1
|
Yao C, Jiang N, Sun X, Zhang Y, Pan R, He Q, Chang Q, Liu X. Effects of inulin-type oligosaccharides (JSO) from Cichorium intybus L. on behavioral deficits induced by chronic restraint stress in mice and associated molecular alterations. Front Pharmacol 2024; 15:1484337. [PMID: 39555096 PMCID: PMC11563967 DOI: 10.3389/fphar.2024.1484337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
Depression and anxiety are serious psychiatric disorders with significant physical and mental health impacts, necessitating the development of safe and effective treatments. This study aimed to evaluate the efficacy of Jiangshi oligosaccharide (JSO), a type of inulin-based oligosaccharide, in alleviating anxiety and depression and to investigate the underlying molecular mechanisms. Using a mouse model of chronic restraint stress (CRS), JSO was administered orally at doses of 50, 100, and 200 mg/kg for 21 days. Behavioral tests, including the novelty-suppressed feeding test (NSFT), open field test (OFT), elevated plus maze test (EPMT), tail suspension test (TST), and forced swimming test (FST), demonstrated that JSO significantly improved anxiety- and depressive-like behaviors (P< 0.05). Notably, JSO reduced feeding latency in the NSFT, increased time spent in the center in the OFT, enhanced time and entries into open arms in the EPMT, and decreased immobility time in the TST and FST (P< 0.01). Histological and molecular analyses revealed that JSO treatment attenuated neuronal loss in the hippocampus (Hip) and medial prefrontal cortex (mPFC) and reduced the expression of inflammatory markers such as Iba-1 and GFAP in these regions. JSO significantly downregulated the mRNA and protein expression of pro-inflammatory factors (IL-1β, TNF-α, IL-6) while increasing anti-inflammatory markers (IL-10, TGF-β) (P< 0.05). Furthermore, JSO inhibited the c-GAS-STING-NLRP3 axis and apoptosis-related proteins (Bax/Bcl-2, Caspase-3/8/9) while promoting the expression of brain-derived neurotrophic factor (BDNF), PSD-95, and synaptophysin (SYP), indicating improved neuronal survival and synaptic plasticity (P< 0.01). These findings suggest that JSO exerts potent anti-anxiety and antidepressant effects by modulating neuroinflammation, synaptic function, and neuronal apoptosis in the Hip and mPFC of CRS mice. This study highlighted JSO as a potential therapeutic agent for stress-induced anxiety and depression.
Collapse
Affiliation(s)
- Caihong Yao
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Jiang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinran Sun
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiwen Zhang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruile Pan
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinghu He
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qi Chang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinmin Liu
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| |
Collapse
|
2
|
Ratajczak P, Martyński J, Zięba JK, Świło K, Kopciuch D, Paczkowska A, Zaprutko T, Kus K. Comparative Efficacy of Animal Depression Models and Antidepressant Treatment: A Systematic Review and Meta-Analysis. Pharmaceutics 2024; 16:1144. [PMID: 39339181 PMCID: PMC11435171 DOI: 10.3390/pharmaceutics16091144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Animal models are critical tools in the study of psychiatric disorders; however, none of the current models fully reflect human stress-related disorders, even though most of the knowledge about the mechanisms of depression comes from animal studies. Animal studies are useful in pharmacological research, whereby we can obtain results that translate into patient treatment by controlling environmental factors, especially in behavioural research. The authors systematically reviewed this issue since medical databases provide access to many primary studies. METHODS A systematic review and meta-analysis were conducted based on 25 primary studies. The studies were identified in databases such as PubMed, Embase, and Web of Science (December 2022) according to the inclusion and exclusion criteria established at the beginning of the research and published in the form of a protocol, following the PRISMA and Cochrane Collaboration methodology for secondary studies and CAMARADES (CAMARADES Berlin, QUEST-BIH Charité) for secondary studies on animals. Forest plot analyses were performed (data presented as Mean Difference, Random Model, Inverse Variance), Risk of Bias assessment (Systematic Review Center for Laboratory animal Experimentation (SYRCLE) evaluation), quality assessment of included studies (Animal research: Reporting of In Vivo Experiments (ARRIVE)), and a range of data from source publications were compiled in tabular form. The study analysed the popularity of both animal depression models (ADM) and rat strains used in pharmacological research to test the efficacy of antidepressant drugs based on the immobility time (IT) factor (Forced Swimming Test). The study examined selective serotonin reuptake inhibitors, namely fluoxetine, sertraline, paroxetine, citalopram, and escitalopram. Additionally, the study addressed issues concerning the "data availability statement" because precise IT data analysis was impossible in the case of 212 papers. RESULTS Our data confirm that the Chronic Unpredictable Mild Stress (CUMS) model is the most popular and versatile model used in preclinical depression research, while the two most popular rat strains were Wistar and Sprague-Dawley. The quality of included papers based on the ARRIVE assessment showed a ratio value equal to 0.63, meaning that studies were of intermediate overall quality. The Risk of Bias assessment based on the SYRCLE tool revealed a high risk related to the blinding and the random outcome assessment. In the meta-analysis, the results indicate that all analysed drugs demonstrated efficacy in reducing IT, and the most analysed drug was fluoxetine (confirmed based on 17 studies (19 models)). The analysis of the efficacy of ADMs showed that the most effective models were CUMS, Flinders Sensitive Line (genetic model), Social Isolation, Restraint Stress, and Low-dose Lipopolysaccharide (pharmacological model). Only 2.35% (5 out of 212) of corresponding authors responded to our data request. CONCLUSIONS The study highlights the dominance of the CUMS model and the Wistar and Sprague-Dawley rat strains in preclinical depression research, affirming the efficacy of SSRIs, particularly fluoxetine, in reducing IT. The findings underscore the need for better data availability and methodological improvements despite intermediate overall study quality and notable bias risks. Enhanced transparency and rigorous assessment standards are essential for advancing the reliability of animal models in depression research.
Collapse
Affiliation(s)
- Piotr Ratajczak
- Department of Pharmacoeconomics and Social Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Jakub Martyński
- Department of Pharmacoeconomics and Social Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Jan Kazimierz Zięba
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznan, Poland
| | - Katarzyna Świło
- Department of Pharmacoeconomics and Social Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Dorota Kopciuch
- Department of Pharmacoeconomics and Social Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Anna Paczkowska
- Department of Pharmacoeconomics and Social Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Tomasz Zaprutko
- Department of Pharmacoeconomics and Social Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| | - Krzysztof Kus
- Department of Pharmacoeconomics and Social Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland
| |
Collapse
|
3
|
Murck H, Karailiev P, Karailievova L, Puhova A, Jezova D. Treatment with Glycyrrhiza glabra Extract Induces Anxiolytic Effects Associated with Reduced Salt Preference and Changes in Barrier Protein Gene Expression. Nutrients 2024; 16:515. [PMID: 38398838 PMCID: PMC10893552 DOI: 10.3390/nu16040515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
We have previously identified that low responsiveness to antidepressive therapy is associated with higher aldosterone/cortisol ratio, lower systolic blood pressure, and higher salt preference. Glycyrrhiza glabra (GG) contains glycyrrhizin, an inhibitor of 11β-hydroxysteroid-dehydrogenase type-2 and antagonist of toll-like receptor 4. The primary hypothesis of this study is that food enrichment with GG extract results in decreased anxiety behavior and reduced salt preference under stress and non-stress conditions. The secondary hypothesis is that the mentioned changes are associated with altered gene expression of barrier proteins in the prefrontal cortex. Male Sprague-Dawley rats were exposed to chronic mild stress for five weeks. Both stressed and unstressed rats were fed a diet with or without an extract of GG roots for the last two weeks. GG induced anxiolytic effects in animals independent of stress exposure, as measured in elevated plus maze test. Salt preference and intake were significantly reduced by GG under control, but not stress conditions. The gene expression of the barrier protein claudin-11 in the prefrontal cortex was increased in control rats exposed to GG, whereas stress-induced rise was prevented. Exposure to GG-enriched diet resulted in reduced ZO-1 expression irrespective of stress conditions. In conclusion, the observed effects of GG are in line with a reduction in the activity of central mineralocorticoid receptors. The treatment with GG extract or its active components may, therefore, be a useful adjunct therapy for patients with subtypes of depression and anxiety disorders with heightened renin-angiotensin-aldosterone system and/or inflammatory activity.
Collapse
Affiliation(s)
- Harald Murck
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, 35039 Marburg, Germany
| | - Peter Karailiev
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Lucia Karailievova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Agnesa Puhova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| | - Daniela Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovakia; (P.K.); (L.K.); (A.P.); (D.J.)
| |
Collapse
|
4
|
Knezevic E, Nenic K, Milanovic V, Knezevic NN. The Role of Cortisol in Chronic Stress, Neurodegenerative Diseases, and Psychological Disorders. Cells 2023; 12:2726. [PMID: 38067154 PMCID: PMC10706127 DOI: 10.3390/cells12232726] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
Cortisol, a critical glucocorticoid hormone produced by the adrenal glands, plays a pivotal role in various physiological processes. Its release is finely orchestrated by the suprachiasmatic nucleus, governing the circadian rhythm and activating the intricate hypothalamic-pituitary-adrenal (HPA) axis, a vital neuroendocrine system responsible for stress response and maintaining homeostasis. Disruptions in cortisol regulation due to chronic stress, disease, and aging have profound implications for multiple bodily systems. Animal models have been instrumental in elucidating these complex cortisol dynamics during stress, shedding light on the interplay between physiological, neuroendocrine, and immune factors in the stress response. These models have also revealed the impact of various stressors, including social hierarchies, highlighting the role of social factors in cortisol regulation. Moreover, chronic stress is closely linked to the progression of neurodegenerative diseases, like Alzheimer's and Parkinson's, driven by excessive cortisol production and HPA axis dysregulation, along with neuroinflammation in the central nervous system. The relationship between cortisol dysregulation and major depressive disorder is complex, characterized by HPA axis hyperactivity and chronic inflammation. Lastly, chronic pain is associated with abnormal cortisol patterns that heighten pain sensitivity and susceptibility. Understanding these multifaceted mechanisms and their effects is essential, as they offer insights into potential interventions to mitigate the detrimental consequences of chronic stress and cortisol dysregulation in these conditions.
Collapse
Affiliation(s)
- Emilija Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (E.K.); (K.N.); (V.M.)
- College of Liberal Arts and Sciences, University of Illinois at Urbana-Champaign, Champaign, IL 61820, USA
| | - Katarina Nenic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (E.K.); (K.N.); (V.M.)
- Department of Psychology, University of Central Florida, Orlando, FL 32826, USA
| | - Vladislav Milanovic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (E.K.); (K.N.); (V.M.)
- College of Medicine Rockford, University of Illinois, Rockford, IL 61107, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL 60657, USA; (E.K.); (K.N.); (V.M.)
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
5
|
Boyle CC, Bower JE, Eisenberger NI, Irwin MR. Stress to inflammation and anhedonia: Mechanistic insights from preclinical and clinical models. Neurosci Biobehav Rev 2023; 152:105307. [PMID: 37419230 DOI: 10.1016/j.neubiorev.2023.105307] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Anhedonia, as evidenced by impaired pleasurable response to reward, reduced reward motivation, and/or deficits in reward-related learning, is a common feature of depression. Such deficits in reward processing are also an important clinical target as a risk factor for depression onset. Unfortunately, reward-related deficits remain difficult to treat. To address this gap and inform the development of effective prevention and treatment strategies, it is critical to understand the mechanisms that drive impairments in reward function. Stress-induced inflammation is a plausible mechanism of reward deficits. The purpose of this paper is to review evidence for two components of this psychobiological pathway: 1) the effects of stress on reward function; and 2) the effects of inflammation on reward function. Within these two areas, we draw upon preclinical and clinical models, distinguish between acute and chronic effects of stress and inflammation, and address specific domains of reward dysregulation. By addressing these contextual factors, the review reveals a nuanced literature which might be targeted for additional scientific inquiry to inform the development of precise interventions.
Collapse
Affiliation(s)
- Chloe C Boyle
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA.
| | - Julienne E Bower
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA; Department of Psychology, UCLA, Los Angeles, CA, USA
| | | | - Michael R Irwin
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA
| |
Collapse
|
6
|
Campeau S, McNulty C, Stanley JT, Gerber AN, Sasse SK, Dowell RD. Determination of steady-state transcriptome modifications associated with repeated homotypic stress in the rat rostral posterior hypothalamic region. Front Neurosci 2023; 17:1173699. [PMID: 37360161 PMCID: PMC10288150 DOI: 10.3389/fnins.2023.1173699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Chronic stress is epidemiologically correlated with physical and psychiatric disorders. Whereas many animal models of chronic stress induce symptoms of psychopathology, repeated homotypic stressors to moderate intensity stimuli typically reduce stress-related responses with fewer, if any, pathological symptoms. Recent results indicate that the rostral posterior hypothalamic (rPH) region is a significant component of the brain circuitry underlying response reductions (habituation) associated with repeated homotypic stress. To test whether posterior hypothalamic transcriptional regulation associates with the neuroendocrine modifications induced by repeated homotypic stress, RNA-seq was performed in the rPH dissected from adult male rats that experienced either no stress, 1, 3, or 7 stressful loud noise exposures. Plasma samples displayed reliable increases of corticosterone in all stressed groups, with the smallest increase in the group exposed to 7 loud noises, indicating significant habituation compared to the other stressed groups. While few or no differentially expressed genes were detected 24-h after one or three loud noise exposures, relatively large numbers of transcripts were differentially expressed between the group exposed to 7 loud noises when compared to the control or 3-stress groups, respectively, which correlated with the corticosterone response habituation observed. Gene ontology analyses indicated multiple significant functional terms related to neuron differentiation, neural membrane potential, pre- and post-synaptic elements, chemical synaptic transmission, vesicles, axon guidance and projection, glutamatergic and GABAergic neurotransmission. Some of the differentially expressed genes (Myt1l, Zmat4, Dlx6, Csrnp3) encode transcription factors that were independently predicted by transcription factor enrichment analysis to target other differentially regulated genes in this study. A similar experiment employing in situ hybridization histochemical analysis in additional animals validated the direction of change of the 5 transcripts investigated (Camk4, Gabrb2, Gad1, Grin2a and Slc32a) with a high level of temporal and regional specificity for the rPH. In aggregate, the results suggest that distinct patterns of gene regulation are obtained in response to a repeated homotypic stress regimen; they also point to a significant reorganization of the rPH region that may critically contribute to the phenotypic modifications associated with repeated homotypic stress habituation.
Collapse
Affiliation(s)
- Serge Campeau
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States
| | - Connor McNulty
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States
| | - Jacob T. Stanley
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States
| | - Anthony N. Gerber
- Department of Medicine, National Jewish Health, Denver, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Sarah K. Sasse
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Robin D. Dowell
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
- BioFrontiers Institute, University of Colorado, Boulder, CO, United States
- Department of Computer Science, University of Colorado, Boulder, CO, United States
| |
Collapse
|
7
|
Elias E, Zhang AY, White AG, Pyle MJ, Manners MT. Voluntary wheel running promotes resilience to the behavioral effects of unpredictable chronic mild stress in male and female mice. Stress 2023; 26:2203769. [PMID: 37125617 DOI: 10.1080/10253890.2023.2203769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Affiliation(s)
- Elias Elias
- Department of Biology. College of Arts and Sciences. St Joseph's University, Philadelphia, PA 19131, United States
- Graduate Program in Cell and Molecular Biology
| | - Ariel Y Zhang
- Department of Biology. College of Arts and Sciences. St Joseph's University, Philadelphia, PA 19131, United States
- Graduate Program in Cell and Molecular Biology
| | - Abigail G White
- Department of Biology. College of Arts and Sciences. St Joseph's University, Philadelphia, PA 19131, United States
- Program in Neuroscience
| | - Matthew J Pyle
- Department of Biology. College of Arts and Sciences. St Joseph's University, Philadelphia, PA 19131, United States
| | - Melissa T Manners
- Department of Biology. College of Arts and Sciences. St Joseph's University, Philadelphia, PA 19131, United States
- Graduate Program in Cell and Molecular Biology
- Program in Neuroscience
- Department of Biological and Biomedical Sciences. College of Science and Mathematics. Rowan University, Glassboro, New Jersey 08028, United States
| |
Collapse
|
8
|
Leite JA, Orellana AM, Andreotti DZ, Matumoto AM, de Souza Ports NM, de Sá Lima L, Kawamoto EM, Munhoz CD, Scavone C. Ouabain Reverts CUS-Induced Disruption of the HPA Axis and Avoids Long-Term Spatial Memory Deficits. Biomedicines 2023; 11:biomedicines11041177. [PMID: 37189795 DOI: 10.3390/biomedicines11041177] [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: 02/13/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 05/17/2023] Open
Abstract
Ouabain (OUA) is a cardiotonic steroid that modulates Na+, K+ -ATPase activity. OUA has been identified as an endogenous substance that is present in human plasma, and it has been shown to be associated with the response to acute stress in both animals and humans. Chronic stress is a major aggravating factor in psychiatric disorders, including depression and anxiety. The present work investigates the effects of the intermittent administration of OUA (1.8 μg/kg) during the chronic unpredictable stress (CUS) protocol in a rat's central nervous system (CNS). The results suggest that the intermittent OUA treatment reversed CUS-induced HPA axis hyperactivity through a reduction in (i) glucocorticoids levels, (ii) CRH-CRHR1 expression, and by decreasing neuroinflammation with a reduction in iNOS activity, without interfering with the expression of antioxidant enzymes. These changes in both the hypothalamus and hippocampus may reflect in the rapid extinction of aversive memory. The present data demonstrate the ability of OUA to modulate the HPA axis, as well as to revert CUS-induced long-term spatial memory deficits.
Collapse
Affiliation(s)
- Jacqueline Alves Leite
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia 74690-900, Brazil
| | - Ana Maria Orellana
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Diana Zukas Andreotti
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Amanda Midori Matumoto
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Larissa de Sá Lima
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Elisa Mitiko Kawamoto
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Carolina Demarchi Munhoz
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Cristoforo Scavone
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
9
|
Murck H, Lehr L, Jezova D. A viewpoint on aldosterone and BMI related brain morphology in relation to treatment outcome in patients with major depression. J Neuroendocrinol 2023; 35:e13219. [PMID: 36539978 DOI: 10.1111/jne.13219] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/06/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
An abundance of knowledge has been collected describing the involvement of neuroendocrine parameters in major depression. The hypothalamic-pituitary-adrenocortical (HPA) axis regulating cortisol release has been extensively studied; however, attempts to target the HPA axis pharmacologically to treat major depression have failed. This review focuses on the importance of the adrenocortical stress hormone aldosterone, which is released by adrenocorticotropic hormone and angiotensin, and the mineralocorticoid receptor (MR) in depression. Depressed patients, in particular those with atypical depression, have signs of central hyperactivation of the aldosterone sensitive MR, potentially as a consequence of a reactive aldosterone release induced by low blood pressure and as a result of low sensitivity of peripheral MR. This is reflected in reduced heart rate variability, increased salt appetite and sleep changes in this group of patients. In addition, enlarged brain ventricles, compressed corpus callosum and changes of the choroid plexus are associated with increased aldosterone (in relation to cortisol). Furthermore, subjects with these features often show obesity. These characteristics are related to a worse antidepressant treatment outcome. Alterations in choroid plexus function as a consequence of increased aldosterone levels, autonomic dysregulation, metabolic changes and/or inflammation may be involved. The characterization of this regulatory system is in its early days but may identify new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Harald Murck
- Philipps-University Marburg, Marburg, Germany
- Murck-Neuroscience LLC Westfield, Westfield, NJ, USA
| | - Lisa Lehr
- Department of Nephrology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Daniela Jezova
- Slovak Academy of Sciences, Biomedical Research Center, Institute of Experimental Endocrinology, Bratislava, Slovakia
| |
Collapse
|
10
|
Sammer G, Neumann E, Blecker C, Pedraz-Petrozzi B. Fractional anisotropy and peripheral cytokine concentrations in outpatients with depressive episode: a diffusion tensor imaging observational study. Sci Rep 2022; 12:17450. [PMID: 36261698 PMCID: PMC9582033 DOI: 10.1038/s41598-022-22437-0] [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: 02/16/2022] [Accepted: 10/14/2022] [Indexed: 01/12/2023] Open
Abstract
Over the past few years, evidence of a positive relationship between inflammation and depression has grown steadily. The aim of the current study was to investigate whether such depression-related inflammation could also be associated with altered microstructural changes in the white matter. FA and serum cytokines (IL-1β, IL-6, TNF-α, and IFN-γ) were measured in 25 patients with depression (DE) and 24 healthy controls (HC). Diffusion tensor imaging was performed. Fractional anisotropy (FA) was calculated using the FSL pipeline for Tract-Based Spatial Statistics (TBSS). Both voxelwise and mean whole-brain FA were analyzed using general linear models (GLM). Higher concentrations of IL-1β were associated with lower whole-brain fractional anisotropy, particularly in people with depression (ρ = - 0.67; p < 0.001). TNF-α shared some variance with IL-1β and also showed a negative relationship between TNF-α concentrations and FA in depression (F1,46 = 11.13, p = 0.002, η2p = 0.21). In detail, the voxelwise analysis showed that the regression slopes of IL-1β on FA were more negative in the DE group than in the HC group, mainly in the corpus callosum (cluster statistics: genu corpus callosum, p = 0.022; splenium of corpus callosum, p = 0.047). Similar effects were not found for the other remaining cytokines. This study clearly demonstrated an association between peripherally measured IL-1β and white matter integrity in depression as assessed by DTI. The results suggest that microstructural changes in the corpus callosum are associated with increased peripheral IL-1β concentrations in depression.
Collapse
Affiliation(s)
- Gebhard Sammer
- grid.8664.c0000 0001 2165 8627Psychiatry, Justus Liebig University Giessen, Klinikstrasse 36, 35392 Giessen, Hessen Germany ,grid.8664.c0000 0001 2165 8627Faculty of Psychology and Sports Science, Justus Liebig University Giessen, Giessen, Hessen Germany ,grid.8664.c0000 0001 2165 8627Bender Institute of Neuroimaging (BION), Faculty of Psychology and Sports Science, Justus Liebig University Giessen, Giessen, Hessen Germany
| | - Elena Neumann
- grid.8664.c0000 0001 2165 8627Internal Medicine and Rheumatology, Campus Kerckhoff, Justus Liebig University Giessen, Giessen, Hessen Germany
| | - Carlo Blecker
- grid.8664.c0000 0001 2165 8627Faculty of Psychology and Sports Science, Justus Liebig University Giessen, Giessen, Hessen Germany ,grid.8664.c0000 0001 2165 8627Bender Institute of Neuroimaging (BION), Faculty of Psychology and Sports Science, Justus Liebig University Giessen, Giessen, Hessen Germany
| | - Bruno Pedraz-Petrozzi
- grid.413757.30000 0004 0477 2235Central Institute of Mental Health, Mannheim, Germany
| |
Collapse
|
11
|
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: 49] [Impact Index Per Article: 16.3] [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
|
12
|
Petković A, Chaudhury D. Encore: Behavioural animal models of stress, depression and mood disorders. Front Behav Neurosci 2022; 16:931964. [PMID: 36004305 PMCID: PMC9395206 DOI: 10.3389/fnbeh.2022.931964] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Animal studies over the past two decades have led to extensive advances in our understanding of pathogenesis of depressive and mood disorders. Among these, rodent behavioural models proved to be of highest informative value. Here, we present a comprehensive overview of the most popular behavioural models with respect to physiological, circuit, and molecular biological correlates. Behavioural stress paradigms and behavioural tests are assessed in terms of outcomes, strengths, weaknesses, and translational value, especially in the domain of pharmacological studies.
Collapse
Affiliation(s)
| | - Dipesh Chaudhury
- Laboratory of Neural Systems and Behaviour, Department of Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
13
|
Abdelmeguid NE, Hammad TM, Abdel-Moneim AM, Salam SA. Effect of Epigallocatechin-3-gallate on Stress-Induced Depression in a Mouse Model: Role of Interleukin-1β and Brain-Derived Neurotrophic Factor. Neurochem Res 2022; 47:3464-3475. [PMID: 35939172 PMCID: PMC9546794 DOI: 10.1007/s11064-022-03707-9] [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: 03/13/2022] [Revised: 07/02/2022] [Accepted: 07/20/2022] [Indexed: 11/29/2022]
Abstract
Epigallocatechin 3-gallate (EGCG) is a natural polyphenolic antioxidant in green tea leaves with well-known health-promoting properties. However, the influence of EGCG on a chronic animal model of depression remains to be fully investigated, and the details of the molecular and cellular changes are still unclear. Therefore, the present study aimed to investigate the antidepressant effect of EGCG in mice subjected to chronic unpredictable mild stress (CUMS). After eight consecutive weeks of CUMS, the mice were treated with EGCG (200 mg/kg b.w.) by oral gavage for two weeks. A forced swimming test (FST) was used to assess depressive symptoms. EGCG administration significantly alleviated CUMS-induced depression-like behavior in mice. EGCG also effectively decreased serum interleukin-1β (IL-1β) and increased the mRNA expression levels of brain-derived neurotrophic factor (BDNF) in the hippocampal CA3 region of CUMS mice. Furthermore, electron microscopic examination of CA3 neurons in CUMS mice showed morphological features of apoptosis, loss or disruption of the myelin sheath, and degenerating synapses. These neuronal injuries were diminished with the administration of EGCG. The treatment effect of EGCG in CUMS-induced behavioral alterations was comparable with that of clomipramine hydrochloride (Anafranil), a tricyclic antidepressant drug. In conclusion, our study demonstrates that the antidepressive action of EGCG involves downregulation of serum IL-1β, upregulation of BDNF mRNA in the hippocampus, and reduction of CA3 neuronal lesions.
Collapse
Affiliation(s)
- Nabila E Abdelmeguid
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Tasneem M Hammad
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.,Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University, Alexandria, Egypt
| | - Ashraf M Abdel-Moneim
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt
| | - Sherine Abdel Salam
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| |
Collapse
|
14
|
Luo X, Zhou Z, Wu J, Zhang L, Zhang J, Li J. Integrated RNA- and miRNA-sequencing analysis identifies molecular basis for stress-induced heart injury in mouse models. Int J Cardiol 2021; 349:115-122. [PMID: 34883141 DOI: 10.1016/j.ijcard.2021.11.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Work stress and its contribution to cardiovascular diseases are well documented in recent years, but its molecular mechanisms are still not clear. In this study, we aimed to explore the potential pathophysiological mechanisms of stress-induced heart injury in mouse models. METHODS The RNA- and miRNA- sequencing profiles from five stress-treated mice and five control mice were performed. After normalization, differentially expressed genes (DEGs) and miRNAs (DEmiRs) were identified using the edgeR method. Then, based on the functional enrichment analysis and protein-protein interaction (PPI) network, as well as miRNA-mRNA interactome, the core DEGs and DEmiRs associated with stress-induced heart injury were marked and validated by qPCR, and the DEmiR targets were validated in vitro. RESULTS A total of 293 genes and 29 miRNAs were identified as DEGs and DEmiRs respectively, and Alb, Stat1, C3, Irf7, Usp18 were hub genes in the PPI network. The enrichment pathways were related to inflammation and immune, coagulation, oxidative phosphorylation, vascular development, cell cycle and extracellular matrix (ECM), which likely mediated the biological injury processes or reflected the results of damage. The target DEGs of DEmiRs were clustered in angiogenesis, immune system process and ECM. After the validation in vitro, we found that miR-29b-3p mimics could down-regulate the expression of its predicted targets, Pxdn and Col15a1. CONCLUSIONS The findings revealed a molecular basis from the gene and miRNA level for the heart injury associated with stress. miR-29b-3p, as a potential target to repair stress-induced ECM disorder in heart, deserves further study.
Collapse
Affiliation(s)
- Xiaoli Luo
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Zhitong Zhou
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Jiawen Wu
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China
| | - Lijuan Zhang
- Tongji University School of Medicine, Shanghai, China
| | - Jie Zhang
- Tongji University School of Medicine, Shanghai, China.
| | - Jue Li
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
15
|
Doremus-Fitzwater TL, Deak T. Adolescent neuroimmune function and its interaction with alcohol. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:167-208. [PMID: 34801169 DOI: 10.1016/bs.irn.2021.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Adolescence is an evolutionarily conserved developmental period associated with behavioral change, including increased risk-taking and alcohol use. Experimentation with alcohol typically begins in adolescence and transitions to binge-like patterns of consumption. Alcohol exposure during adolescence can alter normative changes in brain structure and function. Understanding mechanisms by which ethanol impacts neurodevelopmental processes is important for preventing and ameliorating the deleterious consequences of adolescent alcohol abuse. This review focuses on the neuroimmune system as a key contributor to ethanol-induced changes in adolescent brain and behavior. After brief review of neuroimmune system development, acute and chronic effects of ethanol on adolescent neuroimmune functioning are addressed. Comparisons between stress/immunological challenges and ethanol on adolescent neuroimmunity are reviewed, as cross-sensitization is relevant during adolescence. The mechanisms by which ethanol alters neuroimmune functioning are then discussed, as they may portend development of neuropathological consequences and thus increase vulnerability to subsequent challenges and potentiate addictive behaviors.
Collapse
Affiliation(s)
- T L Doremus-Fitzwater
- Department of Psychology, Ithaca College, Ithaca, NY, United States; Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States.
| | - T Deak
- Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States; Binghamton University-SUNY, Binghamton, NY, United States
| |
Collapse
|
16
|
Chen L, Wang X, Zhang Y, Zhong H, Wang C, Gao P, Li B. Daidzein Alleviates Hypothalamic-Pituitary-Adrenal Axis Hyperactivity, Ameliorates Depression-Like Behavior, and Partly Rectifies Circulating Cytokine Imbalance in Two Rodent Models of Depression. Front Behav Neurosci 2021; 15:671864. [PMID: 34733143 PMCID: PMC8559531 DOI: 10.3389/fnbeh.2021.671864] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/16/2021] [Indexed: 11/29/2022] Open
Abstract
Depression is one very common mental health disorder which can cause morbidity and mortality if not addressed. Recent studies have provided strong evidence that depression may be accompanied by immune activation, secondary inflammatory reaction, and hyperactivity of the Hypothalamic Pituitary Adrenal (HPA) axis. It is well-known that it takes at least 2 weeks for conventional antidepressants, especially SSRIs (Selective serotonin reuptake inhibitors) to produce effects. To better understand the mechanism of antidepressant effects on depression and subsequently further elucidate the pathogenesis of depression, we selected phytestrogen daidzein (DD) to observe its effects on the depression-like and anxiety-like behavior in two different rodent models of depression which were induced by learned helplessness and chronic mild stress (CMS) and then simultaneous evaluation of the depression-like behavior, the activity of HPA axis, and circulatory cytokines. Our results showed that daidzein attenuated depression-like behaviors through alleviating HPA axis hyperactivity, decreasing the levels of stress-related hormones, and partly rectifying some inflammatory cytokines imbalance in both the rodent models of depression.
Collapse
Affiliation(s)
- Long Chen
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaokun Wang
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Yunpeng Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Hequan Zhong
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Cuiting Wang
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Pengfei Gao
- Department of Traditional Chinese Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Bing Li
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
17
|
MacLellan A, Fureix C, Polanco A, Mason G. Can animals develop depression? An overview and assessment of ‘depression-like’ states. BEHAVIOUR 2021. [DOI: 10.1163/1568539x-bja10132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abstract
Describing certain animal behaviours as ‘depression-like’ or ‘depressive’ has become common across several fields of research. These typically involve unusually low activity or unresponsiveness and/or reduced interest in pleasure (anhedonia). While the term ‘depression-like’ carefully avoids directly claiming that animals are depressed, this narrative review asks whether stronger conclusions can be legitimate, with animals developing the clinical disorder as seen in humans (cf., DSM-V/ICD-10). Here, we examine evidence from animal models of depression (especially chronically stressed rats) and animals experiencing poor welfare in conventional captive conditions (e.g., laboratory mice and production pigs in barren environments). We find troubling evidence that animals are indeed capable of experiencing clinical depression, but demonstrate that a true diagnosis has yet to be confirmed in any case. We thus highlight the importance of investigating the co-occurrence of depressive criteria and discuss the potential welfare and ethical implications of animal depression.
Collapse
Affiliation(s)
- Aileen MacLellan
- Department of Integrative Biology, University of Guelph, Guelph, ON, Canada N1G 2W1
| | - Carole Fureix
- Bristol Veterinary School, University of Bristol, Langford, UK
| | - Andrea Polanco
- Department of Integrative Biology, University of Guelph, Guelph, ON, Canada N1G 2W1
| | - Georgia Mason
- Department of Integrative Biology, University of Guelph, Guelph, ON, Canada N1G 2W1
| |
Collapse
|
18
|
Inflammation, Anxiety, and Stress in Attention-Deficit/Hyperactivity Disorder. Biomedicines 2021; 9:biomedicines9101313. [PMID: 34680430 PMCID: PMC8533349 DOI: 10.3390/biomedicines9101313] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a prevalent and serious neurodevelopmental disorder characterized by symptoms of inattention and/or hyperactivity/impulsivity. Chronic and childhood stress is involved in ADHD development, and ADHD is highly comorbid with anxiety. Similarly, inflammatory diseases and a pro-inflammatory state have been associated with ADHD. However, while several works have studied the relationship between peripheral inflammation and stress in affective disorders such as depression or bipolar disorder, fewer have explored this association in ADHD. In this narrative review we synthetize evidence showing an interplay between stress, anxiety, and immune dysregulation in ADHD, and we discuss the implications of a potential disrupted neuroendocrine stress response in ADHD. Moreover, we highlight confounding factors and limitations of existing studies on this topic and critically debate multidirectional hypotheses that either suggest inflammation, stress, or anxiety as a cause in ADHD pathophysiology or inflammation as a consequence of this disease. Untangling these relationships will have diagnostic, therapeutic and prognostic implications for ADHD patients.
Collapse
|
19
|
Qiu W, Cai X, Zheng C, Qiu S, Ke H, Huang Y. Update on the Relationship Between Depression and Neuroendocrine Metabolism. Front Neurosci 2021; 15:728810. [PMID: 34531719 PMCID: PMC8438205 DOI: 10.3389/fnins.2021.728810] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/11/2021] [Indexed: 12/27/2022] Open
Abstract
Through the past decade of research, the correlation between depression and metabolic diseases has been noticed. More and more studies have confirmed that depression is comorbid with a variety of metabolic diseases, such as obesity, diabetes, metabolic syndrome and so on. Studies showed that the underlying mechanisms of both depression and metabolic diseases include chronic inflammatory state, which is significantly related to the severity. In addition, they also involve endocrine, immune systems. At present, the effects of clinical treatments of depression is limited. Therefore, exploring the co-disease mechanism of depression and metabolic diseases is helpful to find a new clinical therapeutic intervention strategy. Herein, focusing on the relationship between depression and metabolic diseases, this manuscript aims to provide an overview of the comorbidity of depression and metabolic.
Collapse
Affiliation(s)
- Wenxin Qiu
- Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaodan Cai
- Fujian Medical University, Fuzhou, Fujian, China
| | | | - Shumin Qiu
- Fujian Medical University, Fuzhou, Fujian, China
| | - Hanyang Ke
- Fujian Medical University, Fuzhou, Fujian, China
| | - Yinqiong Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
20
|
Westfall S, Caracci F, Estill M, Frolinger T, Shen L, Pasinetti GM. Chronic Stress-Induced Depression and Anxiety Priming Modulated by Gut-Brain-Axis Immunity. Front Immunol 2021; 12:670500. [PMID: 34248950 PMCID: PMC8264434 DOI: 10.3389/fimmu.2021.670500] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/08/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic stress manifests as depressive- and anxiety-like behavior while recurrent stress elicits disproportionate behavioral impairments linked to stress-induced immunological priming. The gut-brain-microbiota-axis is a promising therapeutic target for stress-induced behavioral impairments as it simultaneously modulates peripheral and brain immunological landscapes. In this study, a combination of probiotics and prebiotics, known as a synbiotic, promoted behavioral resilience to chronic and recurrent stress by normalizing gut microbiota populations and promoting regulatory T cell (Treg) expansion through modulation of ileal innate lymphoid cell (ILC)3 activity, an impact reflecting behavioral responses better than limbic brain region neuroinflammation. Supporting this conclusion, a multivariate machine learning model correlatively predicted a cross-tissue immunological signature of stress-induced behavioral impairment where the ileal Treg/T helper17 cell ratio associated to hippocampal chemotactic chemokine and prefrontal cortex IL-1β production in the context of stress-induced behavioral deficits. In conclusion, stress-induced behavioral impairments depend on the gut-brain-microbiota-axis and through ileal immune regulation, synbiotics attenuate the associated depressive- and anxiety-like behavior.
Collapse
Affiliation(s)
- Susan Westfall
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Francesca Caracci
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Molly Estill
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tal Frolinger
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Li Shen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| |
Collapse
|
21
|
Zhao X, Kong D, Zhou Q, Wei G, Song J, Liang Y, Du G. Baicalein alleviates depression-like behavior in rotenone- induced Parkinson's disease model in mice through activating the BDNF/TrkB/CREB pathway. Biomed Pharmacother 2021; 140:111556. [PMID: 34087694 DOI: 10.1016/j.biopha.2021.111556] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/23/2021] [Accepted: 03/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder in the world. In addition to motor symptoms, a variety of non-motor symptoms seriously affect the life quality of PD patients. Baicalein, a flavonoid extracted from the herb Scutellaria baicalensis Georgi, exhibits anti-PD activity through alleviation of its motor symptoms. However, its effects on non-motor symptoms were barely reported. This study aimed to investigate the therapeutic effects of baicalein on PD-related depression. METHODS After a 2-week injection of rotenone, mice with PD-related depression behavior were selected, divided into three groups, and administrated saline, baicalein, or madopar orally for four weeks. Behavior, neuroinflammation, neurotransmitters, and synaptic plasticity were evaluated. RESULTS Our results showed that 4-week baicalein treatment significantly alleviated the depression-like behavior in the rotenone-induced mice model. Repeated baicalein treatment reduced α-synuclein aggregation, inhibited neuroinflammation, and maintained neurotransmitters homeostasis. Moreover, we found that baicalein treatment could remarkably protect the synaptic plasticity and activate the BDNF/TrkB/CREB pathway in the PD-related depression mice model. As traditional dopamine replacement therapy unleashed few effects on depression-like symptom amelioration and synaptic function protection, baicalein might be a more appropriate choice for PD-related depression. CONCLUSIONS The current results suggested that baicalein could act as a treatment for PD-related depression.
Collapse
Affiliation(s)
- Xiaoyue Zhao
- Beijing Key Laboratory of Drug Targets Research and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nongtan Street, Beijing, 100050, China
| | - Dewen Kong
- Beijing Key Laboratory of Drug Targets Research and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nongtan Street, Beijing, 100050, China
| | - Qimeng Zhou
- Beijing Key Laboratory of Drug Targets Research and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nongtan Street, Beijing, 100050, China
| | - Guangyi Wei
- Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Junke Song
- Beijing Key Laboratory of Drug Targets Research and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nongtan Street, Beijing, 100050, China
| | - Yu Liang
- Beijing Key Laboratory of Drug Targets Research and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nongtan Street, Beijing, 100050, China
| | - Guanhua Du
- Beijing Key Laboratory of Drug Targets Research and New Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nongtan Street, Beijing, 100050, China.
| |
Collapse
|
22
|
Saccaro LF, Schilliger Z, Dayer A, Perroud N, Piguet C. Inflammation, anxiety, and stress in bipolar disorder and borderline personality disorder: A narrative review. Neurosci Biobehav Rev 2021; 127:184-192. [PMID: 33930472 DOI: 10.1016/j.neubiorev.2021.04.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 04/11/2021] [Accepted: 04/18/2021] [Indexed: 11/30/2022]
Abstract
Bipolar disorder (BD) and borderline personality disorder (BPD) are serious and prevalent psychiatric diseases that share common phenomenological characteristics: symptoms (such as anxiety, affective lability or emotion dysregulation), neuroimaging features, risk factors and comorbidities. While several studies have focused on the link between stress and peripheral inflammation in other affective disorders such as anxiety or depression, fewer have explored this relationship in BD and BPD. This review reports on evidence showing an interplay between immune dysregulation, anxiety and stress, and how an altered acute neuroendocrine stress response may exist in these disorders. Moreover, we highlight limitations and confounding factors of these existing studies and discuss multidirectional hypotheses that either suggest inflammation or stress and anxiety as the primum movens in BD and BPD pathophysiology, or inflammation as a consequence of the pathophysiology of these diseases. Untangling these associations and implementing a transdiagnostic approach will have diagnostic, therapeutic and prognostic implications for BD and BPD patients.
Collapse
Affiliation(s)
- L F Saccaro
- Department of Clinical Neurosciences, Division of Neurology, Geneva University Hospital, University of Geneva, Geneva, Switzerland
| | - Z Schilliger
- Centre for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Prilly-Lausanne, Switzerland
| | - A Dayer
- Psychiatry Department, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Neuroscience Department, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - N Perroud
- Psychiatry Department, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - C Piguet
- Psychiatry Department, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Child and Adolescent Psychiatry Division, Geneva University Hospital, Geneva, Switzerland.
| |
Collapse
|
23
|
Zefferino R, Di Gioia S, Conese M. Molecular links between endocrine, nervous and immune system during chronic stress. Brain Behav 2021; 11:e01960. [PMID: 33295155 PMCID: PMC7882157 DOI: 10.1002/brb3.1960] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/17/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The stress response is different in various individuals, however, the mechanisms that could explain these distinct effects are not well known and the molecular correlates have been considered one at the time. Particular harmful conditions occur if the subject, instead to cope the stressful events, succumb to them, in this case, a cascade reaction happens that through different signaling causes a specific reaction named "sickness behaviour." The aim of this article is to review the complex relations among important molecules belonging to Central nervous system (CNS), immune system (IS), and endocrine system (ES) during the chronic stress response. METHODS After having verified the state of art concerning the function of cortisol, norepinephrine (NE), interleukin (IL)-1β and melatonin, we describe as they work together. RESULTS We propose a speculative hypothesis concerning the complex interplay of these signaling molecules during chronic stress, highlighting the role of IL-1β as main biomarker of this effects, indeed, during chronic stress its increment transforms this inflammatory signal into a nervous signal (NE), in turn, this uses the ES (melatonin and cortisol) to counterbalance again IL-1β. During cortisol resistance, a vicious loop occurs that increments all mediators, unbalancing IS, ES, and CNS networks. This IL-1β increase would occur above all when the individual succumbs to stressful events, showing the Sickness Behaviour Symptoms. IL-1β might, through melatonin and vice versa, determine sleep disorders too. CONCLUSION The molecular links here outlined could explain how stress plays a role in etiopathogenesis of several diseases through this complex interplay.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
24
|
β-blockade prevents coronary macro- and microvascular dysfunction induced by a high salt diet and insulin resistance in the Goto-Kakizaki rat. Clin Sci (Lond) 2021; 135:327-346. [PMID: 33480422 DOI: 10.1042/cs20201441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 01/01/2023]
Abstract
A high salt intake exacerbates insulin resistance, evoking hypertension due to systemic perivascular inflammation, oxidative-nitrosative stress and endothelial dysfunction. Angiotensin-converting enzyme inhibitor (ACEi) and angiotensin receptor blockers (ARBs) have been shown to abolish inflammation and redox stress but only partially restore endothelial function in mesenteric vessels. We investigated whether sympatho-adrenal overactivation evokes coronary vascular dysfunction when a high salt intake is combined with insulin resistance in male Goto-Kakizaki (GK) and Wistar rats treated with two different classes of β-blocker or vehicle, utilising synchrotron-based microangiography in vivo. Further, we examined if chronic carvedilol (CAR) treatment preserves nitric oxide (NO)-mediated coronary dilation more than metoprolol (MET). A high salt diet (6% NaCl w/w) exacerbated coronary microvessel endothelial dysfunction and NO-resistance in vehicle-treated GK rats while Wistar rats showed modest impairment. Microvascular dysfunction was associated with elevated expression of myocardial endothelin, inducible NO synthase (NOS) protein and 3-nitrotyrosine (3-NT). Both CAR and MET reduced basal coronary perfusion but restored microvessel endothelium-dependent and -independent dilation indicating a role for sympatho-adrenal overactivation in vehicle-treated rats. While MET treatment reduced myocardial nitrates, only MET treatment completely restored microvessel dilation to dobutamine (DOB) stimulation in the absence of NO and prostanoids (combined inhibition), indicating that MET restored the coronary flow reserve attributable to endothelium-derived hyperpolarisation (EDH). In conclusion, sympatho-adrenal overactivation caused by high salt intake and insulin resistance evoked coronary microvessel endothelial dysfunction and diminished NO sensitivity, which were restored by MET and CAR treatment in spite of ongoing inflammation and oxidative-nitrosative stress presumably caused by uninhibited renin-angiotensin-aldosterone system (RAAS) overactivation.
Collapse
|
25
|
Dudek KA, Dion‐Albert L, Kaufmann FN, Tuck E, Lebel M, Menard C. Neurobiology of resilience in depression: immune and vascular insights from human and animal studies. Eur J Neurosci 2021; 53:183-221. [PMID: 31421056 PMCID: PMC7891571 DOI: 10.1111/ejn.14547] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a chronic and recurrent psychiatric condition characterized by depressed mood, social isolation and anhedonia. It will affect 20% of individuals with considerable economic impacts. Unfortunately, 30-50% of depressed individuals are resistant to current antidepressant treatments. MDD is twice as prevalent in women and associated symptoms are different. Depression's main environmental risk factor is chronic stress, and women report higher levels of stress in daily life. However, not every stressed individual becomes depressed, highlighting the need to identify biological determinants of stress vulnerability but also resilience. Based on a reverse translational approach, rodent models of depression were developed to study the mechanisms underlying susceptibility vs resilience. Indeed, a subpopulation of animals can display coping mechanisms and a set of biological alterations leading to stress resilience. The aetiology of MDD is multifactorial and involves several physiological systems. Exacerbation of endocrine and immune responses from both innate and adaptive systems are observed in depressed individuals and mice exhibiting depression-like behaviours. Increasing attention has been given to neurovascular health since higher prevalence of cardiovascular diseases is found in MDD patients and inflammatory conditions are associated with depression, treatment resistance and relapse. Here, we provide an overview of endocrine, immune and vascular changes associated with stress vulnerability vs. resilience in rodents and when available, in humans. Lack of treatment efficacy suggests that neuron-centric treatments do not address important causal biological factors and better understanding of stress-induced adaptations, including sex differences, could contribute to develop novel therapeutic strategies including personalized medicine approaches.
Collapse
Affiliation(s)
- Katarzyna A. Dudek
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Laurence Dion‐Albert
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Fernanda Neutzling Kaufmann
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Ellen Tuck
- Smurfit Institute of GeneticsTrinity CollegeDublinIreland
| | - Manon Lebel
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Caroline Menard
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| |
Collapse
|
26
|
Costa R, Carvalho MSM, Brandão JDP, Moreira RP, Cunha TS, Casarini DE, Marcondes FK. Modulatory action of environmental enrichment on hormonal and behavioral responses induced by chronic stress in rats: Hypothalamic renin-angiotensin system components. Behav Brain Res 2020; 397:112928. [PMID: 32987059 DOI: 10.1016/j.bbr.2020.112928] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 09/10/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023]
Abstract
Environmental enrichment (EE) has been studied as a protocol that can improve brain plasticity and may protect against negative insults such as chronic stress. The aim of this study was to evaluate the effects of EE on the hormonal and behavioral responses induced by chronic mild unpredictable stress (CMS) in rats, considering the involvement of the renin-angiotensin system. Male adult rats were divided into 4 groups: control, CMS, EE, and CMS + EE, and the experimental protocol lasted for 7 weeks. EE was performed during 7 weeks, 5 days per week, 2 h per day. CMS was applied during weeks 3, 4, and 5. After the CMS (week 6), depression-like behavior was evaluated by forced swimming and sucrose consumption tests, anxiety level was evaluated using the elevated plus-maze test, and memory was evaluated using the Y-maze test. On week 7, the animals were euthanized and basal plasma levels of corticosterone and catecholamines were determined. The hypothalamus was isolated and tissue levels of angiotensin peptides were evaluated. CMS increased plasma corticosterone, norepinephrine, and epinephrine basal concentrations, induced depression-like behaviors, impaired memory, and increased hypothalamic angiotensin I, II, and IV concentrations. EE decreased stress hormones secretion, depression-like behaviors, memory impairment, and hypothalamic angiotensin II induced by stress. Reductions of anxiety-like behavior and norepinephrine secretion were observed in both stressed and unstressed groups. The results indicated that EE seemed to protect adult rats against hormonal and behavioral CMS effects, and that the reduction of angiotensin II could contribute to these effects.
Collapse
Affiliation(s)
- Rafaela Costa
- Department of Biosciences, Laboratory of Stress, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | - Maeline Santos Morais Carvalho
- Department of Biosciences, Laboratory of Stress, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | | | - Roseli Peres Moreira
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Tatiana Sousa Cunha
- Science and Technology Institute, Federal University of São Paulo, São José Dos Campos, SP, Brazil
| | - Dulce Elena Casarini
- Nephrology Division, Department of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Fernanda Klein Marcondes
- Department of Biosciences, Laboratory of Stress, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil.
| |
Collapse
|
27
|
Bruno A, Dolcetti E, Rizzo FR, Fresegna D, Musella A, Gentile A, De Vito F, Caioli S, Guadalupi L, Bullitta S, Vanni V, Balletta S, Sanna K, Buttari F, Stampanoni Bassi M, Centonze D, Mandolesi G. Inflammation-Associated Synaptic Alterations as Shared Threads in Depression and Multiple Sclerosis. Front Cell Neurosci 2020; 14:169. [PMID: 32655374 PMCID: PMC7324636 DOI: 10.3389/fncel.2020.00169] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
In the past years, several theories have been advanced to explain the pathogenesis of Major Depressive Disorder (MDD), a neuropsychiatric disease that causes disability in general population. Several theories have been proposed to define the MDD pathophysiology such as the classic "monoamine-theory" or the "glutamate hypothesis." All these theories have been recently integrated by evidence highlighting inflammation as a pivotal player in developing depressive symptoms. Proinflammatory cytokines have been indeed claimed to contribute to stress-induced mood disturbances and to major depression, indicating a widespread role of classical mediators of inflammation in emotional control. Moreover, during systemic inflammatory diseases, peripherally released cytokines circulate in the blood, reach the brain and cause anxiety, anhedonia, social withdrawal, fatigue, and sleep disturbances. Accordingly, chronic inflammatory disorders, such as the inflammatory autoimmune disease multiple sclerosis (MS), have been associated to higher risk of MDD, in comparison with overall population. Importantly, in both MS patients and in its experimental mouse model, Experimental Autoimmune Encephalomyelitis (EAE), the notion that depressive symptoms are reactive epiphenomenon to the MS pathology has been recently challenged by the evidence of their early manifestation, even before the onset of the disease. Furthermore, in association to such mood disturbance, inflammatory-dependent synaptic dysfunctions in several areas of MS/EAE brain have been observed independently of brain lesions and demyelination. This evidence suggests that a fine interplay between the immune and nervous systems can have a huge impact on several neurological functions, including depressive symptoms, in different pathological conditions. The aim of the present review is to shed light on common traits between MDD and MS, by looking at inflammatory-dependent synaptic alterations associated with depression in both diseases.
Collapse
Affiliation(s)
- Antonio Bruno
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Ettore Dolcetti
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Francesca Romana Rizzo
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | | | - Francesca De Vito
- Unit of Neurology, Mediterranean Neurological Institute IRCCS Neuromed, Pozzilli, Italy
| | - Silvia Caioli
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Silvia Bullitta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Sara Balletta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Krizia Sanna
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology, Mediterranean Neurological Institute IRCCS Neuromed, Pozzilli, Italy
| | | | - Diego Centonze
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
- Unit of Neurology, Mediterranean Neurological Institute IRCCS Neuromed, Pozzilli, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| |
Collapse
|
28
|
Petralia MC, Mazzon E, Fagone P, Basile MS, Lenzo V, Quattropani MC, Di Nuovo S, Bendtzen K, Nicoletti F. The cytokine network in the pathogenesis of major depressive disorder. Close to translation? Autoimmun Rev 2020; 19:102504. [PMID: 32173514 DOI: 10.1016/j.autrev.2020.102504] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 12/25/2019] [Indexed: 12/18/2022]
Abstract
Major depressive disorder (MDD) is a common condition that afflicts the general population across a broad spectrum of ages and social backgrounds. MDD has been identified by the World Health Organization as a leading cause of disability worldwide. Approximately 30% of patients are poor responsive to standard of care (SOC) treatment and novel therapeutic approaches are warranted. Since chronic inflammation, as it is often observed in certain cancers, type 2 diabetes, psoriasis and chronic arthritis, are accompanied by depression, it has been suggested that immunoinflammatory processes may be involved in the pathogenesis of MDD. Cytokines are a group of glycoproteins secreted from lymphoid and non-lymphoid cells that orchestrate immune responses. It has been suggested that a dysregulated production of cytokines may be implicated in the pathogenesis and maintenance of MDD. On the basis of their functions, cytokines can be subdivided in pro-inflammatory and anti-inflammatory cytokines. Since abnormal blood and cerebrospinal fluid of both pro and anti-inflammatory cytokines are altered in MDD, it has been suggested that abnormal cytokine homeostasis may be implicated in the pathogenesis of MDD and possibly to induction of therapeutic resistance. We review current data that indicate that cytokines may represent a useful tool to identify MDD patients that may benefit from tailored immunotherapeutic approaches and may represent a potential tailored therapeutic target.
Collapse
Affiliation(s)
| | | | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Vittorio Lenzo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Santo Di Nuovo
- Department of Educational Sciences, University of Catania, Catania, Italy
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
29
|
Gideon A, Sauter C, Fieres J, Berger T, Renner B, Wirtz PH. Kinetics and Interrelations of the Renin Aldosterone Response to Acute Psychosocial Stress: A Neglected Stress System. J Clin Endocrinol Metab 2020; 105:5618777. [PMID: 31711229 PMCID: PMC7034950 DOI: 10.1210/clinem/dgz190] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022]
Abstract
CONTEXT The renin-angiotensin-aldosterone system (RAAS) plays an important role in cardiovascular homeostasis and its dysfunction relates to negative health consequences. Acute psychosocial stress seems to activate the RAAS in humans, but stress kinetics and interrelations of RAAS parameters compared with a nonstress control group remain inconclusive. OBJECTIVE We systematically investigated in a randomized placebo-controlled design stress kinetics and interrelations of the reactivity of RAAS parameters measured in plasma and saliva to standardized acute psychosocial stress induction. METHODS 58 healthy young men were assigned to either a stress or a placebo control group. The stress group underwent the Trier Social Stress Test (TSST), while the control group underwent the placebo TSST. We repeatedly assessed plasma renin, and plasma and salivary aldosterone before and up to 3 hours after stress/placebo. We simultaneously assessed salivary cortisol to validate successful stress induction and to test for interrelations. RESULTS Acute psychosocial stress induced significant increases in all endocrine measures compared with placebo-stress (all P ≤ .041). Highest renin levels were observed 1 minute after stress, and highest aldosterone and cortisol levels 10 and 20 minutes after stress, with salivary aldosterone starting earlier at 1 minute after stress. Renin completed recovery at 10 minutes, cortisol at 60 minutes, salivary aldosterone at 90 minutes, and plasma aldosterone at 180 minutes after stress. Stress increase scores of all endocrine measures related to each other, as did renin and cortisol areas under the curve with respect to increase (AUCi) and salivary and plasma aldosterone AUCi (all P ≤ .047). CONCLUSIONS Our findings suggest that in humans acute psychosocial stress induces a differential and interrelated RAAS parameter activation pattern. Potential implications for stress-related cardiovascular risk remain to be elucidated.
Collapse
Affiliation(s)
- Angelina Gideon
- Biological Work and Health Psychology, University of Konstanz, Germany
| | - Christine Sauter
- Biological Work and Health Psychology, University of Konstanz, Germany
| | - Judy Fieres
- Biological Work and Health Psychology, University of Konstanz, Germany
| | - Thilo Berger
- Biological Work and Health Psychology, University of Konstanz, Germany
| | - Britta Renner
- Health Psychology, University of Konstanz, Germany
- Centre for the Advanced Study of Collective Behavior, University of Konstanz, Germany
| | - Petra H Wirtz
- Biological Work and Health Psychology, University of Konstanz, Germany
- Centre for the Advanced Study of Collective Behavior, University of Konstanz, Germany
- Correspondence and Reprint Requests: Petra H. Wirtz, PhD, Biological Work and Health Psychology, University of Konstanz Universitaetsstrasse 10, 78457 Konstanz, Germany. E-mail:
| |
Collapse
|
30
|
Wsol A, Wojno O, Puchalska L, Wrzesien R, Szczepanska-Sadowska E, Cudnoch-Jedrzejewska A. Impaired hypotensive effects of centrally acting oxytocin in SHR and WKY rats exposed to chronic mild stress. Am J Physiol Regul Integr Comp Physiol 2020; 318:R160-R172. [DOI: 10.1152/ajpregu.00050.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The present study was designed to determine the role of centrally acting oxytocin (OT) in the regulation of blood pressure during chronic mild stress (CMS) in spontaneously hypertensive (SHR; n = 36) and normotensive Wistar-Kyoto (WKY; n = 38) rats. The rats were implanted with osmotic minipumps for intracerebroventricular infusions of 0.9% NaCl, OT, and oxytocin receptor antagonist (OTANT) and divided into two groups: SHR and WKY 1) exposed to 4-wk CMS and 2) not exposed to stress (controls). After 4 wk, hemodynamic parameters were recorded at rest and after an application of acute stressor [air-jet stress (AJS)]. Resting mean arterial blood pressure (MAP) was significantly lower in CMS-exposed SHR and WKY infused with OT than in the corresponding groups receiving saline. Exposure to CMS exaggerated the AJS-dependent pressor response in WKY receiving saline but not in the corresponding group of SHR. OT infusion reduced the AJS-dependent pressor response in both CMS-exposed and not exposed SHR and in CMS-exposed WKY. Intracerebroventricular infusion of OTANT potentiated the AJS-dependent pressor response in both stressed and not stressed WKY rats but not in SHR. The results show that centrally delivered OT decreases resting MAP during CMS in both SHR and WKY rats and that in SHR it reduces pressor responses to AJS under control and CMS conditions, whereas in WKY this effect is significant only after CMS exposure. The study indicates that endogenous centrally acting OT may play an essential role in buffering pressor responses to AJS in CMS-exposed and not exposed WKY rats and that this function is significantly impaired in SHR.
Collapse
Affiliation(s)
- A. Wsol
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - O. Wojno
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - L. Puchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - R. Wrzesien
- Department of Animal Breeding, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - E. Szczepanska-Sadowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - A. Cudnoch-Jedrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
31
|
Bollinger J, Wohleb E. The formative role of microglia in stress-induced synaptic deficits and associated behavioral consequences. Neurosci Lett 2019; 711:134369. [PMID: 31422099 PMCID: PMC9875737 DOI: 10.1016/j.neulet.2019.134369] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 01/27/2023]
Abstract
Psychological stress can precipitate depression, and emerging preclinical data suggest a link between stress-induced alterations in microglia function and development of depressive-like behaviors. Microglia are highly dynamic, and play an integral role in maintaining neuronal homeostasis and synaptic plasticity. In this capacity, microglial dysfunction represents a compelling avenue through which stress might disrupt neuronal integrity and induce psychopathology. This review examines preclinical and clinical postmortem findings that indicate microglia-neuron interactions contribute to stress-induced synaptic deficits and associated behavioral and cognitive consequences. We focus on pathways that are implicated in microglia-mediated neuronal remodeling, including CSF1-CSF1R, CX3CL1-CX3CR1, and CD11b (CR3)-C3, as well as purinergic signaling via P2RX7 and P2RY12. We also highlight sex differences in stress effects on microglia, and the potential for microglia in the development of sex-specific treatments for depressive disorders.
Collapse
Affiliation(s)
| | - E.S. Wohleb
- Corresponding author at: Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, 2120 East Galbraith Road, Cincinnati, OH, 45237, USA. (E.S. Wohleb)
| |
Collapse
|
32
|
Zhao M, Harris BN, Nguyen CTY, Saltzman W. Effects of single parenthood on mothers' behavior, morphology, and endocrine function in the biparental California mouse. Horm Behav 2019; 114:104536. [PMID: 31153926 DOI: 10.1016/j.yhbeh.2019.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/31/2019] [Accepted: 05/24/2019] [Indexed: 12/29/2022]
Abstract
Motherhood is energetically costly for mammals and is associated with pronounced changes in mothers' physiology, morphology and behavior. In ~5% of mammals, fathers assist their mates with rearing offspring and can enhance offspring survival and development. Although these beneficial consequences of paternal care can be mediated by direct effects on offspring, they might also be mediated indirectly, through beneficial effects on mothers. We tested the hypothesis that fathers in the monogamous, biparental California mouse (Peromyscus californicus) reduce the burden of parental care on their mates, and therefore, that females rearing offspring with and without assistance from their mates will show differences in endocrinology, morphology and behavior, as well as in the survival and development of their pups. We found that pups' survival and development in the lab did not differ between those raised by a single mother and those reared by both mother and father. Single mothers spent more time in feeding behaviors than paired mothers. Both single and paired mothers had higher lean mass and/or lower fat mass and showed more anxiety-like behavior in open-field tests and tail-suspension tests, compared to non-breeding females. Single mothers had higher body-mass-corrected liver and heart masses, but lower ovarian and uterine masses, than paired mothers and/or non-breeding females. Mass of the gastrointestinal tract did not differ between single and paired mothers, but single mothers had heavier gastrointestinal tract compared to non-breeding females. Single motherhood also induced a flattened diel corticosterone rhythm and a blunted corticosterone response to stress, compared to non-breeding conditions. These findings suggest that the absence of a mate induces morphological and endocrine changes in mothers, which might result from increased energetic demands of pup care and could potentially help maintain normal survival and development of pups.
Collapse
Affiliation(s)
- Meng Zhao
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, United States of America
| | - Breanna N Harris
- Department of Biological Sciences, Texas Tech University, United States of America
| | - Catherine T Y Nguyen
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, United States of America
| | - Wendy Saltzman
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, United States of America.
| |
Collapse
|
33
|
Behavioral effects of toll-like receptor-4 antagonist 'eritoran' in an experimental model of depression: role of prefrontal and hippocampal neurogenesis and γ-aminobutyric acid/glutamate balance. Behav Pharmacol 2019; 29:413-425. [PMID: 29561292 DOI: 10.1097/fbp.0000000000000390] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Depression is the disease of the modern era. The lack of response to the available antidepressants, which were developed on the basis of the monoaminergic deficit hypothesis of depression, has encouraged scientists to think about new mechanisms explaining the pathogenesis of depression. In this context, the inflammatory theory has emerged to clarify many aspects of depression that the previous theories have failed to explain. Toll-like receptor-4 (TLR-4) has a regulatory role in the brain's immune response to stress, and its activation is suggested to play a pivotal role in the pathophysiology of depression. In this study, we tested eritoran (ERI), a TLR-4 receptor-4 antagonist, as a potential antidepressant. We investigated the effect of long-term administration of ERI in three different doses on behavioral changes, hippocampal and prefrontal cortex (PFC) neurogenesis, and γ-aminobutyric acid (GABA)/glutamate balance in male Wistar rats exposed to chronic restraint stress (CRS). Long-term administration of ERI ameliorated CRS-induced depressive-like symptoms and hypothalamic-pituitary-adrenal axis hyperactivity alongside reducing levels of hippocampal and PFC inflammatory cytokines, restoring GABA and glutamate balance, and enhancing PFC and hippocampal neurogenesis, by increasing BDNF gene and protein expression in a dose-dependent manner. The results demonstrate an antidepressant-like activity of ERI in Wistar rats exposed to CRS, which may be largely mediated by its ability to reduce neuroinflammation, increase BDNF, and restore GABA/glutamate balance in prefrontal cortex and hippocampus. Nonetheless, further studies are needed to characterize the mechanism of the antidepressant effect of ERI.
Collapse
|
34
|
Konkle ATM, Keith SE, McNamee JP, Michaud D. Chronic noise exposure in the spontaneously hypertensive rat. Noise Health 2019; 19:213-221. [PMID: 28937015 PMCID: PMC5644380 DOI: 10.4103/nah.nah_15_17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Introduction: Epidemiological studies have suggested an association between the relative risk for developing cardiovascular disease (CVD) and long-term exposure to elevated levels of transportation noise. The contention is that this association is largely owing to an increase in stress-related biomarkers that are thought to be associated with CVD. Animal models have demonstrated that acute noise exposure is capable of triggering a stress response; however, similar studies using chronic noise models are less common. Materials and Methods: The current study assessed the effects of intermittent daily exposure to broadband 80 kHz bandwidth noise of 87.3 dBA for a period of 21 consecutive days in spontaneously hypertensive rats. Results: Twenty-one days of exposure to noise significantly reduced body weight relative to the sham and unhandled control groups; however, noise had no statistically significant impact on plasma adrenocorticotropic hormone (or adrenal gland weights). Noise was associated with a significant, albeit modest, increase in both corticosterone and aldosterone concentrations following the 21 days of exposure. Interleukin 1 and interleukin 6 levels were unchanged in the noise group, whereas both tumour necrosis factor alpha and C-reactive protein were significantly reduced in noise exposed rats. Tail blood sampling for corticosterone throughout the exposure period showed no appreciable difference between the noise and sham exposed animals, largely due to the sizeable variation for each group as well as the observed fluctuations over time. Discussion: The current pilot study provides only modest support that chronic noise may promote stress-related biological and/or developmental effects. More research is required to verify the current findings and resolve some of the unexpected observations.
Collapse
Affiliation(s)
- Anne T M Konkle
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario; School of Psychology, Faculty of Social Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Stephen E Keith
- Healthy Environments and Consumer Safety Branch, Environmental and Radiation Health Sciences Directorate, Consumer and Clinical Radiation Protection Bureau, Health , Ottawa, Ontario, Canada
| | - James P McNamee
- Healthy Environments and Consumer Safety Branch, Environmental and Radiation Health Sciences Directorate, Consumer and Clinical Radiation Protection Bureau, Health , Ottawa, Ontario, Canada
| | - David Michaud
- Healthy Environments and Consumer Safety Branch, Environmental and Radiation Health Sciences Directorate, Consumer and Clinical Radiation Protection Bureau, Health , Ottawa, Ontario, Canada
| |
Collapse
|
35
|
Tsyglakova M, McDaniel D, Hodes GE. Immune mechanisms of stress susceptibility and resilience: Lessons from animal models. Front Neuroendocrinol 2019; 54:100771. [PMID: 31325456 DOI: 10.1016/j.yfrne.2019.100771] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/17/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022]
Abstract
Stress has an impact on the brain and the body. A growing literature demonstrates that feedback between the peripheral immune system and the brain contributes to individual differences in the behavioral response to stress. Here we examine preclinical literature to demonstrate a holistic vision of risk and resilience to stress. We identify a variety of cellular, cytokine and molecular mechanisms in adult animals that act in concert to produce a stress susceptible individual response. We discuss how cross talk between immune cells in the brain and in the periphery act together to increase permeability across the blood brain barrier or block it, resulting in susceptible or stress resilient phenotype. These preclinical studies have importance for understanding how individual differences in the immune response to stress may be contributing to mood related disorders such as depression, anxiety and posttraumatic stress disorders.
Collapse
Affiliation(s)
- Mariya Tsyglakova
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA, USA
| | - Dylan McDaniel
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Georgia E Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
36
|
Antoniuk S, Bijata M, Ponimaskin E, Wlodarczyk J. Chronic unpredictable mild stress for modeling depression in rodents: Meta-analysis of model reliability. Neurosci Biobehav Rev 2019; 99:101-116. [DOI: 10.1016/j.neubiorev.2018.12.002] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 01/01/2023]
|
37
|
Kovačević S, Nestorov J, Matić G, Elaković I. Chronic Stress Combined with a Fructose Diet Reduces Hypothalamic Insulin Signaling and Antioxidative Defense in Female Rats. Neuroendocrinology 2019; 108:278-290. [PMID: 30572328 DOI: 10.1159/000496391] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/19/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND Increased fructose consumption and chronic exposure to stress have been associated with the development of obesity and insulin resistance. In the hypothalamus, a crossroad of stress responses and energy balance, insulin and glucocorticoids regulate the expression of orexigenic neuropeptides, neuropeptide Y (NPY) and agouti-related protein (AgRP), and anorexigenic neuropeptides, proopio-melanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART). OBJECTIVES We investigated whether chronic stress and fructose diet disrupt these hormonal signaling pathways and appetite control in the hypothalamus, contributing to the development of insulin resistance and obesity. Potential roles of hypothalamic inflammation and oxidative stress in the development of insulin resistance were also analyzed. METHODS Insulin, glucocorticoid, and leptin signaling, expression of orexigenic and anorexigenic neuropeptides, and antioxidative and inflammatory statuses in the whole hypothalamus of fructose-fed female rats exposed to unpredictable stress for 9 weeks were analyzed using quantitative PCR and Western blotting. RESULTS Chronic stress combined with a fructose-enriched diet reduced protein content and stimulatory phosphorylation of Akt kinase, and elevated 11β-hydroxysteroid dehydrogenase 1 and glucocorticoid receptor expression, while alterations in appetite regulation (NPY, AgRP, POMC, CART, leptin receptor, and SOCS3 expression) were not observed. The expression of antioxidative defense enzymes (mitochondrial manganese superoxide dismutase 2, glutathione reductase, and catalase) and proinflammatory cytokines (IL-1β, IL-6, and TNFα) was reduced. CONCLUSIONS Our results underline the combination of long-term stress exposure and fructose overconsumption as more detrimental for hypothalamic function than for either of the factors separately, as it enhanced glucocorticoid and impaired insulin signaling, antioxidative -defense, and inflammatory responses of this homeostasis- regulating center.
Collapse
Affiliation(s)
- Sanja Kovačević
- Department of Biochemistry, Institute for Biological Research Siniša Stanković, University of Belgrade, Belgrade, Serbia
| | - Jelena Nestorov
- Department of Biochemistry, Institute for Biological Research Siniša Stanković, University of Belgrade, Belgrade, Serbia
| | - Gordana Matić
- Department of Biochemistry, Institute for Biological Research Siniša Stanković, University of Belgrade, Belgrade, Serbia
| | - Ivana Elaković
- Department of Biochemistry, Institute for Biological Research Siniša Stanković, University of Belgrade, Belgrade, Serbia,
| |
Collapse
|
38
|
Abstract
The causes of essential hypertension remain an enigma. Interactions between genetic and external factors are generally recognized to act as aetiological mechanisms that trigger the pathogenesis of high blood pressure. However, the questions of which genes and factors are involved, and when and where such interactions occur, remain unresolved. Emerging evidence indicates that the hypertensive response to pressor stimuli, like many other physiological and behavioural adaptations, can become sensitized to particular stimuli. Studies in animal models show that, similarly to other response systems controlled by the brain, hypertensive response sensitization (HTRS) is mediated by neuroplasticity. The brain circuitry involved in HTRS controls the sympathetic nervous system. This Review outlines evidence supporting the phenomenon of HTRS and describes the range of physiological and psychosocial stressors that can produce a sensitized hypertensive state. Also discussed are the cellular and molecular changes in the brain neural network controlling sympathetic tone involved in long-term storage of information relating to stressors, which could serve to maintain a sensitized state. Finally, this Review concludes with a discussion of why a sensitized hypertensive response might previously have been beneficial and increased biological fitness under some environmental conditions and why today it has become a health-related liability.
Collapse
Affiliation(s)
- Alan Kim Johnson
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA.
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA, USA.
- Department of Pharmacology, University of Iowa, Iowa City, IA, USA.
- The François M. Abboud Cardiovascular Center, Iowa City, IA, USA.
| | - Baojian Xue
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- The François M. Abboud Cardiovascular Center, Iowa City, IA, USA
| |
Collapse
|
39
|
Lou YX, Li J, Wang ZZ, Xia CY, Chen NH. Glucocorticoid receptor activation induces decrease of hippocampal astrocyte number in rats. Psychopharmacology (Berl) 2018; 235:2529-2540. [PMID: 30069586 DOI: 10.1007/s00213-018-4936-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 05/30/2018] [Indexed: 11/24/2022]
Abstract
RATIONALE The decrease of astrocyte number and hypothalamic-pituitary-adrenal (HPA) axis overactivity are observed in individuals with major depressive disorder. Elevated levels of glucocorticoids induced by hyperactivation of the HPA axis may result in glucocorticoid receptor (GR) activation. However, it is unclear whether there is a direct link between GR activation and the decrease of astrocyte number. METHODS Animals were exposed to chronic unpredictable stress (CUS) for 28 days and treated with continuous subcutaneous injections of vehicle or corticosterone (CORT; 40 mg/kg/day) for 21 days. We then administered mifepristone on day 21 after CUS and on day 18 after the CORT treatment. We observed behavioral deficits in the sucrose preference test, open field test, and forced swim test. Protein expression was analyzed using immunofluorescence (IF) and western blot (WB). RESULTS Animals exposed to CUS exhibited behavioral deficits in tests measuring anhedonia, anxiety, and despair state. They also had decreases in glial fibrillary acidic protein (GFAP) expression and numbers of GFAP-positive cells in the hippocampus. The behavioral and cellular alterations induced by CUS were reversed by subchronic treatment with the GR antagonist mifepristone. We also found that the subcutaneous injection of glucocorticoids may induce depression-like behavior and reduce GFAP protein expression in rats, which was similarly reversed by mifepristone. CONCLUSIONS These findings provide experimental evidence that GR activation due to elevated CORT levels induces the decrease of hippocampal astrocyte number in rats.
Collapse
Affiliation(s)
- Yu-Xia Lou
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Jing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Cong-Yuan Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China. .,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
40
|
Ambrée O, Ruland C, Scheu S, Arolt V, Alferink J. Alterations of the Innate Immune System in Susceptibility and Resilience After Social Defeat Stress. Front Behav Neurosci 2018; 12:141. [PMID: 30057531 PMCID: PMC6053497 DOI: 10.3389/fnbeh.2018.00141] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/21/2018] [Indexed: 12/12/2022] Open
Abstract
Dysregulation of innate immune responses has frequently been reported in stress-associated psychiatric disorders such as major depression. In mice, enhanced circulating cytokine levels as well as altered innate immune cell numbers have been found after stress exposure. In addition, stress-induced recruitment of peripheral monocytes to the brain has been shown to promote anxiety-like behavior. However, it is yet unclear whether specific differences in the innate immune system are associated with stress susceptibility or resilience in mice. Utilizing chronic social defeat, a model of depression and stress vulnerability, we characterized peripheral and brain-invading myeloid cells in stress-susceptible and resilient animals. In all defeated animals, we found reduced percentages of CD11c+ dendritic cells (DCs) by flow cytometry in the spleen when compared to non-defeated controls. Exclusively in susceptible mice conventional DCs of the spleen showed up-regulated expression of MHC class II and co-stimulatory CD80 molecules pointing toward an enhanced maturation phenotype of these cells. Susceptible, but not resilient animals further exhibited an increase in inflammatory Ly6Chi monocytes and higher numbers of spleen-derived CD11b+ cells that produced the proinflammatory cytokine tumor necrosis factor (TNF) upon lipopolysaccharide (LPS) stimulation. Increased percentages of peripheral CD45hi CD11b+ cells immigrated into the brain of defeated mice, regardless of resilience or susceptibility. However, cellular infiltrates in the brain of susceptible mice contained higher percentages of CC chemokine receptor 2 (CCR2+) Ly6Chi monocytes representing an inflammatory phenotype. Thus, we defined specific stress-related immune signatures involving conventional DCs and inflammatory Ly6Chi monocytes in susceptible and resilient mice. Together, our findings suggest an impact of the innate immune system in vulnerability to stress-related disorders such as major depression.
Collapse
Affiliation(s)
- Oliver Ambrée
- Department of Psychiatry, University of Münster, Münster, Germany.,Department of Behavioral Biology, University of Osnabrück, Osnabrück, Germany
| | - Christina Ruland
- Department of Psychiatry, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion, University of Münster, Münster, Germany
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Volker Arolt
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Judith Alferink
- Department of Psychiatry, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion, University of Münster, Münster, Germany
| |
Collapse
|
41
|
Doremus-Fitzwater TL, Paniccia JE, Gano A, Vore A, Deak T. Differential effects of acute versus chronic stress on ethanol sensitivity: Evidence for interactions on both behavioral and neuroimmune outcomes. Brain Behav Immun 2018; 70:141-156. [PMID: 29458194 PMCID: PMC5953812 DOI: 10.1016/j.bbi.2018.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/05/2018] [Accepted: 02/15/2018] [Indexed: 02/07/2023] Open
Abstract
Acute alcohol intoxication induces significant alterations in brain cytokines. Since stress challenges also profoundly impact central cytokine expression, these experiments examined the influence of acute and chronic stress on ethanol-induced brain cytokine responses. In Experiment 1, adult male rats were exposed to acute footshock. After a post-stress recovery interval of 0, 2, 4, or 24 h, rats were administered ethanol (4 g/kg; intragastric), with trunk blood and brains collected 3 h later. In non-stressed controls, acute ethanol increased expression of Il-6 and IκBα in the hippocampus. In contrast, rats exposed to footshock 24 h prior to ethanol demonstrated potentiation of hippocampal Il-6 and IκBα expression relative to ethanol-exposed non-stressed controls. Experiment 2 subsequently examined the effects of chronic stress on ethanol-related cytokine expression. Following a novel chronic escalating stress procedure, rats were intubated with ethanol. As expected, acute ethanol increased Il-6 expression in all structures examined, yet the Il-6 response was attenuated exclusively in the hippocampus in chronically stressed rats. Later experiments determined that neither acute nor chronic stress affected ethanol pharmacokinetics. When ethanol hypnosis was examined, however, rats exposed to chronic stress awoke at significantly lower blood ethanol levels compared to acutely stressed rats, despite similar durations of ethanol-induced sedation. These data indicate that chronic stress may increase sensitivity to ethanol hypnosis. Together, these experiments demonstrate an intriguing interaction between recent stress history and ethanol-induced increases in hippocampal Il-6, and may provide insight into novel pharmacotherapeutic targets for prevention and treatment of alcohol-related health outcomes based on stress susceptibility.
Collapse
Affiliation(s)
| | - Jacqueline E. Paniccia
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000
| | - Anny Gano
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000
| | - Andrew Vore
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton NY 13902-6000
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States.
| |
Collapse
|
42
|
Burrage E, Marshall KL, Santanam N, Chantler PD. Cerebrovascular dysfunction with stress and depression. Brain Circ 2018; 4:43-53. [PMID: 30276336 PMCID: PMC6126243 DOI: 10.4103/bc.bc_6_18] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/11/2022] Open
Abstract
Maintenance of adequate tissue perfusion through a dense network of cerebral microvessels is critical for the perseveration of normal brain function. Regulation of the cerebral blood flow has to ensure adequate delivery of nutrients and oxygen with moment-to-moment adjustments to avoid both hypo- and hyper-perfusion of the brain tissue. Even mild impairments of cerebral blood flow regulation can have significant implications on brain function. Evidence suggests that chronic stress and depression elicits multifaceted functional impairments to the cerebral microcirculation, which plays a critical role in brain health and the pathogenesis of stress-related cognitive impairment and cerebrovascular events. Identifying the functional and structural changes to the brain that are induced by stress is crucial for achieving a realistic understanding of how related illnesses, which are highly disabling and with a large economic cost, can be managed or reversed. This overview discusses the stress-induced alterations in neurovascular coupling with specific attention to cerebrovascular regulation (endothelial dependent and independent vasomotor function, microvessel density). The pathophysiological consequences of cerebral microvascular dysfunction with stress and depression are explored.
Collapse
Affiliation(s)
- Emily Burrage
- Department of Neuroscience, West Virginia University Rockefeller Neuroscience Institute, Morgantown, WV, USA
| | - Kent L. Marshall
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Nalini Santanam
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Paul D. Chantler
- Division of Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| |
Collapse
|
43
|
Brooks SD, Hileman SM, Chantler PD, Milde SA, Lemaster KA, Frisbee SJ, Shoemaker JK, Jackson DN, Frisbee JC. Protection from vascular dysfunction in female rats with chronic stress and depressive symptoms. Am J Physiol Heart Circ Physiol 2018; 314:H1070-H1084. [PMID: 29451821 DOI: 10.1152/ajpheart.00647.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The increasing prevalence and severity of clinical depression are strongly correlated with vascular disease risk, creating a comorbid condition with poor outcomes but demonstrating a sexual disparity whereby female subjects are at lower risk than male subjects for subsequent cardiovascular events. To determine the potential mechanisms responsible for this protection against stress/depression-induced vasculopathy in female subjects, we exposed male, intact female, and ovariectomized (OVX) female lean Zucker rats to the unpredictable chronic mild stress (UCMS) model for 8 wk and determined depressive symptom severity, vascular reactivity in ex vivo aortic rings and middle cerebral arteries (MCA), and the profile of major metabolites regulating vascular tone. While all groups exhibited severe depressive behaviors from UCMS, severity was significantly greater in female rats than male or OVX female rats. In all groups, endothelium-dependent dilation was depressed in aortic rings and MCAs, although myogenic activation and vascular (MCA) stiffness were not impacted. Higher-resolution results from pharmacological and biochemical assays suggested that vasoactive metabolite profiles were better maintained in female rats with normal gonadal sex steroids than male or OVX female rats, despite increased depressive symptom severity (i.e., higher nitric oxide and prostacyclin and lower H2O2 and thromboxane A2 levels). These results suggest that female rats exhibit more severe depressive behaviors with UCMS but are partially protected from the vasculopathy that afflicts male rats and female rats lacking normal sex hormone profiles. Determining how female sex hormones afford partial vascular protection from chronic stress and depression is a necessary step for addressing the burden of these conditions on cardiovascular health. NEW & NOTEWORTHY This study used a translationally relevant model for chronic stress and elevated depressive symptoms to determine how these factors impact conduit and resistance arteriolar function in otherwise healthy rats. While chronic stress leads to an impaired vascular reactivity associated with elevated oxidant stress, inflammation, and reduced metabolite levels, we demonstrated partial protection from vascular dysfunction in female rats with normal sex hormone profiles compared with male or ovariectomized female rats.
Collapse
Affiliation(s)
- Steven D Brooks
- Department of Physiology and Pharmacology, West Virginia University , Morgantown, West Virginia
| | - Stanley M Hileman
- Department of Physiology and Pharmacology, West Virginia University , Morgantown, West Virginia
| | - Paul D Chantler
- Department of Exercise Physiology, West Virginia University , Morgantown, West Virginia
| | - Samantha A Milde
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Faculty of Health Sciences, University of Western Ontario , London, Ontario , Canada
| | - Kent A Lemaster
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Faculty of Health Sciences, University of Western Ontario , London, Ontario , Canada
| | - Stephanie J Frisbee
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Faculty of Health Sciences, University of Western Ontario , London, Ontario , Canada.,Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Faculty of Health Sciences, University of Western Ontario , London, Ontario , Canada
| | - J Kevin Shoemaker
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Faculty of Health Sciences, University of Western Ontario , London, Ontario , Canada.,School of Kinesiology, University of Western Ontario , London, Ontario , Canada
| | - Dwayne N Jackson
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Faculty of Health Sciences, University of Western Ontario , London, Ontario , Canada
| | - Jefferson C Frisbee
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Faculty of Health Sciences, University of Western Ontario , London, Ontario , Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Faculty of Health Sciences, University of Western Ontario , London, Ontario , Canada
| |
Collapse
|
44
|
Cai CY, Wu HY, Luo CX, Zhu DY, Zhang Y, Zhou QG, Zhang J. Extracellular regulated protein kinaseis critical for the role of 5-HT1a receptor in modulating nNOS expression and anxiety-related behaviors. Behav Brain Res 2017; 357-358:88-97. [PMID: 29246772 DOI: 10.1016/j.bbr.2017.12.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/04/2017] [Accepted: 12/11/2017] [Indexed: 12/20/2022]
Abstract
Our previous study found that serotonin 1A receptor (5-HT1aR) is an endogenous suppressor of nNOS expression in the hippocampus, which accounts for anxiolytic effect of fluoxetine. However, the precise molecular mechanism remains unknown. By using 8-OH-DPAT, a selective 5-HT1aR agonist, NAN-190, a selective 5-HT1aR antagonist, and U0126, an Extracellular Regulated Protein Kinases (ERK) phosphorylation inhibitor, we investigated the role of ERK in 5-HT1aR-nNOS pathway. Western blots analysis demonstrated that 5-HT1aR activation up-regulated the level of phosphorylated ERK (P-ERK) beginning at 5 min and down-regulated the expression of nNOS beginning at 20 min. Meanwhile, blockage of 5-HT1aR resulted in a decrease in P-ERK beginning at 20 min and caused an increase in nNOS expression beginning at 6 h. Although U0126 itself did not alter nNOS expression and activity, NO level, and anxiety-related behaviors, the treatment totally reversed 8-OH-DPAT-induced reduction in nNOS expression and function, and anxiolytic effect. Besides, our data showed that ERK phosphorylation was essential for 5-HT1aR activation-induced cAMP responsive element binding protein (CREB) phosphorylation, hippocampal neurogenesis and synaptogenesis of newborn neuron. Our study suggests a crucial role of ERK phosphorylation in the regulation of nNOS expression by 5-HT1aR, which is helpful for understanding the mechanism of 5-HT1aR-based anxiolytic treatment.
Collapse
Affiliation(s)
- Cheng-Yun Cai
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Hai-Yin Wu
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Chun-Xia Luo
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Dong-Ya Zhu
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China; The Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, 211166, People's Republic of China
| | - Yu Zhang
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| | - Qi-Gang Zhou
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| | - Jing Zhang
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
45
|
DeVallance E, Riggs D, Jackson B, Parkulo T, Zaslau S, Chantler PD, Olfert IM, Bryner RW. Effect of chronic stress on running wheel activity in mice. PLoS One 2017; 12:e0184829. [PMID: 28926614 PMCID: PMC5604985 DOI: 10.1371/journal.pone.0184829] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 08/31/2017] [Indexed: 11/29/2022] Open
Abstract
Acute and chronic stress have been reported to have differing effects on physical activity in rodents, but no study has examined a chronic stress protocol that incorporates stressors often experienced by rodents throughout a day. To examine this, the effects of the Unpredictable Chronic Mild Stress (UCMS) protocol on voluntary running wheel activity at multiple time points, and/or in response to acute removal of chronic stress was determined. Twenty male Balb/c mice were given access and accustomed to running wheels for 4 weeks, after which they were randomized into 2 groups; exercise (EX, n = 10) and exercise with chronic stress using a modified UCMS protocol for 7 hours/day (8:00 a.m.-3:00p.m.), 5 days/week for 8 weeks (EXS, n = 10). All mice were given access to running wheels from approximately 3:30 p.m. to 7:30 a.m. during the weekday, however during weekends mice had full-time access to running wheels (a time period of no stress for the EXS group). Daily wheel running distance and time were recorded. The average running distance, running time, and work each weekday was significantly lower in EXS compared to EX mice, however, the largest effect was seen during week one. Voluntary wheel running deceased in all mice with increasing age; the pattern of decline appeared to be similar between groups. During the weekend (when no stress was applied), EXS maintained higher distance compared to EX, as well as higher daily distance, time, and work compared to their weekday values. These results indicate that mild chronic stress reduces total spontaneous wheel running in mice during the first week of the daily stress induction and maintains this reduced level for up to 8 consecutive weeks. However, following five days of UCMS, voluntary running wheel activity rebounds within 2–3 days.
Collapse
Affiliation(s)
- Evan DeVallance
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, West Virginia, United States of America
| | - Dale Riggs
- Department of Surgery, WVU School of Medicine, Morgantown, West Virginia, United States of America
| | - Barbara Jackson
- Department of Surgery, WVU School of Medicine, Morgantown, West Virginia, United States of America
| | - Travis Parkulo
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, West Virginia, United States of America
| | - Stanley Zaslau
- Department of Surgery, WVU School of Medicine, Morgantown, West Virginia, United States of America
| | - Paul D. Chantler
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, West Virginia, United States of America
| | - I. Mark Olfert
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, West Virginia, United States of America
| | - Randy W. Bryner
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, West Virginia, United States of America
- * E-mail:
| |
Collapse
|
46
|
Kumar M, Nayak PK. Psychological sequelae of myocardial infarction. Biomed Pharmacother 2017; 95:487-496. [PMID: 28866415 DOI: 10.1016/j.biopha.2017.08.109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/08/2017] [Accepted: 08/23/2017] [Indexed: 01/10/2023] Open
Abstract
Patient with myocardial infarction (MI) are often affected by psychological disorders such as depression, anxiety, and post-traumatic stress disorder. Psychological disorders are disabling and have a negative influence on recovery, reduce the quality of life and causes high mortality rate in MI patients. Despite tremendous advancement in technologies, screening scales, and treatment strategies, psychological sequelae of MI are currently understudied, underestimated, underdiagnosed, and undertreated. Depression is highly prevalent in MI patients followed by anxiety and post-traumatic stress disorder. Pathophysiological factors involved in psychopathologies observed in patients with MI are sympathetic over-activity, hypothalamic-pituitary-adrenal axis dysfunction, and inflammation. Numerous preclinical and clinical studies evidenced a positive association between MI and psychopathologies with a common molecular pathophysiology. This review provides an update on diagnostic feature, prevalence, pathophysiology, clinical outcomes, and management strategies of psychopathologies associated with MI. Moreover, preclinical research findings on molecular mechanisms involved in post-MI psychopathologies and future therapeutic strategies have been outlined in the review.
Collapse
Affiliation(s)
- Mukesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.
| | - Prasanta Kumar Nayak
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.
| |
Collapse
|
47
|
Watanasriyakul WT, Wardwell J, McNeal N, Schultz R, Woodbury M, Dagner A, Cox M, Grippo AJ. Voluntary physical exercise protects against behavioral and endocrine reactivity to social and environmental stressors in the prairie vole. Soc Neurosci 2017; 13:602-615. [PMID: 28786739 DOI: 10.1080/17470919.2017.1365761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Physical activity can combat detrimental effects of stress. The current study examined the potential protective effects of exercise against a combination of social isolation and chronic mild stress (CMS) in a prairie vole model. Female voles were isolated for 4 weeks, with the addition of CMS during the final 2 weeks. Half of the voles were allowed access to a running wheel during this final 2 weeks, while the other half remained sedentary. Animals underwent behavioral tests to assess depressive- and anxiety-behaviors. In a subset of animals, plasma was collected 10 minutes after behavioral testing for corticosterone analysis. In a separate subset, brains were collected 2 hours after behavioral testing for cFos analysis in the paraventricular nucleus (PVN). Voles in the exercise group displayed significantly lower depressive- and anxiety-behaviors, and displayed significantly lower corticosterone levels, compared to animals in the sedentary group. There was no difference in PVN cFos activity between groups. Interestingly, animals that moderately exercised displayed lower levels of depressive-behavior and attenuated corticosterone reactivity compared to animals in the low and high activity subgroups. These findings suggest that physical activity can protect against a combination of social and environmental stressors.
Collapse
Affiliation(s)
| | - Joshua Wardwell
- a Department of Psychology , Northern Illinois University , DeKalb , IL , USA
| | - Neal McNeal
- a Department of Psychology , Northern Illinois University , DeKalb , IL , USA
| | - Rachel Schultz
- a Department of Psychology , Northern Illinois University , DeKalb , IL , USA
| | - Matthew Woodbury
- a Department of Psychology , Northern Illinois University , DeKalb , IL , USA
| | - Ashley Dagner
- a Department of Psychology , Northern Illinois University , DeKalb , IL , USA
| | - Miranda Cox
- a Department of Psychology , Northern Illinois University , DeKalb , IL , USA
| | - Angela J Grippo
- a Department of Psychology , Northern Illinois University , DeKalb , IL , USA
| |
Collapse
|
48
|
Ma L, Demin KA, Kolesnikova TO, Kharsko SL, Zhu X, Yuan X, Song C, Meshalkina DA, Leonard BE, Tian L, Kalueff AV. Animal inflammation-based models of depression and their application to drug discovery. Expert Opin Drug Discov 2017; 12:995-1009. [DOI: 10.1080/17460441.2017.1362385] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Li Ma
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | | | - Xiaokang Zhu
- School of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital, North China University of Science and Technology, Tangshan, China
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China
- Graduate Institute of Biomedical Sciences, College of Medicine, and Department of Medical Research, China Medical University and Hospital, Taichung, Taiwan
| | - Darya A. Meshalkina
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia
| | - Brian E. Leonard
- Department of Pharmacology, National University of Ireland, Galway, Ireland
| | - Li Tian
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University, Beijing, China
| | - Allan V. Kalueff
- School of Pharmaceutical Sciences, Southwest University, Chongqing, China
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg, Russia
- Institute of Chemical Technologies, Ural Federal University, Ekaterinburg, Russia
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| |
Collapse
|
49
|
Aalling N, Hageman I, Miskowiak K, Orlowski D, Wegener G, Wortwein G. Erythropoietin prevents the effect of chronic restraint stress on the number of hippocampal CA3c dendritic terminals-relation to expression of genes involved in synaptic plasticity, angiogenesis, inflammation, and oxidative stress in male rats. J Neurosci Res 2017; 96:103-116. [PMID: 28752903 DOI: 10.1002/jnr.24107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/19/2017] [Accepted: 06/08/2017] [Indexed: 11/09/2022]
Abstract
Stress-induced allostatic load affects a variety of biological processes including synaptic plasticity, angiogenesis, oxidative stress, and inflammation in the brain, especially in the hippocampus. Erythropoietin (EPO) is a pleiotropic cytokine that has shown promising neuroprotective effects. Recombinant human EPO is currently highlighted as a new candidate treatment for cognitive impairment in neuropsychiatric disorders. Because EPO enhances synaptic plasticity, attenuates oxidative stress, and inhibits generation of proinflammatory cytokines, EPO may be able to modulate the effects of stress-induced allostatic load at the molecular level. The aim of this study was therefore to investigate how EPO and repeated restraint stress, separately and combined, influence (i) behavior in the novelty-suppressed feeding test of depression/anxiety-related behavior; (ii) mRNA levels of genes encoding proteins involved in synaptic plasticity, angiogenesis, oxidative stress, and inflammation; and (iii) remodeling of the dendritic structure of the CA3c area of the hippocampus in male rats. As expected, chronic restraint stress lowered the number of CA3c apical dendritic terminals, and EPO treatment reversed this effect. Interestingly, these effects seemed to be mechanistically distinct, as stress and EPO had differential effects on gene expression. While chronic restraint stress lowered the expression of spinophilin, tumor necrosis factor α, and heat shock protein 72, EPO increased expression of hypoxia-inducible factor-2α and lowered the expression of vascular endothelial growth factor in hippocampus. These findings indicate that the effects of treatment with EPO follow different molecular pathways and do not directly counteract the effects of stress in the hippocampus.
Collapse
Affiliation(s)
- Nadia Aalling
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Rigshospitalet and Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida Hageman
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Rigshospitalet and Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kamilla Miskowiak
- Copenhagen Affective Disorder Research Centre, Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Dariusz Orlowski
- Center for Experimental Neuroscience (Cense), Department of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, University of Aarhus, Aarhus, Denmark.,Center of Excellence for Pharmaceutical Sciences, North-West University (Potchefstroom Campus), Potchefstroom, South Africa
| | - Gitta Wortwein
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen, Rigshospitalet and Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Thakare VN, Aswar MK, Kulkarni YP, Patil RR, Patel BM. Silymarin ameliorates experimentally induced depressive like behavior in rats: Involvement of hippocampal BDNF signaling, inflammatory cytokines and oxidative stress response. Physiol Behav 2017; 179:401-410. [PMID: 28711395 DOI: 10.1016/j.physbeh.2017.07.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 05/05/2017] [Accepted: 07/06/2017] [Indexed: 12/12/2022]
Abstract
Silymarin is a polyphenolic flavonoid of Silybum marianum, exhibited neuroprotection and antidepressant like activity in acute restraint stressed mice. The main objective of the present study is to investigate possible antidepressant like activity of silymarin in experimentally induced depressive behavior in rats. The depressive behaviors were induced in rats by olfactory bulbectomized (OBX) technique. Wistar rats were administered with silymarin at a dose of 100mg/kg and 200mg/kg, by per oral in OBX and sham operated rats. Behavioral (ambulatory and rearing activity and immobility time), neurochemical [serotonin (5-HT), dopamine (DA), norepinephrine (NE) and brain derived neurotrophic factor (BDNF) level], biochemical (MDA formation, IL-6, TNF-α and antioxidants) changes in hippocampus and cerebral cortex along with serum corticosterone were investigated. Rats subjected to OBX elicited significant increase in immobility time, ambulatory and rearing behaviors, reduced BDNF level, 5-HT, DA, NE and antioxidant parameters along with increased serum corticosterone, MDA formation, IL-6, and TNF-α in hippocampus and cerebral cortex compared to sham operated rats. Administration of with silymarin significantly attenuated immobility time, ambulatory and rearing behaviors, serum corticosterone and improved BDNF expression, 5-HT, DA, NE and antioxidant paradigms in cerebral cortex as well as hippocampus. In addition, silymarin attenuated IL-6, and TNF-α significantly in hippocampus and cerebral cortex in OBX rats. Thus, silymarin exhibits anti-depressant-like activity in OBX rats due to alterations in several neurotransmitters, endocrine and immunologic systems, including BDNF, 5-HT, DA, NE, MDA formation, IL-6, and TNF-α in hippocampus and cerebral cortex as well as serum corticosterone.
Collapse
Affiliation(s)
- Vishnu N Thakare
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala 410401, Maharashtra, India; Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382 481, Gujarat, India
| | - Manoj K Aswar
- Department of Pharmacology, Sinhgad Institute of Pharmacy, Nerhe, Pune, Maharashtra, India
| | - Yogesh P Kulkarni
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala 410401, Maharashtra, India
| | - Rajesh R Patil
- Department of Pharmacology, Sinhgad Institute of Pharmaceutical Sciences, Lonavala 410401, Maharashtra, India
| | - Bhoomika M Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad 382 481, Gujarat, India.
| |
Collapse
|