days. On the final day of treatment, brain regions and plasma were collected for analysis of cortical and plasma escitalopram concentrations, and to determine the hippocampal expression of genes previously reported to be altered by chronic antidepressant treatment. Verapamil treatment resulted in a greater than twofold increase in brain levels of escitalopram, without altering plasma levels. Neither gene expression analysis nor behavioral testing revealed an augmentation of responses to escitalopram treatment due to verapamil administration. Taken together, these data demonstrate for the first time that P‐gp inhibition can yield elevated brain concentrations of an antidepressant after chronic treatment. The functional relevance of these increased brain levels requires further elaboration.
Collapse
Affiliation(s)
- Fionn E O'Brien
- APC Microbiome Institute University College Cork CorkIreland; Pharmacodelivery Group School of Pharmacy University College Cork CorkIreland; Department of Anatomy & Neuroscience University College Cork CorkIreland; Present address: UCL School of Pharmacy University College London London United Kingdom
| | - Gerard M Moloney
- Department of Anatomy & Neuroscience University College Cork Cork Ireland
| | - Karen A Scott
- Department of Anatomy & Neuroscience University College Cork Cork Ireland
| | - Richard M O'Connor
- Department of Anatomy & Neuroscience University College Cork Cork Ireland; Present address: Department of Pharmacology and Systems Therapeutics Icahn School of Medicine Mount Sinai Hospital NY USA
| | - Gerard Clarke
- APC Microbiome Institute University College Cork Cork Ireland; Department of Psychiatry University College Cork Cork Ireland
| | - Timothy G Dinan
- APC Microbiome Institute University College Cork Cork Ireland; Department of Psychiatry University College Cork Cork Ireland
| | - Brendan T Griffin
- Pharmacodelivery Group School of Pharmacy University College Cork Cork Ireland
| | - John F Cryan
- APC Microbiome Institute University College Cork Cork Ireland; Department of Anatomy & Neuroscience University College Cork Cork Ireland
| |
Collapse
|
41
|
O'Mahony SM, Clarke G, Dinan TG, Cryan JF. Early-life adversity and brain development: Is the microbiome a missing piece of the puzzle? Neuroscience 2015; 342:37-54. [PMID: 26432952 DOI: 10.1016/j.neuroscience.2015.09.068] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 12/12/2022]
Abstract
The prenatal and postnatal early-life periods are both dynamic and vulnerable windows for brain development. During these important neurodevelopmental phases, essential processes and structures are established. Exposure to adverse events that interfere with this critical sequence of events confers a high risk for the subsequent emergence of mental illness later in life. It is increasingly accepted that the gastrointestinal microbiota contributes substantially to shaping the development of the central nervous system. Conversely, several studies have shown that early-life events can also impact on this gut community. Due to the bidirectional communication between the gut and the brain, it is possible that aberrant situations affecting either organ in early life can impact on the other. Studies have now shown that deviations from the gold standard trajectory of gut microbiota establishment and development in early life can lead not only to disorders of the gastrointestinal tract but also complex metabolic and immune disorders. These are being extended to disorders of the central nervous system and understanding how the gut microbiome shapes brain and behavior during early life is an important new frontier in neuroscience.
Collapse
Affiliation(s)
- S M O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.
| | - G Clarke
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - T G Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - J F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.
| |
Collapse
|
42
|
De Palma G, Blennerhassett P, Lu J, Deng Y, Park AJ, Green W, Denou E, Silva MA, Santacruz A, Sanz Y, Surette MG, Verdu EF, Collins SM, Bercik P. Microbiota and host determinants of behavioural phenotype in maternally separated mice. Nat Commun 2015. [DOI: 10.1038/ncomms8735] [Citation(s) in RCA: 299] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
43
|
Abstract
The wide spectrum of disruptions that characterizes major depressive disorder (MDD) and bipolar disorder (BD) highlights the difficulties researchers are posed with as they try to mimic these disorders in the laboratory. Nonetheless, numerous attempts have been made to create rodent models of mood disorders or at least models of the symptoms of MDD and BD. Present antidepressants are all descendants of the serendipitous findings in the 1950s that the monoamine oxidase inhibitor iproniazid and the tricyclic antidepressant imipramine were effective antidepressants. Thus, the need for improved animal models to provide insights into the neuropathology underlying the disease is critical. Such information is in turn crucial for identifying new antidepressants and mood stabilisers. Currently, there is a shift away from traditional animal models to more focused research dealing with an endophenotype-style approach, genetic models, and incorporation of new findings from human neuroimaging and genetic studies. Such approaches are opening up more tractable avenues for understanding the neurobiological and genetic bases of these disorders. Further, such models promise to yield better translational animal models and hence more fruitful therapeutic targets. This overview focuses on such animal models and tests and how they can be used to assess MDD and BD in rodents.
Collapse
|
44
|
Investigating the mechanism(s) underlying switching between states in bipolar disorder. Eur J Pharmacol 2015; 759:151-62. [PMID: 25814263 DOI: 10.1016/j.ejphar.2015.03.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/03/2015] [Accepted: 03/12/2015] [Indexed: 12/12/2022]
Abstract
Bipolar disorder (BD) is a unique disorder that transcends domains of function since the same patient can exhibit depression or mania, states with polar opposite mood symptoms. During depression, people feel helplessness, reduced energy, and risk aversion, while with mania behaviors include grandiosity, increased energy, less sleep, and risk preference. The neural mechanism(s) underlying each state are gaining clarity, with catecholaminergic disruption seen during mania, and cholinergic dysfunction during depression. The fact that the same patient cycles/switches between these states is the defining characteristic of BD however. Of greater importance therefore, is the mechanism(s) underlying cycling from one state - and its associated neural changes - to another, considered the 'holy grail' of BD research. Herein, we review studies investigating triggers that induce switching to these states. By identifying such triggers, researchers can study neural mechanisms underlying each state and importantly how such mechanistic changes can occur in the same subject. Current animal models of this switch are also discussed, from submissive- and dominant-behaviors to kindling effects. Focus however, is placed on how seasonal changes can induce manic and depressive states in BD sufferers. Importantly, changing photoperiod lengths can induce local switches in neurotransmitter expression in normal animals, from increased catecholaminergic expression during periods of high activity, to increased somatostatin and corticotrophin releasing factor during periods of low activity. Identifying susceptibilities to this switch would enable the development of targeted animal models. From animal models, targeted treatments could be developed and tested that would minimize the likelihood of switching.
Collapse
|
45
|
Caldarone BJ, Zachariou V, King SL. Rodent models of treatment-resistant depression. Eur J Pharmacol 2014; 753:51-65. [PMID: 25460020 DOI: 10.1016/j.ejphar.2014.10.063] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/16/2014] [Accepted: 10/09/2014] [Indexed: 01/06/2023]
Abstract
Major depression is a prevalent and debilitating disorder and a substantial proportion of patients fail to reach remission following standard antidepressant pharmacological treatment. Limited efficacy with currently available antidepressant drugs highlights the need to develop more effective medications for treatment- resistant patients and emphasizes the importance of developing better preclinical models that focus on treatment- resistant populations. This review discusses methods to adapt and refine rodent behavioral models that are predictive of antidepressant efficacy to identify populations that show reduced responsiveness or are resistant to traditional antidepressants. Methods include separating antidepressant responders from non-responders, administering treatments that render animals resistant to traditional pharmacological treatments, and identifying genetic models that show antidepressant resistance. This review also examines pharmacological and non-pharmacological treatments regimes that have been effective in refractory patients and how some of these approaches have been used to validate animal models of treatment-resistant depression. The goals in developing rodent models of treatment-resistant depression are to understand the neurobiological mechanisms involved in antidepressant resistance and to develop valid models to test novel therapies that would be effective in patients that do not respond to traditional monoaminergic antidepressants.
Collapse
Affiliation(s)
- Barbara J Caldarone
- Department of Neurology, Brigham and Women's Hospital and NeuroBehavior Laboratory, Harvard NeuroDiscovery Center, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | - Venetia Zachariou
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
| | - Sarah L King
- School of Psychology, University of Sussex, Brighton, East Sussex, UK
| |
Collapse
|
46
|
Levone BR, Cryan JF, O'Leary OF. Role of adult hippocampal neurogenesis in stress resilience. Neurobiol Stress 2014; 1:147-55. [PMID: 27589664 PMCID: PMC4721321 DOI: 10.1016/j.ynstr.2014.11.003] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/30/2014] [Accepted: 11/03/2014] [Indexed: 02/04/2023] Open
Abstract
There is a growing appreciation that adult hippocampal neurogenesis plays a role in emotional and cognitive processes related to psychiatric disorders. Although many studies have investigated the effects of stress on adult hippocampal neurogenesis, most have not focused on whether stress-induced changes in neurogenesis occur specifically in animals that are more resilient or more susceptible to the behavioural and neuroendocrine effects of stress. Thus, in the present review we explore whether there is a clear relationship between stress-induced changes in adult hippocampal neurogenesis, stress resilience and antidepressant-induced recovery from stress-induced changes in behaviour. Exposure to different stressors is known to reduce adult hippocampal neurogenesis, but some stressors have also been shown to exert opposite effects. Ablation of neurogenesis does not lead to a depressive phenotype, but it can enhance responsiveness to stress and affect stress susceptibility. Monoaminergic-targeted antidepressants, environmental enrichment and adrenalectomy are beneficial for reversing stress-induced changes in behaviour and have been shown to do so in a neurogenesis-dependant manner. In addition, stress and antidepressants can affect hippocampal neurogenesis, preferentially in the ventral hippocampus. Together, these data show that adult hippocampal neurogenesis may play a role in the neuroendocrine and behavioural responses to stress, although it is not yet fully clear under which circumstances neurogenesis promotes resilience or susceptibility to stress. It will be important that future studies carefully examine how adult hippocampal neurogenesis can contribute to stress resilience/susceptibility so that it may be appropriately exploited for the development of new and more effective treatments for stress-related psychiatric disorders. Ablation of adult hippocampal NG can affect stress susceptibility. Increased hippocampal NG is associated with both susceptibility and resilience. Adult hippocampal NG can influence stress-induced alterations in HPA-axis activity. Behavioural effects of some but not all antidepressants are neurogenesis-dependent. Stress and antidepressants can affect NG preferentially in the ventral hippocampus.
Collapse
Affiliation(s)
- Brunno R Levone
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Ireland
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Ireland
| |
Collapse
|
47
|
Jackson E, Demarest K, Eckert WJ, Cates-Gatto C, Nadav T, Cates LN, Howard H, Roberts AJ. Aspen shaving versus chip bedding: effects on breeding and behavior. Lab Anim 2014; 49:46-56. [DOI: 10.1177/0023677214553320] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The choice of laboratory cage bedding material is often based on both practical and husbandry issues, whereas behavioral outcomes rarely appear to be considered. It has been noted that a breeding success difference appears to be associated with the differential use of aspen chip and aspen shaving bedding in our facility; therefore, we sought to analyze breeding records maintained over a 20-month period. In fact, in all four mouse strains analyzed, shaving bedding was associated with a significant increase in average weanlings per litter relative to chip bedding. To determine whether these bedding types also resulted in differences in behaviors associated with wellbeing, we examined nest building, anxiety-like, depressive-like (or helpless-like), and social behavior in mice housed on chip versus shaving bedding. We found differences in the nests built, but no overall effect of bedding type on the other behaviors examined. Therefore, we argue that breeding success, perhaps especially in more challenging strains, is improved on shaving bedding and this is likely due to improved nest-building potential. For standard laboratory practices, however, these bedding types appear equivalent.
Collapse
Affiliation(s)
- E Jackson
- Mouse Behavioral Assessment Core Facility, The Scripps Research Institute, LA Jolla, CA, USA
| | - K Demarest
- Department of Animal Resources, The Scripps Research Institute, La Jolla, CA, USA
| | - W J Eckert
- Department of Animal Resources, The Scripps Research Institute, La Jolla, CA, USA
| | - C Cates-Gatto
- Mouse Behavioral Assessment Core Facility, The Scripps Research Institute, LA Jolla, CA, USA
| | - T Nadav
- Mouse Behavioral Assessment Core Facility, The Scripps Research Institute, LA Jolla, CA, USA
| | - L N Cates
- Mouse Behavioral Assessment Core Facility, The Scripps Research Institute, LA Jolla, CA, USA
| | - H Howard
- Department of Animal Resources, The Scripps Research Institute, La Jolla, CA, USA
| | - A J Roberts
- Mouse Behavioral Assessment Core Facility, The Scripps Research Institute, LA Jolla, CA, USA
| |
Collapse
|
48
|
O'Leary OF, O'Brien FE, O'Connor RM, Cryan JF. Drugs, genes and the blues: Pharmacogenetics of the antidepressant response from mouse to man. Pharmacol Biochem Behav 2014; 123:55-76. [PMID: 24161683 DOI: 10.1016/j.pbb.2013.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 10/04/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022]
|
49
|
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: 90] [Impact Index Per Article: 8.2] [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
|
50
|
Blanchard DC, Summers CH, Blanchard RJ. The role of behavior in translational models for psychopathology: functionality and dysfunctional behaviors. Neurosci Biobehav Rev 2013; 37:1567-77. [PMID: 23791787 PMCID: PMC3800172 DOI: 10.1016/j.neubiorev.2013.06.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 05/31/2013] [Accepted: 06/10/2013] [Indexed: 01/05/2023]
Abstract
The history of science has frequently included a problem-based impetus toward research that can be translated expeditiously into solutions. A current problem is that psychopathologies, typically chronic, contribute hugely to the economic and social burden of medical care, especially in the United States. For behavioral neuroscientists a psychopathology-aimed translational research emphasis particularly involves animal models to facilitate the experimental and invasive work necessary to an understanding of the biology of normal and aberrant behavior. When the etiology of a particular psychopathology is unknown, and there are no specific biomarkers, behavioral parallels between the focal disorder and its putative models become crucial elements in assessing model validity. Evaluation of these parallels is frequently neglected, reflecting in part the lack of a systematic conceptualization of the organization of behavior and how this may be conserved across species. Recent work specifically attempting to bridge this gap suggests that analysis of behaviors that are functional - adaptive in crucial situations such as danger or social contexts - can facilitate an understanding of the parallels between behaviors of human and nonhuman species, including the dysfunctional behaviors of psycho pathologies. As research with animal models comes to provide a more systematic analysis of particular behaviors and their adaptive functions, cross-talk between model and focal psychopathology may be advantageous to understanding both.
Collapse
Affiliation(s)
- D Caroline Blanchard
- Pacific Biosciences Research Center, 1993 East West Road, Honolulu, HI 96822, USA.
| | | | | |
Collapse
|